1
|
Chen L, Qu Y, Cui H, Zhang W, Wu X, Zhao X, Xiao J, Tang M, Wang Y, Zou Y, Qiu L, Tan Z, Lei B, Ma X, Zhang D, Liu Y, Fan M, Li J, Zhang B, Jiang X. Genomic correlation, shared loci, and causal association between obesity, periodontitis and tooth loss. Sci Rep 2025; 15:5155. [PMID: 39934647 PMCID: PMC11814315 DOI: 10.1038/s41598-025-89782-8] [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: 09/13/2024] [Accepted: 02/07/2025] [Indexed: 02/13/2025] Open
Abstract
Observational studies have reported an association of obesity with periodontitis and tooth loss, yet findings remain inconsistent. We aim to investigate the genetic link underlying obesity-related traits (BMI [body mass index], WHR [waist-to-hip ratio], WHRadjBMI and childhood BMI), periodontitis and tooth loss. Leveraging summary statistics from large-scale genome-wide association studies, we comprehensively investigated the pair-wise genetic correlation using linkage disequilibrium score regression (LDSC) and SUPERGNOVA, identified shared loci using cross-phenotype association analysis (CPASSOC), and estimated causal association using Mendelian randomization. We identified a significant genetic correlation of obesity with tooth loss, but not with periodontitis. Partitioning the genome into LD-independent regions revealed 10 significantly shared local signals across six regions. Genome-wide cross-trait analysis uncovered 14 shared loci, four of which were colocalized: rs2064044 (PP4 = 0.94), rs6000329 (PP4 = 0.86), rs7134628 (PP4 = 0.86), and rs1286769 (PP4 = 0.90). Notably, rs1286769, identified via CPASSOC and validated through colocalization analysis, is located near RARβ, a gene associated with both BMI and denture use. Mendelian randomization revealed a nominally-significant causal association of obesity with periodontitis (P = 0.045) but a robust causal association with tooth loss represented by number of teeth (BMI: beta = [Formula: see text]0.20, 95%CI = [Formula: see text]0.26 to [Formula: see text]0.14, P = 5.27 × 10-11; WHR: beta = [Formula: see text]0.16, 95%CI = [Formula: see text]0.24 to [Formula: see text]0.08, P = 3.71 × 10-5). Results of CAUSE were consistent with main findings. From a genetic perspective, our work highlights an intrinsic link between obesity, periodontitis and tooth loss, which may add new lines of evidence and provide insights for clinical and public oral health applications.
Collapse
Affiliation(s)
- Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yang Qu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Xuan Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Jinyu Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Lingli Qiu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Zhixin Tan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Bowen Lei
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Xiaofeng Ma
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Di Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Mengyu Fan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China
| | - Ben Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China.
- Departments of Cardiology, Neurology, and Oncology, Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China.
- Department of Occupational and Environmental Health, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China.
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, China-PUMC C. C. Chen Institute of Health, Sichuan University, Chengdu, Sichuan, China.
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
2
|
Replogle MR, Thompson S, Reis LM, Semina EV. A De Novo Noncoding RARB Variant Associated with Complex Microphthalmia Alters a Putative Regulatory Element. Hum Mutat 2024; 2024:6619280. [PMID: 39450403 PMCID: PMC11501074 DOI: 10.1155/2024/6619280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Retinoic acid receptor beta (RARB) is a transcriptional regulator crucial for coordinating retinoic acid- (RA-) mediated morphogenic movements, cell growth, and differentiation during eye development. Loss- or gain-of-function RARB coding variants have been associated with microphthalmia, coloboma, and anterior segment defects. We identified a de novo variant c.157+1895G>A located within a conserved region (CR1) in the first intron of RARB in an individual with complex microphthalmia and significant global developmental delay. Based on the phenotypic overlap, we further investigated the possible effects of the variant on mRNA splicing and/or transcriptional regulation through in silico and functional studies. In silico analysis identified the possibility of alternative splicing, suggested by one out of three (HSF, SpliceAI, and MaxEntScan) splicing prediction programs, and a strong indication of regulatory function based on publicly available DNase hypersensitivity, histone modification, chromatin folding, and ChIP-seq data sets. Consistent with the predictions of SpliceAI and MaxEntScan, in vitro minigene assays showed no effect on RARB mRNA splicing. Evaluation of CR1 for a regulatory role using luciferase reporter assays in human lens epithelial cells demonstrated a significant increase in the activity of the RARB promoter in the presence of wild-type CR1. This activity was further significantly increased in the presence of CR1 carrying the c.157+1895G>A variant, suggesting that the variant may promote RARB overexpression in human cells. Induction of RARB overexpression in human lens epithelial cells resulted in increased cell proliferation and elevated expression of FOXC1, a known downstream target of RA signaling and a transcription factor whose down- and upregulation is associated with ocular phenotypes overlapping the RARB spectrum. These results support a regulatory role for the CR1 element and suggest that the de novo c.157+1895G>A variant affecting this region may alter the proper regulation of RARB and, as a result, its downstream genes, possibly leading to abnormal development.
Collapse
Affiliation(s)
- Maria R. Replogle
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samuel Thompson
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Linda M. Reis
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Elena V. Semina
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Hospital of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
3
|
Haggagy MG, Ahmed LA, Sharaky M, Elhefnawi MM, Omran MM. SIRT1 as a potential key regulator for mediating apoptosis in oropharyngeal cancer using cyclophosphamide and all-trans retinoic acid. Sci Rep 2024; 14:41. [PMID: 38167952 PMCID: PMC10761886 DOI: 10.1038/s41598-023-50478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Although cyclophosphamide (CTX) has been used for recurrent or metastatic head and neck cancers, resistance is usually expected. Thus, we conducted this study to examine the effect of adding all-trans retinoic acid (ATRA) to CTX, to increase efficacy of CTX and reduce the risk of resistance developed. In this study, we investigated the combined effect of ATRA and CTX on the expression of apoptotic and angiogenesis markers in oropharyngeal carcinoma cell line (NO3), and the possible involved mechanisms. ATRA and CTX in combination significantly inhibited the proliferation of NO3 cells. Lower dose of CTX in combination with ATRA exhibited significant cytotoxicity than that of CTX when used alone, implying lower expected toxicity. Results showed that ATRA and CTX modulated oxidative stress; increased NOx and MDA, reduced GSH, and mRNA expression of Cox-2, SIRT1 and AMPK. Apoptosis was induced through elevating mRNA expressions of Bax and PAR-4 and suppressing that of Bcl-xl and Bcl-2, parallel with increased caspases 3 and 9 and decreased VEGF, endothelin-1 and CTGF levels. The primal action of the combined regimen on inflammatory signaling highlights its impact on cell death in NO3 cell line which was mediated by oxidative stress associated with apoptosis and suppression of angiogenesis.
Collapse
Affiliation(s)
- Mahitab G Haggagy
- Clinical Pharmacy Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Clinical Pharmacy Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Lamiaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Marwa Sharaky
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Mahmoud M Elhefnawi
- Biomedical Informatics and Chemoinformatic Group, Informatics and Systems Department, National Research Centre, Cairo, Egypt
| | - Mervat M Omran
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt.
| |
Collapse
|
4
|
DiKun KM, Gudas LJ. Vitamin A and retinoid signaling in the kidneys. Pharmacol Ther 2023; 248:108481. [PMID: 37331524 PMCID: PMC10528136 DOI: 10.1016/j.pharmthera.2023.108481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
Vitamin A (VA, retinol) and its metabolites (commonly called retinoids) are required for the proper development of the kidney during embryogenesis, but retinoids also play key roles in the function and repair of the kidney in adults. Kidneys filter 180-200 liters of blood per day and each kidney contains approximately 1 million nephrons, which are often referred to as the 'functional units' of the kidney. Each nephron consists of a glomerulus and a series of tubules (proximal tubule, loop of Henle, distal tubule, and collecting duct) surrounded by a network of capillaries. VA is stored in the liver and converted to active metabolites, most notably retinoic acid (RA), which acts as an agonist for the retinoic acid receptors ((RARs α, β, and γ) to regulate gene transcription. In this review we discuss some of the actions of retinoids in the kidney after injury. For example, in an ischemia-reperfusion model in mice, injury-associated loss of proximal tubule (PT) differentiation markers occurs, followed by re-expression of these differentiation markers during PT repair. Notably, healthy proximal tubules express ALDH1a2, the enzyme that metabolizes retinaldehyde to RA, but transiently lose ALDH1a2 expression after injury, while nearby myofibroblasts transiently acquire RA-producing capabilities after injury. These results indicate that RA is important for renal tubular injury repair and that compensatory mechanisms exist for the generation of endogenous RA by other cell types upon proximal tubule injury. ALDH1a2 levels also increase in podocytes, epithelial cells of the glomeruli, after injury, and RA promotes podocyte differentiation. We also review the ability of exogenous, pharmacological doses of RA and receptor selective retinoids to treat numerous kidney diseases, including kidney cancer and diabetic kidney disease, and the emerging genetic evidence for the importance of retinoids and their receptors in maintaining or restoring kidney function after injury. In general, RA has a protective effect on the kidney after various types of injuries (eg. ischemia, cytotoxic actions of chemicals, hyperglycemia related to diabetes). As more research into the actions of each of the three RARs in the kidney is carried out, a greater understanding of the actions of vitamin A is likely to lead to new insights into the pathology of kidney disorders and the development of new therapies for kidney diseases.
Collapse
Affiliation(s)
- Krysta M DiKun
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA; Department of Urology, Weill Cornell Medicine, New York, NY, USA; New York Presbyterian Hospital, New York, NY, USA; Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
5
|
Butsri S, Kukongviriyapan V, Senggunprai L, Kongpetch S, Prawan A. All- trans-retinoic acid induces RARB-dependent apoptosis via ROS induction and enhances cisplatin sensitivity by NRF2 downregulation in cholangiocarcinoma cells. Oncol Lett 2022; 23:179. [PMID: 35464301 PMCID: PMC9025595 DOI: 10.3892/ol.2022.13299] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
All-trans-retinoic acid (ATRA) has been clinically used to treat acute promyelocytic leukemia and is being studied to treat other types of cancer; however, the therapeutic role and mechanism of ATRA against cholangiocarcinoma (CCA) remain unclear. The present study investigated the cytotoxic effect and underlying mechanisms of ATRA on CCA cell lines. Cell viability was evaluated by sulforhodamine B assay. Intracellular reactive oxygen species (ROS) levels were assessed by dihydroethidium assay. Apoptosis analysis was performed by flow cytometry. The pathways of apoptotic cell death induction were examined using enzymatic caspase activity assay. Proteins associated with apoptosis were evaluated by western blotting. The effects on gene expression were analyzed by reverse transcription-quantitative PCR analysis. ATRA induced a concentration- and time-dependent toxicity in CCA cells. Furthermore, when the cytotoxicity of ATRA against retinoic acid receptor (RAR)-deficient cells was assessed, it was revealed that ATRA cytotoxicity was RARB-dependent. Following ATRA treatment, there was a significant accumulation of cellular ROS and ATRA-induced ROS generation led to an increase in the expression levels of apoptosis-inducing proteins and intrinsic apoptosis. Pre-treatment with ROS scavengers could diminish the apoptotic effect of ATRA, suggesting that ROS and mitochondria may have an essential role in the induction of apoptosis. Furthermore, following ATRA treatment, an increase in cellular ROS content was associated with suppressing nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2) and NRF2-downstream active genes. ATRA also suppressed cisplatin-induced NRF2 expression, suggesting that the enhancement of cisplatin cytotoxicity by ATRA may be associated with the downregulation of NRF2 signaling. In conclusion, the results of the present study demonstrated that ATRA could be repurposed as an alternative drug for CCA therapy.
Collapse
Affiliation(s)
- Siriwoot Butsri
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Veerapol Kukongviriyapan
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Laddawan Senggunprai
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Auemduan Prawan
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
6
|
Gangwar SK, Kumar A, Jose S, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Nuclear receptors in oral cancer-emerging players in tumorigenesis. Cancer Lett 2022; 536:215666. [DOI: 10.1016/j.canlet.2022.215666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 12/24/2022]
|
7
|
Wu H, Zhao D, Wang C, Zhang D, Tang M, Qian S, Xu L, Xia T, Zhou J, Wang G, He Y, Gao L, Chen W, Li L, Yang W, Zhao Q, Hu C, Hu A. All-Trans Retinoic Acid Prevents the Progression of Gastric Precancerous Lesions by Regulating Disordered Retinoic Acid Metabolism. Nutr Cancer 2022; 74:3351-3362. [PMID: 35225106 DOI: 10.1080/01635581.2022.2044062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Hanhan Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Didi Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Chen Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Daoming Zhang
- Department of Gastroenterology, Lujiang County People’s Hospital, Hefei, Anhui, PR China
| | - Min Tang
- Department of Gastroenterology and Hepatology, the Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China
| | - Shiqing Qian
- Department of Pathology, Lujiang County People’s Hospital, Hefei, Anhui, PR China
| | - Lina Xu
- Department of Gastroenterology, Lujiang County People’s Hospital, Hefei, Anhui, PR China
| | - Tao Xia
- Department of Gastroenterology, Lujiang County People’s Hospital, Hefei, Anhui, PR China
| | - Juanyan Zhou
- Department of Gastroenterology and Hepatology, the Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, PR China
| | - Guangjun Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Yue He
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Lei Gao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Wenjun Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Li Li
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Wanshui Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Qihong Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Chuanlai Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| | - Anla Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui, PR China
| |
Collapse
|
8
|
Giuli MV, Hanieh PN, Giuliani E, Rinaldi F, Marianecci C, Screpanti I, Checquolo S, Carafa M. Current Trends in ATRA Delivery for Cancer Therapy. Pharmaceutics 2020; 12:E707. [PMID: 32731612 PMCID: PMC7465813 DOI: 10.3390/pharmaceutics12080707] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/23/2020] [Accepted: 07/25/2020] [Indexed: 12/12/2022] Open
Abstract
All-Trans Retinoic Acid (ATRA) is the most active metabolite of vitamin A. It is critically involved in the regulation of multiple processes, such as cell differentiation and apoptosis, by activating specific genomic pathways or by influencing key signaling proteins. Furthermore, mounting evidence highlights the anti-tumor activity of this compound. Notably, oral administration of ATRA is the first choice treatment in Acute Promyelocytic Leukemia (APL) in adults and NeuroBlastoma (NB) in children. Regrettably, the promising results obtained for these diseases have not been translated yet into the clinics for solid tumors. This is mainly due to ATRA-resistance developed by cancer cells and to ineffective delivery and targeting. This up-to-date review deals with recent studies on different ATRA-loaded Drug Delivery Systems (DDSs) development and application on several tumor models. Moreover, patents, pre-clinical, and clinical studies are also reviewed. To sum up, the main aim of this in-depth review is to provide a detailed overview of the several attempts which have been made in the recent years to ameliorate ATRA delivery and targeting in cancer.
Collapse
Affiliation(s)
- Maria Valeria Giuli
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (M.V.G.); (E.G.); (I.S.)
| | - Patrizia Nadia Hanieh
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| | - Eugenia Giuliani
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (M.V.G.); (E.G.); (I.S.)
| | - Federica Rinaldi
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| | - Carlotta Marianecci
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy; (M.V.G.); (E.G.); (I.S.)
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, 04100 Latina, Italy
| | - Maria Carafa
- Department of Drug Chemistry and Technology, Sapienza University of Rome, 00185 Rome, Italy; (P.N.H.); (F.R.); (C.M.); (M.C.)
| |
Collapse
|
9
|
Park J, Zhang X, Lee SK, Song NY, Son SH, Kim KR, Shim JH, Park KK, Chung WY. CCL28-induced RARβ expression inhibits oral squamous cell carcinoma bone invasion. J Clin Invest 2020; 129:5381-5399. [PMID: 31487270 DOI: 10.1172/jci125336] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 09/03/2019] [Indexed: 12/30/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) frequently invades the maxillary or mandibular bone, and this bone invasion is closely associated with poor prognosis and survival. Here, we show that CCL28 functions as a negative regulator of OSCC bone invasion. CCL28 inhibited invasion and epithelial-mesenchymal transition (EMT), and its inhibition of EMT was characterized by induced E-cadherin expression and reduced nuclear localization of β-catenin in OSCC cells with detectable RUNX3 expression levels. CCL28 signaling via CCR10 increased retinoic acid receptor-β (RARβ) expression by reducing the interaction between RARα and HDAC1. In addition, CCL28 reduced RANKL production in OSCC and osteoblastic cells and blocked RANKL-induced osteoclastogenesis in osteoclast precursors. Intraperitoneally administered CCL28 inhibited tumor growth and osteolysis in mouse calvaria and tibia inoculated with OSCC cells. RARβ expression was also increased in tumor tissues. In patients with OSCC, low CCL28, CCR10, and RARβ expression levels were highly correlated with bone invasion. Patients with OSCC who had higher expression of CCL28, CCR10, or RARβ had significantly better overall survival. These findings suggest that CCL28, CCR10, and RARβ are useful markers for the prediction and treatment of OSCC bone invasion. Furthermore, CCL28 upregulation in OSCC cells or CCL28 treatment can be a therapeutic strategy for OSCC bone invasion.
Collapse
Affiliation(s)
- Junhee Park
- Department of Dentistry and.,Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Xianglan Zhang
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea.,Department of Pathology, Yanbian University Hospital, Yanji city, China
| | - Sun Kyoung Lee
- Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Na-Young Song
- Department of Dentistry and.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Seung Hwa Son
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Ki Rim Kim
- Department of Dental Hygiene, College of Science and Engineering, Kyungpook National University, Sangju, Korea
| | - Jae Hoon Shim
- Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea
| | - Kwang-Kyun Park
- Department of Dentistry and.,Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Won-Yoon Chung
- Department of Dentistry and.,Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea.,Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| |
Collapse
|
10
|
Mechanistic understanding of β-cryptoxanthin and lycopene in cancer prevention in animal models. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158652. [PMID: 32035228 DOI: 10.1016/j.bbalip.2020.158652] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/26/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
To better understand the potential function of carotenoids in the chemoprevention of cancers, mechanistic understanding of carotenoid action on genetic and epigenetic signaling pathways is critically needed for human studies. The use of appropriate animal models is the most justifiable approach to resolve mechanistic issues regarding protective effects of carotenoids at specific organs and tissue sites. While the initial impetus for studying the benefits of carotenoids in cancer prevention was their antioxidant capacity and pro-vitamin A activity, significant advances have been made in the understanding of the action of carotenoids with regards to other mechanisms. This review will focus on two common carotenoids, provitamin A carotenoid β-cryptoxanthin and non-provitamin A carotenoid lycopene, as promising chemopreventive agents or chemotherapeutic compounds against cancer development and progression. We reviewed animal studies demonstrating that β-cryptoxanthin and lycopene effectively prevent the development or progression of various cancers and the potential mechanisms involved. We highlight recent research that the biological functions of β-cryptoxanthin and lycopene are mediated, partially via their oxidative metabolites, through their effects on key molecular targeting events, such as NF-κB signaling pathway, RAR/PPARs signaling, SIRT1 signaling pathway, and p53 tumor suppressor pathways. The molecular targets by β-cryptoxanthin and lycopene, offer new opportunities to further our understanding of common and distinct mechanisms that involve carotenoids in cancer prevention. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
|
11
|
Mitochondrial dysfunction is a key determinant of the rare disease lymphangioleiomyomatosis and provides a novel therapeutic target. Oncogene 2018; 38:3093-3101. [PMID: 30573768 PMCID: PMC6484686 DOI: 10.1038/s41388-018-0625-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/04/2018] [Accepted: 11/20/2018] [Indexed: 01/15/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare and progressive systemic disease affecting mainly young women of childbearing age. A deterioration in lung function is driven by neoplastic growth of atypical smooth muscle-like LAM cells in the pulmonary interstitial space that leads to cystic lung destruction and spontaneous pneumothoraces. Therapeutic options for preventing disease progression are limited and often end with lung transplantation temporarily delaying an inevitable decline. To identify new therapeutic strategies for this crippling orphan disease, we have performed array based and metabolic molecular analysis on patient-derived cell lines. Our results point to the conclusion that mitochondrial biogenesis and mitochondrial dysfunction in LAM cells provide a novel target for treatment.
Collapse
|
12
|
Bierbaumer L, Schwarze UY, Gruber R, Neuhaus W. Cell culture models of oral mucosal barriers: A review with a focus on applications, culture conditions and barrier properties. Tissue Barriers 2018; 6:1479568. [PMID: 30252599 PMCID: PMC6389128 DOI: 10.1080/21688370.2018.1479568] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Understanding the function of oral mucosal epithelial barriers is essential for a plethora of research fields such as tumor biology, inflammation and infection diseases, microbiomics, pharmacology, drug delivery, dental and biomarker research. The barrier properties are comprised by a physical, a transport and a metabolic barrier, and all these barrier components play pivotal roles in the communication between saliva and blood. The sum of all epithelia of the oral cavity and salivary glands is defined as the blood-saliva barrier. The functionality of the barrier is regulated by its microenvironment and often altered during diseases. A huge array of cell culture models have been developed to mimic specific parts of the blood-saliva barrier, but no ultimate standard in vitro models have been established. This review provides a comprehensive overview about developed in vitro models of oral mucosal barriers, their applications, various cultivation protocols and corresponding barrier properties.
Collapse
Affiliation(s)
- Lisa Bierbaumer
- a Competence Unit Molecular Diagnostics, Center Health and Bioresources, Austrian Institute of Technology (AIT) GmbH , Vienna , Austria
| | - Uwe Yacine Schwarze
- b Department of Oral Biology , School of Dentistry, Medical University of Vienna , Vienna , Austria.,c Austrian Cluster for Tissue Regeneration , Vienna , Austria
| | - Reinhard Gruber
- b Department of Oral Biology , School of Dentistry, Medical University of Vienna , Vienna , Austria.,c Austrian Cluster for Tissue Regeneration , Vienna , Austria.,d Department of Periodontology , School of Dental Medicine, University of Bern , Bern , Switzerland
| | - Winfried Neuhaus
- a Competence Unit Molecular Diagnostics, Center Health and Bioresources, Austrian Institute of Technology (AIT) GmbH , Vienna , Austria
| |
Collapse
|
13
|
Jiang L, Qin Y, Lei F, Chen X, Zhou Z. Retinoic acid receptors α and γ are involved in antioxidative protection in renal tubular epithelial cells injury induced by hypoxia/reoxygenation. Free Radic Res 2017; 51:873-885. [PMID: 29096559 DOI: 10.1080/10715762.2017.1387655] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Renal interstitial fibrosis (RIF) is a common outcome in various chronic kidney diseases. Injury to renal tubular epithelial cell (RTEC) is major link in RIF. Hypoxia is one of the common factors for RTEC damage. Retinoic acid receptors (RARs), RARα, RARβ and RARγ, are evolutionary conserved and pleiotropic proteins that have been involved in various cellular functions, including proliferation, differentiation, apoptosis, and transcription. Recently, we discovered that aberrant expression of RARs was involved in the development of RIF in rats. Here, we investigated the role of RARs in the hypoxia/reoxygenation (HR) damage model in RTEC with virus-based delivery vectors to knockdown or overexpress RARs. Relevant indicators were detected. Our results showed that HR inhibited RARα and RARγ expressions in a time-dependent manner in RTECs; however, the expression of RARβ was not changed obviously. RARα and RARγ overexpression could protect cells from oxidative stress-induced injury by inhibiting HR-induced intracellular superoxide anion (O2-) generation, cell viability and mitochondria membrane potential (MMP) decrease and transforming growth factor β1 (TGF-β1) expression and promoting endogenous antioxidant defense components, superoxide dismutase (SOD) and glutathione (GSH). Meanwhile, inhibition of RARα and RARγ expressions by small interference RNAs (siRNA) resulted in a less resistance of RTEC to HR as shown in increased O2- production and TGF-β1 expression and decreased cell viability, MMP, SOD and GSH levels. These data indicates that RARα and RARγ act as positive regulators to offset oxidative damage and profibrosis cytokine accumulation and therefore has an antioxidative effect.
Collapse
Affiliation(s)
- Ling Jiang
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , PR China
| | - Yuanhan Qin
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , PR China
| | - Fengying Lei
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , PR China
| | - Xiuping Chen
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , PR China
| | - Zhiqiang Zhou
- a Department of Pediatrics , The First Affiliated Hospital of Guangxi Medical University , Nanning , PR China
| |
Collapse
|
14
|
Azarnezhad A, Mehdipour P. Cancer Genetics at a Glance: The Comprehensive Insights. CANCER GENETICS AND PSYCHOTHERAPY 2017:79-389. [DOI: 10.1007/978-3-319-64550-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
15
|
Gutiérrez J, García-Villa E, Ocadiz-Delgado R, Cortés-Malagón EM, Vázquez J, Roman-Rosales A, Alvarez-Rios E, Celik H, Romano MC, Üren A, Lambert PF, Gariglio P. Human papillomavirus type 16 E7 oncoprotein upregulates the retinoic acid receptor-beta expression in cervical cancer cell lines and K14E7 transgenic mice. Mol Cell Biochem 2015; 408:261-272. [PMID: 26173416 DOI: 10.1007/s11010-015-2504-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 07/04/2015] [Indexed: 12/22/2022]
Abstract
Persistent infection with high-risk human papillomaviruses is the main etiological factor in cervical cancer (CC). The human papillomavirus type 16 (HPV16) E7 oncoprotein alters several cellular processes, regulating the expression of many genes in order to avoid cell cycle control. Retinoic acid receptor beta (RARB) blocks cell growth, inducing differentiation and apoptosis. This tumor suppressor gene is gradually silenced in late passages of foreskin keratinocytes immortalized with HPV16 and in various tumors, including CC, mainly by epigenetic modifications. We investigated the effect of E7 oncoprotein on RARB gene expression. We found that HPV16 E7 increases RARB mRNA and RAR-beta protein expression both in vitro and in the cervix of young K14E7 transgenic mice. In E7-expressing cells, RARB overexpression is further increased in the presence of the tumor suppressor p53 (TP53) R273C mutant. This effect does not change when either C33-A or E7-expressing C33-A cell line is treated with Trichostatin A, suggesting that E7 enhances RARB expression independently of histone deacetylases inhibition. These findings indicate that RARB overexpression is part of the early molecular events induced by the E7 oncoprotein.
Collapse
Affiliation(s)
- Jorge Gutiérrez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Enrique García-Villa
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Rodolfo Ocadiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Enoc M Cortés-Malagón
- Laboratorio de Biología Molecular del Cáncer, Unidad de Investigación, Hospital Juárez de México, Av. IPN 5160, Magdalena de Las Salinas, Gustavo A. Madero, 07760, Ciudad de México, México
| | - Juan Vázquez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Alejandra Roman-Rosales
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Elizabeth Alvarez-Rios
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Haydar Celik
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057-1469, USA
| | - Marta C Romano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México
| | - Aykut Üren
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 20057-1469, USA
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Ciudad de México, México.
| |
Collapse
|
16
|
Li M, Sun Y, Guan X, Shu X, Li C. Advanced progress on the relationship between RA and its receptors and malignant tumors. Crit Rev Oncol Hematol 2014; 91:271-82. [DOI: 10.1016/j.critrevonc.2014.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/20/2014] [Accepted: 04/02/2014] [Indexed: 12/27/2022] Open
|
17
|
Kanai F, Obi S, Fujiyama S, Shiina S, Tamai H, Mochizuki H, Koike Y, Imamura J, Yamaguchi T, Saida I, Yokosuka O, Omata M. An open-label phase I/II study of tamibarotene in patients with advanced hepatocellular carcinoma. Hepatol Int 2013. [PMID: 26202410 DOI: 10.1007/s12072-013-9459-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIM Tamibarotene is a synthetic retinoid expected to inhibit tumor-cell proliferation and to induce apoptosis by selective interaction with retinoic acid receptor α/β. We conducted an open-label phase I/II study to determine the maximum tolerated dose (MTD) and recommended dose (RD), and to evaluate the pharmacokinetics, efficacy, and safety profiles for advanced hepatocellular carcinoma (HCC). METHODS Patients with histologically confirmed, measurable, unresectable HCC of Child-Pugh classification A or B and with no effective systemic or local therapies were eligible. In phase I, patients were assigned based on the 3 + 3 dose escalation criteria to receive tamibarotene at 8, 12, and 16 mg/day. The RD determined in phase I was employed for phase II. The planned sample size in phase II was 25, including the RD-treated patients in phase I. RESULTS Thirty-six patients were enrolled. No patients experienced dose-limiting toxicity (DLT) at 8 mg/day. However, two out of six patients experienced the DLTs at 12 mg/day: one experienced thrombosis in a limb vein and pulmonary artery, and the other experienced an increase of γ-GTP. The MTD and RD were determined as 12 and 8 mg/day, respectively. In phase II, one patient achieved partial response, and seven achieved stable disease. The disease control rate was 32 % (95 % CI: 15.0-53.5). The following drug-related serious adverse events were reported: thrombosis in a limb vein, pulmonary artery, and portal vein; interstitial lung disease; and vomiting. CONCLUSIONS Tamibarotene demonstrated the inhibition of tumor cell growth in advanced HCC with acceptable tolerance.
Collapse
Affiliation(s)
- Fumihiko Kanai
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba, 260-8670, Japan.
| | - Shuntaro Obi
- Department of Hepatology, Kyoundo Hospital, 1-8 Kandasurugadai, Chiyoda-ku, Tokyo, 101-0062, Japan.
| | - Shigetoshi Fujiyama
- Department of Gastroenterology and Hepatology, Kumamoto Shinto General Hospital, 1-17-27 Shinyashiki, Kumamoto City, Kumamoto, 862-8655, Japan.
| | - Shuichiro Shiina
- Department of Gastroenterology, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Hideyuki Tamai
- Second Department of Internal Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama City, Wakayama, 641-0012, Japan.
| | - Hitoshi Mochizuki
- Department of Gastroenterology, Yamanashi Prefectural Central Hospital, 1-1-1 Fujimi, Kofu City, Yamanashi, 400-8506, Japan.
| | - Yukihiro Koike
- Department of Gastroenterology, Kanto Central Hospital, 6-25-1 Kamiyouga, Setagaya-ku, Tokyo, 158-8531, Japan.
| | - Jun Imamura
- Division of Hepatology, Department of Internal Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan.
| | - Takayoshi Yamaguchi
- Clinical Research 1, Zeria Pharmaceutical Co., Ltd, 10-11 Nihonbashi Kobuna-cho, Chuo-ku, Tokyo, 103-8351, Japan.
| | - Isamu Saida
- Clinical Research 1, Zeria Pharmaceutical Co., Ltd, 10-11 Nihonbashi Kobuna-cho, Chuo-ku, Tokyo, 103-8351, Japan.
| | - Osamu Yokosuka
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba City, Chiba, 260-8670, Japan.
| | - Masao Omata
- Yamanashi Prefectural Hospital Organization, 1-1-1 Fujimi, Kofu City, Yamanashi, 400-8506, Japan.
| |
Collapse
|
18
|
Cheng ASL, Li MS, Kang W, Cheng VY, Chou JL, Lau SS, Go MY, Lee CC, Ling TK, Ng EK, Yu J, Huang TH, To KF, Chan MW, Sung JJY, Chan FKL. Helicobacter pylori causes epigenetic dysregulation of FOXD3 to promote gastric carcinogenesis. Gastroenterology 2013; 144:122-133.e9. [PMID: 23058321 DOI: 10.1053/j.gastro.2012.10.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 08/25/2012] [Accepted: 10/03/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Deregulation of forkhead box (Fox) proteins, an evolutionarily conserved family of transcriptional regulators, leads to tumorigenesis. Little is known about their regulation or functions in the pathogenesis of gastric cancer. Promoter hypermethylation occurs during Helicobacter pylori-induced gastritis. We investigated whether the deregulated genes contribute to gastric tumorigenesis. METHODS We used integrative genome-wide scans to identify concomitant hypermethylated genes in mice infected with H pylori and human gastric cancer samples. We also analyzed epigenetic gene silencing in gastric tissues from patients with H pylori infection and gastritis, intestinal metaplasia, gastric tumors, or without disease (controls). Target genes were identified by chromatin immunoprecipitation microarrays and expression and luciferase reporter analyses. RESULTS Methylation profile analyses identified the promoter of FOXD3 as the only genomic region with increased methylation in mice and humans during progression of H pylori-associated gastric tumors. FOXD3 methylation also correlated with shorter survival times of patients with gastric cancer. Genome demethylation reactivated FOXD3 expression in gastric cancer cell lines. Transgenic overexpression of FOXD3 significantly inhibited gastric cancer cell proliferation and invasion, and reduced growth of xenograft tumors in mice, at least partially, by promoting tumor cell apoptosis. FOXD3 bound directly to the promoters of, and activated transcription of, genes encoding the cell death regulators CYFIP2 and RARB. Levels of FOXD3, CYFIP2, and RARB messenger RNAs were reduced in human gastric tumor samples, compared with control tissues. CONCLUSIONS FOXD3-mediated transcriptional control of tumor suppressors is deregulated by H pylori infection-induced hypermethylation; this could perturb the balance between cell death and survival. These findings identify a pathway by which epigenetic changes affect gastric tumor suppression.
Collapse
Affiliation(s)
- Alfred S L Cheng
- Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Sabatino L, Fucci A, Pancione M, Carafa V, Nebbioso A, Pistore C, Babbio F, Votino C, Laudanna C, Ceccarelli M, Altucci L, Bonapace IM, Colantuoni V. UHRF1 coordinates peroxisome proliferator activated receptor gamma (PPARG) epigenetic silencing and mediates colorectal cancer progression. Oncogene 2012; 31:5061-72. [PMID: 22286757 DOI: 10.1038/onc.2012.3] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARG) inactivation has been identified as an important step in colorectal cancer (CRC) progression, although the events involved have been partially clarified. UHRF1 is emerging as a cofactor that coordinates the epigenetic silencing of tumor suppressor genes, but its role in CRC remains elusive. Here, we report that UHRF1 negatively regulates PPARG and is associated with a higher proliferative, clonogenic and migration potential. Consistently, UHRF1 ectopic expression induces PPARG repression through its recruitment on the PPARG promoter fostering DNA methylation and histone repressive modifications. In agreement, UHRF1 knockdown elicits PPARG re-activation, accompanied by positive histone marks and DNA demethylation, corroborating its role in PPARG silencing. UHRF1 overexpression, as well as PPARG-silencing, imparts higher growth rate and phenotypic features resembling those occurring in the epithelial-mesenchymal transition. In our series of 110 sporadic CRCs, high UHRF1-expressing tumors are characterized by an undifferentiated phenotype, higher proliferation rate and poor clinical outcome only in advanced stages III-IV. In addition, the inverse relationship with PPARG found in vitro is detected in vivo and UHRF1 prognostic significance appears closely related to PPARG low expression, as remarkably validated in an independent dataset. The results demonstrate that UHRF1 regulates PPARG silencing and both genes appear to be part of a complex regulatory network. These findings suggest that the relationship between UHRF1 and PPARG may have a relevant role in CRC progression.
Collapse
Affiliation(s)
- L Sabatino
- Department of Biological, Geological and Environmental Sciences, University of Sannio, Benevento, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Yang M, Park JY. DNA methylation in promoter region as biomarkers in prostate cancer. Methods Mol Biol 2012; 863:67-109. [PMID: 22359288 DOI: 10.1007/978-1-61779-612-8_5] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The prostate gland is the most common site of cancer and the second leading cause of cancer death in American men. Recent emerging molecular biological technologies help us to know that epigenetic alterations such as DNA methylation within the regulatory (promoter) regions of genes are associated with transcriptional silencing in cancer. Promoter hypermethylation of critical pathway genes could be potential biomarkers and therapeutic targets for prostate cancer. In this chapter, we updated current information on methylated genes associated with the development and progression of prostate cancer. Over 40 genes have been investigated for methylation in promoter region in prostate cancer. These methylated genes are involved in critical pathways, such as DNA repair, metabolism, and invasion/metastasis. The role of hypermethylated genes in regulation of critical pathways in prostate cancer is discussed. These findings may provide new information of the pathogenesis, the exciting potential to be predictive and to provide personalized treatment of prostate cancer. Indeed, some epigenetic alterations in prostate tumors are being translated into clinical practice for therapeutic use.
Collapse
Affiliation(s)
- Mihi Yang
- Division of Cancer Prevention and Controls, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | |
Collapse
|
21
|
Dai Y, Wang J, Xia J, Hong Y, Chen N, Cheng B. Genome-wide transcriptional profiling analysis of all trans retinoic acid-treated tongue carcinoma SCC-9 cells. J Surg Oncol 2011; 104:830-5. [PMID: 21725974 DOI: 10.1002/jso.21998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 05/27/2011] [Indexed: 11/05/2022]
Abstract
BACKGROUND All trans retinoic acid (ATRA) is used as standard of care in promyelocytic leukemia. Not much is known about the gene expression profile in ATRA-treated tongue cancer cells. We performed a genome-wide transcriptional profiling of ATRA-treated tongue cancer cells to understand the pathways that mediate ATRA action in tongue cancer. METHODS We measured the effects of ATRA on the proliferation of SCC-9 human tongue carcinoma cells. The differential gene expression profile was measured by microarray analysis of untreated and ATRA-treated cells and expression of key genes was validated by real-time RT-PCR. RESULTS ATRA treatment (24 and 48 hr) significantly inhibited SCC-9 cell proliferation in a dose-dependent manner. SCC-9 cells treated for 48 hr with ATRA showed upregulation of 276 genes, including ANGPTL4, GDF15, ICAM1 and TUSC4, and downregulation of 43 genes, including CXCL10. Validation by real-time PCR showed a significant upregulation of intracellular adhesion molecule 1 (ICAM1) and downregulation of CXCL10 and IL32. CONCLUSIONS ATRA had an anti-tumor effect in tongue cancer cells. This effect is likely mediated via upregulation of ICAM1 and downregulation of CXCL10 and IL32.
Collapse
Affiliation(s)
- Yaohui Dai
- Department of Oral Medicine, Guanghua School of Stomatology and Institute of Stomatological Research, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
22
|
Acquafreda T, Nunes FD, Soprano DR, Soprano KJ. Expression of homeobox genes in oral squamous cell carcinoma cell lines treated with all-trans retinoic acid. J Cell Biochem 2011; 111:1437-44. [PMID: 20830740 DOI: 10.1002/jcb.22871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Oral squamous cell carcinoma (OSCC) may arise from potentially malignant oral lesions. All-trans retinoic acid (atRA), which plays a role in cell growth and differentiation, has been studied as a possible chemotherapeutic agent in the prevention of this progression. While the mechanism by which atRA suppresses cell growth has not been completely elucidated, it is known that homeobox genes are atRA targets. To determine if these genes are involved in the atRA-mediated OSCC growth inhibition, PCR array was performed to evaluate the expression of 84 homeobox genes in atRA-sensitive SCC-25 cells compared to atRA-resistant SCC-9 cells following 7 days with atRA treatment. Results showed that the expression of 8 homeobox genes was downregulated and expression of 4 was upregulated in SCC-25 cells but not in SCC-9 cells. Gene expression levels were confirmed for seven of these genes by RT-qPCR. Expression of three genes that showed threefold downregulation was evaluated in SCC-25 cells treated with atRA for 3, 5, and 7 days. Three different patterns of atRA-dependent gene expression were observed. ALX1 showed downregulation only on day 7. DLX3 showed reduced expression on day 3 and further reduced on day 7. TLX1 showed downregulation only on days 5 and 7. Clearly the expression of homeobox genes is modulated by atRA in OSCC cell lines. However, the time course of this modulation suggests that these genes are not direct targets of atRA mediating OSCC growth suppression. Instead they appear to act as downstream effectors of atRA signaling.
Collapse
Affiliation(s)
- Thais Acquafreda
- Department of Oral Pathology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
23
|
Abstract
BACKGROUND The prostate gland is the most common site of cancer and the second leading cause of cancer mortality in American men. It is well known that epigenetic alterations such as DNA methylation within the regulatory (promoter) regions of genes are associated with transcriptional silencing in cancer. Promoter hypermethylation of critical pathway genes could be potential biomarkers and therapeutic targets for prostate cancer. METHODS This review discusses current information on methylated genes associated with prostate cancer development and progression. RESULTS Over 30 genes have been investigated for promoter methylation in prostate cancer. These methylated genes are involved in critical pathways, such as DNA repair, metabolism, and invasion/metastasis. The role of hypermethylated genes in regulation of critical pathways in prostate cancer is reviewed. CONCLUSIONS These findings may provide new information of the pathogenesis of prostate cancer. Certain epigenetic alterations in prostate tumors are being translated into clinical practice for therapeutic use.
Collapse
Affiliation(s)
- Jong Y Park
- Division of Cancer Prevention and Control, Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
24
|
Bushue N, Wan YJY. Retinoid pathway and cancer therapeutics. Adv Drug Deliv Rev 2010; 62:1285-98. [PMID: 20654663 DOI: 10.1016/j.addr.2010.07.003] [Citation(s) in RCA: 245] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2010] [Revised: 07/08/2010] [Accepted: 07/14/2010] [Indexed: 12/18/2022]
Abstract
The retinoids are a class of compounds that are structurally related to vitamin A. Retinoic acid, which is the active metabolite of retinol, regulates a wide range of biological processes including development, differentiation, proliferation, and apoptosis. Retinoids exert their effects through a variety of binding proteins including cellular retinol-binding protein (CRBP), retinol-binding proteins (RBP), cellular retinoic acid-binding protein (CRABP), and nuclear receptors i.e. retinoic acid receptor (RAR) and retinoid x receptor (RXR). Because of the pleiotropic effects of retinoids, understanding the function of these binding proteins and nuclear receptors assists us in developing compounds that have specific effects. This review summarizes our current understanding of how retinoids are processed and act with an emphasis on the application of retinoids in cancer treatment and prevention.
Collapse
Affiliation(s)
- Nathan Bushue
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | | |
Collapse
|
25
|
Siddikuzzaman, Guruvayoorappan C, Berlin Grace VM. All trans retinoic acid and cancer. Immunopharmacol Immunotoxicol 2010; 33:241-9. [PMID: 20929432 DOI: 10.3109/08923973.2010.521507] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
All-trans retinoic acid (ATRA) is an active metabolite of vitamin A under the family retinoid. Retinoids, through their cognate nuclear receptors, exert potent effects on cell growth, differentiation and apoptosis, and have significant promise for cancer therapy and chemoprevention. Differentiation therapy with ATRA has marked a major advance and become the first choice drug in the treatment of acute promyelocytic leukemia (APL). Conversions of 13-cis-retinoic acid and 9-cis-retinoic acid to all-trans-retinoic acid is very rapid. Currently, two distinct families of retinoid responsive nuclear receptors have been identified and characterized: retinoic acid receptors (RARs) and retinoid receptors (RXRs), each of which include three isoforms, α,β,and γ. ATRA is being increasingly included in anti-tumour therapeutical schemes for the treatment of various tumoral diseases such as Kaposi's sarcoma, head and neck squamous cell carcinoma, ovarian carcinoma, bladder cancer, neuroblastoma and has shown antiangiogenic effects in several systems, inhibiting proliferation in vascular smooth muscle cells (VSMCs) and anti-inflammatory in rheumatoid arthritis. This review helps to understand in details about the ATRA and its role on cancer and it is predicted that modulating the activity of ATRA will soon provide novel prevention and treatment approaches for the cancer patients.
Collapse
Affiliation(s)
- Siddikuzzaman
- Department of Biotechnology, School of Biotechnology & Health Sciences, Karunya University, Karunya Nagar, Coimbatore - 641 114, Tamil Nadu, India
| | | | | |
Collapse
|
26
|
Hong TK, Lee-Kim YC. Effects of retinoic acid isomers on apoptosis and enzymatic antioxidant system in human breast cancer cells. Nutr Res Pract 2009; 3:77-83. [PMID: 20016705 PMCID: PMC2788174 DOI: 10.4162/nrp.2009.3.2.77] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/16/2009] [Accepted: 04/30/2009] [Indexed: 11/04/2022] Open
Abstract
Retinoic acids (RAs) modulate growth, differentiation, and apoptosis in normal, pre-malignant & malignant cells. In the present study, the effects of RA isomers (all-trans RA, 13-cis RA, and 9-cis RA) on the cell signal transduction of human breast cancer cells have been studied. The relationship between RAs and an enzymatic antioxidant system was also determined. Estrogen-receptor (ER) positive MCF-7 and ER-negative MDA-MB-231 human breast cancer cells were treated with different doses of each RA isomers, all-trans RA, 13-cis RA, or 9-cis RA. Treatment of RA isomers inhibited cell viability and induced apoptosis of MCF-7 cells as a result of increased caspase activity in cytoplasm and cytochrome C released from mitochondria. All-trans RA was the most effective RA isomer in both cell growth inhibition and induction of apoptosis in MCF-7 cells. However, no significant effect of RA isomers was observed on the cell growth or apoptosis in ER-negative MDA-MB-231 cells. In addition, activities of antioxidant enzymes such as catalase and glutathione peroxidase were decreased effectively after treatment of RA in MCF-7 cells, whereas SOD activity was rarely affected. Thus, the present data suggest that all-trans RA is the most potential inducer of apoptosis and modulator of antioxidant enzymes among RA isomers in MCF-7 human breast cancer cells.
Collapse
Affiliation(s)
- Tae-Kyong Hong
- Department of Food and Nutrition, Yonsei University, 134 Shinchon-dong, Seodaemun-gu, Seoul 120-749, Korea
| | | |
Collapse
|
27
|
Tatebe H, Shimizu M, Shirakami Y, Sakai H, Yasuda Y, Tsurumi H, Moriwaki H. Acyclic retinoid synergises with valproic acid to inhibit growth in human hepatocellular carcinoma cells. Cancer Lett 2009; 285:210-7. [PMID: 19520494 DOI: 10.1016/j.canlet.2009.05.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 05/18/2009] [Accepted: 05/18/2009] [Indexed: 01/09/2023]
Abstract
A malfunction of retinoid X receptor-alpha (RXRalpha) due to phosphorylation is associated with the development of hepatocellular carcinoma (HCC) and acyclic retinoid (ACR), which targets RXRalpha, can prevent the development of second primary HCC. Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, induces apoptosis and cell cycle arrest in cancer cells. VPA can also enhance the sensitivity of cancer cells to retinoids. The present study examined the possible combined effects of ACR plus VPA in HepG2 human HCC cell line. The combination of 5muM ACR and 1mM VPA, about the IC(25) value for both compounds, synergistically inhibited the growth of HepG2 cells without affecting the growth of Hc normal human hepatocytes. The combined treatment with ACR plus VPA also acted synergistically to induce apoptosis and G(0)-G(1) cell cycle arrest in HepG2 cells. This combination further exerted a synergistic inhibition of the phosphorylation of RXRalpha, ERK, Akt and GSK-3beta proteins and caused an accumulation of acetylated histones H3 and H4 proteins. VPA enhanced the ability of ACR to raise the cellular levels of RARbeta and p21(CIP1). The combination of these agents markedly increased both the RARE and RXRE promoter activities in HepG2 cells. These results suggest that ACR and VPA cooperatively increase the expression of RARbeta and p21(CIP1), while inhibiting the phosphorylation of RXRalpha, and these effects were associated with induction of apoptosis and the inhibition of cell growth in HepG2 cells. This combination might therefore be an effective regimen for the chemoprevention and chemotherapy of HCC.
Collapse
Affiliation(s)
- Hideharu Tatebe
- Department of Medicine, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Tekade RK, Dutta T, Tyagi A, Bharti AC, Das BC, Jain NK. Surface-engineered dendrimers for dual drug delivery: A receptor up-regulation and enhanced cancer targeting strategy. J Drug Target 2008; 16:758-72. [DOI: 10.1080/10611860802473154] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
29
|
Ran L, Tan W, Tan S, Zhang R, Wang W, Zeng W. Effects of ATRA, acitretin and tazarotene on growth and apoptosis of Tca8113 cells. ACTA ACUST UNITED AC 2007; 25:393-6. [PMID: 16196285 DOI: 10.1007/bf02828205] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To investigate the effects of ATRA, acitretin and tazarotene on the growth and apoptosis of human tongue squamous cell carcinoma cell line Tca8113. The effect of retinoids on growth of Tca8113 cells in vitro was examined by MTT assay and Trypan blue exclusion assay. Cell cycle analysis, early apoptosis analysis with double staining with Annexin V-FITC and PI, and active caspase-3 analysis with the staining of FITC-conjugated monoclonal rabbit anti-active caspase-3 antibody were made by flow cytometer. Streptavidin-biotin complex (SABC) immunocytochemical assays were employed for the detections of Bax/Bcl-2 proteins expressions. Our results showed that the retinoids inhibited growth of Tca8113 cells in a dose- and time-dependent manner with maximal inhibition 24 h after treatment of 10(-5) mol/L. 10(-5) mol/L retinoids altered cell cycle distribution of Tca8113 cells, revealing an increase in G0/G1-phase population, a decrease in S-phase population and the inhibition of G1/S switching. 10(-5) mol/L retinoids significantly induced apoptosis of Tca8113 cells (all P < 0.05), elevated the cells population with detectable active caspase-3 (P < 0.05 for all), increased the number of cells forming Bax and decreased the number of cells forming Bcl-2 significantly (all P < 0.05). Acitretin played a most prominent role among the retinoids. It is concluded that the inhibition of cell cycle progress of Tca8113 cells by ATRA, acitretin and tazarotene is one of the possible mechanisms for proliferation arrest of Tca8113 cells elicited by the retinoids. The retinoids mediate apoptosis in Tca8113 cells that may be caspase-dependent through mitochondria pathway. High concentration retinoids inhibit growth of Tca8113 cells in vitro by interfering with proliferation and inducing apoptosis of cells. Acitretin may be an alternative medicine for the prevention and treatment of tongue squamous cell carcinoma.
Collapse
Affiliation(s)
- Liwei Ran
- Department of Dermatology and Venereology, the Second Hospital, Xian Jiaotong University, Xian 710004, China
| | | | | | | | | | | |
Collapse
|
30
|
Kada N, Suzuki T, Aizawa K, Matsumura T, Ishibashi N, Suzuki N, Takeda N, Munemasa Y, Sawaki D, Ishikawa T, Nagai R. Acyclic Retinoid Inhibits Neointima Formation Through Retinoic Acid Receptor Beta-Induced Apoptosis. Arterioscler Thromb Vasc Biol 2007; 27:1535-41. [PMID: 17478760 DOI: 10.1161/atvbaha.106.134114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objectives—
Acyclic retinoid (ACR) is a synthetic retinoid with a high safety profile that has been pursued with high expectations for therapeutic use in prevention (recurrence) and treatment of malignancies. With the objective of addressing the therapeutic potential in the cardiovasculature, namely neointima formation, effects of ACR on neointima formation and the involved mechanisms were investigated.
Methods and Results—
ACR was administered to cuff-injured mice which showed inhibition of neointima formation. Investigation of involved mechanisms at the cellular and molecular levels showed that ACR induces apoptosis of neointimal cells and this to be mediated by selective induction of retinoic-acid receptor β (RARβ) which shows growth inhibitory and proapoptotic effects on smooth muscle cells.
Conclusion—
We show that ACR inhibits neointima formation by inducing RARβ which in turn inhibits cell growth and induces apoptosis. The retinoid, ACR, may be potentially exploitable for treatment and prevention of neointima formation.
Collapse
Affiliation(s)
- Nanae Kada
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kawakami S, Suzuki S, Yamashita F, Hashida M. Induction of apoptosis in A549 human lung cancer cells by all-trans retinoic acid incorporated in DOTAP/cholesterol liposomes. J Control Release 2006; 110:514-21. [PMID: 16360957 DOI: 10.1016/j.jconrel.2005.10.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 10/25/2005] [Accepted: 10/31/2005] [Indexed: 01/29/2023]
Abstract
All-trans retinoic acid (ATRA) has been shown to exert anti-cancer activities in a number of types of cancer cells. However, it has been reported that many NSCLC exhibited resistance to ATRA treatment. In the present study, we hypothesized that intracellular delivery of ATRA would overcome the ATRA resistance in A549 cells. Here, we investigated the induction of apoptosis by ATRA incorporated in cationic liposomes composed of DOTAP/cholesterol in A549 human lung cancer cells, which are insensitive (resistant) to the growth inhibitory effects of ATRA. The zeta potentials of DOTAP/cholesterol liposomes and DSPC/cholesterol liposomes were about +50 and -3 mV. In A549 cells, [(3)H]ATRA incorporated in DOTAP liposomes showed increased cellular association compared with [(3)H]ATRA or [(3)H]ATRA incorporated in DSPC/cholesterol liposomes. ATRA incorporated in DOTAP/cholesterol liposomes showed much higher cytotoxic effects and apoptosis-inducing activity compared with ATRA or ATRA incorporated in DSPC/cholesterol liposomes. The enhanced expression of TIG3 mRNA tumor suppressor gene by ATRA incorporation into DOTAP/cholesterol liposomes might partly explain the mechanism of enhanced cytotoxicity and/or apoptosis. These observations provide valuable information to help in the design of differentiation therapy by ATRA in non-small cell lung carcinoma.
Collapse
Affiliation(s)
- Shigeru Kawakami
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | | | | | | |
Collapse
|
32
|
Ralhan R, Chakravarti N, Kaur J, Sharma C, Kumar A, Mathur M, Bahadur S, Shukla NK, Deo SVS. Clinical significance of altered expression of retinoid receptors in oral precancerous and cancerous lesions: Relationship with cell cycle regulators. Int J Cancer 2005; 118:1077-89. [PMID: 16161051 DOI: 10.1002/ijc.21483] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Alterations in expression of retinoid receptors are implicated in human cancers. We hypothesized that altered expression of retinoic acid receptors (RARalpha,beta,gamma) and retinoid X receptor RXRalpha and their relationship with cell cycle regulators (p53, p16, p21) is associated with development, progression and prognosis of oral cancer. Immunohistochemical analysis of RAR alpha, beta, gamma and RXRalpha proteins was carried out on serial sections from 244 oral squamous cell carcinomas (OSCCs), 102 potentially malignant lesions (65 hyperplasias, 37 dysplasias), 83 matched histologically normal oral tissues and 29 normal mucosa from non-exposed individuals without oral lesions and correlated with expression of cell cycle regulators p53, p16 and p21 as well as with clinicopathological parameters. Expression of retinoid receptors RARbeta, RARgamma, RXRalpha and cell cycle regulators p16 and p21 was decreased in majority of oral SCCs as well as in potentially malignant lesions. Multivariate stepwise logistic regression analysis carried out for comparison of non-exposed normal oral mucosa with histologically normal oral tissues from patients with oral lesions showed significant loss of RARbeta or p53 accumulation (RARbeta(-)/p53(+) Odd's ratio, OR = 266.6, p = 0.000); non-exposed normal mucosa from individuals without oral lesions with potentially malignant lesion was RARbeta(-)/p21(-)/p53(+) (OR = 215.7, p = 0.000); matched normal to potentially malignant stage was RARalpha(+)/p21(-) (OR = 4.414, p = 0.005); hyperplasia to dysplasia was RARalpha(+)/p53(+) (OR = 4.72, p = 0.005) and potentially malignant to malignant phenotype was RARalpha(+) (OR = 2.061, p = 0.004). The prognostic relevance of these factors was assessed in 115 of these SCC patients who were followed-up for a maximum period of 94 months (median 21 months). Multivariate analysis using Cox's proportional Hazard's model showed that RARalpha(+)/p21(-) phenotype was associated with shorter disease-free survival (Hazard's ratio, HR = 1.863, p = 0.0471). To our knowledge, this is the first large study showing alterations in expression of retinoid receptors at the protein level at different stages in development and progression of oral SCC. It also underscored the prognostic significance of retinoid receptors and their interactions with cell cycle regulators in multistep oral tumorigenesis.
Collapse
Affiliation(s)
- Ranju Ralhan
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi-100029, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
AIM: To investigate the pathological characteristics and carcinogenesis mechanism of benign lymphoadenosis of oral mucosa (BLOM).
METHODS: The expressions of Ki-67, CD34 and apoptosis were evaluated by immunohistochemical SP staining in 64 paraffin-embedded tissue samples. Of them, 9 were from BLOM with dysplasia, 15 from BLOM without dysplasia, 15 from oral squamous cell carcinoma (OSCC), 15 from oral precancerosis, and 10 from normal tissues. Cell proliferation, apoptosis and angiogenesis of tissue samples were also analyzed.
RESULTS: The expression of Ki-67 in BLOM with dysplasia, oral precancerosis and OSCC was significantly higher than in BLOM without dysplasia and normal mucosa. The microvascular density (MVD) in BLOM with and without dysplasia, oral precancerosis, and OSCC was significantly higher than in normal mucosa. Apoptosis in BLOM and oral precancerosis was significantly higher than in OSCC and normal mucosa.
CONCLUSION: Benign lymphoadenosis of oral mucosa has potentialities of cancerization.
Collapse
Affiliation(s)
- Shu-Xia Li
- Department of Oral Pathology, School of Stomatology, Peking University, Beijing 100081, China.
| | | | | |
Collapse
|
34
|
Shimizu M, Suzui M, Deguchi A, Lim JTE, Xiao D, Hayes JH, Papadopoulos KP, Weinstein IB. Synergistic effects of acyclic retinoid and OSI-461 on growth inhibition and gene expression in human hepatoma cells. Clin Cancer Res 2005; 10:6710-21. [PMID: 15475462 DOI: 10.1158/1078-0432.ccr-04-0659] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hepatoma is one of the most frequently occurring cancers worldwide. However, effective chemotherapeutic agents for this disease have not been developed. Acyclic retinoid, a novel synthetic retinoid, can reduce the incidence of postsurgical recurrence of hepatoma and improve the survival rate. OSI-461, a potent derivative of exisulind, can increase intracellular levels of cyclic GMP, which leads to activation of protein kinase G and induction of apoptosis in cancer cells. In the present study, we examined the combined effects of acyclic retinoid plus OSI-461 in the HepG2 human hepatoma cell line. We found that the combination of as little as 1.0 micromol/L acyclic retinoid and 0.01 micromol/L OSI-461 exerted synergistic inhibition of the growth of HepG2 cells. Combined treatment with low concentrations of these two agents also acted synergistically to induce apoptosis in HepG2 cells through induction of Bax and Apaf-1, reduction of Bcl-2 and Bcl-xL, and activation of caspase-3, -8, and -9. OSI-461 enhanced the G0-G1 arrest caused by acyclic retinoid, and the combination of these agents caused a synergistic decrease in the levels of expression of cyclin D1 protein and mRNA, inhibited cyclin D1 promoter activity, decreased the level of hyperphosphorylated forms of the Rb protein, induced increased cellular levels of the p21(CIP1) protein and mRNA, and stimulated p21(CIP1) promoter activity. Moreover, OSI-461 enhanced the ability of acyclic retinoid to induce increased cellular levels of retinoic acid receptor beta and to stimulate retinoic acid response element-chloramphenicol acetyltransferase activity. A hypothetical model involving concerted effects on p21(CIP1) and retinoic acid receptor beta expression is proposed to explain these synergistic effects. Our results suggest that the combination of acyclic retinoid plus OSI-461 might be an effective regimen for the chemoprevention and chemotherapy of human hepatoma and possibly other malignancies.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Adhesion Molecules/metabolism
- Cell Cycle Proteins/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Chloramphenicol O-Acetyltransferase/genetics
- Chloramphenicol O-Acetyltransferase/metabolism
- Cyclic GMP/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Cyclin-Dependent Kinase Inhibitor p21
- Cytoskeletal Proteins/metabolism
- Dose-Response Relationship, Drug
- Drug Synergism
- G1 Phase/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Intracellular Space/drug effects
- Intracellular Space/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Microfilament Proteins
- Models, Biological
- Phosphoproteins/metabolism
- Phosphorylation/drug effects
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Response Elements/genetics
- Resting Phase, Cell Cycle/drug effects
- Retinoblastoma Protein/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sulindac/analogs & derivatives
- Sulindac/pharmacology
- Trans-Activators/metabolism
- Transfection
- Tretinoin/analogs & derivatives
- Tretinoin/pharmacology
- Tumor Suppressor Protein p53/metabolism
- beta Catenin
Collapse
Affiliation(s)
- Masahito Shimizu
- Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Studies utilizing experimental animals, epidemiological approaches, cellular models, and clinical trials all provide evidence that retinoic acid and some of its synthetic derivatives (retinoids) are useful pharmacological agents in cancer therapy and prevention. In this chapter, we first review the current knowledge of retinoic acid receptors (RARs) and their role in mediating the actions of retinoic acid. We then focus on a discussion of RARalpha and acute promyelocytic leukemia followed by a discussion of the role of RARs, in particular RARbeta expression, in other cancer types. Loss of normal RAR function in the presence of physiological levels of RA (either due to alterations in the protein structure or level of expression) is associated with a variety of different cancers. In some cases treatment with pharmacological doses of RA can be effective.
Collapse
Affiliation(s)
- Dianne Robert Soprano
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA.
| | | | | |
Collapse
|
36
|
Youssef EM, Lotan D, Issa JP, Wakasa K, Fan YH, Mao L, Hassan K, Feng L, Lee JJ, Lippman SM, Hong WK, Lotan R. Hypermethylation of the retinoic acid receptor-beta(2) gene in head and neck carcinogenesis. Clin Cancer Res 2004; 10:1733-42. [PMID: 15014026 DOI: 10.1158/1078-0432.ccr-0989-3] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Retinoic acid receptor-beta(2) (RAR-beta(2)) expression is suppressed in oral premalignant lesions and head and neck squamous cell carcinomas (HNSCCs). This study was conducted to determine whether RAR-beta(2) gene expression in such lesions can be silenced by promoter methylation. EXPERIMENTAL DESIGN RAR-beta(2) methylation was analyzed in DNA samples from 22 pairs of primary HNSCC and adjacent normal epithelium, 124 samples of oral leukoplakia, and 18 HNSCC cell lines using methylation-specific PCR. RAR-beta(2) promoter was methylated in 67, 56, and 53% of HNSCC tumors, HNSCC cell lines, and microdissected oral leukoplakia specimens, respectively. RAR-beta(2) hypermethylation was confirmed by sodium bisulfite-PCR combined with restriction enzyme digestion analysis and by random cloning and sequencing of bisulfite-treated DNA isolates. RESULTS Significantly higher RAR-beta(2) hypermethylation levels were found in tumor tissue compared with adjacent normal tissue (P = 0.002). RAR-beta(2) methylation in the cell lines was correlated with loss of RAR-beta(2) expression (P = 0.013) and inversely related to the presence of mutated p53 (P = 0.025). The demethylating agent 5-aza-2'-deoxycytidine (5-aza-CdR) restored RAR-beta(2) inducibility by all-trans-retinoic acid (ATRA) in some of the cell lines, which posses a methylated RAR-beta(2) promoter. In some cell lines, this effect was associated with increased growth inhibition after combined treatment with 5-aza-CdR and ATRA. CONCLUSIONS RAR-beta(2) silencing by methylation is an early event in head and neck carcinogenesis; 5-Aza-CdR can restore RAR-beta(2) inducibility by ATRA in most cell lines, and the combination of 5-aza-CdR and ATRA is more effective in growth inhibition than single agents.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Base Sequence
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Cell Division
- Cell Line, Tumor
- Cloning, Molecular
- DNA Methylation
- DNA Primers
- DNA, Neoplasm/genetics
- Female
- Gene Silencing
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/pathology
- Humans
- Leukoplakia, Oral/genetics
- Leukoplakia, Oral/pathology
- Male
- Middle Aged
- Molecular Sequence Data
- Mouth Neoplasms/genetics
- Mouth Neoplasms/pathology
- Polymerase Chain Reaction
- Precancerous Conditions/genetics
- Precancerous Conditions/pathology
- Promoter Regions, Genetic/genetics
- Receptors, Retinoic Acid/drug effects
- Receptors, Retinoic Acid/genetics
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- Emile M Youssef
- Departments of Thoracic/Head and Neck Medical Oncology,The University of Texas, MD Anderson Cancer Center, Houston, Texas 77345, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Toyoshiba H, Yamanaka T, Sone H, Parham FM, Walker NJ, Martinez J, Portier CJ. Gene interaction network suggests dioxin induces a significant linkage between aryl hydrocarbon receptor and retinoic acid receptor beta. ENVIRONMENTAL HEALTH PERSPECTIVES 2004; 112:1217-24. [PMID: 15345368 PMCID: PMC1277115 DOI: 10.1289/txg.7020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Gene expression arrays (gene chips) have enabled researchers to roughly quantify the level of mRNA expression for a large number of genes in a single sample. Several methods have been developed for the analysis of gene array data including clustering, outlier detection, and correlation studies. Most of these analyses are aimed at a qualitative identification of what is different between two samples and/or the relationship between two genes. We propose a quantitative, statistically sound methodology for the analysis of gene regulatory networks using gene expression data sets. The method is based on Bayesian networks for direct quantification of gene expression networks. Using the gene expression changes in HPL1A lung airway epithelial cells after exposure to 2,3,7,8-tetrachlorodibenzo-(Italic)p(/Italic)-dioxin at levels of 0.1, 1.0, and 10.0 nM for 24 hr, a gene expression network was hypothesized and analyzed. The method clearly demonstrates support for the assumed network and the hypothesis linking the usual dioxin expression changes to the retinoic acid receptor system. Simulation studies demonstrated the method works well, even for small samples.
Collapse
Affiliation(s)
- Hiroyoshi Toyoshiba
- Laboratory of Computational Biology and Risk Analysis, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Hsu S, Singh B, Schuster G. Induction of apoptosis in oral cancer cells: agents and mechanisms for potential therapy and prevention. Oral Oncol 2004; 40:461-73. [PMID: 15006617 DOI: 10.1016/j.oraloncology.2003.09.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2003] [Accepted: 09/24/2003] [Indexed: 12/23/2022]
Abstract
Oral cancer is one of the most disfiguring types of cancer, since the surgical removal of the tumor may result in facial distortion. Oral cancer is also known to exhibit "field cancerization", resulting in the development of a second primary tumor. Furthermore, the five-year survival rate of this disease has remained approximately 50% during the past 30 years. Prevention and early detection/treatment of oral cancer could significantly improve the quality of life for individuals at risk. Recently, the targeted elimination of oral squamous cell carcinoma cells by inducing apoptosis has emerged as a valued strategy to combat oral cancer. Studies utilizing a variety of chemical or biological interventions demonstrated promising results for induction of apoptosis in oral malignant cells. This review summarizes the results of a number of investigations focused specifically on induction of apoptosis in oral cancer cells by synthetic compounds and naturally occurring chemopreventive agents with apoptotic potential.
Collapse
Affiliation(s)
- Stephen Hsu
- Department of Oral Biology and Maxillofacial Pathology, School of Dentistry, Medical College of Georgia, AD 1443, Augusta, GA 30912-1125, USA.
| | | | | |
Collapse
|
39
|
Dhillon VS, Young AR, Husain SA, Aslam M. Promoter hypermethylation of MGMT, CDH1, RAR-beta and SYK tumour suppressor genes in granulosa cell tumours (GCTs) of ovarian origin. Br J Cancer 2004; 90:874-81. [PMID: 14970867 PMCID: PMC2410165 DOI: 10.1038/sj.bjc.6601567] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Ovarian carcinoma (OC) is a leading cause of death among women throughout the world. A number of cancer-associated genes have been shown to be inactivated by hypermethylation of CpG islands during tumorigenesis. We tested the hypothesis that methylation status of MGMT, CDH1, RAR-β and SYK could be important in the ovarian tumorigenic process and can lead to the gene(s) inactivation. Therefore, we assessed the promoter hypermethylation of MGMT, CDH1, RAR-β and SYK in 43 ovarian granulosa cell tumours (GCTs) (adult type) using methylation-specific PCR. These tumours are relatively rare, accounting for approximately 3% of all ovarian cancers. Hypermethylation of MGMT (in 14 tumours), CDH1 (in nine tumours), RAR-β (in eight tumours) and SYK (in seven tumours) have been found. Selective loss of RAR-β and RAR-β2 mRNA has been found in seven patients, while that of MGMT and SYK in three patients who also show aberrant methylation in promoter region of RAR-β in addition to MGMT, SYK and CDH1 genes. Promoter CpG hypermethylation may be an alternative to mutation(s) to inactivate tumour suppressor genes such as MGMT, CDH1, RAR-β and SYK, and this can also be an early event in the pathogenesis of OCs. Moreover, hypermethylation of the MGMT and CDH1, MGMT and RAR-β and CDH1 and RAR-β promoters occurred concordantly (P< 0.001, 0.0421 and 0.0005 respectively; Fischer's exact test). In addition to this, monosomy 22 and trisomy 14 have also been found in 10 tumours. It is clear from the results that hypermethylation of the promoter region of these tumour suppressor genes, monosomy 22 and trisomy 14, may be critical steps in the tumorigenesis, which consequently play a permissive role for tumour aggressiveness. All these events might play an important role in the early clinical diagnosis of the disease. Our results, therefore, suggest a potential role for epigenetic modification of these critical tumour suppressor genes in pathways relevant to the transformation and differentiation of rare type of ovarian cancer (GCTs).
Collapse
Affiliation(s)
- V S Dhillon
- Department of Molecular and Clinical Genetics, Royal Prince Alfred Hospital, Camperdown NSW 2050, Australia.
| | | | | | | |
Collapse
|
40
|
Li R, Faria TN, Boehm M, Nabel EG, Gudas LJ. Retinoic acid causes cell growth arrest and an increase in p27 in F9 wild type but not in F9 retinoic acid receptor β2 knockout cells. Exp Cell Res 2004; 294:290-300. [PMID: 14980522 DOI: 10.1016/j.yexcr.2003.11.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2003] [Revised: 11/10/2003] [Indexed: 11/16/2022]
Abstract
We have previously shown that an F9 teratocarcinoma retinoic acid receptor beta(2) (RARbeta(2)) knockout cell line exhibits no growth arrest in response to all-trans-retinoic acid (RA), whereas F9 wild type (Wt), F9 RARalpha(-/-), and F9 RARgamma(-/-) cell lines do growth arrest in response to RA. To examine the role of RARbeta(2) in growth inhibition, we analyzed the cell cycle regulatory proteins affected by RA in F9 Wt and F9 RARbeta(2)(-/-) cells. Flow microfluorimetry analyses revealed that RA treatment of F9 Wt cells greatly increased the percentage of cells in the G1/G0 phase of the cell cycle. In contrast, RA did not alter the cell cycle distribution profile of RARbeta(2)(-/-) cells. In F9 Wt cells, cyclin D1, D3, and cyclin E protein levels decreased, while cyclin D2 and p27 levels increased after RA treatment. Compared to the F9 Wt cells, the F9 RARbeta(2)(-/-) cells exhibited lower levels of cyclins D1, D2, D3, and E in the absence of RA, but did not exhibit further changes in the levels of these cell cycle regulators after RA addition. Since RA significantly increased the level of p27 protein (approximately 24-fold) in F9 Wt as compared to the F9 RARbeta(2)(-/-) cells, we chose to study p27 in greater detail. The p27 mRNA level and the rate of p27 protein synthesis were increased in RA-treated F9 Wt cells, but not in F9 RARbeta(2)(-/-) cells. Moreover, RA increased the half-life of p27 protein in F9 Wt cells. Reduced expression of RARbeta(2) is associated with the process of carcinogenesis and RARbeta(2) can mediate the growth arrest induced by RA in a variety of cancer cells. Using both genetic and molecular approaches, we have identified some of the molecular mechanisms, such as the large elevation of p27, through which RARbeta(2) mediates these growth inhibitory effects of RA in F9 cells.
Collapse
Affiliation(s)
- Rong Li
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
41
|
Shimizu M, Suzui M, Deguchi A, Lim JTE, Weinstein IB. Effects of Acyclic Retinoid on Growth, Cell Cycle Control, Epidermal Growth Factor Receptor Signaling, and Gene Expression in Human Squamous Cell Carcinoma Cells. Clin Cancer Res 2004; 10:1130-40. [PMID: 14871993 DOI: 10.1158/1078-0432.ccr-0714-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We described recently the growth inhibitory effects of the novel compound acyclic retinoid (ACR) in human hepatoma cell lines (M. Suzui et al., Cancer Res., 62: 3997-4006, 2002). In this study we examined the cellular and molecular effects of ACR on human squamous cell carcinoma (SCC) cells. ACR inhibited growth of the esophageal SCC cell line HCE7, and the head and neck SCC cell lines YCU-N861 and YCU-H891, with IC(50) values of approximately 10, 25, and 40 microM, respectively. Detailed studies were then done with HCE7 cells. Treatment of these cells with 10 microM ACR caused an increase of cells in G(0)-G(1) and induced apoptosis. This was associated with two phases of molecular events. During phase 1, which occurred within 6-12 h, there was an increase in the retinoic acid receptor beta (RARbeta) and p21(CIP1) proteins, and their corresponding mRNAs, and a decrease in the hyperphosphorylated form of the retinoblastoma protein. During phase 2, which occurred at approximately 24 h, there was a decrease in the cellular level of transforming growth factor alpha, and the phosphorylated (i.e., activated) forms of the epidermal growth factor receptor, Stat3, and extracellular signal-regulated kinase proteins, and a decrease in both cyclin D1 protein and mRNA. Reporter assays indicated that ACR inhibited the transcriptional activity of the cyclin D1, c-fos, and activator protein promoters. On the other hand, ACR markedly stimulated the activity of a retinoic acid response element-CAT reporter when the cells were cotransfected with a RARbeta expression vector. A hypothetical model explaining these two phases is presented. The diverse effects that we obtained with ACR suggest that this agent might be useful in the chemoprevention and/or therapy of human SCCs.
Collapse
Affiliation(s)
- Masahito Shimizu
- Herbert Irving Comprehensive Cancer Center, College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York, NY 10032-2704, USA
| | | | | | | | | |
Collapse
|
42
|
Kaur J, Ralhan R. Establishment and characterization of a cell line from smokeless tobacco associated oral squamous cell carcinoma. Oral Oncol 2003; 39:806-20. [PMID: 13679204 DOI: 10.1016/s1368-8375(03)00084-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A cell line, AMOS-III has been established from the surgically resected specimen of an untreated primary human oral squamous cell carcinoma of the floor of mouth from a chronic smokeless tobacco consumer. Immunocytochemical analysis showed epithelial specific antigen, cytokeratins 5, 10, 13 and 16 and integrin alpha(6) markers in AMOS-III cells, confirming the epithelial lineage of the cell line. Analyses of morphology, ultrastructure, karyotype, anchorage independent growth and immunocytochemical properties of the cell line demonstrated the transformed phenotype of epithelial cells. AMOS-III cells have doubling time of 42-44 h. Giemsa-banding patterns of chromosomes confirmed the human origin of the AMOS-III cells. Molecular analysis of cancer-related gene products, p53 and p21(cip1/waf1) showed the presence of wild type p21(cip1/waf1) and truncated p53 proteins. The molecular mechanism underlying the action of retinoids in preventing the occurrence of second primary tumors in oral cancer patients remain to be clearly defined. Treatment of AMOS-III cells with all-trans retinoic acid (ATRA) at 10(-4) microM resulted in 81% cell death. ATRA treatment resulted in enhanced expression of p21(cip1/waf1), nuclear translocation of retinoic acid receptors and apoptotic cell death. Thus, this cell line provides an in vitro model for elucidating the mechanism involving p53 inactivation and p21(cip1/waf1) overexpression in smokeless tobacco-induced oral cancer. Furthermore, the ATRA responsiveness of the cell line underscores its potential utility in identifying the retinoid responsive molecular targets in oral cancer cells.
Collapse
Affiliation(s)
- Jatinder Kaur
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | | |
Collapse
|
43
|
Liu JD, Wang YJ, Chen CH, Yu CF, Chen LC, Lin JK, Liang YC, Lin SY, Ho YS. Molecular mechanisms of G0/G1 cell-cycle arrest and apoptosis induced by terfenadine in human cancer cells. Mol Carcinog 2003; 37:39-50. [PMID: 12720299 DOI: 10.1002/mc.10118] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Terfenadine (TF), a highly potent histamine H1 receptor antagonist, has been shown to exert no significant central nervous system side effects in clinically effective doses. In this study, we demonstrated that TF induced significant growth inhibition of human cancer cells, including Hep G2, HT 29, and COLO 205 cells, through induction of G(0)/G(1) phase cell-cycle arrest. The minimal dose of TF induced significant G(0)/G(1) arrest in these cells was 1-3 microM. The protein levels of p53, p21/Cip1, and p27/Kip1 were significantly elevated, whereas the kinase activities of cyclin-dependent kinase 2 (CDK2) and CDK4 were inhibited simultaneously in the TF-treated cells. On the other hand, significant apoptosis, but not G(0)/G(1) arrest, was induced in the HL 60 (p53-null) or Hep 3B (with deleted p53) cells when treated with TF (3-5 microM). To clarify the roles of p21/Cip1 and p27/Kip1 protein expression, which was involved in G(0)/G(1) arrest and apoptosis induced by TF in human cancer cells, antisense oligodeoxynucleotides (ODNs) specific to p21/Cip1 and p27/Kip1 were used, and the expression of the p21/Cip1 and p27/Kip1 were monitored by immunoblotting analysis. Our data demonstrated that the percentage of the apoptotic cells detected by annexin V/PI analysis in the TF-treated group was clearly attenuated by pretreatment with p27/Kip1-specific ODNs. These results indicated that p27/Kip1 (but not p21/Cip1) protein indeed played a critical role in the TF-induced apoptosis. We also demonstrated that the TF-induced G(0)/G(1) cell-cycle arrest effect was not reversed by TF removal, and this growth inhibition lasted for at least 7 d. Importantly, the occurrence of apoptosis and cell growth arrest was not observed in the TF-treated normal human fibroblast, even at a dose as high as 25 microM. Our study showed the molecular mechanisms for TF-induced cell growth inhibition and the occurrence of apoptosis in human cancer cells.
Collapse
Affiliation(s)
- Jean-Dean Liu
- Department of Internal Medicine, School of Medicine, Taipei Medical University and Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Soria JC, Xu X, Liu DD, Lee JJ, Kurie J, Morice RC, Khuri F, Mao L, Hong WK, Lotan R. Retinoic acid receptor beta and telomerase catalytic subunit expression in bronchial epithelium of heavy smokers. J Natl Cancer Inst 2003; 95:165-8. [PMID: 12529350 DOI: 10.1093/jnci/95.2.165] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Expression of retinoic acid receptor beta (RAR beta) and telomerase catalytic subunit (human telomerase reverse transcriptase [hTERT]) was analyzed in 285 bronchial biopsy specimens from 53 heavy cigarette smokers and four former smokers by using in situ hybridization. Of the 191 biopsy specimens that were positive for RAR beta, 69% expressed hTERT, whereas only 54% of the 94 RAR beta negative biopsy specimens expressed hTERT (P =.014). Because hTERT expression in bronchial tissue has been previously associated with increased risk of lung cancer, the association between RAR beta and hTERT expression suggests that RAR beta expression may be an indicator of increased risk of lung cancer in heavy smokers.
Collapse
Affiliation(s)
- Jean-Charles Soria
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dietze EC, Caldwell LE, Marcom K, Collins SJ, Yee L, Swisshelm K, Hobbs KB, Bean GR, Seewaldt VL. Retinoids and retinoic acid receptors regulate growth arrest and apoptosis in human mammary epithelial cells and modulate expression of CBP/p300. Microsc Res Tech 2002; 59:23-40. [PMID: 12242694 DOI: 10.1002/jemt.10174] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Retinoids and retinoic acid receptors (RARs) are important mediators of normal epithelial cell homeostasis. To assess the role of retinoids and RARs in regulating growth arrest and apoptosis in benign and malignant mammary epithelial cells, two model systems were developed: 1) RAR function was suppressed in retinoid-sensitive normal human mammary epithelial cells (HMECs) by the dominant-negative retinoic acid receptor, RARalpha403 (DNRAR), and 2) retinoid-resistant MCF-7 breast cancer cells were transduced with a functional RARbeta2. Inhibition of RAR function by the DNRAR in HMECs resulted in retinoid-resistance, increased proliferation, and dysregulated growth when cells were cultured in reconstituted extracellular matrix (rECM). Expression of RARbeta2 in MCF-7 cells resulted in sensitivity to retinoid-induced growth arrest and apoptosis. The CREB-binding protein (CBP) and the homologous protein p300 are tightly regulated, rate-limiting integrators of diverse signaling pathways and are recruited during retinoid-mediated transcriptional activation. The relationship between retinoid receptor expression, growth regulation, and transcriptional regulation of CBP/p300 is poorly understood. Inhibition of RAR function in HMECs by DNRAR suppressed expression of CBP/p300 and expression of RARbeta2 in MCF-7 cells promoted induction of CBP/p300 when cells were treated with 1.0 microM all-trans-retinoic acid (ATRA). These results suggest that ATRA and RARs regulate growth arrest of HMECs and modulate CBP/p300 protein expression. Since CBP and p300 are normally present in limiting amounts, their regulation by ATRA and RARs may be an important element in the control of transcriptional activation of genes regulating growth arrest and apoptosis.
Collapse
Affiliation(s)
- Eric C Dietze
- Division of Medical Oncology and Transplantation, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Klaassen I, Braakhuis BJM. Anticancer activity and mechanism of action of retinoids in oral and pharyngeal cancer. Oral Oncol 2002; 38:532-42. [PMID: 12167430 DOI: 10.1016/s1368-8375(01)00118-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Retinoids are the natural and synthetic derivatives of vitamin A. Epidemiological studies indicate that a low intake of vitamin A is associated with an increased risk of squamous cancer. In vitro studies on cancer cells show that exposure to retinoids results in the inhibition of growth, by blocking the cell cycle or by inducing apoptosis. With respect to the clinical efficacy of retinoids some positive effects have been observed in early stage oral and oropharyngeal cancer. Administration of retinoids has been shown to elicit responses in leukoplakia, a premalignant lesion of the oral mucosa that frequently develops into invasive cancer. Furthermore, it has been possible with a retinoid, 13-cis-retinoic acid, to delay or inhibit the development of second primary tumors in patients who have been curatively treated for a first primary tumor in the oral cavity or oropharynx. Recent trials, however, failed to show protective effects on the development of second primary tumors. Because of the short duration of the response, the intrinsic resistance to retinoids and the toxic side effects, the treatment with this class of compounds has not become a standard therapy. Recent studies have shed light on how preneoplastic and neoplastic cells defend themselves against the growth inhibiting action of retinoids. An increased retinoid breakdown and an inactivation of nuclear retinoid receptor appear to be the cause of acquired or intrinsic resistance. This knowledge can be used to develop novel tumor-selective strategies. This review gives an update on the role of retinoids in oral and oropharyngeal cancer and their precursor lesions. The focus will be on the anticancer activity, the mechanism of action and future directions.
Collapse
Affiliation(s)
- Ingeborg Klaassen
- Section Tumor Biology, Department of Otolaryngology/Head and Neck Surgery, Vrije Universiteit Medical Center, PO Box 7057, The, Amsterdam, Netherlands
| | | |
Collapse
|
47
|
Wang YJ, Jeng JH, Chen RJ, Tseng H, Chen LC, Liang YC, Lin CH, Chen CH, Chu JS, Ho WL, Ho YS. Ketoconazole potentiates the antitumor effects of nocodazole: In vivo therapy for human tumor xenografts in nude mice. Mol Carcinog 2002; 34:199-210. [PMID: 12203371 DOI: 10.1002/mc.10066] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Our previous studies demonstrated that the oral antifungal agent ketoconazole (KT) induces apoptosis and G0/G1 phase cell cycle arrest in human cancer cell lines. In this study, we first demonstrated that KT (1 microM) potentiated the apoptotic effects of nocodazole (ND, 1 nM) in COLO 205 cancer cells. We further demonstrated the therapeutic efficacy of a combined treatment of KT (50 mg/kg/three times per week) and ND (5 mg/kg/three times per week) in vivo by treating athymic mice bearing COLO 205 tumor xenografts. The antitumor effects of ND were significantly potentiated by KT in mice after 6 wk of treatment. No gross signs of toxicity were observed in mice receiving these treatment regimens. The apoptotic cells were detected in a microscopic view of the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and by observation of DNA fragmentation in KT + ND-treated tumor tissues. The levels of cell cycle regulatory proteins were determined by Western blot analysis. Treatment with KT inhibits tumor growth through elevation of p53, p21/CIP1, and p27/KIP1 as well as inhibition of cyclin D3 and cyclin-dependent kinase 4 protein expression. Immunohistochemical staining analysis showed that p53, p21/CIP1, and p27/KIP1 immunoreactivity were induced in the tumor tissues. To clarify the roles of the p21/CIP1 and p27/KIP1 protein expression involved in G(0)/G(1) arrest and/or apoptosis induced by a combined treatment with KT and ND, antisense oligodeoxynucleotides (ODNs) specific to p21/CIP1 and p27/KIP1 were used. Our results demonstrated that apoptotic phenomena, including BAX induction and cytochrome C released from mitochondria induced by KT + ND, were significantly attenuated by pretreatment the cells with the p27/KIP1-specific antisense ODNs. These results indicate that p27/KIP1 protein does indeed play a critical role in the KT + ND-induced apoptosis. Our study revealed the molecular mechanism of KT + ND in regression of the tumor growth. The apoptotic effects of KT in a great variety of cancer cells make it a very attractive agent for cancer chemotherapy.
Collapse
Affiliation(s)
- Ying-Jan Wang
- Department of Environmental and Occupational Health, National Cheng Kung University Medical College, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Masuda M, Toh S, Koike K, Kuratomi Y, Suzui M, Deguchi A, Komiyama S, Weinstein IB. The roles of JNK1 and Stat3 in the response of head and neck cancer cell lines to combined treatment with all-trans-retinoic acid and 5-fluorouracil. Jpn J Cancer Res 2002; 93:329-39. [PMID: 11927016 PMCID: PMC5926966 DOI: 10.1111/j.1349-7006.2002.tb02176.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
We have used a combination of vitamin A (all-trans-retinyl palmitate), 5-fluorouracil (5-FU) and radiation to treat human head and neck squamous cell carcinoma (HNSCC). This chemoradiotherapy is called "FAR therapy." In this study we examined the effects of all-trans-retinoic acid (ATRA), the active metabolite of vitamin A, and ATRA plus 5-FU on two HNSCC cell lines (YCU-N861 and YCU-H891) to gain insight into the molecular mechanisms of FAR therapy. ATRA at 1 mM (the order of concentration found in HNSCC tumors treated with FAR therapy) inhibited cell proliferation and caused G1 cell cycle arrest in both cell lines. This was associated with a decrease in cyclin D1, an increase in p27(Kip1) and a reduction in the hyperphosphorylated form of retinoblastoma protein (pRB). With YCU-N861 cells, ATRA also caused a decrease in Bcl-2 and Bcl-X(L) and an increase in Bax. Both ATRA and 5-FU activated c-Jun N-terminal kinase (JNK) 1 and the combination of both agents resulted in additive or synergistic activation of JNK1, and also enhanced the induction of apoptosis. The YCU-H891 cells, in which the epidermal growth factor receptor (EGFR)-signal transducer and activator of transcription 3 (Stat3) pathway is constitutively activated, were more resistant to treatments with ATRA, 5-FU and the combination of both agents than YCU-N861 cells. A dominant negative Stat3 construct strongly enhanced the cellular sensitivity of this cell line to 5-FU but not to ATRA. In addition there is evidence that activation of Stat3 is associated with cellular resistance to radiation in HNSCC. Therefore, the addition to FAR therapy of agents that inhibit activation of the Stat3 pathway may enhance the clinical response of patients with HNSCC to FAR therapy.
Collapse
Affiliation(s)
- Muneyuki Masuda
- Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, Kyushu University, Fukuoka 812-0052, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zou CP, Youssef EM, Zou CC, Carey TE, Lotan R. Differential effects of chromosome 3p deletion on the expression of the putative tumor suppressor RAR beta and on retinoid resistance in human squamous carcinoma cells. Oncogene 2001; 20:6820-7. [PMID: 11687961 DOI: 10.1038/sj.onc.1204846] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2001] [Revised: 07/09/2001] [Accepted: 07/17/2001] [Indexed: 11/09/2022]
Abstract
Retinoids' effects on cell growth and differentiation are mediated by nuclear retinoid receptors, which are ligand-activated transcription enhancing factors. Because the expression of the retinoic acid receptor beta (RARbeta) gene, which is located on chromosome 3p24, is diminished in premalignant and malignant tissues it has been proposed that it acts as a tumor suppressor. To test the hypothesis that RARbeta loss leads to retinoid resistance, we studied several karyotyped head and neck squamous carcinoma (HNSCC) cell lines (UMSCC-17A, -17B, -22A, -22B, and -38) with deletion of one chromosome 3p arm. RARbeta mRNA was neither detected nor induced by retinoic acid in these cells, whereas it was expressed and induced by retinoic acid in two other HNSCC cell lines (1483 and 183) without 3p deletion. Methylation of the RARbeta gene promoter was detected in the 17B and 22B cells that failed to express RARbeta but no methylation was found in 183A cells that did express RARbeta mRNA. Responsiveness of HNSCC cells to several retinoids in assays of growth inhibition and colony formation, was rank ordered as: 22B>1483>38>183>17B. Additionally, retinoid response elements were transactivated in 22B more efficiently than in 17B cells. These results indicate that loss of RARbeta expression does not necessarily lead to loss of growth inhibition by retinoids or to a block of retinoid signaling.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Division
- Chromosome Deletion
- Chromosomes, Human, Pair 3
- DNA, Neoplasm/genetics
- Drug Resistance, Neoplasm
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/metabolism
- Head and Neck Neoplasms/pathology
- Humans
- Promoter Regions, Genetic
- RNA, Neoplasm/biosynthesis
- Receptors, Retinoic Acid/biosynthesis
- Receptors, Retinoic Acid/genetics
- Retinoids/pharmacology
- Transcriptional Activation
- Tretinoin/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C P Zou
- Gynecologic Oncology, Department of Obstetrics, Gynecology & Reproductive Science, The University of Texas, Medical School, Houston, Texas, TX 77030, USA
| | | | | | | | | |
Collapse
|
50
|
Hayashi K, Yokozaki H, Goodison S, Oue N, Suzuki T, Lotan R, Yasui W, Tahara E. Inactivation of retinoic acid receptor beta by promoter CpG hypermethylation in gastric cancer. Differentiation 2001; 68:13-21. [PMID: 11683489 DOI: 10.1046/j.1432-0436.2001.068001013.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Inactivation of nuclear retinoic acid receptor beta (RARbeta) expression is implicated in tumorigenesis. We hypothesized that loss of RARbeta in gastric cancer cells may occur as a result of multiple factors, including epigenetic modifications which alter RARbeta promoter chromatin structure. We examined hypermethylation of CpG islands present in the RARbeta promoter by methylation-specific PCR and the expression of RARbeta in gastric cancer cell lines and tissues. Three (MKN-28, -45 and -74) out of eight gastric cancer cell lines had a loss of RAR expression associated with promoter methylation. RARbeta expression was retrieved in these cell lines by treatment with 5-azacytidine or by the histone deacetylase inhibitor trichostatin A. Promoter hypermethylation was detected in 64% (7/11) of gastric carcinoma tissues with reduced expression of RARbeta, whereas it was detected in 22% (2/9) of tumors with retained RARbeta expression. To investigate the functions of exogenous RARbeta in gastric cancer cells, we transfected a retroviral RARbeta expression vector (LNSbeta) into MKN-28 cells that have hypermethylation of the RARbeta promoter. Overexpression of RAR in MKN-28 cells appeared to regulate the expression of DNA methyltransferase and DNA demethylase and the acetylation of hitone H4. These results suggest that the transcriptional inactivation of the RARbeta by promoter CpG hypermethylation is frequently associated with gastric carcinoma. Our data also suggests that DNA methylation plays a pivotal role in establishing and maintaining an inactive state of RARbeta by rendering the chromatin structure inaccessible to the transcription machinery.
Collapse
Affiliation(s)
- K Hayashi
- First Department of Pathology, Hiroshima University School of Medicine, Japan.
| | | | | | | | | | | | | | | |
Collapse
|