1
|
Chen X, Wang Y, Liu Z, He Y, Gao H, Guan X, Chen L, Chen S, Xing X, Chen W, Li D, Wang Q. Role of the USP7/FOXO3A axis in environmentally relevant doses of arsenic-induced lung carcinogenesis: Insights from bioinformatics analysis and model of human epithelial cell malignant transformation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118224. [PMID: 40273609 DOI: 10.1016/j.ecoenv.2025.118224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/05/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025]
Abstract
Arsenic (As) is classified as a Group 1 carcinogen by the International Agency for Research on Cancer (IARC). Exposure to As has been associated with an increased risk of various cancers, particularly lung cancer. However, the precise molecular mechanisms contributing to this carcinogenesis are not well understood. In our study, we analyzed transcriptomic data from the GEO database (GSE36684), identifying 764 differentially expressed genes (DEGs) in BEAS-2B cells treated with environmentally relevant doses of As for 8 weeks. A KEGG pathway enrichment analysis suggested that the FoxO pathway activation might be a novel key signaling event in As-induced carcinogenesis. We further analyzed the expression of 11 DEGs involved in the FoxO pathway using the TCGA-LUSC dataset. The findings revealed that four genes displayed expression patterns in tumor tissues consistent with those observed after As treatment in GEO dataset. Among them, USP7 was upregulated, while ATM, S1PR1, and PLK2 were downregulated in cancer tissues. High USP7 expression was specifically linked to a poor prognosis in lung squamous cell carcinoma (LUSC). To explore the role of USP7 in As-induced malignant transformation, BEAS-2B cells were exposed to NaAsO2 concentrations of 0.2 μM and 2 μM for up to 20 weeks. Experimental results confirmed that NaAsO2 treatment suppressed the FoxO transcriptional activity by upregulating USP7 expression, subsequently downregulating ATM and PLK2 expression, which led to abnormalities in cell cycle regulation and apoptosis. Notably, knocking down USP7 in As-transformed cells resulted in significant reductions in cell proliferation, colony formation, and tumor formation ability in nude mice, indicating the USP7-regulated FOXO3A pathway could be central to As-induced lung carcinogenesis. Moreover, our research demonstrated that USP7 inhibited FOXO3A's ability to translocate from the cytoplasm to the nucleus by affecting its monoubiquitination status. Additionally, we speculated that As-induced the elevation of USP7 expression due to the excessive inflammatory cytokines secretion and the activation of mTORC1/WTAP pathway. These findings offer novel insights into the molecular mechanisms underlying As-mediated lung cancer.
Collapse
Affiliation(s)
- Xiaoyu Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuqing Wang
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ziqi Liu
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Yujie He
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Huan Gao
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xinchao Guan
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shen Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiumei Xing
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Qing Wang
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
2
|
Zhang Y, Luo X, Zhu M, Wu Y. Evaluation the role of insulin signaling pathway in reproductive toxicity of dispersed diesel particulate extract under environmental conditions. Comp Biochem Physiol C Toxicol Pharmacol 2024; 283:109959. [PMID: 38866378 DOI: 10.1016/j.cbpc.2024.109959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/27/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
Diesel particulate extract (DPE), which is a significant constituent of airborne particle pollution, has a strong association with the development of cancer and respiratory diseases. Fulvic acid (FA), a plentiful organic macromolecule found in water, has the capability to modify particle surface charge and adsorption capacity when combined with minerals. Nevertheless, there is a scarcity of data regarding the influence of their interaction on DPE toxicity. To examine the impact of environmental factor on the toxic effects of DPE, we used the Caenorhabditis elegans (C. elegans) model to investigate the reproductive toxicity of DPE and FA on insulin signaling pathway. C. elegans were subjected to a semi-fluid medium (NGG) containing different concentrations of DPE or DPE + FA in order to assess germline apoptosis and the expression of important genes in the insulin signaling pathway. Through several mutant strains, we found that daf-2, age-1, pdk-1, akt-1 and daf-16 were involved in DPE-induced apoptosis. Furthermore, and the expression levels of these genes significantly altered. The ratio of daf-16 translocation to nucleation, as well as the amount of reactive oxygen species (ROS), exhibited a dose-response relationship, however, the presence of FA could altered these effects. The results revealed that the insulin signaling pathway plays a vital role in mediating the harmful effects caused by DPE, whereas environmental factors have a substantial impact on its toxicity. Moreover, it was noted that semi-fluid medium could effectively replicate three-dimensional exposure circumstances closely resembling those observed in actual situations.
Collapse
Affiliation(s)
- Yajun Zhang
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Health and Safety, Ministry of Education, Anhui University of Science & Technology, Huainan 232001, China; School of Public Health, Anhui University of Science & Technology, Hefei 231131, China.
| | - Xun Luo
- School of Biological Engineering, Huainan Normal University, Huainan 232038, China.
| | - Mengyun Zhu
- School of Biological Engineering, Huainan Normal University, Huainan 232038, China
| | - Yu Wu
- School of Biological Engineering, Huainan Normal University, Huainan 232038, China
| |
Collapse
|
3
|
Li M, Muhammad JS, Zhao QL, Zakki SA, Hiraku Y, Hatta H, Tong X, Cui ZG, Wu C. Protective effects of baicalin against phenylarsine oxide-induced cytotoxicity in human skin keratinocytes. Bioorg Chem 2024; 150:107535. [PMID: 38865859 DOI: 10.1016/j.bioorg.2024.107535] [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: 09/26/2023] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
Phenylarsine oxide (PAO) is a known environmental pollutant and skin keratinocytes are most seriously affected. Baicalin (BCN) was reported to have antioxidant and anti-inflammatory effects, but its protective effect against PAO toxicity is unknown. This study aimed at exploring whether baicalin can reverse the toxicity of human epidermal keratinocytes that are subjected to PAO exposure and underlying mechanisms. In silico analysis from a publicly accessible HaCaT cell transcriptome dataset exposed to chronic Arsenic showed significant differential expression of several genes, including the genes related to DNA replication. Later, we performed in vitro experiments, in which HaCaT cells were exposed to PAO (500 nM) in the existence of BCN (10-50 µM). Treatment of PAO alone induces the JNK, p38 and caspase-3 activation, which were engaged in the apoptosis induction, while the activity of AKT was significantly inhibited, which was engaged in the suppression of apoptosis. PAO suppressed SIRT3 expression and induced intracellular reactive oxygen species (ROS), causing a marked reduce in cell viability and apoptosis. However, BCN treatment restored the PAO-induced suppression of SIRT3 and AKT expression, reduced intracellular ROS generation, and markedly suppressed both caspase-3 activation and apoptosis induction. However, the protective effect of BCN was significantly attenuated after pretreatment with nicotinamide, an inhibitor of SIRT3. These findings indicate that BCN protects against cell death induced by PAO via inhibiting excessive intracellular ROS generation via restoring SIRT3 activity and reactivating downstream AKT pathway. In this study, we firstly shown that BCN is an efficient drug to prevent PAO-induced skin cytotoxicity, and these findings need to be confirmed by in vivo and clinical investigations.
Collapse
Affiliation(s)
- Mengling Li
- School of Medicine, Xizang Minzu University, No. 6 Wenhui East Road, Weicheng District, Xianyang, Shaanxi, 712082, China; Department of Environmental Health, University of Fukui School of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Jibran Sualeh Muhammad
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Qing-Li Zhao
- Department of Radiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 910-0194, Japan
| | - Shahbaz Ahmad Zakki
- Department of Public Health & Nutrition, The University of Haripur, Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Yusuke Hiraku
- Department of Environmental Health, University of Fukui School of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Hideki Hatta
- Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Xiaopeng Tong
- School of Medicine, Xizang Minzu University, No. 6 Wenhui East Road, Weicheng District, Xianyang, Shaanxi, 712082, China
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Sciences, University of Fukui, Fukui 910-1193, Japan.
| | - Chengai Wu
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| |
Collapse
|
4
|
Obata T, Tsutsumi K, Ueta E, Oda T, Kikuchi T, Ako S, Fujii Y, Yamazaki T, Uchida D, Matsumoto K, Horiguchi S, Kato H, Okada H, Otsuka M. MicroRNA-451a inhibits gemcitabine-refractory biliary tract cancer progression by suppressing the MIF-mediated PI3K/AKT pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102054. [PMID: 38111913 PMCID: PMC10726424 DOI: 10.1016/j.omtn.2023.102054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/11/2023] [Indexed: 12/20/2023]
Abstract
Gemcitabine is an effective chemotherapeutic agent for biliary tract cancers (BTCs), including gallbladder cancer (GBC) and cholangiocarcinoma (CCA). However, few other effective agents are currently available, particularly for GEM-refractory BTCs. We previously identified microRNA-451a (miR-451a) as a potential therapeutic target in GBC. To elucidate the antineoplastic effects of miR-451a and its underlying mechanisms, we transfected miR-451a into GBC, gemcitabine-resistant GBC (GR-GBC), and gemcitabine-resistant CCA (GR-CCA) cell lines. Furthermore, mimicking in vivo conditions, tumorigenic GBC organoids and three-dimensional (3D) cell culture systems were employed to investigate the anti-proliferative effects of miR-451a on BTCs, and its effect on stem cell properties. We found that miR-451a significantly inhibited cell proliferation, induced apoptosis, and reduced chemoresistant phenotypes, such as epithelial-mesenchymal transition, in both GBC and GR-GBC. The principal mechanism is probably the negative regulation of the phosphatidylinositol 3-kinase/AKT pathway, partially accomplished by directly downregulating macrophage migration inhibitory factor. The Gene Expression Omnibus database revealed that miR-451a was the most significantly downregulated microRNA in CCA tissues. The introduction of miR-451a resulted in similar antineoplastic effects in GR-CCA. Furthermore, miR-451a reduced cell viability in 3D spheroid models and tumorigenic GBC organoids. These findings suggest that the supplementation of miR-451a is a potential treatment strategy for GEM-refractory BTCs.
Collapse
Affiliation(s)
- Taisuke Obata
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama 700-8558, Japan
| | - Koichiro Tsutsumi
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Eijiro Ueta
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Takashi Oda
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama 700-8558, Japan
| | - Tatsuya Kikuchi
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama 700-8558, Japan
| | - Soichiro Ako
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Yuki Fujii
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Tatsuhiro Yamazaki
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Daisuke Uchida
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Kazuyuki Matsumoto
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Shigeru Horiguchi
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Hironari Kato
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Hiroyuki Okada
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama 700-8558, Japan
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Okayama 700-8558, Japan
- Department of Gastroenterology and Hepatology, Okayama University Hospital, Okayama 700-8558, Japan
| |
Collapse
|
5
|
Gao M, Xiao H, Liang Y, Cai H, Guo X, Lin J, Zhuang S, Xu J, Ye S. The Hyperproliferation Mechanism of Cholesteatoma Based on Proteomics: SNCA Promotes Autophagy-Mediated Cell Proliferation Through the PI3K/AKT/CyclinD1 Signaling Pathway. Mol Cell Proteomics 2023; 22:100628. [PMID: 37532176 PMCID: PMC10495652 DOI: 10.1016/j.mcpro.2023.100628] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/19/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023] Open
Abstract
Cholesteatoma is a chronic inflammatory ear disease with abnormal keratinized epithelium proliferation and tissue damage. However, the mechanism of keratinized epithelium hyperproliferation in cholesteatoma remains unknown. Hence, our study sought to shed light on mechanisms affecting the pathology and development of cholesteatoma, which could help develop adjunctive treatments. To investigate molecular changes in cholesteatoma pathogenesis, we analyzed clinical cholesteatoma specimens and paired ear canal skin with mass spectrometry-based proteomics and bioinformatics. From our screen, alpha-synuclein (SNCA) was overexpressed in middle ear cholesteatoma and might be a key hub protein associated with inflammation, proliferation, and autophagy in cholesteatoma. SNCA was more sensitive to lipopolysaccharide-induced inflammation, and autophagy marker increase was accompanied by autophagy activation in middle ear cholesteatoma tissues. Overexpression of SNCA activated autophagy and promoted cell proliferation and migration, especially under lipopolysaccharide inflammatory stimulation. Moreover, inhibiting autophagy impaired SNCA-mediated keratinocyte proliferation and corresponded with inhibition of the PI3K/AKT/CyclinD1 pathways. Also, 740Y-P, a PI3K activator reversed the suppression of autophagy and PI3K signaling by siATG5 in SNCA-overexpressing cells, which restored proliferative activity. Besides, knockdown of SNCA in RHEK-1 and HaCaT cells or knockdown of PI3K in RHEK-1 and HaCaT cells overexpressing SNCA both resulted in attenuated cell proliferation. Our studies indicated that SNCA overexpression in cholesteatoma might maintain the proliferative ability of cholesteatoma keratinocytes by promoting autophagy under inflammatory conditions. This suggests that dual inhibition of SNCA and autophagy may be a promising new target for treating cholesteatoma.
Collapse
Affiliation(s)
- Miao Gao
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Heng Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yonglan Liang
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huimin Cai
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaojing Guo
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianwei Lin
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Suling Zhuang
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jianhua Xu
- Department of Pharmacology, School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, China.
| | - Shengnan Ye
- Department of Otorhinolaryngology Head and Neck Surgery, Fujian Institute of Otorhinolaryngology, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Otorhinolaryngology Head and Neck Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
6
|
Wang B, Zheng Z, Chen L, Zhang W, He Y, Wu B, Ji R. Transcriptomics reveals key regulatory pathways and genes associated with skin diseases induced by face paint usage. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 890:164374. [PMID: 37236445 DOI: 10.1016/j.scitotenv.2023.164374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/13/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023]
Abstract
The use of face paint cosmetics can cause skin diseases in opera performers due to the presence of heavy metals and other toxic ingredients in the cosmetics. However, the underlying molecular mechanism for these diseases remains unknown. Here we examined the transcriptome gene profile of human skin keratinocytes exposed to artificial sweat extracts of face paints, and identified the key regulatory pathways and genes, using RNA sequencing technique. Bioinformatics analyses suggested that the face paint exposure induced the differentially expression of 1531 genes and enriched inflammation-relevant TNF and IL-17 signaling pathways after just 4 h of exposure. Inflammation-relevant genes CREB3L3, FOS, FOSB, JUN, TNF, and NFKBIA were identified as the potential regulatory genes, and SOCS3 capable to prevent inflammation-induced carcinogenesis as the hub-bottleneck gene. Long-term exposure (24 h) could exacerbate inflammation, accompanied by interference in cellular metabolism pathways, and the potential regulatory genes (ATP1A1, ATP1B1, ATP1B2, FXYD2, IL6, and TNF) and hub-bottleneck genes (JUNB and TNFAIP3) were all related to inflammation induction and other adverse responses. We proposed that the exposure to face paint might cause the inflammatory factors TNF and IL-17, which are encoded by the genes TNF and IL17, to bind to receptors and activate TNF and IL-17 signaling pathways, leading to the expression of cell proliferation factors (CREB and AP-1) and proinflammatory mediators including transcription factors (FOS, JUN, and JUNB), inflammatory factors (TNF-α and IL6), and intracellular signaling factors (TNFAIP3). This finally resulted in cell inflammation, apoptosis, and other skin diseases. TNF was identified as the key regulator and connector in all the enriched signaling pathways. Our study provides the first insights into the cytotoxicity mechanism of face paints to skin cells and highlights the need for stricter regulations in face paint safety.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Zhaohao Zheng
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China; Quanzhou Institute for Environment Protection Industry, Nanjing University, Quanzhou 362000, China
| | - Ling Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Wenhui Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Yujie He
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China; Quanzhou Institute for Environment Protection Industry, Nanjing University, Quanzhou 362000, China.
| | - Bing Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Rong Ji
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China; Quanzhou Institute for Environment Protection Industry, Nanjing University, Quanzhou 362000, China
| |
Collapse
|
7
|
Zhao T, Sun D, Xiong W, Man J, Zhang Q, Zhao M, Zhang Z. N 6-methyladenosine plays a dual role in arsenic carcinogenesis by temporal-specific control of core target AKT1. JOURNAL OF HAZARDOUS MATERIALS 2023; 445:130468. [PMID: 36444808 DOI: 10.1016/j.jhazmat.2022.130468] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/11/2022] [Accepted: 11/23/2022] [Indexed: 06/16/2023]
Abstract
High-profile RNA epigenetic modification N6-methyladenosine (m6A), as a double-edged sword for cancer, can either promote or inhibit arsenic-induced skin carcinogenesis. However, the core m6A-target gene determining the duality of m6A and the regulatory mechanism of m6A on the core gene are still poorly understood. Based on m6A microarray detection, integrated multi-omics analysis, and further experiments in vitro and in vivo, we explored the molecular basis for the dual role of m6A in cancer induced by environmental pollutants using models in different stages of arsenic carcinogenesis, including As-treated, As-transformed, and As-tumorigenic cell models. We found that the key proliferative signaling node AKT1 is in the center of the m6A-regulatory network in arsenic carcinogenicity. The m6A level on AKT1 mRNA (3'UTR, CDS, and 5'UTR) dynamically changed in different stages of arsenic carcinogenesis. The m6A writer METTL3-catalyzed upregulation of m6A promotes AKT1 expression by elevating m6A reader YTHDF1-mediated AKT1 mRNA stability in As-treated and As-transformed cells, while the m6A eraser FTO-catalyzed downregulation of m6A promotes AKT1 expression mainly by inhibiting m6A reader YTHDF2-mediated AKT1 mRNA degradation in As-tumorigenic cells. Furthermore, upregulation of m6A inhibits the expression of AKT1 negative regulator PHLPP2 and promotes the expression of AKT1 positive regulator PDK1. These changes in AKT1 regulators result in AKT1 activation by upregulating AKT1 phosphorylation at S473 and T308. Interestingly, the FTO-catalyzed decrease in m6A prevents AKT upregulation in As-treated cells but promotes AKT upregulation in As-tumorigenic cells. Both inhibitors targeting the m6A writer and eraser can inhibit the AKT1-mediated proliferation of As-tumorigenic cells by breaking the balance of m6A regulators. Our results demonstrated that AKT1 is the core hub determining m6A as a double-edged sword. Changed m6A dynamically upregulates the expression and activity of AKT1 in different stages of arsenic carcinogenesis. This study can advance our understanding of the dual role and precise time-specific mechanism of RNA epigenetics involved in the carcinogenesis of hazardous materials.
Collapse
Affiliation(s)
- Tianhe Zhao
- Department of Environmental and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Donglei Sun
- Department of Environmental and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenxiao Xiong
- Department of Environmental and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Man
- Department of Environmental and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Zhang
- Department of Environmental and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Manyu Zhao
- Department of Environmental and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zunzhen Zhang
- Department of Environmental and Occupational Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Lou Q, Chen F, Li B, Zhang M, Yin F, Liu X, Zhang Z, Zhang X, Fan C, Gao Y, Yang Y. Malignant growth of arsenic-transformed cells depends on activated Akt induced by reactive oxygen species. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2023; 33:284-298. [PMID: 34974760 DOI: 10.1080/09603123.2021.2023113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Arsenic is an identified carcinogen for humans.In this study, chronic exposure of human hepatocyte L-02 to low-doses of inorganic arsenic caused cell malignant proliferation. Meanwhile, compared with normal L-02 cells, arsenic-transformed malignant cells, L-02-As displayed more ROS and significantly higher Cyclin D1 expression as well as aerobic glycolysis. Moreover, Akt activation is followed by the upregulation of Cyclin D1 and HK2 expression in L-02-As cells, since inhibition of Akt activity by Ly294002 attenuated the colony formation in soft agar and decreased the levels of Cyclin D1 and HK2. In addition, scavenging of ROS by NAC resulted in a decreased expression of phospho-Akt, HK2 and Cyclin D1, and attenuates the ability of anchorage-independent growth ofL-02-As cells, suggested that ROS mediated the Akt activation in L-02-As cells. In summary, our results demonstrated that ROS contributes to the malignant phenotype of arsenic-transformed human hepatocyte L-02-As via the activation of Akt pathway.
Collapse
Affiliation(s)
- Qun Lou
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fuxun Chen
- Yantai Center for Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yantai, Shandong, China
| | - Bingyang Li
- Yantai Center for Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Yantai, Shandong, China
| | - Meichen Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fanshuo Yin
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xiaona Liu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Zaihong Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xin Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chenlu Fan
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
9
|
Luo X, Zhang Y, Lu C, Zhang J. Role of insulin signaling pathway in apoptosis induced by food chain delivery of nano-silver under the action of environmental factors. Comp Biochem Physiol C Toxicol Pharmacol 2022; 261:109429. [PMID: 35944823 DOI: 10.1016/j.cbpc.2022.109429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To investigate how the environmental factor affects the delivery of nano silver through food chain, we set up a two-stage food delivery chain model of Escherichia coli and Caenorhabditis elegans system. METHODS Through a two-stage food delivery chain model of E. coli and C. elegans, the mRNA expression levels of DAF-2, age-1, PDK-1, Akt-1 and DAF-16 in the insulin growth factor 1 signaling pathway in nematode gonad cells which occurs AgNPs induced apoptosis were evaluated and the apoptosis of gonad cells in the mutant strains of the above key genes were detected. RESULTS DAF-2, age-1, PDK-1 and Akt-1 could significantly negatively regulate the apoptosis of nematode cells induced by AgNPs, while DAF-16 could significantly promote the apoptosis induced by AgNPs. The DAF-16 up-regulated expression was a protective effect on the body and the phenomenon of DNA double-strand breaks was significantly increased. The damage effect induced by AgNPs was significantly enhanced in the presence of the environmental factor fulvic acid. CONCLUSION The damage effect induced by AgNPs after food delivery involves the regulation of the insulin growth factor 1 signaling pathway and environmental factors have a significant impact on the biological effects.
Collapse
Affiliation(s)
- Xun Luo
- School of Biological Engineering, Huainan Normal University, China.
| | - Yajun Zhang
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Health and Safety, Ministry of Education, China; Medicine School, Anhui University of Science & Technology, China.
| | - Changjie Lu
- School of Biological Engineering, Huainan Normal University, China
| | - Jiaming Zhang
- School of Biological Engineering, Huainan Normal University, China
| |
Collapse
|
10
|
Li X, Zuo C, Wu M, Zhang Z. Linc-ROR promotes arsenite-transformed keratinocyte proliferation by inhibiting P53 activity. Metallomics 2021; 12:963-973. [PMID: 32373892 DOI: 10.1039/d0mt00076k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Linc-ROR is an oncogenic long non-coding RNA over-expressed in many kinds of cancer that promotes cancer cell proliferation. Arsenite is a determined carcinogen that increases the risk of skin cancer, but the carcinogenic mechanism of arsenite remains unclear. To explore whether and how linc-ROR plays a role in arsenite-induced carcinogenesis of skin cancer, we established arsenite-transformed keratinocyte HaCaT cells by exposing them to 1 μM arsenite for 50 passages. Then we examined the linc-ROR expression during the transformation and explored the effect of linc-ROR on the cell proliferation of arsenite-transformed HaCaT cells. We found that the linc-ROR level in HaCaT cells was gradually increased during arsenite-induced malignant transformation, and the activity of P53 was decreased, but the P53 expression was not significantly altered, indicating that linc-ROR may play a role in arsenite-induced HaCaT cell transformation that is associated with P53 activity but not P53 expression. We further demonstrated that linc-ROR down-regulation by siRNA significantly inhibited the cellular proliferation and restored P53 activity in arsenite-transformed HaCaT cells, suggesting that linc-ROR promotes proliferation of arsenite-transformed HaCaT cells by inhibiting P53 activity. Moreover, linc-ROR siRNA also down-regulated the PI3K/AKT pathway in arsenite-transformed HaCaT cells, and treatment with AKT inhibitor wortmannin restored P53 activity, implying that linc-ROR inhibits P53 activity by activating the PI3K/AKT pathway. Taken together, the present study shows that linc-ROR promotes arsenite-transformed keratinocyte proliferation by inhibiting P53 activity through activating PI3K/AKT, providing a novel carcinogenic mechanism of arsenite-induced skin cancer.
Collapse
Affiliation(s)
- Xinyang Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, Chengdu 610041, People's Republic of China.
| | - Chao Zuo
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, Chengdu 610041, People's Republic of China.
| | - Mei Wu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, Chengdu 610041, People's Republic of China.
| | - Zunzhen Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin Nanlu, Chengdu 610041, People's Republic of China.
| |
Collapse
|
11
|
Kim C, Ceresa BP. Using In Vitro Models to Dissect the Molecular Effects of Arsenic Exposure in Skin and Lung Cell Lines. APPLIED IN VITRO TOXICOLOGY 2021; 7:71-88. [DOI: 10.1089/aivt.2020.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Affiliation(s)
- Christine Kim
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Brian P. Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
12
|
Kabir R, Sinha P, Mishra S, Ebenebe OV, Taube N, Oeing CU, Keceli G, Chen R, Paolocci N, Rule A, Kohr MJ. Inorganic arsenic induces sex-dependent pathological hypertrophy in the heart. Am J Physiol Heart Circ Physiol 2021; 320:H1321-H1336. [PMID: 33481702 PMCID: PMC8260381 DOI: 10.1152/ajpheart.00435.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/15/2021] [Accepted: 01/15/2021] [Indexed: 01/17/2023]
Abstract
Arsenic exposure though drinking water is widespread and well associated with adverse cardiovascular outcomes, yet the pathophysiological mechanisms by which iAS induces these effects are largely unknown. Recently, an epidemiological study in an American population with a low burden of cardiovascular risk factors found that iAS exposure was associated with altered left ventricular geometry. Considering the possibility that iAS directly induces cardiac remodeling independently of hypertension, we investigated the impact of an environmentally relevant iAS exposure on the structure and function of male and female hearts. Adult male and female C56BL/6J mice were exposed to 615 μg/L iAS for 8 wk. Males exhibited increased systolic blood pressure via tail cuff photoplethysmography, left ventricular wall thickening via transthoracic echocardiography, and increased plasma atrial natriuretic peptide via enzyme immunoassay. RT-qPCR revealed increased myocardial RNA transcripts of Acta1, Myh7, and Nppa and decreased Myh6, providing evidence of pathological hypertrophy in the male heart. Similar changes were not detected in females, and nitric oxide-dependent mechanisms of cardioprotection in the heart appeared to remain intact. Further investigation found that Rcan1 was upregulated in male hearts and that iAS activated NFAT in HEK-293 cells via luciferase assay. Interestingly, iAS induced similar hypertrophic gene expression changes in neonatal rat ventricular myocytes, which were blocked by calcineurin inhibition, suggesting that iAS may induce pathological cardiac hypertrophy in part by targeting the calcineurin-NFAT pathway. As such, these results highlight iAS exposure as an independent cardiovascular risk factor and provide biological impetus for its removal from human consumption.NEW & NOTEWORTHY This investigation provides the first mechanistic link between an environmentally relevant dose of inorganic arsenic (iAS) and pathological hypertrophy in the heart. By demonstrating that iAS exposure may cause pathological cardiac hypertrophy not only by increasing systolic blood pressure but also by potentially activating calcineurin-nuclear factor of activated T cells and inducing fetal gene expression, these results provide novel mechanistic insight into the theat of iAS exposure to the heart, which is necessary to identify targets for medical and public health intervention.
Collapse
MESH Headings
- Animals
- Arsenites/toxicity
- Calcineurin/metabolism
- Female
- Gene Expression Regulation
- HEK293 Cells
- Humans
- Hypertrophy, Left Ventricular/chemically induced
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Isolated Heart Preparation
- Male
- Mice, Inbred C57BL
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- NFATC Transcription Factors/metabolism
- Sex Factors
- Signal Transduction
- Sodium Compounds/toxicity
- Time Factors
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
- Water Pollutants, Chemical/toxicity
- Mice
Collapse
Affiliation(s)
- Raihan Kabir
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Prithvi Sinha
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Sumita Mishra
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Obialunanma V Ebenebe
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Nicole Taube
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Chistian U Oeing
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rui Chen
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Ana Rule
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Mark J Kohr
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
13
|
Chen Y, Liu X, Wang H, Liu S, Hu N, Li X. Akt Regulated Phosphorylation of GSK-3β/Cyclin D1, p21 and p27 Contributes to Cell Proliferation Through Cell Cycle Progression From G1 to S/G2M Phase in Low-Dose Arsenite Exposed HaCat Cells. Front Pharmacol 2019; 10:1176. [PMID: 31680960 PMCID: PMC6798184 DOI: 10.3389/fphar.2019.01176] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022] Open
Abstract
Arsenic is a toxic environmental contaminant. Long-term exposure to arsenic through drinking water induces human cancers. However, it is as yet uncertain about the mechanisms of arsenic induced carcinogenesis. Although the effects of low-dose arsenicals on proliferation and cell cycle have been revealed by short time exposure, the evidences for long-term exposure were seldom reported. The detailed mechanism has been unclear and supplemented constantly. In the present study, we used normal human keratinocytes (HaCat) to study the effects of long-term, low-dose sodium arsenite (NaAsO2) exposure on cell proliferation with emphasis on the Akt regulated cell cycle signaling pathways. Treatment of NaAsO2 resulted in increased cell proliferation and promotion of cell cycle progression from G1 to S/G2M phase, both of which could be attenuated by MK2206, a highly selective inhibitor of Akt. Along with the increased expression of phospho-Akt (p-Akt, Ser 473), increased expression of p-GSK-3β (Ser 9), p-p21 (Thr 145), p-p27 (Thr 157) and total cyclin D1, and decreased expression of p-cyclin D1 (Thr 286), p21 and p27 were also found in the NaAsO2 exposed cells. Treatment of MK2206 markedly reversed the expression of all of the above proteins. Our findings indicated that the phosphorylated activation of Akt played a role in the proliferation of HaCat cells upon long-term, low-dose NaAsO2 exposure through the phosphorylative regulation of its downstream cell cycle regulating factors of GSK-3β/cyclin D1, p21 and p27, which could induce the promotion of cell cycle progression from G1 to S/G2M phase.
Collapse
Affiliation(s)
- Yao Chen
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, Shenyang, China
| | - Xudan Liu
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, Shenyang, China
| | - Huanhuan Wang
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, Shenyang, China
| | - Shiyi Liu
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, Shenyang, China
| | - Nannan Hu
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, Shenyang, China
| | - Xin Li
- Department of Occupational and Environmental Health, Key Laboratory of Arsenic-related Biological Effects and Prevention and Treatment in Liaoning Province, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
14
|
Zheng L, Dong H, Zhao W, Zhang X, Duan X, Zhang H, Liu S, Sui G. An Air-Liquid Interface Organ-Level Lung Microfluidics Platform for Analysis on Molecular Mechanisms of Cytotoxicity Induced by Cancer-Causing Fine Particles. ACS Sens 2019; 4:907-917. [PMID: 30843693 DOI: 10.1021/acssensors.8b01672] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fine particulate matter less than 2.5 μm in diameter (PM2.5) is regarded as a carcinogenic factor, but the mechanism has been left unexplored. Our goal was to reveal the carcinogenic mechanism at the gene and protein level under the inhalational air-liquid interface (ALI) condition. Herein, we developed an ALI organ-level lung microfluidic platform (ALI-OLMP) carrying lung epithelial cell line BEAS-2B and human pulmonary microvascular endothelial cells (HPMEC); the cell viability was above 98% within 14 days on this system, which was used to mimic the practical alveolar microenvironment for the multiomics analysis, to identify the global gene and protein expression after exposure to PM2.5 in Shanghai, China from 2014 to 2015. The combined RNA-Seq and iTRAQ analysis indicated that the unique set was 2532 genes at 10 μg/cm2 of PM2.5, and there were also at least 25 identical activated signal transduction cascades including bladder cancer, transcriptional dysregulation in cancer, the TP53 (p53) signaling pathway, Jak-STAT signaling pathway, and PI3K-Akt signaling pathway, which could lead to blocking of differentiation, cell proliferation and survival, and sustained angiogenesis. The images obtained by the transmission electron microscopy (TEM) showed that the particles could enter the mitochondria, and even get into the nucleus. The Pearson's correlation coefficient test elucidated that inorganics (EC), organics (OC, PAHs, and alkane), and metals (Cr, Mn, and Sb) were significantly correlated to the dysregulated oncoproteins (VEGF, IL6, MDM2, AKT1, STAT, and P53). The findings may to some extent explain the molecular mechanism of carcinogenicity caused by fine-particle exposure.
Collapse
Affiliation(s)
- Lulu Zheng
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
- Engineering Research Center of Optical Instrument and System, Ministry of Education, Shanghai Key Lab of Modern Optical System, University of Shanghai for Science and Technology, 516 Jungong Road, Shanghai 200093, China
| | - Heng Dong
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Wang Zhao
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Xinlian Zhang
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Xiaoxiao Duan
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Hao Zhang
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Sixiu Liu
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Guodong Sui
- Shanghai Key laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science & Engineering, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| |
Collapse
|
15
|
XIAP RING domain mediates miR-4295 expression and subsequently inhibiting p63α protein translation and promoting transformation of bladder epithelial cells. Oncotarget 2018; 7:56540-56557. [PMID: 27447744 PMCID: PMC5302933 DOI: 10.18632/oncotarget.10645] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/13/2016] [Indexed: 12/12/2022] Open
Abstract
The X-linked inhibitor of apoptosis protein (XIAP) contains three N-terminal BIR domains that mediate anti-apoptosis and one C-terminal RING finger domain whose function(s) are not fully defined. Here we show that the RING domain of XIAP strongly inhibits the expression of p63α, a known tumor suppressor. XIAP knockdown in urothelial cells or RING deletion in knockin mice markedly upregulates p63α expression. This RING-mediated p63α downregulation is critical for the malignant transformation of normal urothelial cells following EGF treatment. We further show that the RING domain promotes Sp1-mediated transcription of miR-4295 which targets the 3′UTR of p63α mRNA and consequently inhibits p63α translation. Our results reveal a previously unknown function of the RING of XIAP in promoting miR-4295 transcription, thereby reducing p63α translation and enhancing urothelial transformation. Our data offer novel insights into the multifunctional effects of the XIAP RING domain on urothelial tumorigenesis and the potential for targeting this frequently overexpressed protein as a therapeutic alternative.
Collapse
|
16
|
Truong VL, Kong AN, Jeong WS. Red Ginseng Oil Inhibits TPA-Induced Transformation of Skin Epidermal JB6 Cells. J Med Food 2017; 21:380-389. [PMID: 29271701 DOI: 10.1089/jmf.2017.4082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Red ginseng oil (RGO) has been shown to possess anti-inflammatory and hepatoprotective activity. In this study, we evaluated the inhibitory effect of RGO on 12-O-tetradecanoylphorbol-13-acetate (TPA)-stimulated neoplastic transformation of JB6 P+ cells. RGO pretreatment abolished the transformation of JB6 P+ cells challenged by TPA. RGO suppressed the transactivation of activator protein-1 (AP-1) and nuclear factor kappa B (NF-κB) transcription factors as well as protein levels of cyclooxygenase-2, cyclin D1, cyclin E, and Bcl-2 in the TPA-treated cells. Additionally, TPA-induced phosphorylations of extracellular signal-regulated kinases, 90 kDa ribosomal S6 kinase 2, c-Jun N-terminal kinases, and glycogen synthase kinase 3β were downregulated in the presence of RGO. Furthermore, RGO induced the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant enzyme heme oxygenase-1 (HO-1) expression, and effectively blocked the overproduction of TPA-induced reactive oxygen species. These results suggest that RGO exerts a potent chemopreventive activity in skin cell model.
Collapse
Affiliation(s)
- Van-Long Truong
- 1 Department of Food and Life Sciences, College of BNIT, Inje University , Gimhae, Korea
| | - Ah Ng Kong
- 2 Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey , Piscataway, New Jersey, USA
| | - Woo-Sik Jeong
- 1 Department of Food and Life Sciences, College of BNIT, Inje University , Gimhae, Korea
| |
Collapse
|
17
|
Liao X, Huang C, Zhang D, Wang J, Li J, Jin H, Huang C. Mitochondrial catalase induces cells transformation through nucleolin-dependent Cox-2 mRNA stabilization. Free Radic Biol Med 2017; 113:478-486. [PMID: 29097213 DOI: 10.1016/j.freeradbiomed.2017.10.387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 12/12/2022]
Abstract
It's well documented that over-production of reactive oxygen species (ROS) causes detrimental damages to cells. While a low level of ROS, such as H2O2, functions as signaling transducer and motivates cell proliferation in both cancer and non-transformed stem cells. As a double-edged sword, the direct evidence for demonstrating the function of H2O2 in the cause of tumor is barely characterized in intact cells. In our current study, we found that targeted expression of mitochondrial catalase (mCAT), but not catalase, could significantly reduce the accumulation of H2O2 in mouse epithelial JB6 Cl41 cells, consequently led to the cell malignant transformation and anchorage-independent cell growth. Further study revealed that this reduction of H2O2 resulted in the translocation of nucleolin from the cytoplasm to nuclear, and maintaining the nucleolin nuclear location status, and in turn stabilizing the cox-2 mRNA and consequently leading to a COX-2 protein upregulation, as well as malignant transforming mCAT-overexpressed Cl41 cells. Collectively, our studies here provide direct experimental evidence demonstrating a novel function and molecular mechanisms of mCAT in transforming mouse Cl41 cells, and high significance insight into understanding the beneficial aspect of H2O2 in circumventing tumor promotion and the theoretical basis for the management of H2O2 in the clinic implementation as a chemotherapeutic strategy.
Collapse
Affiliation(s)
- Xin Liao
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA; Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Huang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA
| | - Dongyun Zhang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA
| | - Jingjing Wang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA; Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University, School of Medicine, Tuxedo Park, NY 10987, USA.
| |
Collapse
|
18
|
Chen C, Jiang X, Gu S, Zhang Z. MicroRNA-155 regulates arsenite-induced malignant transformation by targeting Nrf2-mediated oxidative damage in human bronchial epithelial cells. Toxicol Lett 2017; 278:38-47. [PMID: 28688901 DOI: 10.1016/j.toxlet.2017.07.215] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/01/2017] [Accepted: 07/04/2017] [Indexed: 12/23/2022]
Abstract
Arsenite is a well-documented human lung carcinogen but the detailed mechanisms of carcinogenesis remain unclear. In this study, human bronchial epithelial (16-HBE) cells were continuously exposed to 2.5μM arsenite for about 13 weeks to induce the phenotypes of malignant transformation. Our results showed that Nrf2 expression was gradually decreased whereas no significant change was observed on NF-κB activation with increased time of arsenite exposure. To test the roles of Nrf2-meidtaed oxidative damage in the arsenite-induced malignant transformation, we compared the levels of cGMP, PKG and oxidative damage-related indicators between arsenic-transformed cells and control cells. Our data demonstrated there were no significantly differences on the contents of cGMP, PKG, MDA and the production of ROS, but the levels of GSH and NO, the activities of SOD, tNOS and iNOS were significantly enhanced in the arsenic-transformed cells. Importantly, Nrf2 inactivation could be modulated by miR-155, and inhibition of miR-155 remarkably attenuated the malignant phenotypes and promoted apoptotic cell death in the arsenic-transformed cells. Together, our findings provide the novel mechanism that miR-155 may regulate arsenite-induced cell malignant transformation by targeting Nrf2-mediated oxidative damage, indicating that inhibition of miR-155 may be a potential strategy against lung carcinogenesis of arsenite.
Collapse
Affiliation(s)
- Chengzhi Chen
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Occupational and Environmental Health, School of Public Health and Management, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xuejun Jiang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China; Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People's Republic of China
| | - Shiyan Gu
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zunzhen Zhang
- Department of Occupational and Environmental Health, West China School of Public Health, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
19
|
Tiwari P, Sahay S, Pandey M, Qadri SSYH, Gupta KP. Preventive effects of butyric acid, nicotinamide, calcium glucarate alone or in combination during the 7, 12-dimethylbenz (a) anthracene induced mouse skin tumorigenesis via modulation of K-Ras-PI3K-AKTpathway and associated micro RNAs. Biochimie 2015; 121:112-22. [PMID: 26655363 DOI: 10.1016/j.biochi.2015.11.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 11/27/2015] [Indexed: 10/22/2022]
Abstract
Skin cancer is among the most common cancers worldwide and identifiable molecular changes for early and late stage of skin tumorigenesis can suggest the better targets for its control. In this study, we investigated the status of K-Ras-PI3K-AKTpathway followed by NF-κB, cyclin D1, MMP-9 and regulatory micro RNA during 7, 12-dimethylbenz[a]anthracene (DMBA) induced mouse skin tumorigenesis and its prevention by butyric acid (BA), nicotinamide (NA) and calcium glucarate (CAG), individually or in combination with respect to time. DMBA upregulated the K-Ras, PI3K, Akt, NF-κB, cyclin D1 and MMP-9, but downregulated the PTEN in a time dependent manner. DMBA also reduced the levels of micoRNA let-7a but induced the levels of miR-21 and miR-20a as a function of time. BA, NA and CAG were found to prevent DMBA induced changes, but they were most effective when used together in a combination. Reduced let-7a and miR-211 were correlated with the overexpression of K-Ras and MMP-9. Overexpression of miR-21 and miR-20a was correlated with the down regulation of PTEN and overexpression of Cyclin D1. Collectively, the enhanced chemopreventive potential of natural compound in combination via regulation of K-Ras-PI3K-AKTpathway along with regulatory micro RNAs provide a newer and effective mean for cancer management.
Collapse
Affiliation(s)
- Prakash Tiwari
- Environmental Carcinogenesis Division, CSIR-Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow, 226001, India; PhD Programme, Academy of Scientific and Innovative Research (AcSIR), India
| | - Satya Sahay
- Environmental Carcinogenesis Division, CSIR-Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow, 226001, India; PhD Programme, Academy of Scientific and Innovative Research (AcSIR), India
| | - Manuraj Pandey
- Environmental Carcinogenesis Division, CSIR-Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow, 226001, India
| | - Syed S Y H Qadri
- Pathology Division, National Institute of Nutrition, Hyderabad, India
| | - Krishna P Gupta
- Environmental Carcinogenesis Division, CSIR-Indian Institute of Toxicology Research, Mahatma Gandhi Marg, Lucknow, 226001, India.
| |
Collapse
|
20
|
Isorhapontigenin (ISO) inhibited cell transformation by inducing G0/G1 phase arrest via increasing MKP-1 mRNA Stability. Oncotarget 2015; 5:2664-77. [PMID: 24797581 PMCID: PMC4058035 DOI: 10.18632/oncotarget.1872] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The cancer chemopreventive property of Chinese herb new isolate isorhapontigenin (ISO) and mechanisms underlying its activity have never been explored. Here we demonstrated that ISO treatment with various concentrations for 3 weeks could dramatically inhibit TPA/EGF-induced cell transformation of Cl41 cells in Soft Agar assay, whereas co-incubation of cells with ISO at the same concentrations could elicit G0/G1 cell-cycle arrest without redundant cytotoxic effects on non-transformed cells. Further studies showed that ISO treatment resulted in cyclin D1 downregulation in dose- and time-dependent manner. Our results indicated that ISO regulated cyclin D1 at transcription level via targeting JNK/C-Jun/AP-1 activation. Moreover, we found that ISO-inhibited JNK/C-Jun/AP-1 activation was mediated by both upregulation of MKP-1 expression through increasing its mRNA stability and deactivating MKK7. Most importantly, MKP-1 knockdown could attenuate ISO-mediated suppression of JNK/C-Jun activation and cyclin D1 expression, as well as G0/G1 cell cycle arrest and cell transformation inhibition, while ectopic expression of FLAG-cyclin D1 T286A mutant also reversed ISO-induced G0/G1 cell-cycle arrest and inhibition of cell transformation. Our results demonstrated that ISO is a promising chemopreventive agent via upregulating mkp-1 mRNA stability, which is distinct from its cancer therapeutic effect with downregulation of XIAP and cyclin D1 expression.
Collapse
|
21
|
Huang H, Ma L, Li J, Yu Y, Zhang D, Wei J, Jin H, Xu D, Gao J, Huang C. NF-κB1 inhibits c-Myc protein degradation through suppression of FBW7 expression. Oncotarget 2015; 5:493-505. [PMID: 24457827 PMCID: PMC3964224 DOI: 10.18632/oncotarget.1643] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
NF-κB is a well-known transcription factor in regulation of multiple gene transcription and biological processes, and most of them are relied on its transcriptional activity of the p65/RelA subunit, while biological function of another ubiquitously expressed subunit NF-κB1 (p50) remains largely unknown due to lack transcriptional activation domain. Here we discovered a novel biological function of p50 as a regulator of oncogenic c-Myc protein degradation upon arsenite treatment in a NF-κB transcriptional-independent mechanism. Our results found that p50 was crucial for c-Myc protein induction following arsenite treatment by using specific knockdown and deletion of p50 in its normal expressed cells as well as reconstituting expression of p50 in its deficient cells. Subsequently we showed that p50 upregulated c-Myc protein expression mainly through inhibiting its degradation. We also identified that p50 exhibited this novel property by suppression of FBW7 expression. FBW7 was profoundly upregulated in p50-defecient cells in comparison to that in p50 intact cells, whereas knockdown of FBW7 in p50-/- cells restored arsenite-induced c-Myc protein accumulation, assuring that FBW7 up-regulation was responsible for defect of c-Myc protein expression in p50-/- cells. In addition, we discovered that p50 suppressed fbw7 gene transcription via inhibiting transcription factor E2F1 transactivation. Collectively, our studies demonstrated a novel function of p50 as a regulator of c-Myc protein degradation, contributing to our notion that p50-regulated protein expression through multiple levels at protein translation and degradation, further providing a significant insight into the understanding of biomedical significance of p50 protein.
Collapse
Affiliation(s)
- Haishan Huang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
TG-interacting factor mediates arsenic-induced malignant transformation of keratinocytes via c-Src/EGFR/AKT/FOXO3A and redox signalings. Arch Toxicol 2014; 89:2229-41. [DOI: 10.1007/s00204-014-1445-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 12/17/2014] [Indexed: 12/13/2022]
|
23
|
The N-terminal region of p27 inhibits HIF-1α protein translation in ribosomal protein S6-dependent manner by regulating PHLPP-Ras-ERK-p90RSK axis. Cell Death Dis 2014; 5:e1535. [PMID: 25412313 PMCID: PMC4260754 DOI: 10.1038/cddis.2014.496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/15/2014] [Accepted: 10/20/2014] [Indexed: 12/15/2022]
Abstract
P27 was identified as a tumor suppressor nearly two decades, being implicated in cell-cycle control, differentiation, senescence, apoptosis and motility. Our present study, for the first time to the best of our knowledge, revealed a potential role of p27 in inhibiting S6-mediated hypoxia-inducible factor-1α (HIF-1α) protein translation, which contributed to the protection from environmental carcinogen (sodium arsenite)-induced cell transformation. Our findings showed that depletion of p27 expression by knockout and knockdown approaches efficiently enhanced S6 phosphorylation in arsenite response via overactivating Ras/Raf/MEK/ERK pathway, which consequently resulted in the stimulation of p90RSK (90 kDa ribosomal S6 kinase), a direct kinase for S6 phosphorylation. Although PI3K/AKT pathway was also involved in S6 activation, blocking AKT and p70S6K activation did not attenuate arsenite-induced S6 activation in p27−/− cells, suggesting p27 specifically targeted Ras/ERK pathway rather than PI3K/AKT pathway for inhibition of S6 activation in response to arsenite exposure. Further functional studies found that p27 had a negative role in cell transformation induced by chronic low-dose arsentie exposure. Mechanistic investigations showed that HIF-1α translation was upregulated in p27-deficient cells in an S6 phosphorylation-dependent manner and functioned as a driving force in arsenite-induced cell transformation. Knockdown of HIF-1α efficiently reversed arsenite-induced cell transformation in p27-depleted cells. Taken together, our findings provided strong evidence showing that by targeting Ras/ERK pathway, p27 provided a negative control over HIF-1α protein synthesis in an S6-dependent manner, and abrogated arsenite-induced cell transformation via downregulation of HIF-1α translation.
Collapse
|
24
|
Zhu J, Zhang J, Huang H, Li J, Yu Y, Jin H, Li Y, Deng X, Gao J, Zhao Q, Huang C. Crucial role of c-Jun phosphorylation at Ser63/73 mediated by PHLPP protein degradation in the cheliensisin a inhibition of cell transformation. Cancer Prev Res (Phila) 2014; 7:1270-81. [PMID: 25281487 DOI: 10.1158/1940-6207.capr-14-0233] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Cheliensisin A (Chel A), as a novel styryl-lactone isolated from Goniothalamus cheliensis Hu, has been demonstrated to have an inhibition of EGF-induced Cl41 cell transformation via stabilizing p53 protein in a Chk1-dependent manner, suggesting its chemopreventive activity in our previous studies. However, its underlying molecular mechanisms have not been fully characterized yet. In the current study, we found that Chel A treatment could increase c-Jun protein phosphorylation and activation, whereas the inhibition of c-Jun phosphorylation, by ectopic expression of a dominant-negative mutant of c-Jun, TAM67, reversed the Chel A inhibition of EGF-induced cell transformation and impaired Chel A induction of p53 protein and apoptosis. Moreover, our results indicated that Chel A treatment led to a PHLPP downregulation by promoting PHLPP protein degradation. We also found that PHLPP could interact with and bind to c-Jun protein, whereas ectopic PHLPP expression blocked c-Jun activation, p53 protein and apoptotic induction by Chel A, and further reversed the Chel A inhibition of EGF-induced cell transformation. With the findings, we have demonstrated that Chel A treatment promotes a PHLPP protein degradation, which can bind to c-Jun and mediates c-Jun phosphorylation, and further leading to p53 protein induction, apoptotic responses, subsequently resulting in cell transformation inhibition and chemopreventive activity of Chel A.
Collapse
Affiliation(s)
- Junlan Zhu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York. Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjie Zhang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| | - Haishan Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York. Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| | - Yonghui Yu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| | - Honglei Jin
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York. Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yang Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York. Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xu Deng
- State Key Laboratory of Phytochemistry and Plant Resources in West China and Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Qinshi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China and Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China.
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York.
| |
Collapse
|
25
|
Wang S, Teng X, Wang Y, Yu HQ, Luo X, Xu A, Wu L. Molecular control of arsenite-induced apoptosis in Caenorhabditis elegans: roles of insulin-like growth factor-1 signaling pathway. CHEMOSPHERE 2014; 112:248-255. [PMID: 25048913 DOI: 10.1016/j.chemosphere.2014.04.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/02/2014] [Accepted: 04/05/2014] [Indexed: 06/03/2023]
Abstract
Apoptosis is one of the main cellular processes in responses to arsenic, the well known environmental carcinogen. By using the nematode Caenorhabditis elegans as an in vivo model, we found that insulin-like growth factor-1 networks and their target protein DAF-16/FOXO, known as key regulators of energy metabolism and growth, played important roles in arsenite-induced apoptosis. Inactivation of DAF-2, AGE-1 and AKT-1 caused worms more susceptible to arsenite-induced apoptosis, which could be attenuated by DAF-16 knockout. Worms with inactivated AKT-2 and SGK-1 or with constitutively activated PDK-1 and AKT-1 showed low levels of apoptosis, which could be elevated by DAF-16 mutation. Our results demonstrated that DAF-2/IGF-1R, AGE-1/PI3K, PDK-1/PDK1 and AKT-1/PKB negatively regulated the arsenite-induced apoptosis, whereas AKT-2 and SGK-1 acted proapoptotically. DAF-16/FOXO antagonized IGF-1 signals in signaling the arsenite-induced apoptosis, and apoptosis promoted by DAF-16 inactivation was attributed to its higher sensitivity to oxidative stress.
Collapse
Affiliation(s)
- Shunchang Wang
- Department of Life Science, Huainan Normal University, Huainan 232001, China.
| | - Xiaoxue Teng
- Department of Life Science, Huainan Normal University, Huainan 232001, China; School of Life Sciences, Anhui University, Hefei 230601, China
| | - Yun Wang
- Department of Life Science, Huainan Normal University, Huainan 232001, China
| | - Han-Qing Yu
- Department of Chemistry, University of Science & Technology of China, Hefei 230026, China
| | - Xun Luo
- Department of Life Science, Huainan Normal University, Huainan 232001, China; Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - An Xu
- Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Lijun Wu
- Key Laboratory of Ion Beam Bioengineering, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
26
|
SUMOylation of RhoGDIα is required for its repression of cyclin D1 expression and anchorage-independent growth of cancer cells. Mol Oncol 2013; 8:285-96. [PMID: 24342356 DOI: 10.1016/j.molonc.2013.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/30/2013] [Accepted: 11/18/2013] [Indexed: 12/19/2022] Open
Abstract
Selective activation of Rho GTPase cascade requires the release of Rho from RhoGDI (GDP-dissociation inhibitors) complexes. Our previous studies identified RhoGDIα SUMOylation at Lys-138 and its function in the regulation of cancer cell invasion. In the current study, we demonstrate that RhoGDIα SUMOylation has a crucial role in the suppression of cancer cell anchorage-independent growth as well as the molecular mechanisms underlying this suppression. We found that ectopic expression of RhoGDIα resulted in marked inhibition of an anchorage-independent growth with induction of G0/G1 cell cycle arrest, while point mutation of RhoGDIα SUMOylation at residue Lys-138 (K138R) abrogated this growth suppression and G0/G1 cell cycle arrest in cancer cells. Further studies showed that SUMOylation at Lys-138 was critical for RhoGDIα down-regulation of cyclin D1 protein expression and that MEK1/2-Erk was a specific downstream target of SUMOylated RhoGDIα for its inhibition of C-Jun/AP-1 cascade, cyclin d1 transcription, and cell cycle progression. These results strongly demonstrate that SUMOylated RhoGDIα suppressed C-Jun/AP-1-dependent transactivation specifically via targeting MEK1/2-Erk, subsequently leading to the down-regulation of cyclin D1 expression and anti-cancer activity. Our results provide new mechanistic insights into the understanding of essential role of SUMOylation at Lys-138 in RhoGDIα's biological function.
Collapse
|
27
|
The role of EGFR/PI3K/Akt/cyclinD1 signaling pathway in acquired middle ear cholesteatoma. Mediators Inflamm 2013; 2013:651207. [PMID: 24311896 PMCID: PMC3839121 DOI: 10.1155/2013/651207] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/24/2013] [Indexed: 12/21/2022] Open
Abstract
Cholesteatoma is a benign keratinizing and hyper proliferative squamous epithelial lesion of the temporal bone. Epidermal growth factor (EGF) is one of the most important cytokines which has been shown to play a critical role in cholesteatoma. In this investigation, we studied the effects of EGF on the proliferation of keratinocytes and EGF-mediated signaling pathways underlying the pathogenesis of cholesteatoma. We examined the expressions of phosphorylated EGF receptor (p-EGFR), phosphorylated Akt (p-Akt), cyclinD1, and proliferating cell nuclear antigen (PCNA) in 40 cholesteatoma samples and 20 samples of normal external auditory canal (EAC) epithelium by immunohistochemical method. Furthermore, in vitro studies were performed to investigate EGF-induced downstream signaling pathways in primary external auditory canal keratinocytes (EACKs). The expressions of p-EGFR, p-Akt, cyclinD1, and PCNA in cholesteatoma epithelium were significantly increased when compared with those of control subjects. We also demonstrated that EGF led to the activation of the EGFR/PI3K/Akt/cyclinD1 signaling pathway, which played a critical role in EGF-induced cell proliferation and cell cycle progression of EACKs. Both EGFR inhibitor AG1478 and PI3K inhibitor wortmannin inhibited the EGF-induced EGFR/PI3K/Akt/cyclinD1 signaling pathway concomitantly with inhibition of cell proliferation and cell cycle progression of EACKs. Taken together, our data suggest that the EGFR/PI3K/Akt/cyclinD1 signaling pathway is active in cholesteatoma and may play a crucial role in cholesteatoma epithelial hyper-proliferation. This study will facilitate the development of potential therapeutic targets for intratympanic drug therapy for cholesteatoma.
Collapse
|
28
|
Fang Y, Cao Z, Hou Q, Ma C, Yao C, Li J, Wu XR, Huang C. Cyclin d1 downregulation contributes to anticancer effect of isorhapontigenin on human bladder cancer cells. Mol Cancer Ther 2013; 12:1492-503. [PMID: 23723126 DOI: 10.1158/1535-7163.mct-12-0922] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Isorhapontigenin (ISO) is a new derivative of stilbene compound that was isolated from the Chinese herb Gnetum Cleistostachyum and has been used for treatment of bladder cancers for centuries. In our current studies, we have explored the potential inhibitory effect and molecular mechanisms underlying isorhapontigenin anticancer effects on anchorage-independent growth of human bladder cancer cell lines. We found that isorhapontigenin showed a significant inhibitory effect on human bladder cancer cell growth and was accompanied with related cell cycle G(0)-G(1) arrest as well as downregulation of cyclin D1 expression at the transcriptional level in UMUC3 and RT112 cells. Further studies identified that isorhapontigenin downregulated cyclin D1 gene transcription via inhibition of specific protein 1 (SP1) transactivation. Moreover, ectopic expression of GFP-cyclin D1 rendered UMUC3 cells resistant to induction of cell-cycle G(0)-G(1) arrest and inhibition of cancer cell anchorage-independent growth by isorhapontigenin treatment. Together, our studies show that isorhapontigenin is an active compound that mediates Gnetum Cleistostachyum's induction of cell-cycle G(0)-G(1) arrest and inhibition of cancer cell anchorage-independent growth through downregulating SP1/cyclin D1 axis in bladder cancer cells. Our studies provide a novel insight into understanding the anticancer activity of the Chinese herb Gnetum Cleistostachyum and its isolate isorhapontigenin.
Collapse
Affiliation(s)
- Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, ZheJiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Cao Z, Zhang R, Li J, Huang H, Zhang D, Zhang J, Gao J, Chen J, Huang C. X-linked inhibitor of apoptosis protein (XIAP) regulation of cyclin D1 protein expression and cancer cell anchorage-independent growth via its E3 ligase-mediated protein phosphatase 2A/c-Jun axis. J Biol Chem 2013; 288:20238-47. [PMID: 23720779 DOI: 10.1074/jbc.m112.448365] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The X-linked inhibitor of apoptosis protein (XIAP) is a well known potent inhibitor of apoptosis; however, it is also involved in other cancer cell biological behavior. In the current study, we discovered that XIAP and its E3 ligase played a crucial role in regulation of cyclin D1 expression in cancer cells. We found that deficiency of XIAP expression resulted in a marked reduction in cyclin D1 expression. Consistently, cell cycle transition and anchorage-independent cell growth were also attenuated in XIAP-deficient cancer cells compared with those of the parental wild-type cells. Subsequent studies demonstrated that E3 ligase activity within the RING domain of XIAP is crucial for its ability to regulate cyclin D1 transcription, cell cycle transition, and anchorage-independent cell growth by up-regulating transactivation of c-Jun/AP-1. Moreover, we found that E3 ligase within RING domain was required for XIAP inhibition of phosphatase PP2A activity by up-regulation of PP2A phosphorylation at Tyr-307 in its catalytic subunit. Such PP2A phosphorylation and inactivation resulted in phosphorylation and activation of its downstream target c-Jun in turn leading to cyclin D1 expression. Collectively, our studies uncovered a novel function of E3 ligase activity of XIAP in the up-regulation of cyclin D1 expression, providing significant insight into the understanding of the biomedical significance of overexpressed XIAP in cancer development, further offering a new molecular basis for utilizing XIAP E3 ligase as a cancer therapeutic target.
Collapse
Affiliation(s)
- Zipeng Cao
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York 10987, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yu Y, Huang H, Li J, Zhang J, Gao J, Lu B, Huang C. GADD45β mediates p53 protein degradation via Src/PP2A/MDM2 pathway upon arsenite treatment. Cell Death Dis 2013; 4:e637. [PMID: 23681232 PMCID: PMC3674369 DOI: 10.1038/cddis.2013.162] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Growth arrest and DNA-damage-inducible, beta (GADD45β) has been reported to inhibit apoptosis via attenuating c-Jun N-terminal kinase (JNK) activation. We demonstrated here that GADD45β mediated its anti-apoptotic effect via promoting p53 protein degradation following arsenite treatment. We found that p53 protein expression was upregulated in GADD45β−/− cells upon arsenite exposure as compared with those in GADD45β+/+ cells. Further studies showed that GADD45β attenuated p53 protein expression through Src/protein phosphatase 2A/murine double minute 2-dependent p53 protein-degradation pathway. Moreover, we identified that GADD45β-mediated p53 protein degradation was crucial for its anti-apoptotic effect due to arsenite exposure, whereas increased JNK activation was not involved in the increased cell apoptotic response in GADD45β−/− cells under same experimental conditions. Collectively, our results demonstrate a novel molecular mechanism responsible for GADD45β protection of arsenite-exposed cells from cell death, which provides insight into our understanding of GADD45β function and a unique compound arsenite as both a cancer therapeutic reagent and an environmental carcinogen. Those novel findings may also enable us to design more effective strategies for utilization of arsenite for the treatment of cancers.
Collapse
Affiliation(s)
- Y Yu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Zuo Z, Ouyang W, Li J, Costa M, Huang C. Cyclooxygenase-2 (COX-2) mediates arsenite inhibition of UVB-induced cellular apoptosis in mouse epidermal Cl41 cells. Curr Cancer Drug Targets 2012; 12:607-16. [PMID: 22463588 DOI: 10.2174/156800912801784802] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 02/24/2012] [Accepted: 02/24/2012] [Indexed: 12/29/2022]
Abstract
Inorganic arsenic is an environmental human carcinogen, and has been shown to act as a co-carcinogen with solar ultraviolet (UV) radiation in mouse skin tumor induction even at low concentrations. However, the precise mechanism of its co-carcinogenic action is largely unknown. Apoptosis plays an essential role as a protective mechanism against neoplastic development in the organism by eliminating genetically damaged cells. Thus, suppression of apoptosis is thought to contribute to carcinogenesis. It is known that cyclooxygenase-2 (COX-2) can promote carcinogenesis by inhibiting cell apoptosis under stress conditions; and our current studies investigated the potential contribution of COX-2 to the inhibitory effect of arsenite in UV-induced cell apoptosis in mouse epidermal Cl41 cells. We found that treatment of cells with low concentration (5 μM) arsenite attenuated cellular apoptosis upon UVB radiation accompanied with a coinductive effect on COX-2 expression and nuclear factor-κB (NFκB) transactivation. Our results also showed that the COX-2 induction by arsenite and UVB depended on an NFκB pathway because COX-2 co-induction could be attenuated in either p65-deficient or p50-deficient cells. Moreover, UVB-induced cell apoptosis could be dramatically reduced by the introduction of exogenous COX-2 expression, whereas the inhibitory effect of arsenite on UVB-induced cell apoptosis could be impaired in COX-2 knockdown C141 cells. Our results indicated that COX-2 mediated the anti-apoptotic effect of arsenite in UVB radiation through an NFκB-dependent pathway. Given the importance of apoptosis evasion during carcinogenesis, we anticipated that COX-2 induction might be at least partially responsible for the co-carcinogenic effect of arsenite on UVB-induced skin carcinogenesis.
Collapse
Affiliation(s)
- Z Zuo
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, 10987, USA
| | | | | | | | | |
Collapse
|
32
|
Li X, Li H, Li S, Zhu F, Kim DJ, Xie H, Li Y, Nadas J, Oi N, Zykova TA, Yu DH, Lee MH, Kim MO, Wang L, Ma W, Lubet RA, Bode AM, Dong Z, Dong Z. Ceftriaxone, an FDA-approved cephalosporin antibiotic, suppresses lung cancer growth by targeting Aurora B. Carcinogenesis 2012; 33:2548-57. [PMID: 22962305 DOI: 10.1093/carcin/bgs283] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Ceftriaxone, an FDA-approved third-generation cephalosporin antibiotic, has antimicrobial activity against both gram-positive and gram-negative organisms. Generally, ceftriaxone is used for a variety of infections such as community-acquired pneumonia, meningitis and gonorrhea. Its primary molecular targets are the penicillin-binding proteins. However, other activities of ceftriaxone remain unknown. Herein, we report for the first time that ceftriaxone has antitumor activity in vitro and in vivo. Kinase profiling results predicted that Aurora B might be a potential 'off' target of ceftriaxone. Pull-down assay data confirmed that ceftriaxone could bind with Aurora B in vitro and in A549 cells. Furthermore, ceftriaxone (500 µM) suppressed anchorage-independent cell growth by targeting Aurora B in A549, H520 and H1650 lung cancer cells. Importantly, in vivo xenograft animal model results showed that ceftriaxone effectively suppressed A549 and H520 lung tumor growth by inhibiting Aurora B. These data suggest the anticancer efficacy of ceftriaxone for the treatment of lung cancers through its inhibition of Aurora B.
Collapse
Affiliation(s)
- Xiang Li
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kim HG, Kim DJ, Li S, Lee KY, Li X, Bode AM, Dong Z. Polycomb (PcG) proteins, BMI1 and SUZ12, regulate arsenic-induced cell transformation. J Biol Chem 2012; 287:31920-8. [PMID: 22843710 DOI: 10.1074/jbc.m112.360362] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Inorganic arsenic is a well-documented human carcinogen associated with cancers of the skin, lung, liver, and bladder. However, the underlying mechanisms explaining the tumorigenic role of arsenic are not well understood. The present study explored a potential mechanism of cell transformation induced by arsenic exposure. Exposure to a low dose (0.5 μm) of arsenic trioxide (As(2)O(3)) caused transformation of BALB/c 3T3 cells. In addition, in a xenograft mouse model, tumor growth of the arsenic-induced transformed cells was dramatically increased. In arsenic-induced transformed cells, polycomb group (PcG) proteins, including BMI1 and SUZ12, were activated resulting in enhanced histone H3K27 tri-methylation levels. On the other hand, tumor suppressor p16(INK4a) and p19(ARF) mRNA and protein expression were dramatically suppressed. Introduction of small hairpin (sh) RNA-BMI1 or -SUZ12 into BALB/c 3T3 cells resulted in suppression of arsenic-induced transformation. Histone H3K27 tri-methylation returned to normal in BMI1- or SUZ12-knockdown BALB/c 3T3 cells compared with BMI1- or SUZ12-wildtype cells after arsenic exposure. As a consequence, the expression of p16(INK4a) and p19(ARF) was recovered in arsenic-treated BMI1- or SUZ12-knockdown cells. Thus, arsenic-induced cell transformation was blocked by inhibition of PcG function. Taken together, these results strongly suggest that the polycomb proteins, BMI1 and SUZ12 are required for cell transformation induced by organic arsenic exposure.
Collapse
Affiliation(s)
- Hong-Gyum Kim
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Zhang J, Ouyang W, Li J, Zhang D, Yu Y, Wang Y, Li X, Huang C. Suberoylanilide hydroxamic acid (SAHA) inhibits EGF-induced cell transformation via reduction of cyclin D1 mRNA stability. Toxicol Appl Pharmacol 2012; 263:218-24. [PMID: 22749963 DOI: 10.1016/j.taap.2012.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/16/2012] [Accepted: 06/19/2012] [Indexed: 02/07/2023]
Abstract
Suberoylanilide hydroxamic acid (SAHA) inhibiting cancer cell growth has been associated with its downregulation of cyclin D1 protein expression at transcription level or translation level. Here, we have demonstrated that SAHA inhibited EGF-induced Cl41 cell transformation via the decrease of cyclin D1 mRNA stability and induction of G0/G1 growth arrest. We found that SAHA treatment resulted in the dramatic inhibition of EGF-induced cell transformation, cyclin D1 protein expression and induction of G0/G1 growth arrest. Further studies showed that SAHA downregulation of cyclin D1 was only observed with endogenous cyclin D1, but not with reconstitutionally expressed cyclin D1 in the same cells, excluding the possibility of SAHA regulating cyclin D1 at level of protein degradation. Moreover, SAHA inhibited EGF-induced cyclin d1 mRNA level, whereas it did not show any inhibitory effect on cyclin D1 promoter-driven luciferase reporter activity under the same experimental conditions, suggesting that SAHA may decrease cyclin D1 mRNA stability. This notion was supported by the results that treatment of cells with SAHA decreased the half-life of cyclin D1 mRNA from 6.95 h to 2.57 h. Consistent with downregulation of cyclin D1 mRNA stability, SAHA treatment also attenuated HuR expression, which has been well-characterized as a positive regulator of cyclin D1 mRNA stability. Thus, our study identifies a novel mechanism responsible for SAHA inhibiting cell transformation via decreasing cyclin D1 mRNA stability and induction of G0/G1 growth arrest in Cl41 cells.
Collapse
Affiliation(s)
- Jingjie Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, 38 Xueyuan Rd, Haidian District, Beijing 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Suzuki T, Kita K, Ochi T. Phosphorylation of histone H3 at serine 10 has an essential role in arsenite-induced expression of FOS, EGR1 and IL8 mRNA in cultured human cell lines. J Appl Toxicol 2012; 33:746-55. [PMID: 22354777 DOI: 10.1002/jat.2724] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 12/28/2011] [Accepted: 12/28/2011] [Indexed: 12/31/2022]
Abstract
Trivalent inorganic arsenite [iAs(III)] is known to alter the expression of a number of genes associated with transcription and cell proliferation, which was thought to be one of the possible mechanisms of arsenical carcinogenesis. However, the detailed mechanisms underlying iAs(III) induction of changes in gene expression are not fully understood. Here we examine the role of histone H3 phosphorylation at serine 10 (Ser(10) ) in gene regulation when the cells were treated with iAs(III). Among the 34 genes tested, iAs(III) induced mRNA expression of JUN, FOS, EGR1, HMOX1, HSPA1A, IL8, GADD45A, GADD45B and GADD153. Phosphorylation of histone H3 Ser(10) was induced by iAs(III) in interphase cells, and was effectively blocked by the ERKs pathway inhibitor (U0126). U0126 treatment significantly reduced constitutive mRNA expression of FOS and EGR1, and dramatically suppressed the induction of FOS, EGR1 and IL8 mRNA in iAs(III)-treated cells. The other genes, which were induced by iAs(III), were not affected by U0126 treatment. When the histone H3 nonphosphorylatable mutant of serine 10 (S10A) was overexpressed in cells, iAs(III) induction of FOS, EGR1and IL8 expression was significantly decreased as compared with wild-type cells. The other genes induced by iAs(III) were not changed in S10A cells nor by U0126 treatment. In addition, S10A cells were more resistant to iAs(III) cytotoxicity. These results indicated that the phosphorylation of histone H3 at Ser(10) through the ERKs pathway in interphase cells is an important regulatory event for iAs(III)-mediated gene expression. Aberrant gene expression seems to be an important cause of cytotoxicity and may have some relation to iAs(III) carcinogenicity.
Collapse
Affiliation(s)
- Toshihide Suzuki
- Faculty of Pharmaceutical Sciences, Teikyo University, 1091-1 Sagamihara, Kanagawa, 252-5195, Japan.
| | | | | |
Collapse
|
36
|
Wang Z, Yang J, Fisher T, Xiao H, Jiang Y, Yang C. Akt activation is responsible for enhanced migratory and invasive behavior of arsenic-transformed human bronchial epithelial cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:92-7. [PMID: 21954225 PMCID: PMC3261952 DOI: 10.1289/ehp.1104061] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 09/27/2011] [Indexed: 05/05/2023]
Abstract
BACKGROUND Arsenic is one of the most common environmental contaminants. Long-term exposure to arsenic causes human bronchial epithelial cell (HBEC) malignant transformation and lung cancer. However, the mechanism of arsenic lung carcinogenesis is not clear, and the migratory and invasive properties of arsenic-transformed cells (As-transformed cells) have rarely been studied. OBJECTIVES This study was designed to investigate the migratory and invasive behavior of As-transformed HBECs and the underlying mechanism. METHODS As-transformed p53lowHBECs were generated by exposing p53-knockdown HBECs to sodium arsenite (2.5 μM) for 16 weeks. Cell migration was assessed by transwell migration and wound-healing assay. Cell invasion was evaluated using Matrigel-coated transwell chambers. Gene overexpression, small interfering RNA (siRNA) knockdowns, and pharmacological inhibitors were used to determine the potential mechanism responsible for enhanced cell migration and invasion. RESULTS Transwell migration and invasion assays revealed that As-transformed p53lowHBECs were highly migratory and invasive. Akt (also known as protein kinase B) and extracellular signal-regulated protein kinase 1/2 (Erk1/2) were strongly activated in As-transformed p53lowHBECs. Stable expression of microRNA 200b in As-transformed p53lowHBECs abolished Akt and Erk1/2 activation and completely suppressed cell migration and invasion. Pharmacological inactivation of Akt but not Erk1/2 significantly decreased cell migration and invasion. Inhibition of Akt reduced the expression of epithelial-to-mesenchymal transition-inducing transcription factors zinc-finger E-box-binding homeobox factor 1 (ZEB1) and ZEB2. siRNA knockdown of ZEB1 and ZEB2 impaired As-transformed p53lowHBEC migration and invasion. CONCLUSIONS Akt activation plays a critical role in enabling As-transformed HBEC migration and invasion by promoting ZEB1 and ZEB2 expression.
Collapse
Affiliation(s)
- Zhishan Wang
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | | | |
Collapse
|
37
|
Carpenter RL, Jiang Y, Jing Y, He J, Rojanasakul Y, Liu LZ, Jiang BH. Arsenite induces cell transformation by reactive oxygen species, AKT, ERK1/2, and p70S6K1. Biochem Biophys Res Commun 2011; 414:533-8. [PMID: 21971544 DOI: 10.1016/j.bbrc.2011.09.102] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 09/20/2011] [Indexed: 11/18/2022]
Abstract
Arsenic is naturally occurring element that exists in both organic and inorganic formulations. The inorganic form arsenite has a positive association with development of multiple cancer types. There are significant populations throughout the world with high exposure to arsenite via drinking water. Thus, human exposure to arsenic has become a significant public health problem. Recent evidence suggests that reactive oxygen species (ROS) mediate multiple changes to cell behavior after acute arsenic exposure, including activation of proliferative signaling and angiogenesis. However, the role of ROS in mediating cell transformation by chronic arsenic exposure is unknown. We found that cells chronically exposed to sodium arsenite increased proliferation and gained anchorage-independent growth. This cell transformation phenotype required constitutive activation of AKT, ERK1/2, mTOR, and p70S6K1. We also observed these cells constitutively produce ROS, which was required for the constitutive activation of AKT, ERK1/2, mTOR, and p70S6K1. Suppression of ROS levels by forced expression of catalase also reduced cell proliferation and anchorage-independent growth. These results indicate cell transformation induced by chronic arsenic exposure is mediated by increased cellular levels of ROS, which mediates activation of AKT, ERK1/2, and p70S6K1.
Collapse
Affiliation(s)
- Richard L Carpenter
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | | | | | | | | | | | | |
Collapse
|
38
|
Zhang Y, Wang Q, Guo X, Miller R, Guo Y, Yang HS. Activation and up-regulation of translation initiation factor 4B contribute to arsenic-induced transformation. Mol Carcinog 2011; 50:528-38. [PMID: 21268130 PMCID: PMC3110507 DOI: 10.1002/mc.20733] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 12/03/2010] [Accepted: 12/08/2010] [Indexed: 01/23/2023]
Abstract
Arsenic is a known human carcinogen. However, the mechanism of how arsenic induces cell transformation remains unclear. In this study, we demonstrated that long-term exposure to sodium arsenite at low-dose (2 µM) increases cell proliferation and neoplastic transformation in a mouse epidermal cell model, JB6 promotion-susceptible cells. The phosphorylation of AKT and its downstream targets, 70-kDa ribosomal protein S6 kinase (p70S6K) and translation initiation factor 4B (eIF4B), are increased in the arsenite treated cells, indicating that long-term arsenite treatment activates AKT-p70S6K signaling pathway. In addition, long-term exposure to arsenite up-regulates eIF4B expression and increases the rate of translation. Knockdown of eIF4B expression resulted in inhibition of arsenic-induced cell proliferation, transformation, and translation. Moreover, the expression of c-Myc which is up-regulated by long-term arsenite treatment is inhibited by eIF4B knockdown. Taken together, these results indicate that activation and up-regulation of eIF4B contributes to arsenic-induced transformation in JB6 cells.
Collapse
Affiliation(s)
- Yong Zhang
- Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY40536, USA
- Institute and Department of Urology, Peking University First Hospital, Beijing, China
| | - Qing Wang
- Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY40536, USA
| | - Xiaoling Guo
- Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY40536, USA
- Department of Oral and Maxillofacial Surgery, and School and Hospital of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Robert Miller
- Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY40536, USA
| | - Yinglu Guo
- Institute and Department of Urology, Peking University First Hospital, Beijing, China
| | - Hsin-Sheng Yang
- Graduate Center for Toxicology, College of Medicine, University of Kentucky, Lexington, KY40536, USA
- Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY40536, USA
| |
Collapse
|
39
|
Tyagi S, George J, Singh R, Bhui K, Shukla Y. Neoplastic Alterations Induced in Mammalian Skin Following Mancozeb Exposure Using In Vivo and In Vitro Models. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2011; 15:155-67. [DOI: 10.1089/omi.2010.0076] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Shilpa Tyagi
- Proteomics Laboratory, Indian Institute of Toxicology Research, Council of Scientific & Industrial Research (CSIR), Lucknow 226001 (U.P), India
| | - Jasmine George
- Proteomics Laboratory, Indian Institute of Toxicology Research, Council of Scientific & Industrial Research (CSIR), Lucknow 226001 (U.P), India
| | - Richa Singh
- Proteomics Laboratory, Indian Institute of Toxicology Research, Council of Scientific & Industrial Research (CSIR), Lucknow 226001 (U.P), India
| | - Kulpreet Bhui
- Proteomics Laboratory, Indian Institute of Toxicology Research, Council of Scientific & Industrial Research (CSIR), Lucknow 226001 (U.P), India
| | - Yogeshwer Shukla
- Proteomics Laboratory, Indian Institute of Toxicology Research, Council of Scientific & Industrial Research (CSIR), Lucknow 226001 (U.P), India
| |
Collapse
|
40
|
Liu J, Zhang D, Mi X, Xia Q, Yu Y, Zuo Z, Guo W, Zhao X, Cao J, Yang Q, Zhu A, Yang W, Shi X, Li J, Huang C. p27 suppresses arsenite-induced Hsp27/Hsp70 expression through inhibiting JNK2/c-Jun- and HSF-1-dependent pathways. J Biol Chem 2010; 285:26058-65. [PMID: 20566634 DOI: 10.1074/jbc.m110.100271] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p27 is an atypical tumor suppressor that can regulate the activity of cyclin-dependent kinases and G(0)-to-S phase transitions. More recent studies reveal that p27 may also exhibit its tumor-suppressive function through regulating many other essential cellular events. However, the molecular mechanisms underlying these anticancer effects of p27 are largely unknown. In this study, we found that depletion of p27 expression by either gene knock-out or knockdown approaches resulted in up-regulation of both Hsp27 and Hsp70 expression at mRNA- and promoter-derived transcription as well as protein levels upon arsenite exposure, indicating that p27 provides a negative signal for regulating the expression of Hsp27 and Hsp70. Consistently, arsenite-induced activation of JNK2/c-Jun and HSF-1 pathways was also markedly elevated in p27 knock-out (p27(-/-)) and knockdown (p27 shRNA) cells. Moreover, interference with the expression or function of JNK2, c-Jun, and HSF-1, but not JNK1, led to dramatic inhibition of arsenite-induced Hsp27 and Hsp70 expression. Collectively, our results demonstrate that p27 suppresses Hsp27 and Hsp70 expression at the transcriptional level specifically through JNK2/c-Jun- and HSF-1-dependent pathways upon arsenite exposure, which provides additional important molecular mechanisms for the tumor-suppressive function of p27.
Collapse
Affiliation(s)
- Jinyi Liu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, New York 10987, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ding J, Ning B, Gong W, Wen W, Wu K, Liang J, He G, Huang S, Sun W, Han T, Huang L, Cao G, Wu M, Xie W, Wang H. Cyclin D1 induction by benzo[a]pyrene-7,8-diol-9,10-epoxide via the phosphatidylinositol 3-kinase/Akt/MAPK- and p70s6k-dependent pathway promotes cell transformation and tumorigenesis. J Biol Chem 2009; 284:33311-9. [PMID: 19801633 DOI: 10.1074/jbc.m109.046417] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Benzo[a]pyrene-7,8-diol-9,10-epoxide (B[a]PDE), the major metabolite of B[a]P, has been well recognized as one ubiquitous carcinogen, but the molecular mechanism involved in its carcinogenic effect remains obscure. In the present study, we found that bronchial epithelial cells (Beas-2B) and hepatocytes treated with B[a]PDE presented a significant increase of cyclin D1 expression. Moreover, Akt, p70(s6k), and MAPKs including JNK, Erks, and p38 were notably activated in B[a]PDE-treated Beas-2B cells, whereas NF-kappaB, NFAT, and Egr-1 were not. Our results demonstrated that JNK and Erks were required in B[a]PDE-induced cyclin D1 expression because the inhibition of JNK or Erks by a selective chemical inhibitor or dominant negative mutant robustly impaired the cyclin D1 induction by B[a]PDE. Furthermore, we found that overexpression of the dominant negative mutant of p85 (regulatory subunit of phosphatidylinositol 3-kinase) or Akt dramatically suppressed B[a]PDE-induced JNK and Erk activation as well as cyclin D1 expression, suggesting that cyclin D1 induction by B[a]PDE is via the phosphatidylinositol 3-kinase/Akt/MAPK-dependent pathway. In addition, we clarified that p70(s6k) is also involved in B[a]PDE-induced cyclin D1 expression because rampamycin pretreatment dramatically reduced cyclin D1 induction by B[a]PDE. More importantly, we demonstrated that up-regulated cyclin D1 by B[a]PDE plays a critical role in oncogenic transformation and tumorigenesis of Beas-2B cells. These results not only broaden our knowledge of the molecular mechanism of B[a]PDE carcinogenicity but also lead to the further study of chemoprevention of B[a]PDE-associated human cancers.
Collapse
Affiliation(s)
- Jin Ding
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Changzheng Hospital, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ding J, Ning B, Huang Y, Zhang D, Li J, Chen CY, Huang C. PI3K/Akt/JNK/c-Jun signaling pathway is a mediator for arsenite-induced cyclin D1 expression and cell growth in human bronchial epithelial cells. Curr Cancer Drug Targets 2009; 9:500-9. [PMID: 19519318 DOI: 10.2174/156800909788486740] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Arsenite exposure is associated with an increased risk of human lung cancer. However, the molecular mechanisms underlying the arsenite-induced human lung carcinogenesis remain elusive. In this study, we demonstrated that arsenite upregulates cyclin D1 expression/activity to promote the growth of human bronchial epithelial Beas-2B cells. In this process, the JNKs (c-Jun N-terminal kinases)/c-Jun cascade is elicited. The inhibition of JNKs or c-Jun by chemical or genetic inhibitors blocks the cyclin D1 induction mediated by arsenite. Furthermore, using a loss of function mutant of p85 (Deltap85, a subunit of PI3K) or dominant-negative Akt (DN-Akt), we showed that PI3K and Akt act as the upstream regulators of JNKs and c-Jun in arsenite-mediated growth promotion. Overall, our data suggest a pathway of PI-3K/Akt/JNK/c-Jun/cylin D1 signaling in response to arsenite in human bronchial epithelial cells.
Collapse
Affiliation(s)
- Jin Ding
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Chien CW, Ho IC, Lee TC. Induction of neoplastic transformation by ectopic expression of human aldo-keto reductase 1C isoforms in NIH3T3 cells. Carcinogenesis 2009; 30:1813-20. [PMID: 19696165 DOI: 10.1093/carcin/bgp195] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have shown previously that chronic low-dose arsenic exposure induces malignant transformation of human skin keratinocyte HaCaT cells. In this study, we found that several isoforms of aldo-keto reductase 1C (AKR1C) were overexpressed in arsenic-exposed HaCaT cells. The AKR1C family of proteins are phase I drug-metabolizing enzymes involved in maintenance of steroid homeostasis, prostaglandin metabolism and metabolic activation of polycyclic aromatic hydrocarbons. To explore the oncogenic potential of AKR1C isoforms, we established mouse NIH3T3 cell lines ectopically and stably expressing human AKR1C1, AKR1C2 or AKR1C3. Our results showed that ectopic expression of human AKR1C1 and AKR1C2, but not AKR1C3, significantly enhanced foci formation. Following subcutaneous injection of these stable cell lines into nude mice, fibrosarcoma were formed from all three cell lines. However, the number and size of tumors formed by the AKR1C3-expressing cell line was fewer and smaller, respectively, than those formed by AKR1C1- and AKR1C2-expressing cells. Inhibitors of AKR1C, genistein and ursodeoxycholic acid, decreased foci formation in AKR1C1- and AKR1C2-expressing NIH3T3 cells in a dose-dependent manner, implying the association of enzymatic activity and oncogenic potential of AKR1C. The requirement of enzymatic ability for neoplastic transformation was confirmed by establishing a NIH3T3 cell line stably expressing a mutant AKR1C1 lacking enzymatic activity, which did not form foci in culture or tumors in nude mice. Our present study reveals that AKR1C enzymatic activity plays crucial roles on induction of neoplastic transformation of mouse NIH3T3 cells.
Collapse
Affiliation(s)
- Chia-Wen Chien
- Graduate Institute of Life Sciences, National Defense Medical Center, Taiwan, Republic of China
| | | | | |
Collapse
|
44
|
Hu N, Yu R, Shikuma C, Shiramizu B, Ostrwoski MA, Yu Q. Role of cell signaling in poxvirus-mediated foreign gene expression in mammalian cells. Vaccine 2009; 27:2994-3006. [PMID: 19428911 DOI: 10.1016/j.vaccine.2009.02.103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 02/15/2009] [Accepted: 02/24/2009] [Indexed: 10/21/2022]
Abstract
Poxviruses have been extensively used as a promising vehicle to efficiently deliver a variety of antigens in mammalian hosts to induce immune responses against infectious diseases and cancer. Using recombinant vaccinia virus (VV) and canarypox virus (ALVAC) expressing enhanced green fluorescent protein (EGFP) or multiple HIV-1 gene products, we studied the role of four cellular signaling pathways, the phosphoinositide-3-OH kinase (PI3K), extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (p38 MAPK), and c-Jun N-terminal kinase (JNK), in poxvirus-mediated foreign gene expression in mammalian cells. In nonpermissive infection (human monocytes), activation of PI3K, ERK, p38 MAPK, and JNK was observed in both VV and ALVAC and blocking PI3K, p38 MAKP, and JNK pathways with their specific inhibitors significantly reduced viral and vaccine antigen gene expression. Whereas, blocking the ERK pathway had no significant effect. Among these cellular signaling pathways studied, PI3K was the most critical pathway involved in gene expression by VV- or ALVAC-infected monocytes. The important role of PI3K in poxvirus-mediated gene expression was further confirmed in mouse epidermal cells stably transfected with dominant-negative PI3K mutant, as poxvirus-mediated targeted gene expression was significantly decreased in these cells when compared with their parental cells. Signaling pathway activation influenced gene expression at the mRNA level rather than virus binding. In permissive mammalian cells, however, VV DNA copies were also significantly decreased in the absence of normal function of the PI3K pathway. Poxvirus-triggered activation of PI3K pathway could be completely abolished by atazanavir, a new generation of antiretroviral protease inhibitors (PIs). As a consequence, ALVAC-mediated EGFP or HIV-1 gag gene expression in infected primary human monocytes was significantly reduced in the presence of atazanavir. These findings implicate that antiretroviral therapy (ART), also known as highly active antiretroviral therapy (HAART), may negatively impact the efficacy of live poxvirus vector-based vaccines and should be carefully considered when administering such live vaccines to individuals on ART.
Collapse
Affiliation(s)
- Ningjie Hu
- Hawaii AIDS Clinical Research Program, University of Hawaii at Manoa, Leahi Hospital, Honolulu, HI 96816, USA
| | | | | | | | | | | |
Collapse
|
45
|
Zhang D, Li J, Gao J, Huang C. c-Jun/AP-1 pathway-mediated cyclin D1 expression participates in low dose arsenite-induced transformation in mouse epidermal JB6 Cl41 cells. Toxicol Appl Pharmacol 2008; 235:18-24. [PMID: 19059425 DOI: 10.1016/j.taap.2008.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Revised: 11/03/2008] [Accepted: 11/10/2008] [Indexed: 01/31/2023]
Abstract
Arsenic is a well-documented human carcinogen associated with skin carcinogenesis. Our previous work reveals that arsenite exposure is able to induce cell transformation in mouse epidermal cell JB6 Cl41 through the activation of ERK, rather than JNK pathway. Our current studies further evaluate downstream pathway in low dose arsenite-induced cell transformation in JB6 Cl41 cells. Our results showed that treatment of cells with low dose arsenite induced activation of c-Jun/AP-1 pathway, and ectopic expression of dominant negative mutant of c-Jun (TAM67) blocked arsenite-induced transformation. Furthermore, our data indicated that cyclin D1 was an important downstream molecule involved in c-Jun/AP-1-mediated cell transformation upon low dose arsenite exposure, because inhibition of cyclin D1 expression by its specific siRNA in the JB6 Cl41 cells resulted in impairment of anchorage-independent growth of cells induced by low dose arsenite. Collectively, our results demonstrate that c-Jun/AP-1-mediated cyclin D1 expression is at least one of the key events implicated in cell transformation upon low dose arsenite exposure.
Collapse
Affiliation(s)
- Dongyun Zhang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | | | | | | |
Collapse
|