1
|
Li W, Li Z, Ma J, Xu X, Wang B, Long P, Jiang Q, You Y, Qu J, Wang Y, Wang Y, He M, Chen W, Yuan Y, Wu T. Circulating MicroRNAs in association with urinary arsenic: A community-based multi-center study in China. ENVIRONMENTAL RESEARCH 2025; 274:121354. [PMID: 40058551 DOI: 10.1016/j.envres.2025.121354] [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: 10/12/2024] [Revised: 03/03/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
The profile of plasma miRNAs in association with arsenic exposure remains largely unclear. We aim to identify plasma miRNAs assoicated with urinary arsenic using a two-stage design in Chinese population. The discovery group, Shimen panel, consists of 19 high vs. low arsenic-exposed pairs selected from 1095 residents in an arsenic-contaminated area. The validation group, Wuhan-Zhuhai panel, consists of 53 community-dwelling participants with moderate arsenic exposure. Plasma miRNAs were measured by microarray in the Shimen panel and by sequencing in the Wuhan-Zhuhai panel. Arsenic levels in urine and plasma were quantified using inductively coupled plasma mass spectrometry. During the discovery stage, 16 miRNAs were found to be differentially expressed between high and low urinary arsenic groups in the Shimen panel (fold change >2, P < 0.05). Seven miRNAs (miR-101-3p, miR-142-3p, miR-148a-3p, miR-15a-5p, miR-199a-3p, miR-27b-3p, and miR-340-5p) were validated to have a positive association with log-transformed urinary arsenic levels in the Wuhan-Zhuhai panel (P < 0.05). Furthermore, five of the seven miRNAs were also associated with arsenic in plasma. The identified miRNAs were primarily associated with cancer-related pathways. These identified miRNAs would serve as crucial biomarkers for arsenic exposure, elucidating the epigenetic mechanisms underlying arsenic-induced toxicity and carcinogenesis.
Collapse
Affiliation(s)
- Wending Li
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Zhaoyang Li
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jixuan Ma
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuedan Xu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bin Wang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pinpin Long
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qin Jiang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yutong You
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingli Qu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yufei Wang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaxin Wang
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meian He
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Yuan
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Tangchun Wu
- Department of Occupational and Environmental Health, Key Laboratory of Environment and Health, Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Zhang Q, Guo P, Zhou X, Yang C, Lu Z, Wu S, Lu X, Yang J, Jin C. Nanoplastic PS and cadmium co-exposure accelerates ferroptosis mediated by HIF-1α-related signaling in spermatogonium. Free Radic Biol Med 2025; 237:11-20. [PMID: 40412570 DOI: 10.1016/j.freeradbiomed.2025.05.402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 05/05/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Microplastics (MPs) in the real environment media often adsorbed toxic substances such as heavy metals and this co-exposure may exert various negative effects on human reproduction. In particular, the adsorption capacity of nanoplastics (NPs) is stronger. However, there are few studies focused on the combined toxicity of NPs and heavy metal co-exposure to germ cells. In this study, mouse spermatogonium GC-1 cells were applied as in vitro model to observe the effects of PS-NPs (0.1 μm, 200 mg/L) and cadmium (Cd) chloride (5 μM) on mouse spermatogonia and the underlying mechanism. The results showed that GC-1 cells turned significantly abnormal in morphology, and cell number and survival rate reduced in the co-exposed group. These injuries were restored after the intervention of ferroptosis inhibitor (Fer-1, 1 μM), HIF-1α specific inhibitor (YC-1, 10 μM) and miR-199a-5p mimic (50 nM). Double luciferase gene reporting experiment showed that there existed binding sites of miR-199a-5p to HIF-1α mRNA. Collectively, we found that PS-NPs + Cd co-exposure induced GC-1 cell ferroptosis, which might be mediated by miR-199a-5p/HIF-1α signaling axis. Hopefully, these findings will shed new light on how PS-NPs may impair male reproductive function in real scenario.
Collapse
Affiliation(s)
- Qingpeng Zhang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China; Leshan Center for Disease Control and Prevention, Leshan, Sichuan, 614000, PR China
| | - Pan Guo
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Xingyue Zhou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Chengying Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Ziwei Lu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Shengwen Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Xiaobo Lu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Jinghua Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China
| | - Cuihong Jin
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Shenyang, 110122, PR China; Department of Toxicology, School of Public Health, China Medical University, Shenyang, 110122, PR China.
| |
Collapse
|
3
|
Wen J, Li A, Wang Z, Guo X, Zhang G, Litzow MR, Liu Q. Hepatotoxicity induced by arsenic trioxide: clinical features, mechanisms, preventive and potential therapeutic strategies. Front Pharmacol 2025; 16:1536388. [PMID: 40051569 PMCID: PMC11882591 DOI: 10.3389/fphar.2025.1536388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
Arsenic trioxide (ATO) has shown substantial efficacy in the treatment of patients with acute promyelocytic leukemia, and the utilization of ATO as a potential treatment for other tumors is currently being investigated; thus, its clinical application is becoming more widespread. However, the toxicity of ATO has prevented many patients from receiving this highly beneficial treatment. The clinical features, mechanisms, and preventive measures for ATO hepatotoxicity, as well as potential curative strategies, are discussed in this review. This review not only discusses existing drugs for the treatment of hepatotoxicity but also focuses on potential future therapeutic agents, providing forward-looking guidance for the clinical use of small molecule extracts, trace elements, antidiabetic drugs, and vitamins.
Collapse
Affiliation(s)
- Jun Wen
- Department of Haematology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Aiwen Li
- Department of Haematology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Ziliang Wang
- Department of Haematology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Guo
- Department of Haematology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Gaoling Zhang
- Center of Hematology, Peking University People’s Hospital Qingdao, Qingdao, China
| | - Mark R. Litzow
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Qiuju Liu
- Department of Haematology, Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
He T, Xiong L, Lin K, Yi J, Duan C, Zhang J. Functional metabolomics reveals arsenic-induced inhibition of linoleic acid metabolism in mice kidney in drinking water. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 349:123949. [PMID: 38636836 DOI: 10.1016/j.envpol.2024.123949] [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: 12/12/2023] [Revised: 01/27/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
Arsenic (As) is a heavy metal known for its detrimental effects on the kidneys, but the precise mechanisms underlying its toxicity remain unclear. In this study, we employed an integrated approach combining traditional toxicology methods with functional metabolomics to explore the nephrotoxicity induced by As in mice. Our findings demonstrated that after 28 days of exposure to sodium arsenite, blood urea nitrogen, serum creatinine levels were significantly increased, and pathological examination of the kidneys revealed dilation of renal tubules and glomerular injury. Additionally, uric acid, total cholesterol, and low-density lipoprotein cholesterol levels were significant increased while triglyceride level was decreased, resulting in renal insufficiency and lipid disorders. Subsequently, the kidney metabolomics analysis revealed that As exposure disrupted 24 differential metabolites, including 14 up-regulated and 10 down-regulated differential metabolites. Ten metabolic pathways including linoleic acid and glycerophospholipid metabolism were significantly enriched. Then, 80 metabolic targets and 168 predicted targets were identified using metabolite network pharmacology analysis. Of particular importance, potential toxicity targets, such as glycine amidinotransferase, mitochondrial (GATM), and nitric oxide synthase, and endothelial (NOS3), were prioritized through the "metabolite-target-pathway" network. Receiver operating characteristics curve and molecular docking analyses suggested that 1-palmitoyl-2-myristoyl-sn-glycero-3-PC, linoleic acid, and L-hydroxyarginine might be functional metabolites associated with GATM and NOS3. Moreover, targeted verification result showed that the level of linoleic acid in As group was 0.4951 μg/mL, which was significantly decreased compared with the control group. And in vivo and in vitro protein expression experiments confirmed that As exposure inhibited the expression of GATM and NOS3. In conclusion, these results suggest that As-induced renal injury may be associated with the inhibition of linoleic acid metabolism through the down-regulation of GATM and NOS3, resulting in decreased levels of linoleic acid, 1-palmitoyl-2-myristoyl-sn-glycero-3-PC, and L-hydroxyarginine metabolites.
Collapse
Affiliation(s)
- Tianmu He
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, China; School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China.
| | - Lijuan Xiong
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi, 563000, China
| | - Kexin Lin
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, China
| | - Jing Yi
- School of Basic Medicine, Zunyi Medical University, Zunyi, 563000, China
| | - Cancan Duan
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi, 563000, China.
| | - Jianyong Zhang
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
5
|
Bixin H, Yuling Z, Ying M, Jinming C, Zhang Z. Regulation of Osteosarcoma Cell Proliferation, Migration, and Invasion by miR-143 and miR-199a Through COX-2 Targeting. Dose Response 2024; 22:15593258241264947. [PMID: 38912334 PMCID: PMC11193354 DOI: 10.1177/15593258241264947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Objective To investigate the biological role of miR-143 and miR-199a in mediating the progression of osteosarcoma (OS) by targeting cyclooxygenase (COX-2). Introduction COX-2 plays a crucial role in the development and progression of OS. However, the specific regulatory mechanisms of COX-2 in OS are still not well understood. Methods The expression levels of COX-2, miR-143 and miR-199a in OS tissues were detected using immunohistochemistry, qPCR, or western blot assays. The targeting relationship between miRNAs and COX-2 was determined. The effect of miRNA and COX-2 on OS cells was evaluated in vitro and in vivo. Results COX-2 expression was upregulated while miR-143 and miR-199a were downregulated in OS tissues. miR-143 and miR-199a suppressed the proliferation, migration, and invasion of OS cells. The dual-luciferase reporter gene assay showed that COX-2 was a direct target of miR-143 and miR-199a. Genetic knockdown of COX-2 significantly suppressed cell proliferation, induced apoptosis, and inhibited migration and invasion of OS cells. The expression levels of COX-2 and PGE2 were decreased after the overexpression of miR-143 and miR-199a. Additionally, COX-2 silencing inhibited the tumorigenesis of OS and the synthesis of PGE2 in vivo. Conclusions miR-143 and miR-199a/COX-2 axis modulates the proliferation, invasion, and migration in osteosarcoma.
Collapse
Affiliation(s)
- Huang Bixin
- Department of Anesthesiology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| | - Zheng Yuling
- Department of Anesthesiology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| | - Mai Ying
- Department of Anesthesiology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| | - Chen Jinming
- Department of Anesthesiology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| | - Zhongqi Zhang
- Department of Anesthesiology, The Affiliated Shunde Hospital of Jinan University, Foshan, China
| |
Collapse
|
6
|
Ghafouri-Fard S, Shoorei H, Dabiri Oskuei S, Hussen BM, Rasool Abdullah S, Taheri M, Jamali E. The interaction between miRNAs and hazardous materials. Noncoding RNA Res 2023; 8:507-519. [PMID: 37497124 PMCID: PMC10365984 DOI: 10.1016/j.ncrna.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Toxic agents are broadly present in the environment, households, and workplaces. Contamination of food and drinking water with these agents results in entry of these materials to the body. The crosstalk between these agents and microRNAs (miRNAs) affects pathoetiology of several disorders. These agents can influence the redox status, release of inflammatory cytokines and mitochondrial function. Altered expression of miRNA is involved in the dysregulation of several pathophysiological conditions and signaling pathways. These molecules are also implicated in the adaption to environmental stimuli. Thus, the interactions between miRNAs and toxic materials might participate in the hazardous effects of these materials in the body. This review describes the effects of the toxic materials on miRNAs and the consequences of these interactions on the human health.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Dabiri Oskuei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bashdar Mahmud Hussen
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Kurdistan Region, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Urology and Nephrology Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elena Jamali
- Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Sun J, Wu L, Wu M, Liu Q, Cao H. Non-coding RNA therapeutics: Towards a new candidate for arsenic-induced liver disease. Chem Biol Interact 2023; 382:110626. [PMID: 37442288 DOI: 10.1016/j.cbi.2023.110626] [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: 03/26/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Arsenic, a metalloid toxicant, has caused serious environmental pollution and is presently a global health issue. Long-term exposure to arsenic causes diverse organ and system dysfunctions, including liver disease. Arsenic-induced liver disease comprises a spectrum of liver pathologies, ranging from hepatocyte damage, steatosis, fibrosis, to hepatocellular carcinoma. Various mechanisms, including an imbalance in redox reactions, mitochondrial dysfunction and epigenetic changes, participate in the pathogenesis of arsenic-induced liver disease. Altered epigenetic processes involved in its initiation and progression. Dysregulated modulations of non-coding RNAs (ncRNAs), including miRNAs, lncRNAs and circRNAs, exert regulating effects on these processes. Here, we have reviewed the underlying pathogenic mechanisms that lead to progressive arsenic-induced liver disease, and we provide a discussion focusing on the effects of ncRNAs on dysfunctions in intercellular communication and on the activation of hepatic stellate cells and malignant transformation of hepatocytes. Further, we have discussed the roles of ncRNAs in intercellular communication via extracellular vesicles and cytokines, and have provided a perspective for the application of ncRNAs as biomarkers in the early diagnosis and evaluation of the pathogenesis of arsenic-induced liver disease. Further investigations of ncRNAs will help us to understand the nature of arsenic-induced liver disease and to identify biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jing Sun
- Department of Nutrition, Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
| | - Lu Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Meng Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| | - Hong Cao
- Department of Nutrition, Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
| |
Collapse
|
8
|
Yuan X, Ma C, Li J, Li J, Yu R, Cai F, Qu G, Yu B, Liu L, Zeng D, Jiao Q, Liao Q, Lv X. Indirect bilirubin impairs invasion of osteosarcoma cells via inhibiting the PI3K/AKT/MMP-2 signaling pathway by suppressing intracellular ROS. J Bone Oncol 2023; 39:100472. [PMID: 36876225 PMCID: PMC9982672 DOI: 10.1016/j.jbo.2023.100472] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Background Osteosarcoma is most prevalently found primary malignant bone tumors, with primary metastatic patients accounting for approximately 25% of all osteosarcoma patients, yet their 5-year OS remains below 30%. Bilirubin plays a key role in oxidative stress-associated events, including malignancies, making the regulation of its serum levels a potential anti-tumor strategy. Herein, we investigated the association of osteosarcoma prognosis with serum levels of TBIL, IBIL and DBIL, and further explored the mechanisms by which bilirubin affects tumor invasion and migration. Methods ROC curve was plotted to assess survival conditions based on the determined optimal cut-off values and the AUC. Then, Kaplan-Meier curves, along with Cox proportional hazards model, was applied for survival analysis. Inhibitory function of IBIL on the malignant properties of osteosarcoma cells was examined using the qRT-PCR, transwell assays, western blotting, and flow cytometry. Results We found that, versus osteosarcoma patients with pre-operative higher IBIL (>8.9 μmol/L), those with low IBIL (≤8.9 μmol/L) had shorter OS and PFS. As indicated by the Cox proportional hazards model, pre-operative IBIL functioned as an independent prognostic factor for OS and PFS in total and gender-stratified osteosarcoma patients (P < 0.05 for all). In vitro experiments further confirmed that IBIL inhibits PI3K/AKT phosphorylation and downregulates MMP-2 expression via reducing intracellular ROS, thereby decreasing the invasion of osteosarcoma cells. Conclusions IBIL may serve as an independent prognostic predictor for osteosarcoma patients. IBIL impairs invasion of osteosarcoma cells through repressing the PI3K/AKT/MMP-2 pathway by suppressing intracellular ROS, thus inhibiting its metastatic potential.
Collapse
Key Words
- AUC, area under curve
- BRNP, PEGylated bilirubin nanoparticles
- CCK-8, cell counting kit-8
- CI, confidence interval
- DBIL, direct bilirubin
- DMSO, dimethyl sulfoxide
- ECM, extracellular matrix
- H2O2, hydrogen peroxide
- HIF-1α, hypoxia inducible factor-1α
- HR, hazard ratio
- IBIL
- IBIL, indirect bilirubin
- Invasion
- MDA, malondialdehyde
- MMP, matrix metalloproteinase
- OS, overall survival
- Osteosarcoma
- PFS, progression-free survival
- PI3K/AKT/MMP-2
- PVDF, polyvinylidene fluoride
- Prognosis
- ROC, receiver operative characteristic
- ROS, reactive oxygen species
- SD, standard deviation
- SOD, superoxide dismutase
- TBIL, total bilirubin
- TIMP, tissue inhibitor of matrix metalloproteinase
- VEGF, vascular endothelial growth factor
- qRT-PCR, real-time quantitative PCR
Collapse
Affiliation(s)
- Xuhui Yuan
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China
| | - Cong Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Jiayu Li
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China
| | - Junhong Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ronghui Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Feng Cai
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Gaoyang Qu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Bo Yu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lang Liu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Duo Zeng
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - QuanHui Jiao
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qi Liao
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China
| | - Xiaobin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330008, China
| |
Collapse
|
9
|
Aschner M, Skalny AV, Lu R, Santamaria A, Zhou JC, Ke T, Karganov MY, Tsatsakis A, Golokhvast KS, Bowman AB, Tinkov AA. The role of hypoxia-inducible factor 1 alpha (HIF-1α) modulation in heavy metal toxicity. Arch Toxicol 2023; 97:1299-1318. [PMID: 36933023 DOI: 10.1007/s00204-023-03483-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/02/2023] [Indexed: 03/19/2023]
Abstract
Hypoxia-inducible factor 1 (HIF-1) is an oxygen-sensing transcriptional regulator orchestrating a complex of adaptive cellular responses to hypoxia. Several studies have demonstrated that toxic metal exposure may also modulate HIF-1α signal transduction pathway, although the existing data are scarce. Therefore, the present review aims to summarize the existing data on the effects of toxic metals on HIF-1 signaling and the potential underlying mechanisms with a special focus on prooxidant effect of the metals. The particular effect of metals was shown to be dependent on cell type, varying from down- to up-regulation of HIF-1 pathway. Inhibition of HIF-1 signaling may contribute to impaired hypoxic tolerance and adaptation, thus promoting hypoxic damage in the cells. In contrast, its metal-induced activation may result in increased tolerance to hypoxia through increased angiogenesis, thus promoting tumor growth and contributing to carcinogenic effect of heavy metals. Up-regulation of HIF-1 signaling is mainly observed upon Cr, As, and Ni exposure, whereas Cd and Hg may both stimulate and inhibit HIF-1 pathway. The mechanisms underlying the influence of toxic metal exposure on HIF-1 signaling involve modulation of prolyl hydroxylases (PHD2) activity, as well as interference with other tightly related pathways including Nrf2, PI3K/Akt, NF-κB, and MAPK signaling. These effects are at least partially mediated by metal-induced ROS generation. Hypothetically, maintenance of adequate HIF-1 signaling upon toxic metal exposure through direct (PHD2 modulation) or indirect (antioxidant) mechanisms may provide an additional strategy for prevention of adverse effects of metal toxicity.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Anatoly V Skalny
- IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico
| | - Ji-Chang Zhou
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, 518100, China
| | - Tao Ke
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | | - Aristides Tsatsakis
- IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia.,Laboratory of Toxicology, Medical School, University of Crete, Voutes, 700 13, Heraklion, Crete, Greece
| | - Kirill S Golokhvast
- Siberian Federal Scientific Centre of Agrobiotechnologies of the Russian Academy of Sciences, Krasnoobsk, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, USA
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia. .,Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003, Yaroslavl, Russia.
| |
Collapse
|
10
|
Li X, Ma TK, Wang M, Zhang XD, Liu TY, Liu Y, Huang ZH, Zhu YH, Zhang S, Yin L, Xu YY, Ding H, Liu C, Shi H, Fan QL. YY1-induced upregulation of LncRNA-ARAP1-AS2 and ARAP1 promotes diabetic kidney fibrosis via aberrant glycolysis associated with EGFR/PKM2/HIF-1α pathway. Front Pharmacol 2023; 14:1069348. [PMID: 36874012 PMCID: PMC9974832 DOI: 10.3389/fphar.2023.1069348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Objectives: Dimeric pyruvate kinase (PK) M2 (PKM2) plays an important role in promoting the accumulation of hypoxia-inducible factor (HIF)-1α, mediating aberrant glycolysis and inducing fibrosis in diabetic kidney disease (DKD). The aim of this work was to dissect a novel regulatory mechanism of Yin and Yang 1 (YY1) on lncRNA-ARAP1-AS2/ARAP1 to regulate EGFR/PKM2/HIF-1α pathway and glycolysis in DKD. Materials and methods: We used adeno-associated virus (AAV)-ARAP1 shRNA to knocked down ARAP1 in diabetic mice and overexpressed or knocked down YY1, ARAP1-AS2 and ARAP1 expression in human glomerular mesangial cells. Gene levels were assessed by Western blotting, RT-qPCR, immunofluorescence staining and immunohistochemistry. Molecular interactions were determined by RNA pull-down, co-immunoprecipitation, ubiquitination assay and dual-luciferase reporter analysis. Results: YY1, ARAP1-AS2, ARAP1, HIF-1α, glycolysis and fibrosis genes expressions were upregulated and ARAP1 knockdown could inhibit dimeric PKM2 expression and partly restore tetrameric PKM2 formation, while downregulate HIF-1α accumulation and aberrant glycolysis and fibrosis in in-vivo and in-vitro DKD models. ARAP1 knockdown attenuates renal injury and renal dysfunction in diabetic mice. ARAP1 maintains EGFR overactivation in-vivo and in-vitro DKD models. Mechanistically, YY1 transcriptionally upregulates ARAP1-AS2 and indirectly regulates ARAP1 and subsequently promotes EGFR activation, HIF-1α accumulation and aberrant glycolysis and fibrosis. Conclusion: Our results first highlight the role of the novel regulatory mechanism of YY1 on ARAP1-AS2 and ARAP1 in promoting aberrant glycolysis and fibrosis by EGFR/PKM2/HIF-1α pathway in DKD and provide potential therapeutic strategies for DKD treatments.
Collapse
Affiliation(s)
- Xin Li
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Tian-Kui Ma
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Min Wang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Xiao-Dan Zhang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Tian-Yan Liu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Yue Liu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Zhao-Hui Huang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Yong-Hong Zhu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Shuang Zhang
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Li Yin
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Yan-Yan Xu
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Hong Ding
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Cong Liu
- Department of General Surgery, First Hospital of Harbin Medical University, Harbin, China
| | - Hang Shi
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiu-Ling Fan
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Islam R, Zhao L, Zhang X, Liu LZ. MiR-218-5p/EGFR Signaling in Arsenic-Induced Carcinogenesis. Cancers (Basel) 2023; 15:1204. [PMID: 36831545 PMCID: PMC9954652 DOI: 10.3390/cancers15041204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Arsenic is a well-known carcinogen inducing lung, skin, bladder, and liver cancer. Abnormal epidermal growth factor receptor (EGFR) expression is common in lung cancer; it is involved in cancer initiation, development, metastasis, and treatment resistance. However, the underlying mechanism for arsenic-inducing EGFR upregulation remains unclear. METHODS RT-PCR and immunoblotting assays were used to detect the levels of miR-218-5p and EGFR expression. The Luciferase assay was used to test the transcriptional activity of EGFR mediated by miR-218-5p. Cell proliferation, colony formation, wound healing, migration assays, tube formation assays, and tumor growth assays were used to study the function of miR-218-5p/EGFR signaling. RESULTS EGFR and miR-218-5p were dramatically upregulated and downregulated in arsenic-induced transformed (As-T) cells, respectively. MiR-218-5p acted as a tumor suppressor to inhibit cell proliferation, migration, colony formation, tube formation, tumor growth, and angiogenesis. Furthermore, miR-218-5p directly targeted EGFR by binding to its 3'-untranslated region (UTR). Finally, miR-218-5p exerted its antitumor effect by inhibiting its direct target, EGFR. CONCLUSION Our study highlights the vital role of the miR-218-5p/EGFR signaling pathway in arsenic-induced carcinogenesis and angiogenesis, which may be helpful for the treatment of lung cancer induced by chronic arsenic exposure in the future.
Collapse
Affiliation(s)
| | | | | | - Ling-Zhi Liu
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
12
|
Arsenic trioxide promotes ERK1/2-mediated phosphorylation and degradation of BIM EL to attenuate apoptosis in BEAS-2B cells. Chem Biol Interact 2023; 369:110304. [PMID: 36509116 DOI: 10.1016/j.cbi.2022.110304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Inorganic arsenic is highly toxic, widely distributed in the human environment and may result in multisystem diseases and several types of cancers. The BCL-2-interacting mediator of cell death protein (BIM) is a key modulator of the intrinsic apoptosis pathway. Interestingly, in the present study, we found that arsenic trioxide (As2O3) decreased BIMEL levels in human bronchial epithelial cell line BEAS-2B and increased BIMEL levels in human lung carcinoma cell line A549 and mouse Sertoli cell line TM4. Mechanismly, the 26S proteasome inhibitors MG132 and bortezomib could effectively inhibit BIMEL degradation induced by As2O3 in BEAS-2B cells. As2O3 activated extracellular signal-regulated kinase (ERK) 1/2, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) signaling pathways, but only the ERK1/2 MAPK inhibitor PD98059 blocked BIMEL degradation induced by As2O3. Furthermore, As2O3 induced-phosphorylation of BIMEL at multiple sites was inhibited by ERK1/2 MAPK inhibitor PD98059. Inhibition of As2O3-induced ERK1/2 MAPK phosphorylation increased the levels of BIMEL and cleaved-caspase-3 proteins and decreased BEAS-2B cell viability. As2O3 also markedly mitigated tunicamycin-induced apoptosis of BEAS-2B cells by increasing ERK1/2 phosphorylation and BIMEL degradation. Our results suggest that As2O3-induced activation of the ERK1/2 MAPK pathway increases phosphorylation of BIMEL and promotes BIMEL degradation, thereby alleviating the role of apoptosis in As2O3-induced cell death. This study provides new insights into how to maintain the survival of BEAS-2B cells before malignant transformation induced by high doses of As2O3.
Collapse
|
13
|
Nail AN, Ferragut Cardoso AP, Montero LK, States JC. miRNAs and arsenic-induced carcinogenesis. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 96:203-240. [PMID: 36858773 PMCID: PMC10184182 DOI: 10.1016/bs.apha.2022.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Arsenic-induced carcinogenesis is a worldwide health problem. Identifying the molecular mechanisms responsible for the induction of arsenic-induced cancers is important for developing treatment strategies. MicroRNA (miRNA) dysregulation is known to affect development and progression of human cancer. Several studies have identified an association between altered miRNA expression in cancers from individuals chronically exposed to arsenic and in cell models for arsenic-induced carcinogenesis. This chapter provides a comprehensive review for miRNA dysregulation in arsenic-induced cancer.
Collapse
Affiliation(s)
- Alexandra N Nail
- Department of Pharmacology and Toxicology, Center for Integrative Environmental Health Science, University of Louisville, Louisville, KY, United States
| | - Ana P Ferragut Cardoso
- Department of Pharmacology and Toxicology, Center for Integrative Environmental Health Science, University of Louisville, Louisville, KY, United States
| | - Lakyn K Montero
- Department of Pharmacology and Toxicology, Center for Integrative Environmental Health Science, University of Louisville, Louisville, KY, United States
| | - J Christopher States
- Department of Pharmacology and Toxicology, Center for Integrative Environmental Health Science, University of Louisville, Louisville, KY, United States.
| |
Collapse
|
14
|
Islam R, Zhao L, Wang Y, Lu-Yao G, Liu LZ. Epigenetic Dysregulations in Arsenic-Induced Carcinogenesis. Cancers (Basel) 2022; 14:4502. [PMID: 36139662 PMCID: PMC9496897 DOI: 10.3390/cancers14184502] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Arsenic is a crucial environmental metalloid whose high toxicity levels negatively impact human health. It poses significant health concerns to millions of people in developed and developing countries such as the USA, Canada, Bangladesh, India, China, and Mexico by enhancing sensitivity to various types of diseases, including cancers. However, how arsenic causes changes in gene expression that results in heinous conditions remains elusive. One of the proposed essential mechanisms that still has seen limited research with regard to causing disease upon arsenic exposure is the dysregulation of epigenetic components. In this review, we have extensively summarized current discoveries in arsenic-induced epigenetic modifications in carcinogenesis and angiogenesis. Importantly, we highlight the possible mechanisms underlying epigenetic reprogramming through arsenic exposure that cause changes in cell signaling and dysfunctions of different epigenetic elements.
Collapse
Affiliation(s)
| | | | | | | | - Ling-Zhi Liu
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
15
|
Li X, Chen S, Wang X, Zhang R, Yang J, Xu H, He W, Lai M, Wu S, Nan A. The pivotal regulatory factor circBRWD1 inhibits arsenic exposure-induced lung cancer occurrence by binding mRNA and regulating its stability. Mol Ther Oncolytics 2022; 26:399-412. [PMID: 36159776 PMCID: PMC9463561 DOI: 10.1016/j.omto.2022.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 08/18/2022] [Indexed: 11/28/2022] Open
Abstract
Multiple studies have indicated that circular RNAs (circRNAs) play a regulatory role in different stages of tumors by interacting with various molecules. With continuous in-depth research on the biological functions of circRNAs, increasing evidence has shown that circRNAs play important roles in carcinogenesis caused by environmental pollutants. However, the function and mechanism of circRNAs in arsenic exposure-induced lung cancer occurrence have not been reported. In this study, RNA sequencing and qPCR assays revealed that the expression of circBRWD1 was decreased in BEAS-2B-As cells and multiple lung cancer cell lines. Silencing circBRWD1 promoted cell viability and proliferation, inhibited cell apoptosis, and accelerated the G0/G1 phase transition in BEAS-2B-As cells; however, these functions were abrogated by circBRWD1 overexpression. Mechanistically, under arsenic exposure, expression of decreased circBRWD1 led to enhanced stability of the mRNA to which it directly binds (c-JUN, c-MYC, and CDK6 mRNA), increasing its expression. This mechanism promotes the malignant transformation of lung cells and ultimately leads to lung cancer. Our findings thus reveal the molecular mechanism of arsenic carcinogenesis.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning 530021, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China.,Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, China
| | - Sixian Chen
- Department of Toxicology, School of Public Health, Guangxi Medical University, Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning 530021, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China.,Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, China
| | - Xin Wang
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Ruirui Zhang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning 530021, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China.,Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, China
| | - Jialei Yang
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Haotian Xu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning 530021, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China.,Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, China
| | - Wanting He
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Mingshuang Lai
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China
| | - Shuilian Wu
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Aruo Nan
- Department of Toxicology, School of Public Health, Guangxi Medical University, Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning 530021, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, China.,Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, China.,Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
16
|
Meng W, Li Y, Chai B, Liu X, Ma Z. miR-199a: A Tumor Suppressor with Noncoding RNA Network and Therapeutic Candidate in Lung Cancer. Int J Mol Sci 2022; 23:8518. [PMID: 35955652 PMCID: PMC9369015 DOI: 10.3390/ijms23158518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/12/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Lung cancer is the leading cause of cancer death worldwide. miR-199a, which has two mature molecules: miR-199a-3p and miR-199a-5p, plays an important biological role in the genesis and development of tumors. We collected recent research results on lung cancer and miR-199a from Google Scholar and PubMed databases. The biological functions of miR-199a in lung cancer are reviewed in detail, and its potential roles in lung cancer diagnosis and treatment are discussed. With miR-199a as the core point and a divergence outward, the interplay between miR-199a and other ncRNAs is reviewed, and a regulatory network covering various cancers is depicted, which can help us to better understand the mechanism of cancer occurrence and provide a means for developing novel therapeutic strategies. In addition, the current methods of diagnosis and treatment of lung cancer are reviewed. Finally, a conclusion was drawn: miR-199a inhibits the development of lung cancer, especially by inhibiting the proliferation, infiltration, and migration of lung cancer cells, inhibiting tumor angiogenesis, increasing the apoptosis of lung cancer cells, and affecting the drug resistance of lung cancer cells. This review aims to provide new insights into lung cancer therapy and prevention.
Collapse
Affiliation(s)
| | | | | | | | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer, School of Life Science, Shanghai University, Shanghai 200444, China; (W.M.); (Y.L.); (B.C.); (X.L.)
| |
Collapse
|
17
|
Molecular mechanisms of reactive oxygen species in regulated cell deaths: Impact of ferroptosis in cancer therapy. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
18
|
Zhao L, Wang YF, Liu J, Jiang BH, Liu LZ. Human endothelial cells promote arsenic-transformed lung epithelial cells to induce tumor growth and angiogenesis through interleukin-8 induction. Aging (Albany NY) 2022; 14:2113-2130. [PMID: 35241635 PMCID: PMC8954972 DOI: 10.18632/aging.203930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/29/2022] [Indexed: 11/25/2022]
Abstract
Arsenic exposure is associated with lung cancer. Angiogenesis is essential for tumor development. However, the role and mechanism of human vascular endothelial cells in tumor growth and angiogenesis induced by arsenic-transformed bronchial epithelial (As-T) cells remain to be elucidated. In this study, we found that endothelial cells significantly increased As-T cell-induced tumor growth compared to those induced by As-T cells alone. To understand the molecular mechanism, we found that endothelial cells co-cultured with As-T cells or cultured in conditioned medium (CM) prepared from As-T cells showed much higher cell migration, proliferation, and tube formation compared to those co-cultured with BEAS-2B (B2B) cells or cultured in CM from B2B. We identified that higher levels of intracellular interleukin 8 (IL-8) were secreted by As-T cells, which activated IL-8/IL-8R signaling to promote endothelial cells migration and tube formation. IL-8 silencing and knockout (KO) in As-T cells, or IL-8 neutralizing antibody dramatically suppressed endothelial cell proliferation, migration, tube formation in vitro, and tumor growth and angiogenesis in vivo, suggesting a key role of IL-8 in As-T cells to induce angiogenesis via a paracrine effect. Finally, blocking of IL-8 receptors C-X-C chemokine receptor type 1 (CXCR1) and CXCR2 with neutralizing antibodies and chemical inhibitors inhibited tube formation, indicating that IL-8Rs on endothelial cells are necessary for As-T cell-induced angiogenesis. Overall, this study reveals an important molecular mechanism of arsenic-induced carcinogenesis, and suggests a new option to prevent and treat arsenic-induced angiogenesis.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yi-Fang Wang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jie Liu
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ling-Zhi Liu
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
19
|
Chakraborty A, Ghosh S, Biswas B, Pramanik S, Nriagu J, Bhowmick S. Epigenetic modifications from arsenic exposure: A comprehensive review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 810:151218. [PMID: 34717984 DOI: 10.1016/j.scitotenv.2021.151218] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 06/13/2023]
Abstract
Arsenic is a notorious element with the potential to harm exposed individuals in ways that include cancerous and non-cancerous health complications. Millions of people across the globe (especially in South and Southeast Asian countries including China, Vietnam, India and Bangladesh) are currently being unknowingly exposed to precarious levels of arsenic. Among the diverse effects associated with such arsenic levels of exposure is the propensity to alter the epigenome. Although a large volume of literature exists on arsenic-induced genotoxicity, cytotoxicity, and inter-individual susceptibility due to active research on these subject areas from the last millennial, it is only recently that attention has turned on the ramifications and mechanisms of arsenic-induced epigenetic changes. The present review summarizes the possible mechanisms involved in arsenic induced epigenetic alterations. It focuses on the mechanisms underlying epigenome reprogramming from arsenic exposure that result in improper cell signaling and dysfunction of various epigenetic components. The mechanistic information articulated from the review is used to propose a number of novel therapeutic strategies with a potential for ameliorating the burden of worldwide arsenic poisoning.
Collapse
Affiliation(s)
- Arijit Chakraborty
- Kolkata Zonal Center, CSIR-National Environmental Engineering Research Institute (NEERI), Kolkata, West Bengal 700107, India
| | - Soma Ghosh
- Kolkata Zonal Center, CSIR-National Environmental Engineering Research Institute (NEERI), Kolkata, West Bengal 700107, India
| | - Bratisha Biswas
- Kolkata Zonal Center, CSIR-National Environmental Engineering Research Institute (NEERI), Kolkata, West Bengal 700107, India
| | - Sreemanta Pramanik
- Kolkata Zonal Center, CSIR-National Environmental Engineering Research Institute (NEERI), Kolkata, West Bengal 700107, India
| | - Jerome Nriagu
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, 109 Observatory Street, Ann Arbor, MI 48109-2029, USA
| | - Subhamoy Bhowmick
- Kolkata Zonal Center, CSIR-National Environmental Engineering Research Institute (NEERI), Kolkata, West Bengal 700107, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
20
|
Wang L, Bayanbold K, Zhao L, Wang Y, Adamcakova-Dodd A, Thorne PS, Yang H, Jiang BH, Liu LZ. Redox sensitive miR-27a/b/Nrf2 signaling in Cr(VI)-induced carcinogenesis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 809:151118. [PMID: 34718002 PMCID: PMC9387726 DOI: 10.1016/j.scitotenv.2021.151118] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/14/2021] [Accepted: 10/17/2021] [Indexed: 05/27/2023]
Abstract
Hexavalent chromium [Cr(VI)] is a well-known carcinogen that can cause several types of cancer including lung cancer. NF-E2-related factor 2 (Nrf2), the redox sensitive transcription factor, can protect normal cells from a variety of toxicants and carcinogens by inducing the expression of cellular protective genes and maintaining redox balance. However, Nrf2 also protects cancer cells from radio- and chemo-therapies and facilitates cancer progression. Although Cr(VI) treatment has been demonstrated to upregulate Nrf2 expression, the mechanisms for Nrf2 regulation upon chronic Cr(VI) exposure remain to be elucidated. We found that Nrf2 was upregulated in BEAS-2B cells exposed to Cr(VI) from 1 to 5 months, and also in Cr(VI)-induced transformed (Cr-T) cells with Cr(VI) treatment for 6 months. We showed that KEAP1, the classic negative regulator of Nrf2, was downregulated after Cr(VI) exposure for 4 months, suggesting that Nrf2 induction by Cr(VI) treatment is through KEAP1 decrease at late stage. To further decipher the mechanisms of Nrf2 upregulation at early stage of Cr(VI) exposure, we demonstrated that miR-27a and miR-27b were redox sensitive miRNAs, since reactive oxygen species (ROS) scavengers induced miR-27a/b expression. After Cr(VI) exposure for 1 month, the expression levels of miR-27a/b was dramatically decreased. The changes of miR-27a/b and their target Nrf2 were confirmed in vivo by mouse model intranasally exposed to Cr(VI) for 12 weeks. Nrf2 was a direct target of miR-27a/b, which acted as tumor suppressors in vitro and in vivo to inhibit tumorigenesis and cancer development of Cr-T cells. The results suggested that the inhibition of miR-27a/b was responsible for Nrf2 upregulation at both early stage and late stage of Cr(VI) exposure. This novel regulation of Nrf2 upon chronic Cr(VI) exposure through redox-regulated miR-27a/b will provide potential targets for preventing and treating Cr(VI)-induced carcinogenesis in the future.
Collapse
Affiliation(s)
- Lin Wang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States; Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Khaliunaa Bayanbold
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Lei Zhao
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Yifang Wang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Andrea Adamcakova-Dodd
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, United States
| | - Peter S Thorne
- Department of Occupational and Environmental Health, University of Iowa, Iowa City, IA, United States
| | - Hushan Yang
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States.
| | - Ling-Zhi Liu
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
21
|
Zuo J, Zhang Z, Li M, Yang Y, Zheng B, Wang P, Huang C, Zhou S. The crosstalk between reactive oxygen species and noncoding RNAs: from cancer code to drug role. Mol Cancer 2022; 21:30. [PMID: 35081965 PMCID: PMC8790843 DOI: 10.1186/s12943-021-01488-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/26/2021] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress (OS), characterized by the excessive accumulation of reactive oxygen species (ROS), is an emerging hallmark of cancer. Tumorigenesis and development driven by ROS require an aberrant redox homeostasis, that activates onco-signaling and avoids ROS-induced programmed death by orchestrating antioxidant systems. These processes are revealed to closely associate with noncoding RNAs (ncRNAs). On the basis of the available evidence, ncRNAs have been widely identified as multifarious modulators with the involvement of several key redox sensing pathways, such as NF-κB and Nrf2 signaling, therefore potentially becoming effective targets for cancer therapy. Furthermore, the vast majority of ncRNAs with property of easy detected in fluid samples (e.g., blood and urine) facilitate clinicians to monitor redox homeostasis, indicating a novel method for cancer diagnosis. Herein, focusing on carcinoma initiation, metastasis and chemoradiotherapy resistance, we aimed to discuss the ncRNAs-ROS network involved in cancer progression, and the potential clinical application as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jing Zuo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Maomao Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Yun Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Bohao Zheng
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Ping Wang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China.
| |
Collapse
|
22
|
Wang L, Ji XB, Wang LH, Xia ZK, Xie YX, Liu WJ, Qiu JG, Jiang BH, Liu LZ. MiRNA-30e downregulation increases cancer cell proliferation, invasion and tumor growth through targeting RPS6KB1. Aging (Albany NY) 2021; 13:24037-24049. [PMID: 34727092 PMCID: PMC8610128 DOI: 10.18632/aging.203665] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/25/2021] [Indexed: 12/28/2022]
Abstract
Human esophagus carcinoma (EC) is one of the most common malignant tumors, especially in Africa and Asia including China. In EC initiation and progression, genetic and epigenetic aberrations have been reported to play a major role, but the underlying molecular mechanisms are largely unknown. In this study, the miR-30e levels were analyzed in human EC tissues and TCGA databases, and the results demonstrated that miR-30e expression in EC tissues was significantly decreased compared to adjacent normal tissues. To further investigate the role of miR-30e in cancer cells, we found that forced expression of miR-30e dramatically inhibited cell proliferation, invasion, tube formation, and colony formation of cancer cells. To determine the underlying mechanism of miR-30e, we found that RPS6KB1 was a direct target of miR-30e by binding to its 3′-UTR, which was verified by luciferase activity assay using reporters with wild-type miR-30e and its seed sequence mutant constructs and Western blotting assay. In vivo experiment showed that miR-30e overexpression significantly inhibited tumor growth and decreased RPS6KB1 expression in xenografts. In EC, high expression of RPS6KB1 in tumor tissues indicated poor prognosis of patients with less survival rate. High levels of RPS6KB1 and low levels of miR-30e closely correlated poor survival of patients with several other types of cancer. These findings show that miR-30e and its target RPS6KB1 are important in cancer development and clinical outcomes, and miR-30e/RPS6KB1 is a potential future therapeutic pathway for EC intervention.
Collapse
Affiliation(s)
- Lin Wang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.,Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Xiang-Bo Ji
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Li-Hong Wang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhong-Kun Xia
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Yun-Xia Xie
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Wen-Jing Liu
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jian-Ge Qiu
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ling-Zhi Liu
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
23
|
Kim C, Chen J, Ceresa BP. Chronic arsenic increases cell migration in BEAS-2B cells by increasing cell speed, cell persistence, and cell protrusion length. Exp Cell Res 2021; 408:112852. [PMID: 34599931 DOI: 10.1016/j.yexcr.2021.112852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
There is a strong association between arsenic exposure and lung cancer development, however, the mechanism by which arsenic exposure leads to carcinogenesis is not clear. In our previous study, we observed that when BEAS-2B cells are chronically exposed to arsenic, there is an increase in secreted TGFα, as well as an increase in EGFR expression and activity. Further, these changes were broadly accompanied with an increase in cell migration. The overarching goal of this study was to acquire finer resolution of the arsenic-dependent changes in cell migration, as well as to understand the role of increased EGFR expression and activity levels in the underlying mechanisms of cell migration. To do this, we used a combination of biochemical and single cell assays, and observed chronic arsenic treatment enhancing cell migration by increasing cell speed, cell persistence and cell protrusion length. All three parameters were further increased by the addition of TGFα, indicating EGFR activity is sufficient to enhance those aspects of cell migration. In contrast, EGFR activity was necessary for the increase in cell speed, as it was reversed with an EGFR inhibitor, AG1478, but was not necessary to enhance persistence and protrusion length. From these data, we were able to isolate both EGFR-dependent and -independent features of cell migration that were enhanced by chronic arsenic exposure.
Collapse
Affiliation(s)
- Christine Kim
- Department of Pharmacology and Toxicology, University of Louisville, USA
| | - Joseph Chen
- Department of Pharmacology and Toxicology, University of Louisville, USA; Department of Bioengineering, University of Louisville, USA
| | - Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, USA.
| |
Collapse
|
24
|
Saintilnord WN, Fondufe-Mittendorf Y. Arsenic-induced epigenetic changes in cancer development. Semin Cancer Biol 2021; 76:195-205. [PMID: 33798722 PMCID: PMC8481342 DOI: 10.1016/j.semcancer.2021.03.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/29/2022]
Abstract
Arsenic is a ubiquitous metalloid whose high levels of toxicity pose major health concerns to millions of people worldwide by increasing susceptibility to various cancers and non-cancer illnesses. Since arsenic is not a mutagen, the mechanism by which it causes changes in gene expression and disease pathogenesis is not clear. One possible mechanism is through generation of reactive oxygen species. Another equally important mechanism still very much in its infancy is epigenetic dysregulation. In this review, we discuss recent discoveries underlying arsenic-induced epigenetic changes in cancer development. Importantly, we highlight the proposed mechanisms targeted by arsenic to drive oncogenic gene expression.
Collapse
Affiliation(s)
- Wesley N Saintilnord
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA.
| | | |
Collapse
|
25
|
Wang L, Liu LZ, Jiang BH. Dysregulation of microRNAs in metal-induced angiogenesis and carcinogenesis. Semin Cancer Biol 2021; 76:279-286. [PMID: 34428550 DOI: 10.1016/j.semcancer.2021.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small endogenous non-coding RNAs that regulate cancer initiation, development, angiogenesis, and therapeutic resistance. Metal exposure widely occurs through air, water, soil, food, and industrial contaminants. Hundreds of millions of people may have metal exposure associated with toxicity, serious health problems, and cancer occurrence. Metal exposure is found to induce oxidative stress, DNA damage and repair, and activation of multiple signaling pathways. However, molecular mechanisms of metal-induced carcinogenesis remain to be elucidated. Recent studies demonstrated that the exposure of metals such as arsenic, hexavalent chromium, cadmium, and nickel caused dysregulation of microRNAs that are implicated to play an important role in cell transformation, tumor growth and angiogenesis. This review focuses on the recent studies that show metal-induced miRNA dysregulation and underlined mechanisms in cell malignant transformation, angiogenesis and tumor growth.
Collapse
Affiliation(s)
- Lin Wang
- Academy of Medical Science, Zhengzhou University, Zhengzhou, 450000, China; Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States
| | - Ling-Zhi Liu
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, United States.
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, 19107, United States.
| |
Collapse
|
26
|
Ren C, Zhou Y, Liu W, Wang Q. Paradoxical effects of arsenic in the lungs. Environ Health Prev Med 2021; 26:80. [PMID: 34388980 PMCID: PMC8364060 DOI: 10.1186/s12199-021-00998-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/15/2021] [Indexed: 11/10/2022] Open
Abstract
High levels (> 100 ug/L) of arsenic are known to cause lung cancer; however, whether low (≤ 10 ug/L) and medium (10 to 100 ug/L) doses of arsenic will cause lung cancer or other lung diseases, and whether arsenic has dose-dependent or threshold effects, remains unknown. Summarizing the results of previous studies, we infer that low- and medium-concentration arsenic cause lung diseases in a dose-dependent manner. Arsenic trioxide (ATO) is recognized as a chemotherapeutic drug for acute promyelocytic leukemia (APL), also having a significant effect on lung cancer. The anti-lung cancer mechanisms of ATO include inhibition of proliferation, promotion of apoptosis, anti-angiogenesis, and inhibition of tumor metastasis. In this review, we summarized the role of arsenic in lung disease from both pathogenic and therapeutic perspectives. Understanding the paradoxical effects of arsenic in the lungs may provide some ideas for further research on the occurrence and treatment of lung diseases.
Collapse
Affiliation(s)
- Caixia Ren
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yang Zhou
- Liaoning Clinical Research Center for Lung Cancer, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Wenwen Liu
- Liaoning Clinical Research Center for Lung Cancer, The Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
27
|
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
|
28
|
Wang Y, Qi H, Liu Y, Duan C, Liu X, Xia T, Chen D, Piao HL, Liu HX. The double-edged roles of ROS in cancer prevention and therapy. Theranostics 2021; 11:4839-4857. [PMID: 33754031 PMCID: PMC7978298 DOI: 10.7150/thno.56747] [Citation(s) in RCA: 367] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/31/2021] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) serve as cell signaling molecules generated in oxidative metabolism and are associated with a number of human diseases. The reprogramming of redox metabolism induces abnormal accumulation of ROS in cancer cells. It has been widely accepted that ROS play opposite roles in tumor growth, metastasis and apoptosis according to their different distributions, concentrations and durations in specific subcellular structures. These double-edged roles in cancer progression include the ROS-dependent malignant transformation and the oxidative stress-induced cell death. In this review, we summarize the notable literatures on ROS generation and scavenging, and discuss the related signal transduction networks and corresponding anticancer therapies. There is no doubt that an improved understanding of the sophisticated mechanism of redox biology is imperative to conquer cancer.
Collapse
|
29
|
Paithankar JG, Saini S, Dwivedi S, Sharma A, Chowdhuri DK. Heavy metal associated health hazards: An interplay of oxidative stress and signal transduction. CHEMOSPHERE 2021; 262:128350. [PMID: 33182141 DOI: 10.1016/j.chemosphere.2020.128350] [Citation(s) in RCA: 308] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/09/2020] [Accepted: 09/13/2020] [Indexed: 05/20/2023]
Abstract
Heavy metal-induced cellular and organismal toxicity have become a major health concern in biomedical science. Indiscriminate use of heavy metals in different sectors, such as, industrial-, agricultural-, healthcare-, cosmetics-, and domestic-sectors has contaminated environment matrices and poses a severe health concern. Xenobiotics mediated effect is a ubiquitous cellular response. Oxidative stress is one such prime cellular response, which is the result of an imbalance in the redox system. Further, oxidative stress is associated with macromolecular damages and activation of several cell survival and cell death pathways. Epidemiological as well as laboratory data suggest that oxidative stress-induced cellular response following heavy metal exposure is linked with an increased risk of neoplasm, neurological disorders, diabetes, infertility, developmental disorders, renal failure, and cardiovascular disease. During the recent past, a relation among heavy metal exposure, oxidative stress, and signaling pathways have been explored to understand the heavy metal-induced toxicity. Heavy metal-induced oxidative stress and its connection with different signaling pathways are complicated; therefore, the systemic summary is essential. Herein, an effort has been made to decipher the interplay among heavy metals/metalloids (Arsenic, Chromium, Cadmium, and Lead) exposures, oxidative stress, and signal transduction, which are essential to mount the cellular and organismal response. The signaling pathways involved in this interplay include NF-κB, NRF2, JAK-STAT, JNK, FOXO, and HIF.
Collapse
Affiliation(s)
- Jagdish Gopal Paithankar
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Sanjay Saini
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Shiwangi Dwivedi
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Anurag Sharma
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India.
| | - Debapratim Kar Chowdhuri
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India.
| |
Collapse
|
30
|
Ijomone OM, Ijomone OK, Iroegbu JD, Ifenatuoha CW, Olung NF, Aschner M. Epigenetic influence of environmentally neurotoxic metals. Neurotoxicology 2020; 81:51-65. [PMID: 32882300 PMCID: PMC7708394 DOI: 10.1016/j.neuro.2020.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
Continuous globalization and industrialization have ensured metals are an increasing aspect of daily life. Their usefulness in manufacturing has made them vital to national commerce, security and global economy. However, excess exposure to metals, particularly as a result of environmental contamination or occupational exposures, has been detrimental to overall health. Excess exposure to several metals is considered environmental risk in the aetiology of several neurological and neurodegenerative diseases. Metal-induced neurotoxicity has been a major health concern globally with intensive research to unravel the mechanisms associated with it. Recently, greater focus has been directed at epigenetics to better characterize the underlying mechanisms of metal-induced neurotoxicity. Epigenetic changes are those modifications on the DNA that can turn genes on or off without altering the DNA sequence. This review discusses how epigenetic changes such as DNA methylation, post translational histone modification and noncoding RNA-mediated gene silencing mediate the neurotoxic effects of several metals, focusing on manganese, arsenic, nickel, cadmium, lead, and mercury.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.
| | - Olayemi K Ijomone
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria; Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Joy D Iroegbu
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Chibuzor W Ifenatuoha
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Nzube F Olung
- The Neuro- Lab, Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology and Neurosciences, Albert Einstein College of Medicine, NY, USA.
| |
Collapse
|
31
|
Jia X, Qiu T, Yao X, Jiang L, Wang N, Wei S, Tao Y, Pei P, Wang Z, Zhang J, Zhu Y, Yang G, Liu X, Liu S, Sun X. Arsenic induces hepatic insulin resistance via mtROS-NLRP3 inflammasome pathway. JOURNAL OF HAZARDOUS MATERIALS 2020; 399:123034. [PMID: 32544768 DOI: 10.1016/j.jhazmat.2020.123034] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/24/2020] [Accepted: 05/24/2020] [Indexed: 06/11/2023]
Abstract
Hepatic insulin resistance (IR) is the key event for arsenic-caused type 2 diabetes (T2D). However, the unequivocal mechanism of arsenic-induced hepatic IR remains unclear. The current study determined the role of NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation in arsenic-induced IR and revealed the underlying mechanism. Three-month NaAsO2 gavage led to glucose intolerance and insulin insensitivity, impaired hepatic insulin signaling. Additionally, NaAsO2 upregulated the level of oxidized mitochondrial DNA (ox-mtDNA) and mitophagy, thereby activating the NLRP3 inflammasome in SD rat liver. In vitro, we demonstrated that NaAsO2-induced IR depended upon the NLRP3 inflammasome activation. Moreover, inhibiting mitophagy mitigated the NLRP3 inflammasome activation and impaired insulin signaling induced by NaAsO2. Furthermore, mitochondrial reactive oxygen species (mtROS) scavenger alleviated the upregulated ox-mtDNA and mitophagy, thereby inhibiting the NLRP3 inflammasome activation, and improving insulin signaling. Taken together, these data demonstrated that mtROS-triggered ox-mtDNA, mitophagy, and the activation of NLRP3 inflammasome was involved in arsenic-induced hepatic IR.
Collapse
Affiliation(s)
- Xue Jia
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Tianming Qiu
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Xiaofeng Yao
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Liping Jiang
- Experimental Teaching Center of Public Health, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Ningning Wang
- Nutrition and Food Hygiene, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Sen Wei
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Ye Tao
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Pei Pei
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Zhidong Wang
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Jingyuan Zhang
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Yuhan Zhu
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Guang Yang
- Nutrition and Food Hygiene, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Xiaofang Liu
- Nutrition and Food Hygiene, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Shuang Liu
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| | - Xiance Sun
- Occupational and Environmental Health Department, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China; Global Health Research Center, Dalian Medical University, 9 Lvshun South Road, Dalian, 116044, PR China.
| |
Collapse
|
32
|
Wang W, Zheng F, Lin C, Zhang A. Changes in energy metabolism and macrophage polarization: Potential mechanisms of arsenic-induced lung injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 204:110948. [PMID: 32739672 DOI: 10.1016/j.ecoenv.2020.110948] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 06/11/2023]
Abstract
Exposure to arsenic is epidemiologically associated with increased lung disease. In detailing the mechanism by which arsenic exposure leads to disease, studies have emphasized that metabolic reprogramming and immune dysfunction are related to arsenic-induced lung injury. However, the association between the mechanisms listed above is not well understood. Thus, the current study aimed to investigate the interaction of energy metabolism and macrophage polarization, by which arsenic exposure adversely induced lung injury in both in vitro and human studies. First, we confirmed a shift to glycolytic metabolism resulting from mitochondrial dysfunction. This shift was accompanied by an increase in the levels of phosphorylated PDHE1α (S293) and PDK1 and a concomitant marked increase in several key markers of the HIF-1α signaling pathway (HIF-1α, p-PKM2, GLUT1 and HK-2). In addition, utilizing an in vitro model in which lung epithelial cells are cultured with macrophages, we determined that arsenic treatment polarizes macrophages towards the M2 phenotype through lactate. In the human study, the serum lactate and TGF-β levels were higher in arsenic-exposed subjects than that in reference subjects (t= 4.50, 6.24, both p < 0.05), while FVC and FEV1 were both lower (t= 5.47, 7.59, both p < 0.05). Pearson correlation analyses showed a significant negative correlation between the serum TGF-β and lactate levels and the lung function parameters (pcorrelation<0.05). In mediation analyses, lactate and TGF-β significantly mediated 24.3% and 9.0%, respectively, of the association between arsenic and FVC (pmediation<0.05), while lactate and TGF-β significantly mediated 22.2% and 12.5%, respectively, of the association between arsenic and FEV1 (pmediation<0.05). Together, the results of the in vitro and human studies indicated that there is complex communication between metabolic reprogramming and immune dysfunction, resulting in exacerbated effects in a feedback loop with increased arsenic-induced lung damage.
Collapse
Affiliation(s)
- Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Fanyan Zheng
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Changhu Lin
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China.
| |
Collapse
|
33
|
Angiogenesis in Wound Healing following Pharmacological and Toxicological Exposures. CURRENT PATHOBIOLOGY REPORTS 2020. [DOI: 10.1007/s40139-020-00212-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Kim C, States JC, Ceresa BP. Chronic and acute arsenic exposure enhance EGFR expression via distinct molecular mechanisms. Toxicol In Vitro 2020; 67:104925. [PMID: 32599262 DOI: 10.1016/j.tiv.2020.104925] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/02/2020] [Accepted: 06/22/2020] [Indexed: 01/06/2023]
Abstract
The impacts of acute arsenic exposure (i.e. vomiting, diarrhea, and renal failure) are distinct from those brought about by sustained, low level exposure from environmental sources or drinking of contaminated well water. Chronic arsenic exposure is a risk factor for the development of pulmonary diseases, including lung cancer. How arsenic exposure leads to pulmonary disease is not fully understood. Both acute versus chronic arsenic exposure increase EGFR expression, but do so via distinct molecular mechanisms. BEAS-2B cells were exposed to either acute sodium arsenite (5 μM for 24 h) or chronic sodium arsenite (100 nM for 24 weeks). Cells treated with acute arsenic exhibited a decrease in viability, changes in morphology, and increased mRNA level of BTC. In contrast, during 24 weeks of arsenic exposure, the cells had increased EGFR expression and activity, and increased mRNA and protein levels of TGFα. Further, chronic arsenic treatment caused an increase in cell migration in the absence of exogenous ligand. Elevated TGFα and EGFR expression are features of many non-small cell lung cancers. We propose that lung epithelial cells chronically exposed to low level arsenic increases EGFR signaling via TGFα production to enhance ligand-independent cell migration.
Collapse
Affiliation(s)
- Christine Kim
- Department of Pharmacology and Toxicology, University of Louisville, USA
| | | | - Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, USA.
| |
Collapse
|
35
|
Wang L, Lu YF, Wang CS, Xie YX, Zhao YQ, Qian YC, Liu WT, Wang M, Jiang BH. HB-EGF Activates the EGFR/HIF-1α Pathway to Induce Proliferation of Arsenic-Transformed Cells and Tumor Growth. Front Oncol 2020; 10:1019. [PMID: 32695675 PMCID: PMC7338480 DOI: 10.3389/fonc.2020.01019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/22/2020] [Indexed: 12/27/2022] Open
Abstract
Arsenic was recently identified as a pollutant that is a major cause of lung cancer. Since heparin-binding EGF-like growth factor (HB-EGF) was reported to be a promising therapeutic target for lung cancer, we investigated the role and mechanism of HB-EGF during arsenic-induced carcinogenesis and development of lung cancer. HB-EGF expression were upregulated in As-T cells, lung cancer cell lines, and in most lung cancer tissue samples; and HB-EGF activated the EGFR/p-ERK/HIF-1α pathway and induced VEGF by regulating HIF-1α transcription. HIF-1α transcriptional stimulation by HB-EGF was facilitated by PKM2 and played an important role in HB-EGF's effect on cells. An HB-EGF inhibitor(CRM197, cross-reacting material 197) slowed cell proliferation and inhibited migration of As-T and A549 cells, and inhibited tumor growth. PKM2 also played an important role in the proliferation and migration in As-T cells. The positive staining ratios of EGFR phosphorylation (Y1068) and PKM2 were significantly higher in most cases of lung cancer than in paired normal tumor-adjacent lung tissues; and HB-EGF expression levels strongly correlated with p-EGFR expression levels. Thus, HB-EGF drives arsenic-induced carcinogenesis, tumor growth, and lung cancer development via the EGFR/PKM2/HIF-1α pathway.
Collapse
Affiliation(s)
- Lin Wang
- Department of Pathology, Nanjing Medical University, Nanjing, China.,The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yi-Fan Lu
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Chao-Shan Wang
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Yun-Xia Xie
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yan-Qiu Zhao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying-Chen Qian
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Wei-Tao Liu
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Bing-Hua Jiang
- Department of Pathology, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
36
|
Climent M, Viggiani G, Chen YW, Coulis G, Castaldi A. MicroRNA and ROS Crosstalk in Cardiac and Pulmonary Diseases. Int J Mol Sci 2020; 21:ijms21124370. [PMID: 32575472 PMCID: PMC7352701 DOI: 10.3390/ijms21124370] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) affect many cellular functions and the proper redox balance between ROS and antioxidants contributes substantially to the physiological welfare of the cell. During pathological conditions, an altered redox equilibrium leads to increased production of ROS that in turn may cause oxidative damage. MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level contributing to all major cellular processes, including oxidative stress and cell death. Several miRNAs are expressed in response to ROS to mediate oxidative stress. Conversely, oxidative stress may lead to the upregulation of miRNAs that control mechanisms to buffer the damage induced by ROS. This review focuses on the complex crosstalk between miRNAs and ROS in diseases of the cardiac (i.e., cardiac hypertrophy, heart failure, myocardial infarction, ischemia/reperfusion injury, diabetic cardiomyopathy) and pulmonary (i.e., idiopathic pulmonary fibrosis, acute lung injury/acute respiratory distress syndrome, asthma, chronic obstructive pulmonary disease, lung cancer) compartments. Of note, miR-34a, miR-144, miR-421, miR-129, miR-181c, miR-16, miR-31, miR-155, miR-21, and miR-1/206 were found to play a role during oxidative stress in both heart and lung pathologies. This review comprehensively summarizes current knowledge in the field.
Collapse
Affiliation(s)
- Montserrat Climent
- Humanitas Clinical and Research Center—IRCCS, Via Manzoni 56, 20089 Rozzano, MI, Italy;
| | - Giacomo Viggiani
- Department of Biomedical Sciences, Humanitas University, 20090 Pieve Emanuele, MI, Italy;
| | - Ya-Wen Chen
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Gerald Coulis
- Department of Physiology and Biophysics, and Institute for Immunology, University of California Irvine, Irvine, CA 92697, USA;
| | - Alessandra Castaldi
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Correspondence:
| |
Collapse
|
37
|
Crosstalk of MicroRNAs and Oxidative Stress in the Pathogenesis of Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2415324. [PMID: 32411322 PMCID: PMC7204110 DOI: 10.1155/2020/2415324] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/02/2020] [Accepted: 02/08/2020] [Indexed: 02/06/2023]
Abstract
Oxidative stress refers to an imbalance between reactive oxygen species (ROS) generation and body's capability to detoxify the reactive mediators or to fix the relating damage. MicroRNAs are considered to be important mediators that play essential roles in the regulation of diverse aspects of carcinogenesis. Growing studies have demonstrated that the ROS can regulate microRNA biogenesis and expression mainly through modulating biogenesis course, transcription factors, and epigenetic changes. On the other hand, microRNAs may in turn modulate the redox signaling pathways, altering their integrity, stability, and functionality, thus contributing to the pathogenesis of multiple diseases. Both ROS and microRNAs have been identified to be important regulators and potential therapeutic targets in cancers. However, the information about the interplay between oxidative stress and microRNA regulation is still limited. The present review is aimed at summarizing the current understanding of molecular crosstalk between microRNAs and the generation of ROS in the pathogenesis of cancer.
Collapse
|
38
|
Jin Y, Wang H, Zhu Y, Feng H, Wang G, Wang S. miR-199a-5p is involved in doxorubicin resistance of non-small cell lung cancer (NSCLC) cells. Eur J Pharmacol 2020; 878:173105. [PMID: 32278855 DOI: 10.1016/j.ejphar.2020.173105] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
Non-small cell lung cancer (NSCLC) is one of the prevalent and deadly cancers worldwide. Chemotherapy resistance is one of the most challenging problems for NSCLC and other cancer treatment. Recent study suggested that miRNAs are involved in therapeutic functions of chemotherapy during cancer treatment. Our present study established doxorubicin (Dox) resistant NSCLC A549 and H460 cells (named A549Dox/R and H460 Dox/R). We found that miR-199a-5p was significantly down regulated in Dox resistant cells. Over expression of miR-199a-5p can increase the Dox sensitivity of resistant cells. Among various targets of miR-199a-5p, chemoresistance can increase the expression of ABCC1 and HIF-1α. Gain and loss of function studies confirmed that both ABCC1 and HIF-1α were involved in the chemoresistance of NSCLC cells. Collectively, our data showed that miR-199a-5p regulated expression of ABCC1 and HIF-1α were involved in Dox resistance of NSCLC.
Collapse
Affiliation(s)
- Yonglong Jin
- Department of Radiotherapy, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Huiyun Wang
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Yingqian Zhu
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Hui Feng
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Guanqun Wang
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Shasha Wang
- Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
39
|
The Role of Reactive Oxygen Species in Arsenic Toxicity. Biomolecules 2020; 10:biom10020240. [PMID: 32033297 PMCID: PMC7072296 DOI: 10.3390/biom10020240] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Arsenic poisoning is a global health problem. Chronic exposure to arsenic has been associated with the development of a wide range of diseases and health problems in humans. Arsenic exposure induces the generation of intracellular reactive oxygen species (ROS), which mediate multiple changes to cell behavior by altering signaling pathways and epigenetic modifications, or cause direct oxidative damage to molecules. Antioxidants with the potential to reduce ROS levels have been shown to ameliorate arsenic-induced lesions. However, emerging evidence suggests that constructive activation of antioxidative pathways and decreased ROS levels contribute to chronic arsenic toxicity in some cases. This review details the pathways involved in arsenic-induced redox imbalance, as well as current studies on prophylaxis and treatment strategies using antioxidants.
Collapse
|
40
|
Wu L, Xi Y, Kong Q. Dexmedetomidine protects PC12 cells from oxidative damage through regulation of miR-199a/HIF-1α. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:506-514. [PMID: 32024386 DOI: 10.1080/21691401.2020.1716780] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Ling Wu
- Department of Clinical Pharmacy, Dalian Central Hospital, Dalian, China
| | - Yalin Xi
- Department of Clinical Pharmacy, Dalian Central Hospital, Dalian, China
| | - Qinglong Kong
- Department of Thoracic Surgery, Dalian Central Hospital, Dalian, China
| |
Collapse
|
41
|
Li K, Liu Z, Shi X, Wei TJ, Ma LQ, Luo J. Novel in situ method based on diffusive gradients in thin-films with lanthanum oxide nanoparticles for measuring As, Sb, and V and in waters. JOURNAL OF HAZARDOUS MATERIALS 2020; 383:121196. [PMID: 31539663 DOI: 10.1016/j.jhazmat.2019.121196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 07/18/2019] [Accepted: 09/09/2019] [Indexed: 06/10/2023]
Abstract
Lanthanum oxide nanoparticles (nano-La2O3) was used to develop a novel binding gel within an in situ passive sampler based on diffusive gradients in thin-films technique (NL-DGT) for measuring As(V), Sb(V), and V(V). Performance characteristics of NL-DGT were independent of pH (pH: 3.1-7.9 for As, 3.1-8.5 for V, and 3.1-6.5 for Sb) and ionic strength (0.1-500 mmol L-1 for As and V, and 0.1-200 mmol L-1 for Sb). No obvious competition effects among As, Sb, and V with different concentration ratios were found for NL-DGT measurement. Long term storage (8-188 d) of the nano-La2O3 gels in 0.01 mol L-1 NaNO3 at 4 °C did not affect their performance. During the field deployments in Yangtze and Jiuxiang River, NL-DGT measured concentrations of As and V were similar to those measured by the grab samples, while some differences were found for Sb between DGT and grab sampling because higher pH (∼8.0) in the studied rivers caused the performance deterioration of NL-DGT. Generally, the newly developed NL-DGT is suitable for monitoring As and V in freshwater from acidic to light alkaline and Sb in acidic and neutral water.
Collapse
Affiliation(s)
- Kexin Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Zhaodong Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Xinyao Shi
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Tian-Jiao Wei
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Lena Q Ma
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China; Soil and Water Science Department, University of Florida, Gainesville, FL, 32611, USA
| | - Jun Luo
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu, 210023, PR China.
| |
Collapse
|
42
|
Lemaire J, Van der Hauwaert C, Savary G, Dewaeles E, Perrais M, Lo Guidice JM, Pottier N, Glowacki F, Cauffiez C. Cadmium-Induced Renal Cell Toxicity Is Associated With MicroRNA Deregulation. Int J Toxicol 2020; 39:103-114. [DOI: 10.1177/1091581819899039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cadmium is an environmental pollutant well known for its nephrotoxic effects. Nevertheless, mechanisms underlying nephrotoxicity continue to be elucidated. MicroRNAs (miRNAs) have emerged in recent years as modulators of xenobiotic-induced toxicity. In this context, our study aimed at elucidating whether miRNAs are involved in renal proximal tubular toxicity induced by cadmium exposure. We showed that cadmium exposure, in 2 distinct renal proximal tubular cell models (renal proximal tubular epithelial cell [RPTEC]/human telomerase reverse transcriptase [hTERT] and human kidney-2), resulted in cytotoxicity associated with morphological changes, overexpression of renal injury markers, and induction of apoptosis and inflammation processes. Cadmium exposure also resulted in miRNA modulation, including the significant upregulation of 38 miRNAs in RPTEC/hTERT cells. Most of these miRNAs are known to target genes whose coding proteins are involved in oxidative stress, inflammation, and apoptosis, leading to tissue remodeling. In conclusion, this study provides a list of dysregulated miRNAs which may play a role in the pathophysiology of cadmium-induced kidney damages and highlights promising cadmium molecular biomarkers that warrants to be further evaluated.
Collapse
Affiliation(s)
- J. Lemaire
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
| | - C. Van der Hauwaert
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
- Département de la Recherche en Santé, CHU Lille, Lille, France
| | - G. Savary
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
| | - E. Dewaeles
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
| | - M. Perrais
- UMR-S 1172-JPArc-Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Université de Lille, Lille, France
| | - J. M. Lo Guidice
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
| | - N. Pottier
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
- Service de Toxicologie et Génopathies, CHU Lille, Lille, France
| | - F. Glowacki
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
- Service de Néphrologie, CHU Lille, Lille, France
| | - C. Cauffiez
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Université de Lille, Lille Cedex, France
| |
Collapse
|
43
|
Wang Y, Liu Y, Liu S, Wu B. Influence of Iron on Cytotoxicity and Gene Expression Profiles Induced by Arsenic in HepG2 Cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16224484. [PMID: 31739468 PMCID: PMC6888336 DOI: 10.3390/ijerph16224484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 01/10/2023]
Abstract
The toxicity of arsenic (As) could be influenced by many environmental factors and elements. Iron (Fe) is one of the elements that could be involved in As-induced toxicity. In this study, the interactive effects of Fe and As in HepG2 cells were analyzed based on cytotoxicity and transcriptomic analyses. The results showed that Fe could decrease cell viability and increase mitochondrial depolarization induced by As exposure. Oxidative stress and damage have been proven to be one of the main mechanisms of As toxicity. Our results showed that Fe increased the generation of reactive oxygen species (ROS) and lipid peroxidation product malondialdehyde (MDA) induced by As exposure. Microarray analysis further verified that Fe increased the alteration of gene expression and biological processes related to oxidative stress, cell proliferation, and the apoptotic signaling pathway caused by As exposure. Both results of cytotoxicity and transcriptomic analyses suggest that an increase of Fe in the human body could increase the As-induced toxicity, which should be considered during the health risk assessment of As.
Collapse
Affiliation(s)
- Yonghua Wang
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China;
- Correspondence:
| | - Yuxuan Liu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lakes of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China;
| | - Su Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China; (S.L.); (B.W.)
| | - Bing Wu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China; (S.L.); (B.W.)
| |
Collapse
|
44
|
Cardoso APF, Al-Eryani L, States JC. Arsenic-Induced Carcinogenesis: The Impact of miRNA Dysregulation. Toxicol Sci 2019; 165:284-290. [PMID: 29846715 DOI: 10.1093/toxsci/kfy128] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Arsenic is a toxic metalloid widely present in the earth's crust, and is a proven human carcinogen. Chronic arsenic exposure mainly through drinking water causes skin, lung, and urinary bladder cancers, and is associated with liver, prostate, and kidney cancers, cardiovascular and neurological disorders, and diabetes. Several modes of action have been suggested in arsenic carcinogenesis. However, the molecular etiology of arsenic-induced cancer remains unclear. Recent evidence clearly indicates that gene expression modifications induced by arsenic may involve epigenetic alterations, including miRNA dysregulation. Many miRNAs have been implicated in different human cancers as a consequence of losses and or gains of miRNA function that contribute to cancer development. Progress in identifying miRNA dysregulation induced by arsenic has been made using different approaches and models. The present review discusses the recent data regarding dysregulated expression of miRNA in arsenic-induced malignant transformation in vitro, gaps in current understanding and deficiencies in current models for arsenic-induced carcinogenesis, and future directions of research that would improve our knowledge regarding the mechanisms involved in arsenic-induced carcinogenesis.
Collapse
Affiliation(s)
- Ana P Ferragut Cardoso
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40202
| | - Laila Al-Eryani
- DNA Repair Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-4262
| | - J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky 40202
| |
Collapse
|
45
|
Feng M, Kim J, Field K, Reid C, Chatzistamou I, Shim M. Aspirin ameliorates the long-term adverse effects of doxorubicin through suppression of cellular senescence. FASEB Bioadv 2019; 1:579-590. [PMID: 32123852 PMCID: PMC6996307 DOI: 10.1096/fba.2019-00041] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 05/17/2019] [Accepted: 08/19/2019] [Indexed: 01/08/2023] Open
Abstract
A number of childhood cancer survivors develop adverse, late onset side effects of earlier cancer treatments, known as the late effects of cancer therapy. As the number of survivors continues to increase, this growing population is at increased risk for a number of health-related problems. In the present study, we have examined the effect of aspirin on the late effects of chemotherapy by treating juvenile mice with doxorubicin (DOX). This novel mouse model produced various long-term adverse effects, some of which resemble premature aging phenotypes. DOX also resulted in the tissue accumulation of senescent cells and up-regulation of cyclooxygenase-2 (COX2) expression. However, treatment with aspirin following juvenile exposure to DOX improved body weight gain, ameliorated the long-term adverse effects, and reduced the levels of senescence markers. Moreover, aspirin reduced p53 and p21 accumulation in DOX-treated human and mouse fibroblasts. However, the suppressive effect of aspirin on DOX-induced p53 accumulation was significantly decreased in COX2 knockout mouse embryonic fibroblasts. Additionally, treatment of senescent fibroblasts with aspirin or celecoxib, a COX2 specific inhibitor, reduced cell viability and decreased the levels of Bcl-xL protein. Taken together, these studies suggest that aspirin may be able to reduce the late effects of chemotherapy through the suppression of cellular senescence.
Collapse
Affiliation(s)
- Mingxiao Feng
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Joohwee Kim
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Kevin Field
- UNC School of MedicineUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Christine Reid
- Department of Biological SciencesUniversity of South CarolinaColumbiaSCUSA
- Center for Colon Cancer ResearchUniversity of South CarolinaColumbiaSCUSA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & ImmunologySchool of MedicineUniversity of South CarolinaColumbiaSCUSA
| | - Minsub Shim
- Department of BiochemistryCollege of Graduate Studies and Arizona College of Osteopathic MedicineMidwestern UniversityGlendaleAZUSA
| |
Collapse
|
46
|
He J, Liu W, Ge X, Wang GC, Desai V, Wang S, Mu W, Bhardwaj V, Seifert E, Liu LZ, Bhushan A, Peiper SC, Jiang BH. Arsenic-induced metabolic shift triggered by the loss of miR-199a-5p through Sp1-dependent DNA methylation. Toxicol Appl Pharmacol 2019; 378:114606. [PMID: 31170415 PMCID: PMC6788774 DOI: 10.1016/j.taap.2019.114606] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/21/2019] [Accepted: 05/31/2019] [Indexed: 12/23/2022]
Abstract
Inorganic arsenic is an environmental carcinogen that poses a major global public health risk. A high percentage of drinking water from wells in the U.S. contains higher-than-normal levels of arsenic, suggesting an increased risk of arsenic-induced deleterious effects. In addition to primary preventive measures, therapeutic strategies need to effectively address and integrate multiple molecular mechanisms underlying arsenic-induced carcinogenesis. We previously showed that the loss of miR-199a-5p in arsenic-transformed cells is pivotal to promote arsenic-induced angiogenesis and tumor growth in lung epithelial cells. In this study, we further showed that subacute or chronic exposure to arsenic diminished miR-199a-5p levels largely due to DNA methylation, which was achieved by increased DNA methyltransferase-1 (DNMT1) activity, mediated by the formation of specific protein 1 (Sp1)/DNMT1 complex. In addition to the DNA hypermethylation, arsenic exposure also repressed miR-199a transcription through a transcriptional repressor Sp1. We further identified an association between miR-199a-5p repression and the arsenic-mediated energy metabolic shift, as reflected by mitochondria defects and a switch to glycolysis, in which a glycolytic enzyme pyruvate kinase 2 (PKM2) was a functional target of miR-199a-5p. Taken together, the repression of miR-199a-5p through both Sp1-dependent DNA methylation and Sp1 transcriptional repression promotes an arsenic-mediated metabolic shift from mitochondria respiration to aerobic glycolysis via PKM2.
Collapse
Affiliation(s)
- Jun He
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States of America.
| | - Weitao Liu
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xin Ge
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Gao-Chan Wang
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Vilas Desai
- Department of Pharmaceutical Sciences, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Shaomin Wang
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Wei Mu
- School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Vikas Bhardwaj
- Department of Pharmaceutical Sciences, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Erin Seifert
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Ling-Zhi Liu
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IW 52242, United States of America
| | - Alok Bhushan
- Department of Pharmaceutical Sciences, College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Stephen C Peiper
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Bing-Hua Jiang
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IW 52242, United States of America.
| |
Collapse
|
47
|
Li Y, Wang D, Li X, Shao Y, He Y, Yu H, Ma Z. MiR-199a-5p suppresses non-small cell lung cancer via targeting MAP3K11. J Cancer 2019; 10:2472-2479. [PMID: 31258753 PMCID: PMC6584351 DOI: 10.7150/jca.29426] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) comprise a class of short, non-coding RNAs that directly target 3'UTR of mRNA, causing subsequent degradation or suppression of translation. Here, we verified that miR-199a-5p was significantly down-regulated in mouse NSCLC tissues and human patient samples. To further study the function of miR-199a-5p, lentivirus system was adopted to construct stably over-expressing miR-199a-5p A549, SPC-A1 and H1299 cell lines. Then, miR-199a-5p played a tumor suppression role via directly targeting MAP3K11 gene in non-small cell lung cancer (NSCLC). Elevated miR-199a-5p suppressed cell proliferation and arrested cell cycle in G1 phase. We found that MAP3K11 was negatively correlated with miR-199a-5p in NSCLC patient tissues and mouse xenograft tumors. Our results suggest that miR-199a-5p together with its target gene MAP3K11 is a key factor and constitutes a complicated regulation network in NSCLC.
Collapse
Affiliation(s)
- Yanli Li
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Detao Wang
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xue Li
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yang Shao
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Yanyun He
- Exprimental Center for Life Sciences, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Huansha Yu
- Experimental Animal Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| |
Collapse
|
48
|
Orang AV, Petersen J, McKinnon RA, Michael MZ. Micromanaging aerobic respiration and glycolysis in cancer cells. Mol Metab 2019; 23:98-126. [PMID: 30837197 PMCID: PMC6479761 DOI: 10.1016/j.molmet.2019.01.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Cancer cells possess a common metabolic phenotype, rewiring their metabolic pathways from mitochondrial oxidative phosphorylation to aerobic glycolysis and anabolic circuits, to support the energetic and biosynthetic requirements of continuous proliferation and migration. While, over the past decade, molecular and cellular studies have clearly highlighted the association of oncogenes and tumor suppressors with cancer-associated glycolysis, more recent attention has focused on the role of microRNAs (miRNAs) in mediating this metabolic shift. Accumulating studies have connected aberrant expression of miRNAs with direct and indirect regulation of aerobic glycolysis and associated pathways. SCOPE OF REVIEW This review discusses the underlying mechanisms of metabolic reprogramming in cancer cells and provides arguments that the earlier paradigm of cancer glycolysis needs to be updated to a broader concept, which involves interconnecting biological pathways that include miRNA-mediated regulation of metabolism. For these reasons and in light of recent knowledge, we illustrate the relationships between metabolic pathways in cancer cells. We further summarize our current understanding of the interplay between miRNAs and these metabolic pathways. This review aims to highlight important metabolism-associated molecular components in the hunt for selective preventive and therapeutic treatments. MAJOR CONCLUSIONS Metabolism in cancer cells is influenced by driver mutations but is also regulated by posttranscriptional gene silencing. Understanding the nuanced regulation of gene expression in these cells and distinguishing rapid cellular responses from chronic adaptive mechanisms provides a basis for rational drug design and novel therapeutic strategies.
Collapse
Affiliation(s)
- Ayla V Orang
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Janni Petersen
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| | - Michael Z Michael
- Flinders Centre for Innovation in Cancer, Flinders University, Flinders Medical Centre, Adelaide, South Australia 5042, Australia.
| |
Collapse
|
49
|
Wang HW, Jiang X, Zhang Y, Wang J, Xie J, Wang YQ, Li YH. FGF21 Protects Against Hypoxia Injury Through Inducing HSP72 in Cerebral Microvascular Endothelial Cells. Front Pharmacol 2019; 10:101. [PMID: 30842736 PMCID: PMC6391338 DOI: 10.3389/fphar.2019.00101] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/25/2019] [Indexed: 11/29/2022] Open
Abstract
Background: Fibroblast growth factor 21 (FGF21), a member of a family of atypical FGFs, functions as cytokine to control endocrinology and metabolism. Recently, the roles of FGF21 in cardio-cerebral-vascular diseases have been gradually uncovered. In the present study, we investigated the effect of FGF21 on bEnd.3 cerebral microvascular endothelial cells (CMECs) upon hypoxia stress. Methods and Results: CMECs were cultured in the condition of 1% O2 for 8 h to induce hypoxia stimuli. For FGF21 treatment, recombinant FGF21 (50 nM) was added into the culture medium. Various biomedical assays were used to evaluate the hypoxia-induced injury in CMECs. Under normoxia condition, FGF21 had no obvious effect on cell viability and did not cause any cytotoxicity on CMECs. Under hypoxia condition, FGF21 significantly attenuated the hypoxia-induced injury, evidenced by the influences of FGF21 on CMEC viability and LDH release. TUNEL staining assay and immunoblotting of caspase-3 showed that FGF21 reduced hypoxia-induced apoptosis in CMECs. Mechanistically, FGF21 treatment compromised the hypoxia-induced changes of reactive oxygen species, malondialdehyde, total antioxidant activity, and total superoxide dismutase levels. FGF21 administration decreased hypoxia-induced matrix metalloprotein 3 and matrix metalloprotein 2/9 activity in CMECs. Activities of cyclooxygenase-2 and NF-κB-p65, two pro-inflammatory factors, were also upregulated by hypoxia but suppressed by FGF21. At last, we found that FGF21 increased heat shock protein family A member 1A (HSP72) mRNA and protein expression. Blockade of HSP72 by a pharmacological inhibitor VER155008 or specific siRNA-mediated knockdown abrogated the protection of FGF21 against hypoxia in CMECs. Conclusion: These data demonstrate that FGF21 protects against hypoxia stress-induced injury in CMECs by inducing HSP72 expression, suggesting a therapeutic value of FGF21 in hypoxia-related brain diseases such as ischemic stroke and acute mountain sickness.
Collapse
Affiliation(s)
- Hao-Wei Wang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xin Jiang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yu Zhang
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jian Wang
- Department of Anesthesiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Xie
- Department of Anesthesiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Qiang Wang
- Department of Anesthesiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Hua Li
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
50
|
Developmental Decline in the MicroRNA 199a (miR-199a)/miR-214 Cluster in Human Fetal Lung Promotes Type II Cell Differentiation by Upregulating Key Transcription Factors. Mol Cell Biol 2018; 38:MCB.00037-18. [PMID: 29507184 DOI: 10.1128/mcb.00037-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 02/27/2018] [Indexed: 02/07/2023] Open
Abstract
The major surfactant protein, SP-A (a product of the SFTPA gene), serves as a marker of type II pneumocyte differentiation and surfactant synthesis. SFTPA expression in cultured human fetal lung (HFL) epithelial cells is upregulated by hormones that increase cyclic AMP (cAMP) and activate TTF-1/NKX2.1 and NF-κB. To further define mechanisms for type II cell differentiation and induction of SP-A, we investigated roles of microRNAs (miRNAs). Using microarray to identify differentially expressed miRNAs in HFL epithelial cells during type II cell differentiation in culture, we observed that members of the miRNA 199a (miR-199a)/miR-214 cluster were significantly downregulated during differentiation. Validated and predicted targets of miR-199a-3p/miR-199a-5p and miR-214, which serve roles in type II cell differentiation (COX-2, NF-κB p50/p65, and CREB1), and the CREB1 target, C/EBPβ, were coordinately upregulated. Accordingly, overexpression of miR-199a-5p, miR-199a-3p, or miR-214 mimics in cultured HFL epithelial cells decreased COX-2, NF-κB p50/p65, CREB1, and C/EBPβ proteins, with an associated inhibition of SP-A expression. Interestingly, overexpression of the EMT factor, ZEB1, which declines during cAMP-induced type II cell differentiation, increased pri-miR-199a and reduced the expression of the targets NF-κB/p50 and COX-2. Collectively, these findings suggest that the developmental decline in miR-199a/miR-214 in HFL causes increased expression of critical targets that enhance type II cell differentiation and SP-A expression.
Collapse
|