1
|
Liu L, Li J, Xue Y, Xie S, Dong N, Chen C. Dihydroartemisinin attenuates PM-induced lung injury by inhibiting inflammation and regulating autophagy. Front Public Health 2025; 13:1548224. [PMID: 40124417 PMCID: PMC11925939 DOI: 10.3389/fpubh.2025.1548224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Objective The study investigates the effects and mechanisms of dihydroartemisinin (DHA) in mitigating lung injury induced by particulate matter (PM). Methods The lung injury model was induced by PM particles in vivo and in vitro. Hematoxylin and Eosin (H&E) staining was utilized for the detection of the thickening of airway wall and the infiltration of inflammatory cells in mouse lung tissue. The expressions of inflammatory factors were detected in alveolar lavage fluid and cell supernatant. TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling) staining, Caspase-1, Bcl-2-associated X protein (Bax), B-cell lymphoma 2 (Bcl-2), microtubule-associated protein 1 light chain 3-II (LC3-II) and Belcin-1 were used to observe the apoptosis and autophagy related expressions in mouse lung tissue, and p-p65 was detected by immunofluorescence. Results H&E staining revealed DHA alleviates PM-induced lung injury in vivo. Moreover, DHA reduced IL-6, IL-8, and IL-1β levels by ~50% (p < 0.05), highlighting its anti-inflammatory effects. Furthermore, immunohistochemistry showed that DHA treatment inhibited the pro-apoptotic expression of Bax/BCL2 and cleaved-Caspase-3, respectively. In addition, immunofluorescence staining revealed that the LC3-II and Beclin-1 levels dramatically increased in the PM group compared to Control group, but greatly reduced by DHA. Further, we found that DHA inhibited the activation of the NF-KB signaling pathway. Conclusion DHA protects against PM-induced lung injury through anti-inflammatory, anti-apoptotic, and autophagy-regulating mechanisms, offering a potential drug option for improving PM-induced lung injury.
Collapse
Affiliation(s)
- Lingjing Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingli Li
- Department of Pulmonary and Critical Care Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Yincong Xue
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuying Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Nian Dong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
2
|
Xiao L, Xu C, Lin P, Mu L, Yang X. Novel dihydroartemisinin derivative Mito-DHA5 induces apoptosis associated with mitochondrial pathway in bladder cancer cells. BMC Pharmacol Toxicol 2022; 23:10. [PMID: 35057867 PMCID: PMC8780396 DOI: 10.1186/s40360-021-00542-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 12/09/2021] [Indexed: 11/18/2022] Open
Abstract
Background Bladder cancer is the second most common genitourinary malignancy and the eleventh most common cancer worldwide. Dihydroartemisinin (DHA), a first-line antimalarial drug, has been found to have potent antitumor activity. In our previous study, a novel dihydroartemisinin derivative Mito-DHA5 synthesized in our laboratory has a stronger anti-tumor activity than DHA. In this study, we investigated the apoptotic effect of Mito-DHA5 on bladder cancer T24 cells and molecular mechanisms underlying. Methods Antitumor activity in vitro was evaluated by MTT, wound healing and cloning formation assays. Mitochondrial membrane potential (MMP) was detected by JC-1 probe and ROS levels were measured by specific kit. The expression of caspase-3, cleaved-caspase3, mitochondrial Cyt-C, Bcl-2, Bax and PARP in T24 cells was evaluated by Western blotting. Results The results showed that Mito-DHA5 reduced cell viability with an IC50 value of 3.2 µM and induced T24 cell apoptosis in a dose-dependent manner, increased the production of ROS and decreased MMP. Mito-DHA5 could down-regulate the expression of Bcl-2, mitochondrial Cyt-C, Caspase-3, PARP and up-regulate the expression of Bax and cleaved Caspase-3. Conclusions These data suggested that Mito-DHA5 had a potent inhibitory effect on T24 bladder cancer cell growth and induced these cells apoptosis associated with mitochondrial pathway.
Collapse
|
3
|
Yu R, Jin G, Fujimoto M. Dihydroartemisinin: A Potential Drug for the Treatment of Malignancies and Inflammatory Diseases. Front Oncol 2021; 11:722331. [PMID: 34692496 PMCID: PMC8529146 DOI: 10.3389/fonc.2021.722331] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
Dihydroartemisinin (DHA) has been globally recognized for its efficacy and safety in the clinical treatment of malaria for decades. Recently, it has been found that DHA inhibits malignant tumor growth and regulates immune system function in addition to anti-malaria. In parasites and tumors, DHA causes severe oxidative stress by inducing excessive reactive oxygen species production. DHA also kills tumor cells by inducing programmed cell death, blocking cell cycle and enhancing anti-tumor immunity. In addition, DHA inhibits inflammation by reducing the inflammatory cells infiltration and suppressing the production of pro-inflammatory cytokines. Further, genomics, proteomics, metabolomics and network pharmacology of DHA therapy provide the basis for elucidating the pharmacological effects of DHA. This review provides a summary of the recent research progress of DHA in anti-tumor, inhibition of inflammatory diseases and the relevant pharmacological mechanisms. With further research of DHA, it is likely that DHA will become an alternative therapy in the clinical treatment of malignant tumors and inflammatory diseases.
Collapse
Affiliation(s)
- Ran Yu
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Guihua Jin
- Department of Immunology and Pathogenic Biology, Yanbian University Medical College, Yanji, China
| | - Manabu Fujimoto
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Laboratory of Cutaneous Immunology, Osaka University Immunology Frontier Research Center, Osaka, Japan
| |
Collapse
|
4
|
Long Noncoding RNA WDFY3-AS2 Represses the Progression of Esophageal Cancer through miR-18a/PTEN Axis. JOURNAL OF ONCOLOGY 2021; 2021:9951010. [PMID: 34194502 PMCID: PMC8203383 DOI: 10.1155/2021/9951010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022]
Abstract
Background Understanding the role of lncRNAs in the development of human malignancies is necessary for the targeted therapy of malignant tumors, including esophageal cancer (EC). Nevertheless, the specific role and regulatory mechanism of lncRNA WDFY3-AS2 in EC are still unclear. Here, we examined the functional role and regulatory mechanism of WDFY3-AS2 in EC. Materials and Methods RT-qPCR assay was applied to measure the expression of WDFY3-AS2 and miR-18a in EC samples and cells. The luciferase reporter and RIP assays were used to check the relationship between WDFY3-AS2, miR-18a, and PTEN. Counting Clock Kit-8 (CCK-8) assay was carried out to detect cell viability, and transwell assays were used for measuring cell migration and invasion. Results Underexpression of WDFY3-AS2 was found in EC specimens and cells, which predicted a poor prognosis of EC patients. Reexpression of WDFY3-AS2 repressed the progression of EC via inhibiting cell proliferation, migration, and invasion. Additionally, WDFY3-AS2 was negatively correlated with miR-18a and positively with PTEN. Furthermore, we discovered that the expression of PTEN decreased by miR-18a mimic was rescued by WDFY3-AS2 overexpression. Conclusions WDFY3-AS2 modulates the expressional level of PTEN as a competitive endogenous RNA via sponging miR-18a in EC, which suggests that the WDFY3-AS2/miR-18a/PTEN pathway might be involved in the progression of EC.
Collapse
|
5
|
Dai X, Zhang X, Chen W, Chen Y, Zhang Q, Mo S, Lu J. Dihydroartemisinin: A Potential Natural Anticancer Drug. Int J Biol Sci 2021; 17:603-622. [PMID: 33613116 PMCID: PMC7893584 DOI: 10.7150/ijbs.50364] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Dihydroartemisinin (DHA) is an active metabolite of artemisinin and its derivatives (ARTs), and it is an effective clinical drug widely used to treat malaria. Recently, the anticancer activity of DHA has attracted increasing attention. Nevertheless, there is no systematic summary on the anticancer effects of DHA. Notably, studies have shown that DHA exerts anticancer effects through various molecular mechanisms, such as inhibiting proliferation, inducing apoptosis, inhibiting tumor metastasis and angiogenesis, promoting immune function, inducing autophagy and endoplasmic reticulum (ER) stress. In this review, we comprehensively summarized the latest progress regarding the anticancer activities of DHA in cancer. Importantly, the underlying anticancer molecular mechanisms and pharmacological effects of DHA in vitro and in vivo are the focus of our attention. Interestingly, new methods to improve the solubility and bioavailability of DHA are discussed, which greatly enhance its anticancer efficacy. Remarkably, DHA has synergistic anti-tumor effects with a variety of clinical drugs, and preclinical and clinical studies provide stronger evidence of its anticancer potential. Moreover, this article also gives suggestions for further research on the anticancer effects of DHA. Thus, we hope to provide a strong theoretical support for DHA as an anticancer drug.
Collapse
Affiliation(s)
- Xiaoshuo Dai
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Xiaoyan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.,Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| | - Wei Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.,Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Yihuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Qiushuang Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Saijun Mo
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.,Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.,Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, Henan Province 450001, PR China.,State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province 450052, PR China
| |
Collapse
|
6
|
Salama L, Pastor ER, Stone T, Mousa SA. Emerging Nanopharmaceuticals and Nanonutraceuticals in Cancer Management. Biomedicines 2020; 8:E347. [PMID: 32932737 PMCID: PMC7554840 DOI: 10.3390/biomedicines8090347] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Nanotechnology is the science of nanoscale, which is the scale of nanometers or one billionth of a meter. Nanotechnology encompasses a broad range of technologies, materials, and manufacturing processes that are used to design and/or enhance many products, including medicinal products. This technology has achieved considerable progress in the oncology field in recent years. Most chemotherapeutic agents are not specific to the cancer cells they are intended to treat, and they can harm healthy cells, leading to numerous adverse effects. Due to this non-specific targeting, it is not feasible to administer high doses that may harm healthy cells. Moreover, low doses can cause cancer cells to acquire resistance, thus making them hard to kill. A solution that could potentially enhance drug targeting and delivery lies in understanding the complexity of nanotechnology. Engineering pharmaceutical and natural products into nano-products can enhance the diagnosis and treatment of cancer. Novel nano-formulations such as liposomes, polymeric micelles, dendrimers, quantum dots, nano-suspensions, and gold nanoparticles have been shown to enhance the delivery of drugs. Improved delivery of chemotherapeutic agents targets cancer cells rather than healthy cells, thereby preventing undesirable side effects and decreasing chemotherapeutic drug resistance. Nanotechnology has also revolutionized cancer diagnosis by using nanotechnology-based imaging contrast agents that can specifically target and therefore enhance tumor detection. In addition to the delivery of drugs, nanotechnology can be used to deliver nutraceuticals like phytochemicals that have multiple properties, such as antioxidant activity, that protect cells from oxidative damage and reduce the risk of cancer. There have been multiple advancements and implications for the use of nanotechnology to enhance the delivery of both pharmaceutical and nutraceutical products in cancer prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
| | | | | | - Shaker A. Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (L.S.); (E.R.P.); (T.S.)
| |
Collapse
|
7
|
Chen X, He LY, Lai S, He Y. Dihydroartemisinin inhibits the migration of esophageal cancer cells by inducing autophagy. Oncol Lett 2020; 20:94. [PMID: 32831913 PMCID: PMC7439151 DOI: 10.3892/ol.2020.11955] [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: 12/26/2019] [Accepted: 06/23/2020] [Indexed: 12/26/2022] Open
Abstract
Esophageal cancer (EC) is a complex gastrointestinal malignancy and its global incidence rate ranks 7th among all cancer types. Due to its aggressive nature and the potential for early metastasis, the survival rates of patients with EC are poor. Dihydroartemisinin (DHA) is the primary active derivative of artemisinin, and, as well as its use as an anti-malarial, DHA has also exhibited antitumor activity in various cancer models, such as cholangiocarcinoma, head and neck carcinoma, and hepatocellular carcinoma cells. However, the molecular mechanisms underlying the antitumor effect of DHA in the treatment of EC remains poorly understood. The results of the present study demonstrated that DHA significantly inhibited the migration of TE-1 and Eca-109 EC cells in a dose-dependent manner by activating autophagy. DHA treatment also significantly reversed epithelial-mesenchymal transition (EMT) by downregulating the EMT-associated markers, N-cadherin and vimentin, and upregulating the expression of E-cadherin. Mechanistically, DHA treatment decreased Akt phosphorylation and inhibited the Akt/mTOR signaling pathway, leading to the activation of autophagy. The levels of the autophagy-associated proteins were suppressed and DHA-mediated inhibition of migration in EC cells was reversed when an active form of Akt was overexpressed. In conclusion, the present study demonstrated the potential value of DHA in the treatment of EC, and revealed the underlying mechanism by which FDHA inhibits cellular migration.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, P.R. China
| | - Lan-Ying He
- Department of Gastroenterology, Tumor Hospital Affiliated to Chongqing University, Chongqing 400030, P.R. China
| | - Shu Lai
- Department of Pharmacy, Jiulongpo District People's Hospital of Chongqing, Chongqing 400050, P.R. China
| | - Yao He
- Department of Pharmacy, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, P.R. China
| |
Collapse
|
8
|
Chinnapaka S, Bakthavachalam V, Munirathinam G. Repurposing antidepressant sertraline as a pharmacological drug to target prostate cancer stem cells: dual activation of apoptosis and autophagy signaling by deregulating redox balance. Am J Cancer Res 2020; 10:2043-2065. [PMID: 32775000 PMCID: PMC7407340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/14/2020] [Indexed: 06/11/2023] Open
Abstract
Cancer stem cells play a major role in tumor initiation, progression, and tumor relapse of prostate cancer (PCa). Recent studies suggest that Translationally Controlled Tumor Protein (TCTP) is a critical survival factor of stem cells including cancer stem cells. Here, we aimed to determine whether the TCTP inhibitor sertraline (STL) could target prostate cancer stem cells (PCSC). In colony formation, spheroidogenesis, angiogenesis, and wound healing assays STL showed a robust inhibition of tumorigenic (colony growth), angiogenic (endothelial tube formation) and metastatic (wound healing and migration) potential of PCSC. Interestingly, antioxidants such as N-acetyl cysteine (NAC), Glutathione (GSH) and catalase effectively blocked the cytotoxicity effect of STL on PCSC implicating oxidative stress as the underlying anti-PCSC targeting mechanism. Cell cycle analysis showed a robust G0 arrest in PCSC exposed to STL. Notably, STL induced both apoptosis and autophagy by activating free radical generation, hydrogen peroxide formation (H2O2), lipid peroxidation (LPO) and depleted the levels of glutathione (GSH). Moreover, surface marker expression analysis using confocal revealed that STL significantly down regulates the expression levels of aldehyde dehydrogenase 1 (ALDH1) and cluster of differentiation 44 (CD44) stem cell markers. Furthermore, in western blot analysis, STL treatment applied in a dose-dependent manner, caused a marked decrease in TCTP, phospho TCTP, anti-apoptotic markers survivin and cellular inhibitor of apoptosis protein 1 (cIAP1) expression as well as a significant increase in cleaved caspase3 and cleaved Poly [ADP-ribose] polymerase 1 (PARP-1) expression. Of note, STL also significantly down regulated the stem cell markers (ALDH1 and CD44) and epithelial to mesenchymal transition (EMT) markers such as transcription factor 8 (TCF8) and lymphoid enhancer-binding factor-1 (LEF1) expression levels. Concurrently, STL increased the levels of autophagy markers such as light chain (LC3), Beclin1 and autophagy-related gene (ATG5). Taken together, our study suggests that STL could be an effective therapeutic agent in eliminating prostate cancer stem cells.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Biomedical Sciences, College of Medicine, University of Illinois Rockford, IL, USA
| | - Velavan Bakthavachalam
- Department of Biomedical Sciences, College of Medicine, University of Illinois Rockford, IL, USA
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, College of Medicine, University of Illinois Rockford, IL, USA
| |
Collapse
|
9
|
Ma Q, Liao H, Xu L, Li Q, Zou J, Sun R, Xiao D, Liu C, Pu W, Cheng J, Zhou X, Huang G, Yao L, Zhong X, Guo X. Autophagy-dependent cell cycle arrest in esophageal cancer cells exposed to dihydroartemisinin. Chin Med 2020; 15:37. [PMID: 32351616 PMCID: PMC7183693 DOI: 10.1186/s13020-020-00318-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/14/2020] [Indexed: 12/19/2022] Open
Abstract
Background Dihydroartemisinin (DHA), a derivate of artemisinin, is an effective antimalarial agent. DHA has been shown to exert anticancer activities to numerous cancer cells in the past few years, while the exact molecular mechanisms remain to be elucidated, especially in esophageal cancer. Methods Crystal violet assay was conducted to determine the cell viability of human esophageal cancer cell line Eca109 treated with DHA. Tumor-bearing nude mice were employed to evaluate the anticancer effect of DHA in vivo. Soft agar and crystal violet assays were used to measure the tumorigenicity of Eca109 cells. Flow cytometry was performed to evaluate ROS or cell cycle distribution. GFP-LC3 plasmids were delivered into Eca109 cells to visualize autophagy induced by DHA under a fluorescence microscope. The mRNA and protein levels of each gene were tested by qRT-PCR and western blot, respectively. Results Our results proved that DHA significantly reduced the viability of Eca109 cells in a dose- and time-dependent manner. Further investigation showed that DHA evidently induced cell cycle arrest at the G2/M phase in Eca109 cells. Mechanistically, DHA induced intracellular ROS generation and autophagy in Eca109 cells, while blocking ROS by an antioxidant NAC obviously inhibited autophagy. Furthermore, we found that telomere shelterin component TRF2 was down-regulated in Eca109 cells exposed to DHA through autophagy-dependent degradation, which could be rescued after autophagy was blocked by ROS inhibition. Moreover, the DNA damage response (DDR) was induced obviously in DHA treated cells. To further explore whether ROS or autophagy played a vital role in DHA induced cell cycle arrest, the cell cycle distribution of Eca109 cells was evaluated after ROS or autophagy blocking, and the results showed that autophagy, but not ROS, was essential for cell cycle arrest in DHA treated cells. Conclusion Taken together, DHA showed anticancer effect on esophageal cancer cells through autophagy-dependent cell cycle arrest at the G2/M phase, which unveiled a novel mechanism of DHA as a chemotherapeutic agent, and the degradation of TRF2 followed by DDR might be responsible for this cell phenotype.
Collapse
Affiliation(s)
- Qiang Ma
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Hebin Liao
- 2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Lei Xu
- 2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Qingrong Li
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Jiang Zou
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Ru Sun
- 3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,4Department of Blood Transfusion, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Dan Xiao
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Chang Liu
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Wenjie Pu
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Jibing Cheng
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Xi Zhou
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Guangcheng Huang
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Lihua Yao
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Xiaowu Zhong
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| | - Xiaolan Guo
- 1Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,2Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637000 People's Republic of China.,3Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637000 People's Republic of China
| |
Collapse
|
10
|
Liu P, Zhang B, Chen Z, He Y, Du Y, Liu Y, Chen X. m 6A-induced lncRNA MALAT1 aggravates renal fibrogenesis in obstructive nephropathy through the miR-145/FAK pathway. Aging (Albany NY) 2020; 12:5280-5299. [PMID: 32203053 PMCID: PMC7138587 DOI: 10.18632/aging.102950] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 03/09/2020] [Indexed: 01/15/2023]
Abstract
Renal fibrosis is a key factor in chronic kidney disease (CKD). Long non-coding RNAs (lncRNAs) play important roles in the physiological and pathological progression of human diseases. However, the roles and underlying mechanisms of lncRNAs in renal fibrosis still need to be discovered. In this study, we first displayed the increased lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) expression in renal fibrosis in patients with obstructive nephropathy (ON). Then we found that transforming growth factor beta 1 (TGF-β1) induced epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) protein deposition, which promoted the viability, proliferation and migration of human renal proximal tubular epithelial (HK2) cells. Next, MALAT1/miR-145/focal adhesion kinase (FAK) pathway was confirmed to play an importment role in TGF-β1-induced renal fibrosis. In addition, the MALAT1/miR-145/FAK pathway was involved in the effect of dihydroartemisinin (DHA) on TGF-β1-induced renal fibrosis in vitro and in vivo. Furthermore, m6A methyltransferase methyltransferase-like 3 (METTL3) was shown to be the main methyltransferase of m6A modification on MALAT1.
Collapse
Affiliation(s)
- Peihua Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Bo Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhi Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yao He
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yongchao Du
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Yuhang Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Xiang Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| |
Collapse
|
11
|
Shen S, Li K, Liu Y, Liu X, Liu B, Ba Y, Xing W. Silencing lncRNA AGAP2-AS1 Upregulates miR-195-5p to Repress Migration and Invasion of EC Cells via the Decrease of FOSL1 Expression. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:331-344. [PMID: 32199129 PMCID: PMC7082499 DOI: 10.1016/j.omtn.2019.12.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 12/25/2022]
Abstract
The interaction of long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and mRNAs has been implicated in various types of cancers, including esophageal cancer (EC). The current study aimed to investigate the role of AGAP2-AS1/miR-195-5p/Fos-like antigen-1 (FOSL1) in EC progression. The expression of AGAP2-AS1, miR-195-5p, and FOSL1 in tumor tissues isolated from EC patients and EC cell lines was determined by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), the results of which illustrated that AGAP2-AS1 and FOSL1 were increased while miR-195-5p was reduced in EC. Next, the ectopic expression, knockdown, and reporter assay experiments were all employed to elucidate the mechanism of AGAP2-AS1/miR-195-5p/FOSL1 in the processes of EC cell proliferation, cell cycle, apoptosis, invasion, and migration as well as tumor growth. Knockdown of AGAP2-AS1 or overexpression of miR-195-5p reduced EC cell proliferation, migration, and invasion, blocked cell cycle entry, and elevated apoptosis. FOSL1 was found to be specifically targeted by miR-195-5p. AGAP2-AS1 was observed to upregulate FOSL1 by binding to miR-195-5p. Silencing of AGAP2-AS1 was observed to restrain the development of EC both in vitro and in vivo through upregulating miR-195-5p and downregulating FOSL1. Taken together, AGAP2-AS1 knockdown exercises suppressive effects on the development of EC through miR-195-5p-dependent downregulation of FOSL1. Therefore, targeting AGAP2-AS1 could be a future direction to develop a novel molecule-targeted therapeutic strategy for EC.
Collapse
Affiliation(s)
- Sining Shen
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450008, P.R. China.
| | - Ke Li
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450008, P.R. China
| | - Ying Liu
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450008, P.R. China
| | - Xianben Liu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450008, P.R. China
| | - Baoxing Liu
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450008, P.R. China
| | - Yufeng Ba
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450008, P.R. China
| | - Wenqun Xing
- Department of Thoracic Surgery, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou 450008, P.R. China
| |
Collapse
|
12
|
Wang T, Luo R, Li W, Yan H, Xie S, Xiao W, Wang Y, Chen B, Bai P, Xing J. Dihydroartemisinin suppresses bladder cancer cell invasion and migration by regulating KDM3A and p21. J Cancer 2020; 11:1115-1124. [PMID: 31956358 PMCID: PMC6959076 DOI: 10.7150/jca.36174] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022] Open
Abstract
Emerging evidences have shown that Dihydroartemisinin (DHA), used in malaria treatment, possess anti-cancer activity. However, the study of its potential functional roles and the anti-cancer mechanisms in bladder cancer is limited. We performed this study to elucidate the influence of DHA in the biological behavior of bladder cancer cells and tried to explore the molecular mechanism. The results of CCK-8 assay showed that DHA significantly inhibited bladder cancer cell 5637, UMUC3 and T24 proliferation and the inhibitory effect is dose- and time- dependent. Further mechanism study showed that DHA performed its function via down-regulating the expression of histone demethylase KDM3A and inducing p21 expression. Moreover, wound healing and transwell migration/invasion assays revealed that DHA inhibited the ability of migration and metastasis in bladder cancer cell line T24. Finally, flow cytometry and colony formation assays demonstrated that DHA significantly promoted apoptosis of T24 cells and suppressed tumorigenesis as expected. Taken together, our study identifies the anti-cancer capacity of DHA in bladder cancer and explores the underlying mechanism.
Collapse
Affiliation(s)
- Tao Wang
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Rongtuan Luo
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Wei Li
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Houyu Yan
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Shunqiang Xie
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Wen Xiao
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Yongfeng Wang
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Bin Chen
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Peide Bai
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| | - Jinchun Xing
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology Surgery, The First Affiliated Hospital, School of Medicine, Xiamen University, Xiamen, China 361003
| |
Collapse
|
13
|
Huang XT, Liu W, Zhou Y, Hao CX, Zhou Y, Zhang CY, Sun CC, Luo ZQ, Tang SY. Dihydroartemisinin attenuates lipopolysaccharide‑induced acute lung injury in mice by suppressing NF‑κB signaling in an Nrf2‑dependent manner. Int J Mol Med 2019; 44:2213-2222. [PMID: 31661121 PMCID: PMC6844637 DOI: 10.3892/ijmm.2019.4387] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 10/11/2019] [Indexed: 12/20/2022] Open
Abstract
Acute lung injury (ALI) is a severe health issue with significant morbidity and mortality. Artemisinin is used for the treatment of fever and malaria in clinical practice. Dihydroartemisinin (DHA), the major active metabolite of artemisinin, plays a role in anti‑organizational fibrosis and anti‑neuronal cell death. However, whether DHA can attenuate ALI remains unclear. The current study thus examined the effects of DHA on ALI and primary macrophages. The results revealed that DHA attenuated lipopolysaccharide (LPS)‑induced pulmonary pathological damage. DHA suppressed the LPS‑induced infiltration of inflammatory cells, the elevation of myeloperoxidase activity, oxidative stress and the production of pro‑inflammatory cytokines, including interleukin (IL)‑1β, tumor necrosis factor‑α, and IL‑6. Furthermore, DHA reduced the LPS‑induced inflammatory response by suppressing the degradation of I‑κB and the nuclear translocation of nuclear factor κ‑light‑chain‑enhancer of activated B cells (NF‑κB)/p65 in vivo and in vitro. DHA activated the nuclear factor‑erythroid 2 related factor 2 (Nrf2) pathway, which was suppressed by LPS treatment. The Nrf2 inhibitor, ML385, diminished the protective effects of DHA against LPS‑induced inflammation in macrophages. On the whole, the findings of this study demonstrate that DHA exerts therapeutic effects against LPS‑induced ALI by inhibiting the Nrf2‑mediated NF‑κB activation in macrophages. The present study also confirmed the therapeutic effects of DHA in mice with LPS‑induced ALI. Thus, these findings demonstrate that DHA exhibits anti‑inflammatory activities and may be a therapeutic candidate for the treatment of ALI.
Collapse
Affiliation(s)
- Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Cai-Xia Hao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zi-Qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, P.R. China
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|