1
|
Deng ZT, Liang SF, Huang GK, Wang YQ, Tu XY, Zhang YN, Li S, Liu T, Cheng BB. Autophagy plays a pro-apoptotic role in arsenic trioxide-induced cell death of liver cancer. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:295-302. [PMID: 38599914 DOI: 10.1016/j.joim.2024.03.011] [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: 08/15/2023] [Accepted: 11/18/2023] [Indexed: 04/12/2024]
Abstract
OBJECTIVE The effects of arsenic trioxide (As2O3) on hepatocellular carcinoma have been documented widely. Autophagy plays dual roles in the survival and death of cancer cells. Therefore, we investigated the exact role of autophagy in As2O3-induced apoptosis in liver cancer cells. METHODS The viability of hepatoma cells was determined using the MTT assay with or without fetal bovine serum. The rate of apoptosis in liver cancer cells treated with As2O3 was evaluated using flow cytometry, Hoechst 33258 staining, and TUNEL assays. The rate of autophagy among liver cancer cells treated with As2O3 was detected using immunofluorescence, Western blot assay and transmission electron microscopy. RESULTS Upon treatment with As2O3, the viability of HepG2 and SMMC-7721 cells was decreased in a time- and dose-dependent manner. The apoptosis rates of both liver cancer cell lines increased with the concentration of As2O3, as shown by flow cytometry. Apoptosis in liver cancer cells treated with As2O3 was also shown by the activation of the caspase cascade and the regulation of Bcl-2/Bax expression. Furthermore, As2O3 treatment induced autophagy in liver cancer cells; this finding was supported by Western blot, immunofluorescence of LC3-II and beclin 1, and transmission electron microscopy. In liver cancer cells, As2O3 inhibited the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signal pathway that plays a vital role in both apoptosis and autophagy. The PI3K activator SC-79 partially reversed As2O3-induced autophagy and apoptosis. Furthermore, inhibiting autophagy with 3-methyladenine partially reversed the negative effects of As2O3 on cell viability. Serum starvation increased autophagy and amplified the effect of As2O3 on cell death. CONCLUSION As2O3 induces apoptosis and autophagy in liver cancer cells. Autophagy induced by As2O3 may have a proapoptotic effect that helps to reduce the viability of liver cancer cells. This study provides novel insights into the effects of As2O3 against liver cancer. Please cite this article as: Deng ZT, Liang SF, Huang GK, Wang YQ, Tu XY, Zhang YN, Li S, Liu T, Cheng BB. Autophagy plays a pro-apoptotic role in arsenic trioxide-induced cell death of liver cancer. J Integr Med. 2024; 22(3): 295-302.
Collapse
Affiliation(s)
- Zheng-Ting Deng
- Department of Respiration, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Department of Febrile Diseases, School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Shu-Fang Liang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Guo-Kai Huang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Yu-Qian Wang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xiao-Yu Tu
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Ya-Ni Zhang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Tao Liu
- Department of Febrile Diseases, School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Bin-Bin Cheng
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
2
|
Yan M, Wang H, Wei R, Li W. Arsenic trioxide: applications, mechanisms of action, toxicity and rescue strategies to date. Arch Pharm Res 2024; 47:249-271. [PMID: 38147202 DOI: 10.1007/s12272-023-01481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023]
Abstract
Arsenical medicine has obtained its status in traditional Chinese medicine for more than 2,000 years. In the 1970s, arsenic trioxide was identified to have high efficacy and potency for the treatment of acute promyelocytic leukemia, which promoted many studies on the therapeutic effects of arsenic trioxide. Currently, arsenic trioxide is widely used to treat acute promyelocytic leukemia and various solid tumors through various mechanisms of action in clinical practice; however, it is accompanied by a series of adverse reactions, especially cardiac toxicity. This review presents a comprehensive overview of arsenic trioxide from preclinical and clinical efficacy, potential mechanisms of action, toxicities, and rescue strategies for toxicities to provide guidance or assistance for the clinical application of arsenic trioxide.
Collapse
Affiliation(s)
- Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Hao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Rui Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Pharmacy Department, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Yang D, Xia X, Xi S. Salvianolic acid A attenuates arsenic-induced ferroptosis and kidney injury via HIF-2α/DUOX1/GPX4 and iron homeostasis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 907:168073. [PMID: 37879473 DOI: 10.1016/j.scitotenv.2023.168073] [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: 09/17/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/27/2023]
Abstract
Arsenic (As) is a prevalent pollutant in the environment and causes a high frequency of kidney disease in areas of high arsenic contamination, but its pathogenic mechanisms have yet to be completely clarified. Ferroptosis is a new form of cell death mainly dependent on lipid peroxidation and iron accumulation. Several reports have suggested that ferroptosis is operative in a spectrum of diseases caused by arsenic exposure, including in the lungs, pancreas, and testis. However, the mechanism and exact role of ferroptosis in arsenic-induced kidney injury is not known. Firstly, by constructing in vivo and in vitro arsenic exposure models, we confirmed the occurrence of ferroptosis based on the identification of the ability of NaASO2 to cause kidney injury. In addition, we found that arsenic exposure could upregulate DUOX1 expression in kidney and HK-2 cells, and after knocking down DUOX1 using siRNA was able to significantly upregulate GPX4 expression and attenuate ferroptosis. Subsequently using bioinformatics, we identified and proved the involvement of HIF-2α in the course of ferroptosis, and further confirmed by dual luciferase assay that HIF-2α promotes DUOX1 transcription to increase its expression. Finally, intervention with the natural ingredient SAA significantly attenuated arsenic-induced ferroptosis and kidney injury. These results showed that arsenic could cause ferroptosis and kidney injury by affecting HIF-2α/DUOX1/GPX4 and iron homeostasis and that SAA was an effective intervention component. This study not only discovered the molecular mechanism of sodium arsenite-induced kidney injury but also explored an active ingredient with intervention potential, providing a scientific basis for the prevention and treatment of sodium arsenite-induced kidney injury.
Collapse
Affiliation(s)
- Desheng Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenicy, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Xinyu Xia
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenicy, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China
| | - Shuhua Xi
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning 110122, China; The Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenicy, Shenyang, Liaoning 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
4
|
Wang M, Yin J, Han Q, Li B, Zhao XW, Xue L. Arsenic Trioxide Suppresses Angiogenesis in Non-small Cell Lung Cancer via the Nrf2-IL-33 Signaling Pathway. Anticancer Agents Med Chem 2024; 24:1142-1150. [PMID: 38847245 DOI: 10.2174/0118715206288348240420174853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/29/2024] [Accepted: 03/13/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Non-Small Cell Lung Cancer (NSCLC) ranks as a leading cause of cancer-related mortality, necessitating the urgent search for cost-effective and efficient anti-NSCLC drugs. Our preliminary research has demonstrated that arsenic trioxide (ATO) significantly inhibits NSCLC angiogenesis, exerting anti-tumor effects. In conjunction with existing literature reports, the Nrf2-IL-33 pathway is emerging as a novel mechanism in NSCLC angiogenesis. OBJECTIVE This study aimed to elucidate whether ATO can inhibit NSCLC angiogenesis through the Nrf2-IL-33 pathway. METHODS Immunohistochemistry was employed to assess the expression of Nrf2, IL-33, and CD31 in tumor tissues from patients with NSCLC. DETA-NONOate was used as a nitric oxide (NO) donor to mimic high levels of NO in the tumor microenvironment. Western blot, quantitative real-time PCR, and enzyme-linked immunosorbent assay were utilized to evaluate the expression of Nrf2 and IL-33 in the NCI-H1299 cell line. Subcutaneous xenograft models were established in nude mice by implanting NCI-H1299 cells to assess the anti-tumor efficacy of ATO. RESULTS High expression levels of Nrf2 and IL-33 were observed in tumor samples from patients with NSCLC, and Nrf2 expression positively correlated with microvascular density in NSCLC. In vitro, NO (released from 1mM DETA-NONOate) promoted activation of the Nrf2-IL-33 signaling pathway in NCI-H1299 cells, which was reversed by ATO. Additionally, both Nrf2 deficiency and ATO treatment significantly attenuated NOinduced IL-33 expression. In vivo, both ATO and the Nrf2 inhibitor ML385 demonstrated significant inhibitory effects on angiogenesis tumor growth. CONCLUSION Nrf2-IL-33 signaling is usually activated in NSCLC and positively correlates with tumor angiogenesis. ATO effectively disrupts the activation of the Nrf2-IL-33 pathway in NSCLC and thus inhibits angiogenesis, suggesting its potential as an anti-angiogenic agent for use in the treatment of NSCLC.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Angiogenesis
- Angiogenesis Inhibitors/pharmacology
- Angiogenesis Inhibitors/chemistry
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Arsenic Trioxide/pharmacology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Interleukin-33/metabolism
- Interleukin-33/antagonists & inhibitors
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice, Inbred BALB C
- Mice, Nude
- Molecular Structure
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/metabolism
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- NF-E2-Related Factor 2/metabolism
- NF-E2-Related Factor 2/antagonists & inhibitors
- Signal Transduction/drug effects
- Structure-Activity Relationship
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Mingdong Wang
- Department of Thoracic Surgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
- Department of Thoracic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai, 200434, China
| | - Jizhong Yin
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Qianyu Han
- Department of Thoracic Surgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| | - Bing Li
- Department of Respiratory and Critical Care Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai, 200434, China
| | - Xue-Wei Zhao
- Department of Thoracic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai, 200434, China
| | - Lei Xue
- Department of Thoracic Surgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai, 200003, China
| |
Collapse
|
5
|
Jiang Y, Li J, Liu Y, Shen X, Li J, Zhi F, Xu J, Li X, Shao T, Xu Y. Open a new epoch of arsenic trioxide investigation: ATOdb. Comput Biol Med 2023; 165:107465. [PMID: 37699323 DOI: 10.1016/j.compbiomed.2023.107465] [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: 06/27/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
Arsenic trioxide (ATO) is a great discovery in the treatment of acute promyelocytic leukemia (APL), which has been used in an increasing number of malignant diseases. Systematic integrative analysis will help to precisely understand the mechanism of ATO and find new combined drugs. Therefore, we developed a one-stop comprehensive database of ATO named ATOdb by manually compiling a wealth of experimentally supported ATO-related data from 3479 articles, and integrated analysis tools. The current version of ATOdb contains 8373 associations among 2300 ATO targets, 80 conditions and 262 combined drugs. Each entry in ATOdb contains detailed information on ATO targets, therapeutic/side effects, systems, cell names, cell types, regulations, detection methods, brief descriptions, references, etc. Furthermore, ATOdb also provides data visualization and analysis results such as the drug similarities, protein-protein interactions, and miRNA-mRNA relationships. An easy-to-use web interface was deployed in ATOdb for users to easily browse, search and download the data. In conclusion, ATOdb will serve as a valuable resource for in-depth study of the mechanism of ATO, discovery of new drug combination strategies, promotion of rational drug use and individualized treatments. ATOdb is freely available at http://bio-bigdata.hrbmu.edu.cn/ATOdb/index.jsp.
Collapse
Affiliation(s)
- Yanan Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| | - Jianing Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yujie Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Xiuyun Shen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Junyi Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Fengnan Zhi
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Tingting Shao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yingqi Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; Hohhot Mongolian Medicine of Traditional Chinese Medicine Hospital, Hohhot, 010110, China.
| |
Collapse
|
6
|
Gargalionis AN, Papavassiliou KA, Papavassiliou AG. Implication of mTOR Signaling in NSCLC: Mechanisms and Therapeutic Perspectives. Cells 2023; 12:2014. [PMID: 37566093 PMCID: PMC10416991 DOI: 10.3390/cells12152014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/28/2023] [Accepted: 08/06/2023] [Indexed: 08/12/2023] Open
Abstract
Mechanistic target of the rapamycin (mTOR) signaling pathway represents a central cellular kinase that controls cell survival and metabolism. Increased mTOR activation, along with upregulation of respective upstream and downstream signaling components, have been established as oncogenic features in cancer cells in various tumor types. Nevertheless, mTOR pathway therapeutic targeting has been proven to be quite challenging in various clinical settings. Non-small cell lung cancer (NSCLC) is a frequent type of solid tumor in both genders, where aberrant regulation of the mTOR pathway contributes to the development of oncogenesis, apoptosis resistance, angiogenesis, cancer progression, and metastasis. In this context, the outcome of mTOR pathway targeting in clinical trials still demonstrates unsatisfactory results. Herewith, we discuss recent findings regarding the mechanisms and therapeutic targeting of mTOR signaling networks in NSCLC, as well as future perspectives for the efficient application of treatments against mTOR and related protein molecules.
Collapse
Affiliation(s)
- Antonios N. Gargalionis
- Department of Biopathology, ‘Eginition’ Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|