1
|
Wu B, Zheng M, Guo Q, Wang N, Zhu C, Zhao W, Xu Y. Early potential metabolic biomarkers of T1 stage lung adenocarcinoma based on serum metabolomics. Front Mol Biosci 2025; 12:1544774. [PMID: 40351532 PMCID: PMC12061714 DOI: 10.3389/fmolb.2025.1544774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Background This study aims to investigate serum metabolite changes in patients with early-stage (T1) lung adenocarcinoma, identify potential diagnostic biomarkers, and establish an early warning mechanism for T1 stage lung adenocarcinoma. Methods The study included two groups: a lung adenocarcinoma group and a healthy control group. Serum samples underwent non-targeted metabolomics analysis. Total ion chromatograms (TIC) were generated to assess system stability. Chromatographic data were analyzed using multivariate statistical methods, including principal component analysis (PCA) for dimensionality reduction. Partial least squares discriminant analysis (PLS-DA) further validated PCA findings. Variables with VIP scores >1.0 in the PLS-DA model were selected, combined with ANOVA and T-tests (P < 0.05), to identify differentially expressed metabolites. Receiver operating characteristic (ROC) curve analysis was conducted to evaluate the diagnostic performance of selected metabolites. Results Serum metabolites significantly differed between the lung adenocarcinoma group and the healthy control group. Multivariate statistical analysis and ROC curve evaluation identified four potential diagnostic biomarkers: Cortisol, 3-Oxo-OPC4-CoA, PE-NMe(14:1(9Z)/14:1(9Z)), and Ceramide (d18:1/9Z-18:1), with AUC values of 0.930, 0.895, 0.890, and 0.795, respectively. Conclusion Cortisol,3-Oxo-OPC4-CoA,PE-NMe(14:1(9Z)/14:1(9Z)), and Ceramide (d18:1/9Z-18:1) exhibit significantly altered metabolic levels in T1 stage lung adenocarcinoma patients and can serve as metabolic biomarkers. These markers may enhance the sensitivity and specificity of early diagnosis, facilitating improved detection of T1 stage lung adenocarcinoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ye Xu
- Department of Thoracic Surgery, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Liang TL, Chen Y, Zhou NJ, Shu X, Mi JN, Ma GY, Xiao Y, Yang X, Huang C, Li JX, Xie Y, Yan PY, Yao XJ, Liu L, Pan HD, Leung ELH, Li RZ. Taurine and proline promote lung tumour growth by co-regulating Azgp1/mTOR signalling pathway. NPJ Precis Oncol 2025; 9:90. [PMID: 40155495 PMCID: PMC11953302 DOI: 10.1038/s41698-025-00872-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
Accurate metabolic biomarkers for lung cancer prognosis remain scarce but crucial. Taurine and proline, two metabolites, are consistently elevated across various cancer stages in previous studies, hinting at their potential role in disease progression. This study is the first to reveal how these metabolites contribute to poor prognosis. Transcriptomic analysis uncovered that taurine and proline downregulated Zinc-α2-glycoprotein (Azgp1), a gene linked to key metabolic pathways. Additionally, Azgp1 could also significantly affect downstream lipid metabolic pathways in lung cancer. Both taurine and proline influenced lipid metabolism via mammalian target of rapamycin (mTOR). When Azgp1 was overexpressed, lung cancer progression slowed significantly, alongside reduced mTOR activity. These findings underscore the pro-cancer role of taurine and proline, highlighting the Azgp1/mTOR axis as a vital, yet overlooked, pathway in lung cancer. This study not only advances our understanding but also identifies new therapeutic avenues.
Collapse
Affiliation(s)
- Tu-Liang Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
| | - Ying Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
| | - Nan-Jie Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
| | - Xiao Shu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
| | - Jia-Ning Mi
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
| | - Gang-Yuan Ma
- Guangzhou Medical University, Guangzhou, 510182, PR China
- Guangzhou Laboratory, Guangzhou, 510005, PR China
| | - Yao Xiao
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
| | - Xi Yang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), PR China
| | - Chen Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), PR China
| | - Jia-Xin Li
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
| | - Pei-Yu Yan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), PR China
| | - Xiao-Jun Yao
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, 999078, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China
- Guangzhou Laboratory, Guangzhou, 510005, PR China
| | - Hu-Dan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China.
| | - Run-Ze Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome/Chinese Medicine, Guangdong Laboratory, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, PR China.
| |
Collapse
|
3
|
Yan HX, Zhang YZ, Niu YQ, Wang YW, Liu LH, Tang YP, Huang JM, Leung ELH. Investigating the interaction between calcium signaling and ferroptosis for novel cancer treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156377. [PMID: 39798340 DOI: 10.1016/j.phymed.2025.156377] [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: 08/22/2024] [Revised: 12/02/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Drug resistance in cancer is steadily rising, making the development of new therapeutic targets increasingly critical for improving treatment outcomes. PURPOSE The mutual regulation of ions is essential for cell growth. Based on this concept, ion interference strategies offer a highly effective approach for cancer treatment. Calcium ions (Ca2+), as major second messengers, are closely associated with ion exchange and homeostasis. Disruptions in this balance can lead to cell death. However, while iron ions are also crucial, the connection between Ca2+and iron-induced cell death (ferroptosis) has not been well established. Therefore, this study suggests that Ca2+ may play a role in the induction of ferroptosis, presenting a novel and efficient target for cancer therapy. STUDY DESIGN PubMed, Google Scholar, and Web of Science databases were systematically searched for articles published in the past 15 years on the mechanisms of calcium ion-induced ferroptosis in cancer and related drugs. RESULTS The analysis highlights how Ca2+regulate ferroptosis. The mechanisms by which Ca2+influence ferroptosis are summarized based on existing literature, and relevant drugs that act on Ca2+/ferroptosis axis are outlined. CONCLUSION Ca2+ regulate ferroptosis primarily through the modulation of reactive oxygen species (ROS) and glutathione (GSH) levels, a mechanism that applies to a wide range of cancer cells as well as paracancerous and normal cells in cancer treatment. Furthermore, plant-derived active compounds exhibit potent anticancer properties and often act on the Ca2+/ferroptosis axis. These natural compounds could play a significant role in the development of new cancer treatment strategies.
Collapse
Affiliation(s)
- Hao-Xin Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), PR China
| | - Yu-Qing Niu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China
| | - Yu-Wei Wang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China
| | - Li-Hua Liu
- Economics and Management Yanbian University, Yanji, PR China
| | - Yu-Ping Tang
- Key Laboratory of Shanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shanxi University of Chinese Medicine, Xianyang 712046, Shanxi Province, PR China.
| | - Ju-Min Huang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), PR China.
| |
Collapse
|
4
|
Zhong ZT, Wang XY, Pan Y, Zhou K, Chen JH, Gao YQ, Dai B, Zhou ZL, Wang RQ. AMPK: An energy sensor for non-small cell lung cancer progression and treatment. Pharmacol Res 2025; 212:107592. [PMID: 39805353 DOI: 10.1016/j.phrs.2025.107592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Lung cancer (LC) is the leading cause of cancer-related morbidity and mortality in China, with non-small cell lung cancer (NSCLC) accounting for 85 % of the overall lung cancer cases. AMP-activated protein kinase (AMPK) is a key regulator of energy balance and homeostasis, and its dysregulation is a common feature in various malignancies, particularly in NSCLC with mutations in Liver kinase B1 (LKB1). Studies have shown that the AMPK signalling pathway has a dual role in NSCLC progression, both inhibiting and promoting the progression of malignant tumours. Therefore, drugs targeting the AMPK signalling pathway may hold significant promise for therapeutic application in NSCLC. This review aims to examine the manifestations and mechanisms by which AMPK and its associated signalling molecules influence NSCLC progression and treatment. Firstly, we discuss the critical importance of AMPK within the mutational context of NSCLC. Secondly, we summarise the drugs and related substances that modulate the AMPK signalling pathway in NSCLC and evaluate the evidence from preclinical studies on combination AMPK-targeted therapies to address the issue of drug resistance in NSCLC under current clinical treatments. In summary, this paper highlights the critical importance of developing AMPK-targeted drugs to enhance therapeutic efficacy in NSCLC, as well as the potential for applying these drugs in clinical therapy to overcome drug resistance.
Collapse
Affiliation(s)
- Zhi-Ting Zhong
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China; College of Pharmacy, Jinan University, Guangzhou, China
| | - Xu-Yan Wang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ying Pan
- Department of Oncology, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ke Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Jing-Hui Chen
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Yu-Qi Gao
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Bo Dai
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan City, Guangdong Province 528200, China.
| | - Zhi-Ling Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| | - Rui-Qi Wang
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| |
Collapse
|
5
|
Wang Q, Xu W, Ying L, Shi H, Sun Y, Feng W, Xu H, Xie J, Wei H, Yang Z, Wang H. Preliminary study of the mechanism of isolinderalactone inhibiting the malignant behavior of bladder cancer. Curr Urol 2025; 19:49-58. [PMID: 40313422 PMCID: PMC12042191 DOI: 10.1097/cu9.0000000000000259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 03/19/2024] [Indexed: 05/03/2025] Open
Abstract
Background Isolinderalactone (ILL), extracted from the dried tubers of Linderae aggregate, has multiple functions, such as antioxidation, antitumor, and anti-infection effects. However, there have been few studies on ILL's antitumor role and no reports on its role in bladder cancer (BC). Materials and methods Human BC cell lines T24 and EJ-1 were treated with different concentrations of ILL (0, 10, 20, 50, 100, 200, 400, 600, 800, and 1000 μmol/L), and the cell proliferation inhibition rate was analyzed using the CCK-8 assay. The effect of ILL on T24 and EJ-1 cell cycle inhibition and apoptosis was examined using flow cytometry. Immunoblotting was used to detect the levels of apoptosis-related proteins, BAX, BAK1, and CYCS, in BC cells of the control and ILL-treated groups, and quantitative PCR experiments were performed to detect the apoptosis-related gene expression of CASP10, CYCS, BAX, BCL-2, CASP8, and BAK1. T24 and EJ-1 tumor-bearing mouse models were established and divided into vehicle control, low-dose (10 mg/kg) and high-dose (20 mg/kg) groups, with 5 mice in each group. Hematoxylin and eosin staining and immunohistochemistry were used to detect changes in apoptosis-related proteins in vivo. Results The CCK-8 assay showed that in vitro, ILL significantly inhibited the proliferation of the T24 and EJ-1 BC cell lines. Flow cytometry and immunoblotting results showed that ILL increased mitochondrial permeability by upregulating proapoptotic proteins BAK1 and BAX, promoting CYCS release and significantly inducing cell cycle arrest at G0/G1 phase. In vivo, on day 25 of administration, tumor inhibition rates in T24 and EJ-1 tumor-bearing mice were up to 75.24% and 47.43%, respectively, in the ILL high-dose-treated and 71.58% and 43.89%, respectively, in the ILL low-dose-treated groups. Conclusions Isolinderalactone controls BC progression by inducing apoptosis, suggesting that ILL may be an effective drug for the treatment of BC.
Collapse
Affiliation(s)
- Qun Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenkai Xu
- Department of Urology, The Second People's Hospital of Xindu District, Chengdu, China
| | - Lu Ying
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
- College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, Xinjiang, China
| | - Hongjin Shi
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuxin Sun
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wei Feng
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haole Xu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Xie
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hairong Wei
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Yang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
- College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar, Xinjiang, China
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
6
|
Du CM, Leu WJ, Jiang YH, Chan SH, Chen IS, Chang HS, Hsu LC, Hsu JL, Guh JH. Cardenolide glycosides sensitize gefitinib-induced apoptosis in non-small cell lung cancer: inhibition of Na +/K +-ATPase serving as a switch-on mechanism. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6533-6550. [PMID: 38451282 DOI: 10.1007/s00210-024-03031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
The treatment of non-small cell lung cancer (NSCLC) is known as a significant level of unmet medical need in spite of the progress in targeted therapy and personalized therapy. Overexpression of the Na+/K+-ATPase contributes to NSCLC progression, suggesting its potentiality in antineoplastic approaches. Epi-reevesioside F, purified from Reevesia formosana, showed potent anti-NSCLC activity through inhibiting the Na+/K+-ATPase, leading to internalization of α1- and α3-subunits in Na+/K+-ATPase and suppression of Akt-independent mTOR-p70S6K-4EBP1 axis. Epi-reevesioside F caused a synergistic amplification of apoptosis induced by gefitinib but not cisplatin, docetaxel, etoposide, paclitaxel, or vinorelbine in both NCI-H460 and A549 cells. The synergism was validated by enhanced activation of the caspase cascade. Bax cleavage, tBid formation, and downregulation of Bcl-xL and Bcl-2 contributed to the synergistic apoptosis induced by the combination treatment of epi-reevesioside F and gefitinib. The increase of membrane DR4 and DR5 levels, intracellular Ca2+ concentrations, and active m-calpain expression were responsible for the caspase-8 activation and Bax cleavage. The increased α-tubulin acetylation and activation of MAPK (i.e., p38 MAPK, Erk, and JNK) depending on cell types contributed to the synergistic mechanism under combination treatment. These signaling pathways that converged on profound c-Myc downregulation led to synergistic apoptosis in NSCLC. In conclusion, the data suggest that epi-reevesioside F inhibits the Na+/K+-ATPase and displays potent anti-NSCLC activity. Epi-reevesioside F sensitizes gefitinib-induced apoptosis through multiple pathways that converge on c-Myc downregulation. The data support the inhibition of Na+/K+-ATPase as a switch-on mechanism to sensitize gefitinib-induced anti-NSCLC activity.
Collapse
Affiliation(s)
- Chi-Min Du
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Rd., Zhongzheng Dist, Taipei, 100, Taiwan
| | - Wohn-Jenn Leu
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Rd., Zhongzheng Dist, Taipei, 100, Taiwan
| | - Yi-Huei Jiang
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Rd., Zhongzheng Dist, Taipei, 100, Taiwan
| | - She-Hung Chan
- Department of Cosmetic Science, Providence University, 200, Sec. 7, Taiwan Boulevard, Shalu Dist, Taichung, 43301, Taiwan
| | - Ih-Sheng Chen
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Taiwan, Kaohsiung, Taiwan
| | - Hsun-Shuo Chang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Taiwan, Kaohsiung, Taiwan
| | - Lih-Ching Hsu
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Rd., Zhongzheng Dist, Taipei, 100, Taiwan
| | - Jui-Ling Hsu
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Rd., Zhongzheng Dist, Taipei, 100, Taiwan.
- Department of Nursing, Division of Basic Medical Sciences, Chang-Gung University of Science and Technology, Taoyuan, 333, Taiwan.
- Department of Medical Oncology, New Taipei Municipal TuCheng Hospital, New Taipei City, 236, Taiwan.
| | - Jih-Hwa Guh
- School of Pharmacy, National Taiwan University, No. 33, Linsen S. Rd., Zhongzheng Dist, Taipei, 100, Taiwan.
| |
Collapse
|
7
|
Yang M, Bi W, Zhang Z. Identification and validation of CCL5 as a key gene in HIV infection and pulmonary arterial hypertension. Front Cardiovasc Med 2024; 11:1417701. [PMID: 39119185 PMCID: PMC11306045 DOI: 10.3389/fcvm.2024.1417701] [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: 04/15/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024] Open
Abstract
Background The relationship between human immunodeficiency virus (HIV) infection and pulmonary arterial hypertension (PAH) has garnered significant scrutiny. Individuals with HIV infection have a higher risk of developing PAH. However, the specific mechanism of HIV-associated PAH remains unclear. Our study aims at investigating the shared biomarkers in HIV infection and PAH and predicting the potential therapeutic target for HIV-associated PAH. Methods Data for HIV infection and PAH were downloaded from Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) analysis was performed to detect shared genes in HIV infection and PAH. Enrichment analysis was conducted to identify the function of common DEGs. Protein-protein interaction (PPI) analysis was used to detect key genes. These crucial genes were subsequently verified by RT-qPCR. Finally, candidate drugs were identified by using the Drug Signatures Database (DSigDB). Results Nineteen common DEGs were identified in HIV infection and PAH. Enrichment analysis exhibited that the functions of these genes were mainly enriched in inflammatory responses, mainly including cellular immunity and interaction between viral proteins and cytokines. By constructing PPI networks, we identified the key gene CC-type chemokine ligand 5 (CCL5), and we verified that CCL5 was highly expressed in hypoxia induced human pulmonary artery endothelial cells (hPAECs) and human pulmonary artery smooth muscle cells (hPASMCs). In addition, we predicted 10 potential drugs targeting CCL5 by Autodock Vina. Conclusion This study revealed that CCL5 might be a common biomarker of HIV infection and PAH and provided a new therapeutic target for HIV-associated PAH. However, further clinical validation is still indispensable.
Collapse
Affiliation(s)
- Mengyue Yang
- Department of Cardiology, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Wen Bi
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhijie Zhang
- Department of Cardiology, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
8
|
Li D, Cheng J, Zhang W, Zhang L, Maghsoudloo M, Fu J, Liu X, Xiao X, Wei C, Fu J. Tripartite motif-containing 28 (TRIM28) expression and cordycepin inhibition in progression, prognosis, and therapeutics of patients with breast invasive carcinoma. J Cancer 2024; 15:4374-4385. [PMID: 38947392 PMCID: PMC11212093 DOI: 10.7150/jca.95876] [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: 03/02/2024] [Accepted: 05/12/2024] [Indexed: 07/02/2024] Open
Abstract
Breast cancer (BC) is the most common tumor in women worldwide. TRIM28 (RNF96) plays pleiotropic biological functions, such as silencing target genes, facilitating DNA repair, stimulating cellular proliferation and differentiation, and contributing to cancer progression. TRIM28 plays an increasingly crucial role in cancer, but its impact on BC, including breast invasive carcinoma, remains poorly understood. In the current study, analyses of online databases, quantitative real-time quantitative PCR, immunohistochemistry, and western blotting were performed on patients with breast invasive carcinoma (BRCA). Cordycepin (CD) was used to monitor BC progression and TRIM28 expression in vivo. As a result, we observed that TRIM28 is highly expressed in breast invasive carcinoma tissues compared with the corresponding normal tissues and is correlated with metastatic / invasive progression. High expression of TRIM28 might serve as a prognostic marker for long-term survival in triple-negative BC, advanced BC, or breast invasive carcinoma. Although TRIM28 methylation in tumor tissues of breast invasive carcinoma is not significantly changed compared to the matched normal tissues, the expressions and methylation of TRIM28 are significantly reversely correlated. TRIM28 expression was inhibited by CD in the mouse model, indicating its role in preventing BC progression. Thus, TRIM28 might be a potentially valuable molecular target for forecasting the progression / prognosis of patients with breast invasive carcinoma. CD, which represses BC growth/metastasis, may be involved partially through suppressing TRIM28 expression.
Collapse
Affiliation(s)
- Dabing Li
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Wenqian Zhang
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lianmei Zhang
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- Department of Pathology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an 223300, Jiangsu Province, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Jiewen Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Xiaoyan Liu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Xiuli Xiao
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
- Department of Pathology, the Affiliated Hospital, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Chunli Wei
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
9
|
Maji A, Paul A, Sarkar A, Nahar S, Bhowmik R, Samanta A, Nahata P, Ghosh B, Karmakar S, Kumar Maity T. Significance of TRAIL/Apo-2 ligand and its death receptors in apoptosis and necroptosis signalling: Implications for cancer-targeted therapeutics. Biochem Pharmacol 2024; 221:116041. [PMID: 38316367 DOI: 10.1016/j.bcp.2024.116041] [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: 10/03/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
The human immune defensesystem routinely expresses the tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), which is the most prevalent element for antitumor immunity. TRAIL associates with its death receptors (DRs), DR4 (TRAIL-R1), and DR5 (TRAIL-R2), in cancer cells to initiate the intracellular apoptosis cascade. Accordingly, numerous academic institutions and pharmaceutical companies havetried to exploreTRAIL's capacity to kill tumourcells by producing recombinant versions of it (rhTRAIL) or TRAIL receptor agonists (TRAs) [monoclonal antibody (mAb), synthetic and natural compounds, etc.] and molecules that sensitize TRAIL signalling pathway for therapeutic applications. Recently, several microRNAs (miRs) have been found to activate or inhibit death receptor signalling. Therefore, pharmacological regulation of these miRs may activate or resensitize the TRAIL DRs signal, and this is a novel approach for developing anticancer therapeutics. In this article, we will discuss TRAIL and its receptors and molecular pathways by which it induces various cell death events. We will unravel potential innovative applications of TRAIL-based therapeutics, and other investigated therapeutics targeting TRAIL-DRs and summarize the current preclinical pharmacological studies and clinical trials. Moreover, we will also emphasizea few situations where future efforts may be addressed to modulate the TRAIL signalling pathway.
Collapse
Affiliation(s)
- Avik Maji
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Abhik Paul
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Arnab Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India; Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata-700032, India.
| | - Sourin Nahar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Rudranil Bhowmik
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India; Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata-700032, India.
| | - Ajeya Samanta
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Pankaj Nahata
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad-500078, India.
| | - Sanmoy Karmakar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India; Bioequivalence Study Centre, Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata-700032, India.
| | - Tapan Kumar Maity
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700 032, India.
| |
Collapse
|
10
|
Xu Y, Xu J, Zhu W, Yan Y, Jiang X, Xie Z, Feng F, Zhang J. Bioassay-Guided Fractionation and Biological Activity of Cardenolides from Streptocaulon juventas. PLANTA MEDICA 2023; 89:1444-1456. [PMID: 37709286 DOI: 10.1055/a-2114-5371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
The discovery that Na/K-ATPase acts as a signal transducer led us to investigate the structural diversity of cardiotonic steroids and study their ligand effects. By applying Na/K-ATPase activity assay-guided fractionation, we isolated a total of 20 cardiotonic steroids from Streptocaulon juventas, including an undescribed juventasoside B (10: ) and 19 known cardiotonic steroids. Their structures have been elucidated. Using our platform of purified Na/K-ATPase and an LLC-PK1 cell model, we found that 10: , at a concentration that induces less than 10% Na/K-ATPase inhibition, can stimulate the Na/K-ATPase/Src receptor complex and selectively activate downstream pathways, ultimately altering prostate cancer cell growth. By assessing the ligand effect of the isolated cardiotonic steroids, we found that the regulation of cell viability by the isolated cardiotonic steroids was not associated with their inhibitory potencies against Na/K-ATPase activity but reflected their ligand-binding affinity to the Na/K-ATPase receptor. Based on this discovery, we identified a unique active cardiotonic steroid, digitoxigenin (1: ), and verified that it can protect LLC-PK1 cells from hypoxic injury, implicating its potential use in ischemia/reperfusion injury and inducing collagen synthesis in primary human dermal fibroblast cells, and implicating that compound 2: is the molecular basis of the wound healing activity of S. juventas.
Collapse
Affiliation(s)
- Yunhui Xu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Jian Xu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Wanfang Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Yanling Yan
- Departments of Clinical & Translational Sciences, Biomedical Sciences and Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Xueyang Jiang
- Department of Medicinal Chemistry, Anhui University of Chinese Medicine, Hefei, China
| | - Zijian Xie
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Jie Zhang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
11
|
Dai H, Li L, Yang Y, Chen H, Dong X, Mao Y, Gao Y. Screening microRNAs as potential prognostic biomarkers for lung adenocarcinoma. Ann Med 2023; 55:2241013. [PMID: 37930873 PMCID: PMC10629414 DOI: 10.1080/07853890.2023.2241013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 07/21/2023] [Indexed: 11/08/2023] Open
Abstract
OBJECTIVE To screen and identify microRNAs (miRNAs) associated with the prognosis of lung adenocarcinoma (LUAD) using clinical samples and construct a prediction model for the prognosis of LUAD. METHODS 160 patient samples were used to screen and identify miRNAs associated with the prognosis of LUAD. Differentially expressed miRNAs were analyzed using gene chip technology. The selected miRNAs were validated using samples from the validation sample group. Cox proportional hazards regression was used to construct the model and Kaplan-Meier was used to plot survival curves. Model power was assessed by testing the prognosis of the constructed model using real-time polymerase chain reaction (RT-PCR) data. RESULTS The data showed that miR-1260b, miR-21-3p and miR-92a-3p were highly expressed in the early recurrence and metastasis group, while miR-2467-3p, miR-4659a-3p, miR-4514, miR-1471 and miR-3621 were lowly expressed. It was further confirmed that miR-21-3p was significantly highly expressed in the early recurrence and metastasis group (p = 0.02). Receiver operating characteristic (ROC) curve results showed cut-off point value of 0.0172, sensitivity of 88.2% and specificity of 100%. The predictive results of the constructed model were in good agreement with the actual prognosis of patients by using the validation sample test (Kappa = 0.426, p < 0.001), with a model sensitivity of 74.4%, a specificity of 68.3%, and an accuracy of 71.3%. CONCLUSION miRNAs associated with the prognosis of patients with stage I LUAD were screened and validated, and a risk model for predicting the prognosis of patients was constructed. This model has good consistency with the actual prognosis of patients.
Collapse
Affiliation(s)
- Hongshuang Dai
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center; National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center;National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College Cancer Hospital, Beijing, China
| | - Yikun Yang
- Department of Thoracic Surgical Oncology, National Cancer Center; National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College Cancer Hospital, Beijing, China
| | - Huang Chen
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Xin Dong
- Department of Clinical Laboratory, National Cancer Center; National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College Cancer Hospital, Beijing, China
| | - Yousheng Mao
- Department of Thoracic Surgical Oncology, National Cancer Center; National Clinical Research Center for Cancer, Chinese Academy of Medical Sciences and Peking Union Medical College Cancer Hospital, Beijing, China
| | - Yanning Gao
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center; National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Hajipour S, Hosseini SM, Irani S, Tavallaie M. Identification of novel potential drugs and miRNAs biomarkers in lung cancer based on gene co-expression network analysis. Genomics Inform 2023; 21:e38. [PMID: 37813634 PMCID: PMC10584645 DOI: 10.5808/gi.23039] [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: 05/11/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 10/11/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is an important cause of cancer-associated deaths worldwide. Therefore, the exact molecular mechanisms of NSCLC are unidentified. The present investigation aims to identify the miRNAs with predictive value in NSCLC. The two datasets were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed miRNAs (DEmiRNA) and mRNAs (DEmRNA) were selected from the normalized data. Next, miRNA-mRNA interactions were determined. Then, co-expression network analysis was completed using the WGCNA package in R software. The co-expression network between DEmiRNAs and DEmRNAs was calculated to prioritize the miRNAs. Next, the enrichment analysis was performed for DEmiRNA and DEmRNA. Finally, the drug-gene interaction network was constructed by importing the gene list to dgidb database. A total of 3,033 differentially expressed genes and 58 DE miRNA were recognized from two datasets. The co-expression network analysis was utilized to build a gene co-expression network. Next, four modules were selected based on the Zsummary score. In the next step, a bipartite miRNA-gene network was constructed and hub miRNAs (let-7a-2-3p, let-7d-5p, let-7b-5p, let-7a-5p, and let-7b-3p) were selected. Finally, a drug-gene network was constructed while SUNITINIB, MEDROXYPROGESTERONE ACETATE, DOFETILIDE, HALOPERIDOL, and CALCITRIOL drugs were recognized as a beneficial drug in NSCLC. The hub miRNAs and repurposed drugs may act a vital role in NSCLC progression and treatment, respectively; however, these results must validate in further clinical and experimental assessments.
Collapse
Affiliation(s)
- Sara Hajipour
- Biology Department, Science and Research Branch, Islamic Azad University, Tehran 14155-4933, Iran
| | - Sayed Mostafa Hosseini
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran 14359-16471, Iran
| | - Shiva Irani
- Biology Department, Science and Research Branch, Islamic Azad University, Tehran 14155-4933, Iran
| | - Mahmood Tavallaie
- Human Genetic Research Center, Baqiyatallah University of Medical Sciences, Tehran 14359-16471, Iran
| |
Collapse
|
13
|
Ainembabazi D, Zhang Y, Turchi JJ. The mechanistic role of cardiac glycosides in DNA damage response and repair signaling. Cell Mol Life Sci 2023; 80:250. [PMID: 37584722 PMCID: PMC10432338 DOI: 10.1007/s00018-023-04910-9] [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: 04/26/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/17/2023]
Abstract
Cardiac glycosides (CGs) are a class of bioactive organic compounds well-known for their application in treating heart disease despite a narrow therapeutic window. Considerable evidence has demonstrated the potential to repurpose CGs for cancer treatment. Chemical modification of these CGs has been utilized in attempts to increase their anti-cancer properties; however, this has met limited success as their mechanism of action is still speculative. Recent studies have identified the DNA damage response (DDR) pathway as a target of CGs. DDR serves to coordinate numerous cellular pathways to initiate cell cycle arrest, promote DNA repair, regulate replication fork firing and protection, or induce apoptosis to avoid the survival of cells with DNA damage or cells carrying mutations. Understanding the modus operandi of cardiac glycosides will provide critical information to better address improvements in potency, reduced toxicity, and the potential to overcome drug resistance. This review summarizes recent scientific findings of the molecular mechanisms of cardiac glycosides affecting the DDR signaling pathway in cancer therapeutics from 2010 to 2022. We focus on the structural and functional differences of CGs toward identifying the critical features for DDR targeting of these agents.
Collapse
Affiliation(s)
- Diana Ainembabazi
- Department of Medicine, School of Medicine, Joseph E Walther Hall, Indiana University, 980 W. Walnut St, C560, R3-C560, Indianapolis, IN 46202 USA
| | - Youwei Zhang
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
| | - John J. Turchi
- Department of Medicine, School of Medicine, Joseph E Walther Hall, Indiana University, 980 W. Walnut St, C560, R3-C560, Indianapolis, IN 46202 USA
| |
Collapse
|
14
|
Chen LC, Mokgautsi N, Kuo YC, Wu ATH, Huang HS. In Silico Evaluation of HN-N07 Small Molecule as an Inhibitor of Angiogenesis and Lymphangiogenesis Oncogenic Signatures in Non-Small Cell Lung Cancer. Biomedicines 2023; 11:2011. [PMID: 37509650 PMCID: PMC10376976 DOI: 10.3390/biomedicines11072011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Tumor angiogenesis and lymphangiogenesis pathways have been identified as important therapeutic targets in non-small cell lung cancer (NSCLC). Bevacizumab, which is a monoclonal antibody, was the initial inhibitor of angiogenesis and lymphangiogenesis that received approval for use in the treatment of advanced non-small cell lung cancer (NSCLC) in combination with chemotherapy. Despite its usage, patients may still develop resistance to the treatment, which can be attributed to various histological subtypes and the initiation of treatment at advanced stages of cancer. Due to their better specificity, selectivity, and safety compared to chemotherapy, small molecules have been approved for treating advanced NSCLC. Based on the development of multiple small-molecule antiangiogenic drugs either in house and abroad or in other laboratories to treat NSCLC, we used a quinoline-derived small molecule-HN-N07-as a potential target drug for NSCLC. Accordingly, we used computational simulation tools and evaluated the drug-likeness properties of HN-N07. Moreover, we identified target genes, resulting in the discovery of the target BIRC5/HIF1A/FLT4 pro-angiogenic genes. Furthermore, we used in silico molecular docking analysis to determine whether HN-N07 could potentially inhibit BIRC5/HIF1A/FLT4. Interestingly, the results of docking HN-N07 with the BIRC5, FLT4, and HIF1A oncogenes revealed unique binding affinities, which were significantly higher than those of standard inhibitors. In summary, these results indicate that HN-N07 shows promise as a potential inhibitor of oncogenic signaling pathways in NSCLC. Ongoing studies that involve in vitro experiments and in vivo investigations using tumor-bearing mice are in progress, aiming to evaluate the therapeutic effectiveness of the HN-N07 small molecule.
Collapse
Affiliation(s)
- Lung-Ching Chen
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan
| | - Ntlotlang Mokgautsi
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Alexander T H Wu
- The PhD Program of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
| | - Hsu-Shan Huang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan
- PhD Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
15
|
Khattab OM, El-Kersh DM, Khalifa SAM, Yosri N, El-Seedi HR, Farag MA. Comparative MS- and NMR-Based Metabolome Mapping of Egyptian Red and White Squill Bulbs F. Liliaceae and in Relation to Their Cytotoxic Effect. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12112078. [PMID: 37299060 DOI: 10.3390/plants12112078] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/29/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023]
Abstract
Urginea maritima L. (squill) species is widely spread at the Mediterranean region as two main varieties, i.e., white squill (WS) and red squill (RS), that are recognized for several health potentials. The major secondary metabolite classes of the squill are cardiac glycosides, mainly, bufadienolides, flavonoids, and anthocyanins. Herein, a multiplex MS and NMR metabolomics approach targeting secondary and aroma compounds in WS and RS was employed for varieties classification. Solid-phase micro extraction-gas chromatography/mass spectroscopy (SPME-GC/MS), ultra-high-performance liquid chromatography/mass spectrometry (UPLC/MS), as well as nuclear magnetic resonance (NMR) provided fingerprinting and structural confirmation of the major metabolites for both types of the squill. For comparison of the different platforms' classification potential, multivariate data analysis was employed. While Bufadienolides, viz. "hydroxy-scilliglaucosidin-O-rhamnoside, desacetylscillirosidin-O-rhamnoside and bufotalidin-O-hexoside" as well as oxylipids, were enriched in WS, flavonoids, i.e., dihydro-kaempferol-O-hexoside and its aglycon, taxifolin derivative, were predominant in RS. A cytotoxicity screening against three cancer cell lines, including breast adenocarcinoma (MCF-7), lung (A-549), and ovarian (SKOV-3) cell lines was conducted. Results revealed that WS was more effective on A-549 and SKOV-3 cell lines (WS IC50 0.11 and 0.4 µg/mL, respectively) owing to its abundance of bufadienolides, while RS recorded IC50 (MCF7 cell line) 0.17 µg/mL since is is rich inflavonoids.
Collapse
Affiliation(s)
- Omar M Khattab
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom 32512, Egypt
| | - Dina M El-Kersh
- Department of Pharmacognosy, Faculty of Pharmacy, The British University in Egypt, Cairo 11837, Egypt
| | - Shaden A M Khalifa
- Psychiatry and Psychology Department, Capio Saint Göran's Hospital, Sankt Göransplan 1, 112 19 Stockholm, Sweden
| | - Nermeen Yosri
- Chemistry Department of Medicinal and Aromatic Plants, Research Institute of Medicinal and Aromatic Plants (RIMAP), Beni-Suef University, Beni-Suef 62514, Egypt
| | - Hesham R El-Seedi
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom 32512, Egypt
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- International Joint Research Laboratory of Intelligent Agriculture and Agri-Products Processing, Jiangsu Education Department, Zhenjiang 212013, China
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr El Aini St., Cairo 11562, Egypt
| |
Collapse
|
16
|
Nagao Y, Yokoi A, Yoshida K, Sugiyama M, Watanabe E, Nakamura K, Kitagawa M, Asano-Inami E, Koya Y, Yoshihara M, Tamauchi S, Shimizu Y, Ikeda Y, Yoshikawa N, Kato T, Yamamoto Y, Kajiyama H. Novel therapeutic strategies targeting UCP2 in uterine leiomyosarcoma. Pharmacol Res 2023; 189:106693. [PMID: 36773710 DOI: 10.1016/j.phrs.2023.106693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Uterine leiomyosarcoma (ULMS) is a malignant stromal tumor arising from the myometrium with a poor prognosis and very limited response to current chemotherapy. This study aimed to identify novel targets for ULMS through a three-step screening process using a chemical library consisting of 1271 Food and Drug Administration-approved drugs. First, we evaluated their inhibitory effects on ULMS cells and identified four candidates: proscillaridin A, lanatoside C, floxuridine, and digoxin. Then, we subcutaneously or orthotopically transplanted SK-UT-1 cells into mice to establish mouse models. In vivo analyses showed that proscillaridin A and lanatoside C exerted a superior antitumor effect. The results of mRNA sequencing showed that uncoupling protein 2 (UCP2) was suppressed in the sirtuin signaling pathway, increasing reactive oxygen species (ROS) and inducing cell death. Moreover, the downregulation of UCP2 induced ROS and suppressed ULMS cell growth. Furthermore, analyses using clinical samples showed that UCP2 expression was significantly upregulated in ULMS tissues than in myoma tissues both at the RNA and protein levels. These findings suggested that UCP2 is a potential therapeutic target and can contribute to the development of novel therapeutic strategies in patients with ULMS.
Collapse
Affiliation(s)
- Yukari Nagao
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan; Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya-shi, Aichi 464-8603, Japan; Japan Science and Technology Agency (JST), FOREST, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan.
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan; Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa-ku, Nagoya-shi, Aichi 464-8603, Japan
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Eri Watanabe
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Kae Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan; Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya-shi, Aichi, 464-8603, Japan
| | - Masami Kitagawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Eri Asano-Inami
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Satoshi Tamauchi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Yusuke Shimizu
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Yoshiki Ikeda
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| | - Tomoyasu Kato
- Department of Gynecologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yusuke Yamamoto
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya-shi, Aichi 466-8550, Japan
| |
Collapse
|
17
|
Yang Y, Chen Y, Wu JH, Ren Y, Liu B, Zhang Y, Yu H. Targeting regulated cell death with plant natural compounds for cancer therapy: A revisited review of apoptosis, autophagy-dependent cell death, and necroptosis. Phytother Res 2023; 37:1488-1525. [PMID: 36717200 DOI: 10.1002/ptr.7738] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 02/01/2023]
Abstract
Regulated cell death (RCD) refers to programmed cell death regulated by various protein molecules, such as apoptosis, autophagy-dependent cell death, and necroptosis. Accumulating evidence has recently revealed that RCD subroutines have several links to many types of human cancer; therefore, targeting RCD with pharmacological small-molecule compounds would be a promising therapeutic strategy. Moreover, plant natural compounds, small-molecule compounds synthesized from plant sources, and their derivatives have been widely reported to regulate different RCD subroutines to improve potential cancer therapy. Thus, in this review, we focus on updating the intricate mechanisms of apoptosis, autophagy-dependent cell death, and necroptosis in cancer. Moreover, we further discuss several representative plant natural compounds and their derivatives that regulate the above-mentioned three subroutines of RCD, and their potential as candidate small-molecule drugs for the future cancer treatment.
Collapse
Affiliation(s)
- Yuanyuan Yang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanmei Chen
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jun Hao Wu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yueting Ren
- Department of Pharmacology and Toxicology, Temerity Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Otolaryngology, Head and Neck Surgery and Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
18
|
Phytochemical Analysis and Anticancer Properties of Drimia maritima Bulb Extracts on Colorectal Cancer Cells. Molecules 2023; 28:molecules28031215. [PMID: 36770882 PMCID: PMC9921424 DOI: 10.3390/molecules28031215] [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: 10/25/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/28/2023] Open
Abstract
Cancer is a worldwide health problem and is the second leading cause of death after heart disease. Due to the high cost and severe side effects associated with chemotherapy treatments, natural products with anticancer therapeutic potential may play a promising role in anticancer therapy. The purpose of this study was to investigate the cytotoxic and apoptotic characteristics of the aqueous Drimia maritima bulb extract on Caco-2 and COLO-205 colorectal cancer cells. In order to reach such a purpose, the chemical composition was examined using the GC-MS method, and the selective antiproliferative effect was determined in colon cancer cell lines in normal gingival fibroblasts. The intracellular ROS, mitochondrial membrane potential, and gene expression changes in selected genes (CASP8, TNF-α, and IL-6 genes) were assessed to determine the molecular mechanism of the antitumor effect of the extract. GC-MS results revealed the presence of fifty-seven compounds, and Proscillaridin A was the predominant secondary metabolite in the extract. The IC50 of D. maritima bulb extract on Caco-2, COLO-205, and the normal human gingival fibroblasts were obtained at 0.9 µg/mL, 2.3 µg/mL, and 13.1 µg/mL, respectively. The apoptotic effect assay indicated that the bulb extract induced apoptosis in both colon cancer cell lines. D. maritima bulb extract was only able to induce statistically significant ROS levels in COLO-205 cells in a dose-dependent manner. The mitochondrial membrane potential (MMP) revealed a significant decrease in the MMP of Caco-2 and COLO-205 to various concentrations of the bulb extract. At the molecular level, RT-qPCR was used to assess gene expression of CASP8, TNF-α, and IL-6 genes in Caco-2 and COLO-205 cancer cells. The results showed that the expression of pro-inflammatory genes TNF-α and IL-6 were upregulated. The apoptotic initiator gene CASP8 was also upregulated in the Caco-2 cell line and did not reach significance in COLO-205 cells. These results lead to the conclusion that D. maritima extract induced cell death in both cell lines and may have the potential to be used in CRC therapy in the future.
Collapse
|
19
|
In silico analysis revealed the potential circRNA-miRNA-mRNA regulative network of non-small cell lung cancer (NSCLC). Comput Biol Med 2023; 152:106315. [PMID: 36495751 DOI: 10.1016/j.compbiomed.2022.106315] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/31/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND The primary source of death in the world is non-small cell lung cancer (NSCLC). However, NSCLCs pathophysiology is still not completely understood. The current work sought to study the differential expression of mRNAs involved in NSCLC and their interactions with miRNAs and circRNAs. METHODS We utilized three microarray datasets (GSE21933, GSE27262, and GSE33532) from the GEO NCBI database to identify the differentially expressed genes (DEGs) in NSCLC. We employed DAVID Functional annotation tool to investigate the underlying GO biological process, molecular functions, and KEGG pathways involved in NSCLC. We performed the Protein-protein interaction (PPI) network, MCODE, and CytoHubba analysis from Cytoscape software to identify the significant DEGs in NSCLC. We utilized miRnet to anticipate and build interaction between miRNAs and mRNAs in NSCLC and ENCORI to predict the miRNA-circRNA relationships and build the ceRNA regulatory network. Finally, we executed the gene expression and Kaplan-Meier survival analysis to validate the significant DEGs in the ceRNA network utilizing TCGA NSCLC and GEPIA data. RESULTS We revealed a total of 156 overlapped DEGs (47 upregulated and 109 downregulated genes) in NSCLC. The PPI network, MCODE, and CytoHubba analysis revealed 12 hub genes (cdkn3, rrm2, ccnb1, aurka, nuf2, tyms, kif11, hmmr, ccnb2, nek2, anln, and birc5) that are associated with NSCLC. We identified that these 12 genes encode 12 mRNAs that are strongly linked with 8 miRNAs, and further, we revealed that 1 circRNA was associated with this 5 miRNA. We constructed the ceRNAs network that contained 1circRNA-5miRNAs-7mRNAs. The expression of these seven significant genes in LUAD & LUSC (NSCLC) was considerably higher in the TCGA database than in normal tissues. Kaplan-Meier survival plot reveals that increased expression of these hub genes was related to a poor survival rate in LUAD. CONCLUSION Overall, we developed a circRNA-miRNA-mRNA regulation network to study the probable mechanism of NSCLC.
Collapse
|
20
|
Song Y, Kelava L, Zhang L, Kiss I. Microarray data analysis to identify miRNA biomarkers and construct the lncRNA-miRNA-mRNA network in lung adenocarcinoma. Medicine (Baltimore) 2022; 101:e30393. [PMID: 36086747 PMCID: PMC10980501 DOI: 10.1097/md.0000000000030393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 07/25/2022] [Indexed: 12/09/2022] Open
Abstract
MicroRNAs (miRNAs), regulatory noncoding RNAs, are involved in gene regulation and may play a role in cancer development. The aim of this study was to identify miRNAs involved in lung adenocarcinoma (LUAD) using bioinformatics analysis. MiRNA (GSE135918), mRNA (GSE136043) and lncRNA (GSE130779) microarray datasets were downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed miRNAs (DEMis), mRNAs (DEMs), and lncRNA (DELs) in LUAD. We used DEMs for functional enrichment analysis. MiRNA expression quantification from The Cancer Genome Atlas (TCGA) was used to validate DEMis. LncBase Predicted v.2, Targetscan, and MiRBase were used to predict lncRNAs and mRNAs. The LUAD data in TCGA were used for overall survival (OS) analysis. We screened the downregulation of 8 DEMis and upregulation of 6 DEMis, and found that 70 signal pathways changed. We chose 3 relevant signaling pathways in lung cancer development, WNT, PI3K-Akt, and Notch, and scanned for mRNAs involved in them that are potential targets of these miRNAs. Then a lncRNA-miRNA-mRNA network was constructed. We also found 7 miRNAs that were associated with poor OS in LUAD. Low expression level of hsa-miR-30a was highly associated with poor OS in LUAD (P < .001) and the target genes of hsa-miR-30a-3p were abundant in the Wnt and AKT signaling pathways. In addition, our results reported for the first time that hsa-miR-3944 and hsa-miR-3652 were highly expressed in LUAD. And the high expression level of hsa-miR-3944 was associated with poor OS (P < .05). Hsa-miR-30a-3p may suppress the occurrence and progression of lung cancer through Wnt and AKT signaling pathways and become a good biomarker in LUAD. Hsa-miR-3944 and hsa-miR-3652 may serve as new biomarkers in LUAD.
Collapse
Affiliation(s)
- Yongan Song
- Department of Public Health Medicine, University of Pécs Medical School, Szigeti str 12, Pécs 7624, Hungary
| | - Leonardo Kelava
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pécs, Szigeti str 12, Pécs 7624, Hungary
| | - Lu Zhang
- Department of Health Science, Doctoral School of Health Science, University of Pécs, Vasvári Pál utca 4, Pécs 7622, Hungary
| | - István Kiss
- Department of Public Health Medicine, University of Pécs Medical School, Szigeti str 12, Pécs 7624, Hungary
| |
Collapse
|
21
|
Hou J, Kang N, Liu NN, Tan D, Zhang S, Liu J, Xie Y. Proscillaridin A induces mitochondrial damage and autophagy in pancreatic cancer and reduces the stability of SMAD4 in Panc-1 cells. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:820. [PMID: 36034984 PMCID: PMC9403942 DOI: 10.21037/atm-22-1085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/23/2022] [Indexed: 12/03/2022]
Abstract
Background Pancreatic cancer (PC) is a highly metastatic and lethal cancer with a very low overall 5-year survival rate. There is an urgent need for identifying new therapeutic agents for this deadly disease. Cardiac glycosides (CGs) have been traditionally used for their potent cardiovascular activities and have also recently been reported to exhibit anti-tumor effects. Proscillaridin A (Pro A), a natural CG, has been shown to display anti-tumor effects on multiple cancer types. Methods The cytotoxic effect of Pro A on PC cells was determined using cell viability assay, colony formation assay and transwell assay in vitro. Cell apoptosis, cell cycle, reactive oxygen species (ROS) generation, intracellular Ca2+ levels and mitochondrial membrane potential (MMP) were assayed by flow cytometry. Panc-1-xenografted mice model was used to evaluate Pro A’s effect in tumor growth. Mitochondria morphology was observed by transmission electron microscopy. LC3 aggregation was assessed by GFP-LC3 fluorescence microscopy. Gene expression was assayed by western blot or real-time quantitative polymerase chain reaction (qPCR). Results Pro A inhibits the proliferation, migration and invasion of Panc-1, BxPC-3 and AsPC-1 PC cells in vitro, and Panc-1 cells display the highest sensitivity with an IC50 at the nano-molar level. In vivo, Pro A treatment inhibits tumor progression in Panc-1 xenograft nude mice. Pro A treatment promotes both cell apoptosis and autophagy, and Pro A-treated PC cells display characteristics of mitochondrial damage including increased ROS generation, intracellular Ca2+ levels and disruption of MMP. In addition, high sensitivity towards Pro A of Panc-1 cells compared to BxPC-3 and AsPC-1 cells could be partially attributed to the loss of endogenous SMAD4 expression in the latter. Conclusions Our findings suggest that Pro A constitutes a promising therapeutic candidate for certain types of PC.
Collapse
Affiliation(s)
- Jia Hou
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ning Kang
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China
| | - Nan-Nan Liu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan Tan
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Liu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China.,Children's Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Analyzing the Systems Biology Effects of COVID-19 mRNA Vaccines to Assess Their Safety and Putative Side Effects. Pathogens 2022; 11:pathogens11070743. [PMID: 35889989 PMCID: PMC9320269 DOI: 10.3390/pathogens11070743] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/11/2022] [Accepted: 06/25/2022] [Indexed: 01/25/2023] Open
Abstract
COVID-19 vaccines have been instrumental tools in reducing the impact of SARS-CoV-2 infections around the world by preventing 80% to 90% of hospitalizations and deaths from reinfection, in addition to preventing 40% to 65% of symptomatic illnesses. However, the simultaneous large-scale vaccination of the global population will indubitably unveil heterogeneity in immune responses as well as in the propensity to developing post-vaccine adverse events, especially in vulnerable individuals. Herein, we applied a systems biology workflow, integrating vaccine transcriptional signatures with chemogenomics, to study the pharmacological effects of mRNA vaccines. First, we derived transcriptional signatures and predicted their biological effects using pathway enrichment and network approaches. Second, we queried the Connectivity Map (CMap) to prioritize adverse events hypotheses. Finally, we accepted higher-confidence hypotheses that have been predicted by independent approaches. Our results reveal that the mRNA-based BNT162b2 vaccine affects immune response pathways related to interferon and cytokine signaling, which should lead to vaccine success, but may also result in some adverse events. Our results emphasize the effects of BNT162b2 on calcium homeostasis, which could be contributing to some frequently encountered adverse events related to mRNA vaccines. Notably, cardiac side effects were signaled in the CMap query results. In summary, our approach has identified mechanisms underlying both the expected protective effects of vaccination as well as possible post-vaccine adverse effects. Our study illustrates the power of systems biology approaches in improving our understanding of the comprehensive biological response to vaccination against COVID-19.
Collapse
|
23
|
Hybrid Pharmacophore- and Structure-Based Virtual Screening Pipeline to Identify Novel EGFR Inhibitors That Suppress Non-Small Cell Lung Cancer Cell Growth. Int J Mol Sci 2022; 23:ijms23073487. [PMID: 35408854 PMCID: PMC8999148 DOI: 10.3390/ijms23073487] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
Dysregulated epidermal growth factor receptor (EGFR) expression is frequently observed in non-small cell lung cancer (NSCLC) growth and metastasis. Despite recent successes in the development of tyrosine kinase inhibitors (TKIs), inevitable resistance to TKIs has led to urgent calls for novel EGFR inhibitors. Herein, we report a rational workflow used to identify novel EGFR-TKIs by combining hybrid ligand- and structure-based pharmacophore models. Three types of models were developed in this workflow, including 3D QSAR-, common feature-, and structure-based EGFR-TK domain-containing pharmacophores. A National Cancer Institute (NCI) compound dataset was adopted for multiple-stage pharmacophore-based virtual screening (PBVS) of various pharmacophore models. The six top-scoring compounds were identified through the PBVS pipeline coupled with molecular docking. Among these compounds, NSC609077 exerted a significant inhibitory effect on EGFR activity in gefitinib-resistant H1975 cells, as determined by an enzyme-linked immunosorbent assay (ELISA). Further investigations showed that NSC609077 inhibited the anchorage-dependent growth and migration of lung cancer cells. Furthermore, NSC609077 exerted a suppressive effect on the EGFR/PI3K/AKT pathway in H1975 cells. In conclusion, these findings suggest that hybrid virtual screening may accelerate the development of targeted drugs for lung cancer treatment.
Collapse
|
24
|
Wu Y, Shen S, Shi Y, Tian N, Zhou Y, Zhang X. Senolytics: Eliminating Senescent Cells and Alleviating Intervertebral Disc Degeneration. Front Bioeng Biotechnol 2022; 10:823945. [PMID: 35309994 PMCID: PMC8924288 DOI: 10.3389/fbioe.2022.823945] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/21/2022] [Indexed: 12/25/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is the main cause of cervical and lumbar spondylosis. Over the past few years, the relevance between cellular senescence and IVDD has been widely studied, and the senescence-associated secretory phenotype (SASP) produced by senescent cells is found to remodel extracellular matrix (ECM) metabolism and destruct homeostasis. Elimination of senescent cells by senolytics and suppression of SASP production by senomorphics/senostatics are effective strategies to alleviate degenerative diseases including IVDD. Here, we review the involvement of senescence in the process of IVDD; we also discuss the potential of senolytics on eliminating senescent disc cells and alleviating IVDD; finally, we provide a table listing senolytic drugs and small molecules, aiming to propose potential drugs for IVDD therapy in the future.
Collapse
Affiliation(s)
- Yuhao Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shiwei Shen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| |
Collapse
|
25
|
Liang TL, Li RZ, Mai CT, Guan XX, Li JX, Wang XR, Ma LR, Zhang FY, Wang J, He F, Pan HD, Zhou H, Yan PY, Fan XX, Wu QB, Neher E, Liu L, Xie Y, Leung ELH, Yao XJ. A method establishment and comparison of in vivo lung cancer model development platforms for evaluation of tumour metabolism and pharmaceutical efficacy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153831. [PMID: 34794861 DOI: 10.1016/j.phymed.2021.153831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Currently, the identification of accurate biomarkers for the diagnosis of patients with early-stage lung cancer remains difficult. Fortunately, metabolomics technology can be used to improve the detection of plasma metabolic biomarkers for lung cancer. In a previous study, we successfully utilised machine learning methods to identify significant metabolic markers for early-stage lung cancer diagnosis. However, a related research platform for the investigation of tumour metabolism and drug efficacy is still lacking. HYPOTHESIS/PURPOSE A novel methodology for the comprehensive evaluation of the internal tumour-metabolic profile and drug evaluation needs to be established. METHODS The optimal location for tumour cell inoculation was identified in mouse chest for the non-traumatic orthotopic lung cancer mouse model. Microcomputed tomography (micro-CT) was applied to monitor lung tumour growth. Proscillaridin A (P.A) and cisplatin (CDDP) were utilised to verify the anti-lung cancer efficacy of the platform. The top five clinically valid biomarkers, including proline, L-kynurenine, spermidine, taurine and palmitoyl-L-carnitine, were selected as the evaluation indices to obtain a suitable lung cancer mouse model for clinical metabolomics research by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). RESULTS The platform was successfully established, achieving 100% tumour development rate and 0% surgery mortality. P.A and CDDP had significant anti-lung cancer efficacy in the platform. Compared with the control group, four biomarkers in the orthotopic model and two biomarkers in the metastatic model had significantly higher abundance. Principal component analysis (PCA) showed a significant separation between the orthotopic/metastatic model and the control/subcutaneous/KRAS transgenic model. The platform was mainly involved in arginine and proline metabolism, tryptophan metabolism, and taurine and hypotaurine metabolism. CONCLUSION This study is the first to simulate clinical metabolomics by comparing the metabolic phenotype of plasma in different lung cancer mouse models. We found that the orthotopic model was the most suitable for tumour metabolism. Furthermore, the anti-tumour drug efficacy was verified in the platform. The platform can very well match the clinical reality, providing better lung cancer diagnosis and securing more precise evidence for drug evaluation in the future.
Collapse
Affiliation(s)
- Tu-Liang Liang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Run-Ze Li
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Chu-Tian Mai
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Xiao-Xiang Guan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Jia-Xin Li
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Xuan-Run Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Lin-Rui Ma
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Fang-Yuan Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Jian Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Fan He
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Hu-Dan Pan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Hua Zhou
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Pei-Yu Yan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Xing-Xing Fan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Qi-Biao Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Erwin Neher
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Liang Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China
| | - Ying Xie
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China.
| | - Elaine Lai-Han Leung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China; Zhuhai Hospital of Traditional Chinese and Western Medicine, Zhuhai City, Guangdong, PR China.
| | - Xiao-Jun Yao
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (S.A.R.), China; State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China.
| |
Collapse
|
26
|
Lyu Q, Zhu W, Wei T, Ding W, Cao M, Wang Q, Guo L, Luo P, Zhang J. High mutations in fatty acid metabolism contribute to a better prognosis of small-cell lung cancer patients treated with chemotherapy. Cancer Med 2021; 10:7863-7876. [PMID: 34564955 PMCID: PMC8559513 DOI: 10.1002/cam4.4290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/30/2021] [Accepted: 08/28/2021] [Indexed: 12/15/2022] Open
Abstract
Background The majority of patients with small‐cell lung cancer (SCLC) show a good response in the early stages of treatment, but more than 90% of patients will develop drug resistance. Therefore, biomarkers are urgently needed to identify patients who can benefit from systemic treatment. Methods We prospectively enrolled 52 extensive‐stage SCLC patients before treatment from a local hospital to identify mutations related to patient prognosis, and verified them in the published Jiang's cohort and George's cohort. Results We found that patients with high mutations (mut‐high) in the fatty acid (FA) metabolism pathway had a longer progression‐free survival (PFS) in the local hospital cohort (HR = 0.446, 95% CI, 0.207–0.959, p = 0.0387) and a longer overall survival (OS) in Jiang's cohort (HR = 0.549, 95% CI, 0.314–0.960, p = 0.0351) than patients with low mutations (mut‐low). Multivariate analysis suggested that mut‐high status was an independent prognostic factor in both cohorts. George's cohort verified that mut‐high status was associated with a longer OS than mut‐low status (HR = 0.730, 95% CI 0.440–1.220, p = 0.2277). The possible mechanisms were as follows: the frequency of mutated FA synthase (FASN) in the mut‐high group was greater than that in the mut‐low group, and pathways related to the cell cycle, DNA repair, and oxidative phosphorylation were enriched in the mut‐high group. Conclusions The prognosis of SCLC patients treated with chemotherapy was better among patients with more mutations in the FA metabolism pathway, and the underlying mechanisms could be found at the genome and transcriptome levels.
Collapse
Affiliation(s)
- Qiong Lyu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weiliang Zhu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weimin Ding
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Manming Cao
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qiongyao Wang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linlang Guo
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Kumavath R, Paul S, Pavithran H, Paul MK, Ghosh P, Barh D, Azevedo V. Emergence of Cardiac Glycosides as Potential Drugs: Current and Future Scope for Cancer Therapeutics. Biomolecules 2021; 11:1275. [PMID: 34572488 PMCID: PMC8465509 DOI: 10.3390/biom11091275] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiac glycosides are natural sterols and constitute a group of secondary metabolites isolated from plants and animals. These cardiotonic agents are well recognized and accepted in the treatment of various cardiac diseases as they can increase the rate of cardiac contractions by acting on the cellular sodium potassium ATPase pump. However, a growing number of recent efforts were focused on exploring the antitumor and antiviral potential of these compounds. Several reports suggest their antitumor properties and hence, today cardiac glycosides (CG) represent the most diversified naturally derived compounds strongly recommended for the treatment of various cancers. Mutated or dysregulated transcription factors have also gained prominence as potential therapeutic targets that can be selectively targeted. Thus, we have explored the recent advances in CGs mediated cancer scope and have considered various signaling pathways, molecular aberration, transcription factors (TFs), and oncogenic genes to highlight potential therapeutic targets in cancer management.
Collapse
Affiliation(s)
- Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (P.O) Kasaragod, Kerala 671320, India;
| | - Sayan Paul
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamilnadu 627012, India;
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine, Bangalore 560065, India
| | - Honey Pavithran
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (P.O) Kasaragod, Kerala 671320, India;
| | - Manash K. Paul
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA;
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Debmalya Barh
- Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, India;
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-001, Brazil;
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Departamento de Genetica, Ecologia e Evolucao, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-001, Brazil;
| |
Collapse
|
28
|
Zhong ZH, Yi ZL, Zhao YD, Wang J, Jiang ZB, Xu C, Xie YJ, He QD, Tong ZY, Yao XJ, Leung ELH, Coghi PS, Fan XX, Chen M. Pyronaridine induces apoptosis in non-small cell lung cancer cells by upregulating death receptor 5 expression and inhibiting epidermal growth factor receptor. Chem Biol Drug Des 2021; 99:83-91. [PMID: 34288496 DOI: 10.1111/cbdd.13926] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/09/2021] [Accepted: 07/11/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer is the leading cause of cancer death. Pyronaridine, a synthetic drug of artemisinin, has been used in China for over 30 years for the treatment of malaria, but its effect on non-small cell lung cancer (NSCLC) cells is rarely reported. In this study, we determined the efficacy of pyronaridine in four different NSCLC cell lines and explored its mechanism in H1975. The data showed that pyronaridine could upregulate the expression of TNF-related apoptosis-inducing ligand (TRAIL)-mediated death receptor 5 to promote cellular apoptosis. Meanwhile, the JNK (c-Jun N-terminal kinase) level was detected to be significantly increased after treating with pyronaridine. We used JNK inhibitor and found that it could partially inhibit cell apoptosis. The results showed that epidermal growth factor receptor (EGFR), PI3K, and AKT were downregulated after the treatment of pyronaridine. In summary, pyronaridine can selectively kill NSCLC by regulating TRAIL-mediated apoptosis and downregulating the protein level of EGFR. It is a promising anticancer drug for NSCLC.
Collapse
Affiliation(s)
- Zheng-Hong Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China.,Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Ze-Lin Yi
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Yi-Dan Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Jue Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Ze-Bo Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Cong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Ya-Jia Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Qi-Da He
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Zi-Yan Tong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Paolo Saul Coghi
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China
| | - Min Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau, China.,Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| |
Collapse
|
29
|
Wang Z, Yang L. Chinese herbal medicine: Fighting SARS-CoV-2 infection on all fronts. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113869. [PMID: 33485973 PMCID: PMC7825841 DOI: 10.1016/j.jep.2021.113869] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/07/2021] [Accepted: 01/18/2021] [Indexed: 05/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes coronavirus disease 2019 (COVID-19), a highly pathogenic virus that has spread rapidly across the entire world. There is a critical need to develop safe and effective drugs, especially broad-spectrum antiviral and organ protection agents in order to treat and prevent this dangerous disease. It is possible that Chinese herbal medicine may play an essential role in the treatment of patients with SARS-CoV-2 infection. AIM OF THE REVIEW We aim to review the use of Chinese herbal medicine in the treatment of COVID-19 both in vitro and in clinical practice. Our goal was to provide a better understanding of the potential therapeutic effects of Chinese herbal medicine and to establish a "Chinese protocol" for the treatment of COVID-19. MATERIALS AND METHODS We systematically reviewed published research relating to traditional Chinese herbal medicines and the treatment of SARS-CoV-2 from inception to the 6th January 2021 by screening a range of digital databases (Web of Science, bioRxiv, medRxiv, China National Knowledge Infrastructure, X-MOL, Wanfang Data, Google Scholar, PubMed, Elsevier, and other resources) and public platforms relating to the management of clinical trials. We included the active ingredients of Chinese herbal medicines, monomer preparations, crude extracts, and formulas for the treatment of COVID-19. RESULTS In mainland China, a range of Chinese herbal medicines have been recognized as very promising anti-SARS-CoV-2 agents, including active ingredients (quercetagetin, osajin, tetrandrine, proscillaridin A, and dihydromyricetin), monomer preparations (xiyanping injection, matrine-sodium chloride injection, diammonium glycyrrhizinate enteric-coated capsules, and sodium aescinate injection), crude extracts (Scutellariae Radix extract and garlic essential oil), and formulas (Qingfei Paidu decoction, Lianhuaqingwen capsules, and Pudilan Xiaoyan oral liquid). All these agents have potential activity against SARS-CoV-2 and have attracted significant attention due to their activities both in vitro and in clinical practice. CONCLUSIONS As a key component of the COVID-19 treatment regimen, Chinese herbal medicines have played an irreplaceable role in the treatment of SARS-CoV-2 infection. The "Chinese protocol" has already demonstrated clear clinical importance. The use of Chinese herbal medicines that are capable of inhibiting SARS-Cov-2 infection may help to address this immediate unmet clinical need and may be attractive to other countries that are also seeking new options for effective COVID-19 treatment. Our analyses suggest that countries outside of China should also consider protocols involving Chinese herbal medicines combat this fast-spreading viral infection.
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, PR China; School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, PR China.
| | - Liyan Yang
- School of Physics and Physical Engineering, Qufu Normal University, Qufu, 273165, PR China.
| |
Collapse
|
30
|
Han C, Wang Z, Chen S, Li L, Xu Y, Kang W, Wei C, Ma H, Wang M, Jin X. Berbamine Suppresses the Progression of Bladder Cancer by Modulating the ROS/NF- κB Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8851763. [PMID: 33520087 PMCID: PMC7817266 DOI: 10.1155/2021/8851763] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/27/2020] [Accepted: 12/22/2020] [Indexed: 01/23/2023]
Abstract
Berbamine (BBM), one of the bioactive ingredients extracted from Berberis plants, has attracted intensive attention because of its significant antitumor activity against various malignancies. However, the exact role and potential molecular mechanism of berbamine in bladder cancer (BCa) remain unclear. In the present study, our results showed that berbamine inhibited cell viability, colony formation, and proliferation. Additionally, berbamine induced cell cycle arrest at S phase by a synergistic mechanism involving stimulation of P21 and P27 protein expression as well as downregulation of CyclinD, CyclinA2, and CDK2 protein expression. In addition to suppressing epithelial-mesenchymal transition (EMT), berbamine rearranged the cytoskeleton to inhibit cell metastasis. Mechanistically, the expression of P65, P-P65, and P-IκBα was decreased upon berbamine treatment, yet P65 overexpression abrogated the effects of berbamine on the proliferative and metastatic potential of BCa cells, which indicated that berbamine attenuated the malignant biological activities of BCa cells by inhibiting the NF-κB pathway. More importantly, berbamine increased the intracellular reactive oxygen species (ROS) level through the downregulation of antioxidative genes such as Nrf2, HO-1, SOD2, and GPX-1. Following ROS accumulation, the intrinsic apoptotic pathway was triggered by an increase in the ratio of Bax/Bcl-2. Furthermore, berbamine-mediated ROS accumulation negatively regulated the NF-κB pathway to a certain degree. Consistent with our in vitro results, berbamine successfully inhibited tumor growth and blocked the NF-κB pathway in our xenograft model. To summarize, our data demonstrated that berbamine exerts antitumor effects via the ROS/NF-κB signaling axis in bladder cancer, which provides a basis for further comprehensive study and presents a potential candidate for clinical treatment strategies against bladder cancer.
Collapse
Affiliation(s)
- Chenglin Han
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zilong Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Shuxiao Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Lin Li
- Department of Orthopedics, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yingkun Xu
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Weiting Kang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Chunxiao Wei
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Hongbin Ma
- Department of Hepatobiliary, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, China
| | - Muwen Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xunbo Jin
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| |
Collapse
|
31
|
Luo M, Liu Y, Liu N, Shao W, Ming L, Liu J, Xie Y. Proscillaridin A inhibits hepatocellular carcinoma progression through inducing mitochondrial damage and autophagy. Acta Biochim Biophys Sin (Shanghai) 2021; 53:19-28. [PMID: 33201987 DOI: 10.1093/abbs/gmaa139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally. At present, drug options for systemic treatment of HCC are very limited. There is an urgent need to develop additional effective drugs for HCC treatment. In the present study, we found that proscillaridin A (ProA), a cardiac glycoside, exerted a strong anticancer effect on multiple HCC cell lines. ProA significantly inhibited the cell proliferation, migration, and invasion of HCC cells. ProA also had a marked inhibitory effect on the progression of HCC in the MHCC97H xenograft nude mouse model. ProA-mediated suppression of HCC was closely related to cell apoptosis. ProA-treated HCC cells displayed significant mitochondrial damage and elevated reactive oxygen species production, resulting in profound cell apoptosis. Meanwhile, ProA also played a role in autophagy induction in HCC cells. Defects in autophagy partially relieved ProA's anticancer effect in HCC cells. Our findings demonstrate that ProA can effectively inhibit HCC progression and may serve as a potential therapeutic agent for HCC treatment.
Collapse
Affiliation(s)
- Mengjun Luo
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Institutes of Biomedical Sciences, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences,Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yanfeng Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nannan Liu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Institutes of Biomedical Sciences, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences,Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weiqing Shao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lijun Ming
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Institutes of Biomedical Sciences, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences,Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jing Liu
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Institutes of Biomedical Sciences, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences,Shanghai Medical College, Fudan University, Shanghai 200032, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (NHC & MOE & CAMS), Institutes of Biomedical Sciences, Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences,Shanghai Medical College, Fudan University, Shanghai 200032, China
- Children's Hospital, Fudan University, Shanghai 201199, China
| |
Collapse
|
32
|
Wang WJ, Mao LF, Lai HL, Wang YW, Jiang ZB, Li W, Huang JM, Xie YJ, Xu C, Liu P, Li YM, Leung ELH, Yao XJ. Dolutegravir derivative inhibits proliferation and induces apoptosis of non-small cell lung cancer cells via calcium signaling pathway. Pharmacol Res 2020; 161:105129. [PMID: 32783976 DOI: 10.1016/j.phrs.2020.105129] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most prevalent type of lung cancer. However, there has been little improvement in its cure rate in the last 30 years, due to its intricate heterogeneity and drug resistance. Accumulating evidences have demonstrated that dysregulation of calcium (Ca2+) homeostasis contributes to oncogenesis and promotes tumor development. Inhibitors of Ca2+ channels/transporters to restore intracellular Ca2+ level were found to arrest tumor cell division, induce apoptosis, and suppress tumor growth both in vitro and in vivo. Dolutegravir (DTG), which is a first-line drug for Acquired Immune Deficiency Syndrome (AIDs) treatment, has been shown to increase intracellular Ca2+ levels and Reactive oxygen species (ROS) levels in human erythrocytes, leading to suicidal erythrocyte death or eryptosis. To explore the potential of DTG as an antitumor agent, we have designed and synthesized a panel of compounds based on the principle of biologically active substructure splicing of DTG. Our data demonstrated that 7-methoxy-4-methyl-6,8-dioxo-N-(3-(1-(2-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-4-yl)phenyl)-3,4,6,8,12,12a-hexahydro-2H-pyrido[1',2':4,5]pyrazino[2,1-b][1,3]oxazine-9-carboxamide (DTHP), a novel derivative of DTG, strongly inhibited the colony-forming ability and proliferation of NSCLC cells, but displayed no cytotoxicity to normal lung cells. DTHP treatment also induced apoptosis and upregulate intracellular Ca2+ level in NSCLC cells significantly. Inhibiting Ca2+ signaling alleviated DTHP-induced apoptosis, suggesting the perturbation of intracellular Ca2+ is responsible for DTHP-induced apoptosis. We further discovered that DTHP activates AMPK signaling pathway through binding to SERCA, a Ca2+-ATPase. On the other hand, DTHP treatment promoted mitochondrial ROS production, causing mitochondrial dysfunction and cell death. Finally, DTHP effectively inhibited tumor growth in the mouse xenograft model of lung cancer with low toxicity to normal organs. Taken together, our work identified DTHP as a superior antitumor agent, which will provide a novel strategy for the treatment of NSCLC with potential clinical application.
Collapse
Affiliation(s)
- Wen-Jun Wang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Long-Fei Mao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China; School of Chemistry and Chemical Engineering, Henan Normal University, Henan Engineering Research Center of Chiral Hydroxyl Pharmaceutical, Xinxiang 453007, China
| | - Huan-Ling Lai
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Yu-Wei Wang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Ze-Bo Jiang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Wei Li
- School of Chemistry and Chemical Engineering, Henan Normal University, Henan Engineering Research Center of Chiral Hydroxyl Pharmaceutical, Xinxiang 453007, China
| | - Ju-Min Huang
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Ya-Jia Xie
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Cong Xu
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Pei Liu
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Yue-Ming Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.
| | - Elaine Lai Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China; Department of Thoracic Surgery, Guangzhou Institute of Respiratory Health and State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Respiratory Medicine Department, Taihe Hospital, Hubei University of Medicine, Hubei, China.
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China.
| |
Collapse
|
33
|
Saleem MZ, Alshwmi M, Zhang H, Din SRU, Nisar MA, Khan M, Alam S, Alam G, Jin L, Ma T. Inhibition of JNK-Mediated Autophagy Promotes Proscillaridin A- Induced Apoptosis via ROS Generation, Intracellular Ca +2 Oscillation and Inhibiting STAT3 Signaling in Breast Cancer Cells. Front Pharmacol 2020; 11:01055. [PMID: 33013353 PMCID: PMC7500466 DOI: 10.3389/fphar.2020.01055] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/29/2020] [Indexed: 01/16/2023] Open
Abstract
Breast cancer is the most heterogenous cancer type among women across the world. Despite concerted efforts, breast cancer management is still unsatisfactory. Interplay between apoptosis and autophagy is an imperative factor in categorizing therapeutics for cancer treatment. Proscillaridin A (PSD-A), a well-known cardiac glycoside used for cardiac arrest and arrythmias, has been unveiled in many cancer types but the underlying mechanism for apoptosis and autophagy in breast cancer is not fully understood. In our study, PSD-A restricted cell growth, inhibited STAT3 activation and induced apoptosis and autophagy in breast cancer cells via ROS generation and Ca+2 oscillation. Pretreatment of NAC and BAPTA-AM restored PSD-A induced cellular events in breast cancer cells. PSD-A induced apoptosis via DNA fragmentation, caspase-cascade activation, PARP cleavage, mitochondrial dysfunction, Bax/Bcl-2 proteins modulation and ER chaperone GRP78 inhibition along with decreased phosphorylation of ERK1/2. Inhibition of STAT3 activation was found to be associated with decreased phosphorylation of SRC. Moreover, PSD-A induced events of autophagy i.e. conversion of LC3-I to LC3-II, and Atg3 expression via JNK activation and decreased mTOR and AKT phosphorylation. In this study, pretreatment of SP600125, a JNK inhibitor, reduced autophagy and enhanced STAT3 inhibition and apoptosis. Additionally, SB203580, a commercial p38 inhibitor, stimulated STAT3 activation and improved autophagic events rate in breast cancer cells, displaying the role of the MAPK signaling pathway in interplay between apoptosis and autophagy. Our data suggest that the rate of apoptotic cell death is improved by blocking JNK-induced autophagy in PSD-A treated MCF-7 and MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
| | - Mohammed Alshwmi
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - He Zhang
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Syed Riaz Ud Din
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | | | - Muhammad Khan
- Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Shahid Alam
- Department of Anatomy, Dalian Medical University, Dalian, China
| | - Gulzar Alam
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Lingling Jin
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University, Dalian, China.,School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Reddy D, Kumavath R, Barh D, Azevedo V, Ghosh P. Anticancer and Antiviral Properties of Cardiac Glycosides: A Review to Explore the Mechanism of Actions. Molecules 2020; 25:E3596. [PMID: 32784680 PMCID: PMC7465415 DOI: 10.3390/molecules25163596] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/19/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiac glycosides (CGs) have a long history of treating cardiac diseases. However, recent reports have suggested that CGs also possess anticancer and antiviral activities. The primary mechanism of action of these anticancer agents is by suppressing the Na+/k+-ATPase by decreasing the intracellular K+ and increasing the Na+ and Ca2+. Additionally, CGs were known to act as inhibitors of IL8 production, DNA topoisomerase I and II, anoikis prevention and suppression of several target genes responsible for the inhibition of cancer cell proliferation. Moreover, CGs were reported to be effective against several DNA and RNA viral species such as influenza, human cytomegalovirus, herpes simplex virus, coronavirus, tick-borne encephalitis (TBE) virus and Ebola virus. CGs were reported to suppress the HIV-1 gene expression, viral protein translation and alters viral pre-mRNA splicing to inhibit the viral replication. To date, four CGs (Anvirzel, UNBS1450, PBI05204 and digoxin) were in clinical trials for their anticancer activity. This review encapsulates the current knowledge about CGs as anticancer and antiviral drugs in isolation and in combination with some other drugs to enhance their efficiency. Further studies of this class of biomolecules are necessary to determine their possible inhibitory role in cancer and viral diseases.
Collapse
Affiliation(s)
- Dhanasekhar Reddy
- Department of Genomic Science, School of Biological Sciences, University of Kerala, Tejaswini Hills, Periya (P.O), Kasaragod, Kerala 671320, India;
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, University of Kerala, Tejaswini Hills, Periya (P.O), Kasaragod, Kerala 671320, India;
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur WB-721172, India;
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal deMinas Gerais (UFMG), Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA;
| |
Collapse
|
35
|
Implications for glycosylated compounds and their anti-cancer effects. Int J Biol Macromol 2020; 163:1323-1332. [PMID: 32622770 DOI: 10.1016/j.ijbiomac.2020.06.281] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 06/20/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Glycosylated compounds are major secondary metabolites of plants, which have various therapeutic effects on human diseases, by acting as anti-cancer, antioxidant, and anti-inflammatory agents. Glycosylation increases stability, bioactivity, and solubility of compounds and improves their pharmacological properties. Two well-known examples of glycosylated compounds include cardiac and flavonoid, the anti-tumor activities of which have been emphasized by several studies. However, little is known about their role in the treatment or prevention of cancer. In this review, recent studies on anti-tumor properties of cardiac and flavonoid glycosides, and their mechanisms of action, have been investigated. More specifically, this review is aimed at focusing on the multifactorial properties of cardiac and flavonoid compounds as well as their correlation with signaling pathways in the treatment of cancer.
Collapse
|
36
|
Chen Y, Wu J, Yan H, Cheng Y, Wang Y, Yang Y, Deng M, Che X, Hou K, Qu X, Zou D, Liu Y, Zhang Y, Hu X. Lymecycline reverses acquired EGFR-TKI resistance in non-small-cell lung cancer by targeting GRB2. Pharmacol Res 2020; 159:105007. [PMID: 32561477 DOI: 10.1016/j.phrs.2020.105007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
Abstract
Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) were first-line treatments for NSCLC patients with EGFR-mutations. However, about 30 % of responders relapsed within six months because of acquired resistance. In this study, we used Connectivity Map (CMap) to discover a drug capable of reversing acquired EGFR-TKIs resistance. To investigate Lymecycline's ability to reverse acquired EGFR-TKIs resistance, two Icotinib resistant cell lines were constructed. Lymecycline's ability to suppress the proliferation of Icotinib resistant cells in vitro and in vivo was then evaluated. Molecular targets were predicted using network pharmacology and used to identify the molecular mechanism. Growth factor receptor-bound protein 2 (GRB2) is an EGFR-binding adaptor protein essential for EGFR phosphorylation and regulation of AKT/ERK/STAT3 signaling pathways. Lymecycline targeted GRB2 and inhibited the resistance of the cell cycle to EGFR-TKI, arresting disease progression and inducing apoptosis in cancer cells. Combined Lymecycline and Icotinib treatment produced a synergistic effect and induced apoptosis in HCC827R5 and PC9R10 cells. Cell proliferation in resistant cancer cells was significantly inhibited by the combined Lymecycline and Icotinib treatment in mouse models. Lymecycline inhibited the resistance of the cell cycle to EGFR-TKI and induced apoptosis in NSCLC by inhibiting EGFR phosphorylation and GRB2-mediated AKT/ERK/STAT3 signaling pathways. This provided strong support that Lymecycline when combined with EGFR targeting drugs, enhanced the efficacy of treatments for drug-resistant NSCLC.
Collapse
Affiliation(s)
- Yang Chen
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jie Wu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China; Department of Oncology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Hongfei Yan
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China; Department of Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yang Cheng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yizhe Wang
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yi Yang
- Laboratory Animal Center, China Medical University, Shenyang, 110001, Liaoning, China
| | - Mingming Deng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaofang Che
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Kezuo Hou
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiujuan Qu
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China; Department of Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Dan Zou
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Yunpeng Liu
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China; Department of Oncology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China.
| | - Xuejun Hu
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
37
|
Jin X, Guan Y, Zhang Z, Wang H. Microarray data analysis on gene and miRNA expression to identify biomarkers in non-small cell lung cancer. BMC Cancer 2020; 20:329. [PMID: 32299382 PMCID: PMC7164187 DOI: 10.1186/s12885-020-06829-x] [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: 11/01/2019] [Accepted: 04/05/2020] [Indexed: 01/22/2023] Open
Abstract
Background The aim of this study was to gain further investigation of non-small cell lung cancer (NSCLC) tumorigenesis and identify biomarkers for clinical management of patients through comprehensive bioinformatics analysis. Methods miRNA and mRNA microarray datasets were downloaded from GEO (Gene Expression Omnibus) database under the accession number GSE102286 and GSE101929, respectively. Genes and miRNAs with differential expression were identified in NSCLC samples compared with controls, respectively. The interaction between differentially expressed genes (DEGs) and differentially expressed miRNAs (DEmiRs) was predicted, followed by functional enrichment analysis, and construction of miRNA-gene regulatory network, protein-protein interaction (PPI) network, and competing endogenous RNA (ceRNA) network. Through comprehensive bioinformatics analysis, we anticipate to find novel therapeutic targets and biomarkers for NSCLC. Results A total of 123 DEmiRs (5 up- and 118 down-regulated miRNAs) and 924 DEGs (309 up- and 615 down-regulated genes) were identified. These genes and miRNAs were significantly involved in different pathways including adherens junction, relaxin signaling pathway, and axon guidance. Furthermore, hsa-miR-9-5p, has-miR-196a-5p and hsa-miR-31-5p, as well as hsa-miR-1, hsa-miR-218-5p and hsa-miR-135a-5p were shown to have higher degree in the miRNA-gene regulatory network and ceRNA network, respectively. Furthermore, BIRC5 and FGF2, as well as RTKN2 and SLIT3 were hubs in the PPI network and ceRNA network, respectively. Conclusion Several pathways (adherens junction, relaxin signaling pathway, and axon guidance) miRNAs (hsa-miR-9-5p, has-miR-196a-5p, hsa-miR-31-5p, hsa-miR-1, hsa-miR-218-5p and hsa-miR-135a-5p) and genes (BIRC5, FGF2, RTKN2 and SLIT3) may play important roles in the pathogenesis of NSCLC.
Collapse
Affiliation(s)
- Xiang Jin
- Department of Respiration, The First Hospital of Jilin University, No. 1 Xinminda Street, Changchun, 130021, China
| | - Yinghui Guan
- Department of Respiration, The First Hospital of Jilin University, No. 1 Xinminda Street, Changchun, 130021, China.
| | - Zhen Zhang
- PICU, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hongyue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
38
|
Wang F, Liu L, Tong Y, Li L, Liu Y, Gao WQ. Proscillaridin A slows the prostate cancer progression through triggering the activation of endoplasmic reticulum stress. Cell Cycle 2020; 19:541-550. [PMID: 32009541 DOI: 10.1080/15384101.2020.1716484] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the second commonly diagnosed malignancy in men over the world. Although androgen deprivation therapy for advanced PCa patients has significantly improved their survival, the majority of these patients eventually develop castration-resistant prostate cancer (CRPC). Proscillaridin A (Pro A), a cardiac glycoside that is clinically used to treat various heart failure diseases, has been reported to have anticancer activity in several cancers. However, whether Pro A exerts an inhibitory effect on PCa progression remains unknown. In this study, we determined possible antitumor effects of Pro A on PCa cells and demonstrated the following: firstly, Pro A selectively inhibited androgen-independent PCa (including PC3 and DU145) cell growth and induced cell apoptosis in vitro; secondly, Pro A significantly decreased cell motility and invasion of androgen-independent PCa cells; thirdly, Pro A enhanced the sensitivity of PCa cells to docetaxel; fourthly, Pro A significantly inhibited the growth of PCa xenografts in vivo and patient-derived organoids (PDO). In addition, RNA-sequencing analysis revealed that the antitumor effects of Pro A on androgen-independent PCa appeared to be achieved via driving the activation of endoplasmic reticulum stress. The antitumor effects of Pro A could be ameliorated by reactive oxygen species scavenger and ER stress inhibitors. Therefore, these data suggest that Pro A may provide a potential therapeutic option for the treatment of PCa, particularly CRPC.
Collapse
Affiliation(s)
- Fan Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Tong
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linfeng Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanfeng Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
39
|
Wang LY, Wu J, Gao YF, Lin DM, Ma J. Medium- and long-chain triglyceride propofol reduces the activity of acetyl-coenzyme A carboxylase in hepatic lipid metabolism in HepG2 and Huh7 cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:19-26. [PMID: 31908571 PMCID: PMC6940496 DOI: 10.4196/kjpp.2020.24.1.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/23/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023]
Abstract
Medium- and long-chain triglyceride (MCT/LCT) propofol is widely used as an intravenous anesthetic, especially in the intensive care unit. The present study aimed to assess whether MCT/LCT propofol is safe in the hyperlipidemic population for long-term use. Free fatty acids (FFAs) were used to establish high-fat stimulation of HepG2 and Huh7 cells. Subsequently, these cells were treated with propofol at the concentration of 0, 4, or 8 µg/ml for 24 and 48 h. The results indicated that the cell viability was notably decreased when the cells were stimulated with 2 mmol/L FFAs and treated with 12 µg/ml MCT/LCT propofol. Accordingly, we chose 2 mmol/L FFAs along with 4 and 8 µg/ml MCT/LCT propofol for the subsequent experiments. Four and 8 µg/ml MCT/LCT propofol inhibited FFA-induced lipid accumulation in the cells and significantly reversed acetyl coenzyme A carboxylase (ACC) activity. In addition, MCT/LCT propofol not only significantly promoted the phosphorylation of AMPK and ACC, but also reversed the FFA-induced decreased phosphorylation of AMPK and ACC. In conclusion, MCT/LCT propofol reverses the negative effects caused by FFAs in HepG2 and Huh7 cells, indicating that MCT/LCT propofol might positively regulate lipid metabolism.
Collapse
Affiliation(s)
- Li-Yuan Wang
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R China
| | - Jing Wu
- North China University of Science and Technology, Tangshan, Hebei 063300, P.R China
| | - Ya-Fen Gao
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R China
| | - Duo-Mao Lin
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R China
| | - Jun Ma
- Center for Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R China
| |
Collapse
|
40
|
Zhang N, Peng F, Wang Y, Yang L, Wu F, Wang X, Ye C, Han B, He G. Shikonin induces colorectal carcinoma cells apoptosis and autophagy by targeting galectin-1/JNK signaling axis. Int J Biol Sci 2020; 16:147-161. [PMID: 31892852 PMCID: PMC6930377 DOI: 10.7150/ijbs.36955] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/20/2019] [Indexed: 02/05/2023] Open
Abstract
Colorectal carcinoma (CRC) is the third most common malignant tumor pathology worldwide. Despite progress in surgical procedures and therapy options, CRC is still a considerable cause of cancer-related mortality. In this study, we tested the antitumor effects of shikonin in CRC and tried to identify its potential mechanism. The potential target, molecular mechanism as well as in vitro and in vivo antitumor effects of shikonin in CRC cells were determined by an integrative protocol including quantitative proteomics, RT-PCR, western blotting, RNA interference and overexpression, apoptosis and autophagy assays, etc. Galectin-1 was a potential target of shikonin from the iTRAQ-based proteomic analysis in shikonin-treated SW620 cell. The overexpression and RNA silencing of galectin-1 in two CRC cells suggested that the shikonin sensitivity was correlation to galectin-1 levels. The ROS accumulation induced by shikonin was important to the formation of galectin-1 dimers. Dimer galectin-1 was found to be associated with the activation of JNK and downstream apoptosis or autophagy. Moreover, through functional in vitro studies, we showed that differences in galectin-1 level affected tumor cell proliferation, migration, and invasion. In summary, shikonin induced CRC cells apoptosis and autophagy by targeting galectin-1 and JNK signaling pathway both in vitro and in vivo, which suggested a potential novel therapy target for CRC.
Collapse
Affiliation(s)
- Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fu Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yujia Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fengbo Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaoyun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Cui Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Gu He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
41
|
Ouabain impairs cancer metabolism and activates AMPK-Src signaling pathway in human cancer cell lines. Acta Pharmacol Sin 2020; 41:110-118. [PMID: 31515527 PMCID: PMC7468359 DOI: 10.1038/s41401-019-0290-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
In addition to the well-known cardiotonic effects, cardiac glycosides (CGs) produce potent anticancer effects with various molecular mechanisms. We previously show that ouabain induces autophagic cell death in human lung cancer cells by regulating AMPK-mediated mTOR and Src-mediated ERK1/2 signaling pathways. However, whether and how AMPK and Src signaling interacts in ouabain-treated cancer cells remains unclear. Given the pivotal role of AMPK in metabolism, whether ouabain affects cancer cell metabolism remains elusive. In this study we showed that treatment with ouabain (25 nM) caused simultaneous activation of AMPK and Src signaling pathways in human lung cancer A549 cells and human breast cancer MCF7 cells. Cotreatment with AMPK inhibitor compound C or siRNA greatly abrogates ouabain-induced Src activation, whereas cotreatment with Src inhibitor PP2 has little effect on ouabain-induced AMPK activity, suggesting that AMPK served as an upstream regulator of the Src signaling pathway. On the other hand, ouabain treatment greatly depletes ATP production in A549 and MCF7 cells, and supplement of ATP (100 μM) blocked ouabain-induced AMPK activation. We further demonstrated that ouabain greatly inhibited the mitochondrial oxidative phosphorylation (OXPHOS) in the cancer cells, and exerted differential metabolic effects on glycolysis depending on cancer cell type. Taken together, this study reveals that the altered cancer cell metabolism caused by ouabain may contribute to AMPK activation, as well as its cytotoxicity towards cancer cells.
Collapse
|
42
|
Wang Y, Tang C, Yao S, Lai H, Li R, Xu J, Wang Q, Fan XX, Wu QB, Leung ELH, Ye Y, Yao X. Discovery of a novel protein kinase C activator from Croton tiglium for inhibition of non-small cell lung cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 65:153100. [PMID: 31648127 DOI: 10.1016/j.phymed.2019.153100] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND The incidence of non-small cell lung cancer (NSCLC) accounts for approximately 85-90% of lung cancer, which has been shown to be challenging for treatment owing to poorly understanding of pathological mechanisms. Natural products serve as a source of almost all pharmaceutical preparations or offer guidance for those chemicals that have entered clinical trials, especially in NSCLC. PURPOSE We investigated the effect of B10G5, a natural products isolated from the Croton tiglium, in human non-small cell lung canceras as a protein kinase C (PKC) activator. METHODS The cell viability assay was evaluated by the MTT assay. The apoptosis and cell cycle distribution were assessed by flow cytometry. Reactive oxygen species (ROS) production was determined by using the fluorescent probe DCFDA. Cell migration ability of H1975 cells was analyzed by using the wound healing assay. The inhibiting effect of B10G5 against the phosphorylation level of the substrate by PKCs was assessed by using homogeneous time-resolved fluorescence (HTRF) technology. The correlation between PKCs and overall survival (OS) of Lung Adenocarcinoma (LUAD) patients was analysis by TCGA portal. The binding mode between B10G5 and the PKC isoforms was explored by molecular docking. Protein expression was detected by western blotting analysis. RESULTS B10G5 suppressed cell proliferation and colony formation, as well as migration ability of NSCLC cells, without significant toxic effect on normal lung cells. B10G5 induced the cell apoptosis through the development of PARP cleavage, which is evidenced by means of the production of mitochondrial ROS. In addition, the B10G5 inhibitory effect was also related to the cell cycle arrest at G2/M phase. Mechanistically, molecular modelling technology suggested that the potential target of B10G5 was associated with PKC family. In vitro PKC kinase assay indicated that B10G5 effectively activated the PKC activity. Western blotting data revealed that B10G5 upregulated PKC to activate PKC-mediated RAF/MEK/ERK pathway. CONCLUSION Our results showed that B10G5, a naturally occurring phorbol ester, considered to be a potential and a valuable therapeutic chemical in the treatment of NSCLC.
Collapse
Affiliation(s)
- Yuwei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China
| | - Chunping Tang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academic of Sciences, Shanghai, China
| | - Sheng Yao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academic of Sciences, Shanghai, China
| | - Huanling Lai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China
| | - Runze Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China
| | - Jiahui Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China
| | - Qianqian Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China
| | - Xing Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China
| | - Qi Biao Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China; Department of Thoracic Surgery, Guangzhou Institute of Respiratory Health and State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Respiratory Medicine Department, Taihe Hospital, Hubei University of Medicine, Hubei, China.
| | - Yang Ye
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academic of Sciences, Shanghai, China.
| | - Xiaojun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau (SAR), China.
| |
Collapse
|
43
|
Fang S, Tao H, Xia K, Guo W. Proscillaridin A induces apoptosis and inhibits the metastasis of osteosarcoma in vitro and in vivo. Biochem Biophys Res Commun 2019; 521:880-886. [PMID: 31708095 DOI: 10.1016/j.bbrc.2019.11.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/02/2019] [Indexed: 01/23/2023]
Abstract
The side effects of chemotherapy, drug resistance, and tumor metastasis hinder the development of treatment for osteosarcoma, leading to poor prognosis of patients with the disease. Proscillaridin A, a kind of cardiac glycoside, has been proven to have anti-proliferative properties in many malignant tumors, but the efficacy of the drug in treating osteosarcoma is unclear. In the present study, we assessed the effects of Proscillaridin A on osteosarcoma and investigated its underlying action mechanism. The cell cytotoxicity assay showed that Proscillaridin A significantly inhibited the proliferation of 143B cells in a dose- and time-dependent manner. Also, flow cytometry and invasion assay revealed that Proscillaridin A induced apoptosis and reduced 143B cell motility. Western blotting and PCR were used to detect the expressions of Bcl-xl and MMP2 and showed that mRNA/protein expression levels decreased significantly in Proscillaridin A-treated osteosarcoma cells. Using a mouse xenograft model, we found that Proscillaridin A treatment significantly inhibited tumor growth and lung metastasis in vivo and decreased the expression levels of Bcl-xl and MMP2. No noticeable side effect was observed in the liver, kidney, and hematological functions. Conclusively, Proscillaridin A suppressed proliferation, induced apoptosis, and inhibited 143B cell metastasis in vitro and in vivo, and these effects could be mediated by downregulating the expressions of Bcl-xl and MMP2.
Collapse
Affiliation(s)
- Shuo Fang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
| | - Hai Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
| | - Kezhou Xia
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China.
| |
Collapse
|
44
|
Wei C, Yao X, Jiang Z, Wang Y, Zhang D, Chen X, Fan X, Xie C, Cheng J, Fu J, Leung ELH. Cordycepin Inhibits Drug-resistance Non-small Cell Lung Cancer Progression by Activating AMPK Signaling Pathway. Pharmacol Res 2019; 144:79-89. [PMID: 30974169 DOI: 10.1016/j.phrs.2019.03.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/08/2019] [Accepted: 03/10/2019] [Indexed: 12/24/2022]
Abstract
Lung cancer is the most commonly diagnosed cancer worldwide and it is also the most leading cause of cancer-related deaths. Although multiple generations of targeted therapeutic drugs such as gefitinib and afatinib specifically targeting the epidermal growth factor receptor (EGFR) pathway are currently available for lung cancer treatment, none of them can escape their eventual drug-resistance. As a key component of Cordyceps Sinensis and widely used in traditional Chinese medicines (TCM), cordycepin (CD) has attracted increasing attention to both scientists and clinicians. We aimed to explore the potential in developing cordycepin (CD) as an anti-lung cancer drug. A systematic analysis was conducted on a panel of non-small cell lung cancer (NSCLC) cell lines to identify the cells sensitive to CD. We found that CD can affect different aspects of lung cancer development including proliferation, migration, invasion, cell cycle, and apoptosis. We then explored the underlying molecular mechanisms of CD-mediated NSCLC cell apoptosis by conducting a series of in vitro and in vivo experiments. We found that in addition to affecting different stages of NSCLC development including tumor growth, migration, and invasion, the CD is capable of inhibiting NSCLC cell cycle progression and inducing cancer cell apoptosis without apparent adverse effect on normal lung cells. Furthermore, we found that the cells containing EGFR mutations are more sensitive to CD treatment than those without. Mechanistically, CD induces NSCLC cell apoptosis by interacting with and activating AMP-activated protein kinase (AMPK). More importantly, we found that the potency of CD's anticancer effect both in vitro and in vivo is comparable to afatinib and even better than gefitinib. Our findings suggest that CD either by itself or in combination with the currently available targeted therapeutic drugs might be additional therapeutic options for drug-resistance NSCLC treatment.
Collapse
Affiliation(s)
- Chunli Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China; Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaojun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Zebo Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Yuwei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Dianzheng Zhang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China; Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, PA, 19131, USA
| | - Xi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China; Department of Pathology, Stony Brook University, Stony Brook, NY 11794, United States
| | - Xingxing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Chun Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Junjiang Fu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China; Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China.
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China; Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, Guangdong, China.
| |
Collapse
|
45
|
El-Seedi HR, Khalifa SAM, Taher EA, Farag MA, Saeed A, Gamal M, Hegazy MEF, Youssef D, Musharraf SG, Alajlani MM, Xiao J, Efferth T. Cardenolides: Insights from chemical structure and pharmacological utility. Pharmacol Res 2019; 141:123-175. [PMID: 30579976 DOI: 10.1016/j.phrs.2018.12.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/07/2018] [Accepted: 12/18/2018] [Indexed: 02/08/2023]
Abstract
Cardiac glycosides (CGs) are a class of naturally occurring steroid-like compounds, and members of this class have been in clinical use for more than 1500 years. They have been used in folk medicine as arrow poisons, abortifacients, heart tonics, emetics, and diuretics as well as in other applications. The major use of CGs today is based on their ability to inhibit the membrane-bound Na+/K+-ATPase enzyme, and they are regarded as an effective treatment for congestive heart failure (CHF), cardiac arrhythmia and atrial fibrillation. Furthermore, increasing evidence has indicated the potential cytotoxic effects of CGs against various types of cancer. In this review, we highlight some of the structural features of this class of natural products that are crucial for their efficacy, some methods of isolating these compounds from natural resources, and the structural elucidation tools that have been used. We also describe their physicochemical properties and several modern biotechnological approaches for preparing CGs that do not require plant sources.
Collapse
Affiliation(s)
- Hesham R El-Seedi
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Box 574, SE-75123, Uppsala, Sweden; Chemistry Department, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; H. E. J. Research Institute of Chemistry, International Center for Chemical Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom, Egypt.
| | - Shaden A M Khalifa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-106 91, Stockholm, Sweden
| | - Eman A Taher
- National Organization for Drug Control and Research (NODCAR), P.O. Box 29, Cairo, Egypt; Department of Chemistry, Royal Institute of Technology, KTH, Sweden
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr el Aini St., 11562 Cairo, Egypt; Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, New Cairo, Egypt
| | - Aamer Saeed
- Chemistry Department, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Mohamed Gamal
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom, Egypt
| | - Mohamed-Elamir F Hegazy
- Chemistry of Medicinal Plants Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt; Department of Pharmaceutical Biology, Institute of Pharmacy, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Diaa Youssef
- Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia
| | - Syed G Musharraf
- H. E. J. Research Institute of Chemistry, International Center for Chemical Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muaaz M Alajlani
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Box 574, SE-75123, Uppsala, Sweden
| | - Jianbo Xiao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Control in Chinese Medicine, University of Macau, Macau, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy, Johannes Gutenberg University, 55128 Mainz, Germany
| |
Collapse
|
46
|
Yang J, Mu X, Wang Y, Zhu D, Zhang J, Liang C, Chen B, Wang J, Zhao C, Zuo Z, Heng X, Zhang C, Zhang L. Dysbiosis of the Salivary Microbiome Is Associated With Non-smoking Female Lung Cancer and Correlated With Immunocytochemistry Markers. Front Oncol 2018; 8:520. [PMID: 30524957 PMCID: PMC6256243 DOI: 10.3389/fonc.2018.00520] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/23/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Association between oral bacteria and increased risk of lung cancer have been reported in several previous studies, however, the potential association between salivary microbiome and lung cancer in non-smoking women have not been evaluated. There is also no report on the relationship between immunocytochemistry markers and salivary microbiota. Method: In this study, we assessed the salivary microbiome of 75 non-smoking female lung cancer patients and 172 matched healthy individuals using 16S rRNA gene amplicon sequencing. We also calculated the Spearman's rank correlation coefficient between salivary microbiota and three immunohistochemical markers (TTF-1, Napsin A and CK7). Result: We analyzed the salivary microbiota of 247 subjects and found that non-smoking female lung cancer patients exhibited oral microbial dysbiosis. There was significantly lower microbial diversity and richness in lung cancer patients when compared to the control group (Shannon index, P < 0.01; Ace index, P < 0.0001). Based on the analysis of similarities, the composition of the microbiota in lung cancer patients also differed from that of the control group (r = 0.454, P < 0.001, unweighted UniFrac; r = 0.113, P < 0.01, weighted UniFrac). The bacterial genera Sphingomonas (P < 0.05) and Blastomonas (P < 0.0001) were relatively higher in non-smoking female lung cancer patients, whereas Acinetobacter (P < 0.001) and Streptococcus (P < 0.01) were higher in controls. Based on Spearman's correlation analysis, a significantly positive correlation can be observed between CK7 and Enterobacteriaceae (r = 0.223, P < 0.05). At the same time, Napsin A was positively associated with genera Blastomonas (r = 0.251, P < 0.05). TTF-1 exhibited a significantly positive correlation with Enterobacteriaceae (r = 0.262, P < 0.05). Functional analysis from inferred metagenomes indicated that oral microbiome in non-smoking female lung cancer patients were related to cancer pathways, p53 signaling pathway, apoptosis and tuberculosis. Conclusions: The study identified distinct salivary microbiome profiles in non-smoking female lung cancer patients, revealed potential correlations between salivary microbiome and immunocytochemistry markers used in clinical diagnostics, and provided proof that salivary microbiota can be an informative source for discovering non-invasive lung cancer biomarkers.
Collapse
Affiliation(s)
- Junjie Yang
- College of Life Science, Shandong Normal University, Jinan, China.,College of Life Science, Qilu Normal University, Jinan, China
| | - Xiaofeng Mu
- Clinical Laboratory and Core Research Laboratory, The Affiliated Central Hospital of Qingdao University, Qingdao, China.,Qingdao Human Microbiome Center, The Affiliated Central Hospital of Qingdao University, Qingdao, China.,Qingdao Institute of Oncology, The Affiliated Central Hospital of Qingdao University, Qingdao, China
| | - Ye Wang
- Clinical Laboratory and Core Research Laboratory, The Affiliated Central Hospital of Qingdao University, Qingdao, China.,Qingdao Human Microbiome Center, The Affiliated Central Hospital of Qingdao University, Qingdao, China.,Qingdao Institute of Oncology, The Affiliated Central Hospital of Qingdao University, Qingdao, China
| | - Dequan Zhu
- Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, China
| | - Jiaming Zhang
- College of Life Science, Shandong Normal University, Jinan, China
| | - Cheng Liang
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
| | - Bin Chen
- Shandong Children's Microbiome Center, Qilu Children's Hospital of Shandong University, Jinan, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Chemistry and Environment, Beihang University, Beijing, China
| | - Jingwen Wang
- College of Life Science, Shandong Normal University, Jinan, China
| | - Changying Zhao
- College of Life Science, Shandong Normal University, Jinan, China
| | - Zhiwen Zuo
- Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, China
| | - Xueyuan Heng
- Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, China
| | - Chunling Zhang
- Qingdao Human Microbiome Center, The Affiliated Central Hospital of Qingdao University, Qingdao, China.,Qingdao Institute of Oncology, The Affiliated Central Hospital of Qingdao University, Qingdao, China.,Department of Respiratory Medicine, The Affiliated Central Hospital of Qingdao University, Qingdao, China
| | - Lei Zhang
- College of Life Science, Shandong Normal University, Jinan, China.,Qingdao Human Microbiome Center, The Affiliated Central Hospital of Qingdao University, Qingdao, China.,Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, China.,Shandong Children's Microbiome Center, Qilu Children's Hospital of Shandong University, Jinan, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Chemistry and Environment, Beihang University, Beijing, China.,Shandong Institutes for Food and Drug Control, Jinan, China
| |
Collapse
|