1
|
Shan M, Xu L, Yang W, Liu S, Cui Z. Identification of hub genes and immune-related pathways in acute myeloid leukemia: insights from bioinformatics and experimental validation. Front Immunol 2025; 15:1511824. [PMID: 39867885 PMCID: PMC11757261 DOI: 10.3389/fimmu.2024.1511824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
Background This study aims to identify the hub genes and immune-related pathways in acute myeloid leukemia (AML) to provide new theories for immunotherapy. Methods We use bioinformatics methods to find and verify the hub gene. At the same time, we use the results of GSEA enrichment analysis to find immune-related mediators. Through Mendelian randomization(MR) analysis, on the one hand, we look for related immune cells, and on the other hand, we use it to determine the causal relationship among immune cells, immune mediators, and AML. Finally, in vitro experiments are conducted to further verify and improve the reliability and physiological functions of the hub gene and its immune-related pathways. Results Complement Factor D(CFD) gene is identified as the highly expressed hub gene and is positively correlated with IL-2. IL-2 is also positively correlated with CD27 on CD24+CD27+B cells, JAK/STAT, and PI3K/Akt. The latter three are positively correlated with the occurrence and development of AML. Conclusion We conclude that CFD gene uses IL-2 as a mediator to promote the disease progression of AML by promoting the CD27 on CD24+CD27+B cells, JAK-STAT, and PI3K-Akt pathways.
Collapse
Affiliation(s)
- Mingliang Shan
- Postdoctoral Workstation, Liaocheng People’s Hospital, Liaocheng, China
- Postdoctoral Mobile Stations, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Management, Shandong Second Medical University, Weifang, China
| | - Li Xu
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenzhe Yang
- Post - Doctoral Innovation Practice Base, Gaomi Maternity and Child Health Hospital, Gaomi, China
| | - Shiguo Liu
- Postdoctoral Mobile Stations, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhaoqing Cui
- Postdoctoral Workstation, Liaocheng People’s Hospital, Liaocheng, China
| |
Collapse
|
2
|
Mhaidly N, Barake N, Trelcat A, Journe F, Saussez S, Descamps G. Bufalin Suppresses Head and Neck Cancer Development by Modulating Immune Responses and Targeting the β-Catenin Signaling Pathway. Cancers (Basel) 2024; 16:2739. [PMID: 39123466 PMCID: PMC11311268 DOI: 10.3390/cancers16152739] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Bufalin, a cardiotonic steroid derived from the Chinese toad (Bufo gargarizans), has demonstrated potent anticancer properties across various cancer types, positioning it as a promising therapeutic candidate. However, comprehensive mechanistic studies specific to head and neck cancers have been lacking. Our study aimed to bridge this gap by investigating bufalin's mechanisms of action in head and neck cancer cells. Using several methods, such as Western blotting, immunofluorescence, and flow cytometry, we observed bufalin's dose-dependent reduction in cell viability, disruption of cell membrane integrity, and inhibition of colony formation in both HPV-positive and HPV-negative cell lines. Bufalin induces apoptosis through the modulation of apoptosis-related proteins, mitochondrial function, and reactive oxygen species production. It also arrests the cell cycle at the G2/M phase and attenuates cell migration while affecting epithelial-mesenchymal transition markers and targeting pivotal signaling pathways, including Wnt/β-catenin, EGFR, and NF-κB. Additionally, bufalin exerted immunomodulatory effects by polarizing macrophages toward the M1 phenotype, bolstering antitumor immune responses. These findings underscore bufalin's potential as a multifaceted therapeutic agent against head and neck cancers, targeting essential pathways involved in proliferation, apoptosis, cell cycle regulation, metastasis, and immune modulation. Further research is warranted to validate these mechanisms and optimize bufalin's clinical application.
Collapse
Affiliation(s)
- Nour Mhaidly
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Noura Barake
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Anne Trelcat
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Fabrice Journe
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium;
| | - Sven Saussez
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Géraldine Descamps
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| |
Collapse
|
3
|
Ren J, Lv L, Tao X, Zhai X, Chen X, Yu H, Zhao X, Kong X, Yu Z, Dong D, Liu J. The role of CBL family ubiquitin ligases in cancer progression and therapeutic strategies. Front Pharmacol 2024; 15:1432545. [PMID: 39130630 PMCID: PMC11310040 DOI: 10.3389/fphar.2024.1432545] [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: 05/14/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
The CBL (Casitas B-lineage lymphoma) family, as a class of ubiquitin ligases, can regulate signal transduction and activate receptor tyrosine kinases through various tyrosine kinase-dependent pathways. There are three members of the family: c-CBL, CBL-b, and CBL-c. Numerous studies have demonstrated the important role of CBL in various cellular pathways, particularly those involved in the occurrence and progression of cancer, hematopoietic development, and regulation of T cell receptors. Therefore, the purpose of this review is to comprehensively summarize the function and regulatory role of CBL family proteins in different human tumors, as well as the progress of drug research targeting CBL family, so as to provide a broader clinical measurement strategy for the treatment of tumors.
Collapse
Affiliation(s)
- Jiaqi Ren
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- School of Pharmacy, Dalian Medical University, Dalian, China
| | - Linlin Lv
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohan Zhai
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xuyang Chen
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hao Yu
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- School of Pharmacy, Dalian Medical University, Dalian, China
| | - Xinya Zhao
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- School of Pharmacy, Dalian Medical University, Dalian, China
| | - Xin Kong
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- School of Pharmacy, Dalian Medical University, Dalian, China
| | - Zhan Yu
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
4
|
Zutshi N, Mohapatra BC, Mondal P, An W, Goetz BT, Wang S, Li S, Storck MD, Mercer DF, Black AR, Thayer SP, Black JD, Lin C, Band V, Band H. Cbl and Cbl-b ubiquitin ligases are essential for intestinal epithelial stem cell maintenance. iScience 2024; 27:109912. [PMID: 38974465 PMCID: PMC11225835 DOI: 10.1016/j.isci.2024.109912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/29/2024] [Accepted: 05/03/2024] [Indexed: 07/09/2024] Open
Abstract
Receptor tyrosine kinases (RTKs) control stem cell maintenance vs. differentiation decisions. Casitas B-lineage lymphoma (CBL) family ubiquitin ligases are negative regulators of RTKs, but their stem cell regulatory roles remain unclear. Here, we show that Lgr5+ intestinal stem cell (ISC)-specific inducible Cbl-knockout (KO) on a Cblb null mouse background (iDKO) induced rapid loss of the Lgr5 Hi ISCs with transient expansion of the Lgr5 Lo transit-amplifying population. LacZ-based lineage tracing revealed increased ISC commitment toward enterocyte and goblet cell fate at the expense of Paneth cells. Functionally, Cbl/Cblb iDKO impaired the recovery from radiation-induced intestinal epithelial injury. In vitro, Cbl/Cblb iDKO led to inability to maintain intestinal organoids. Single-cell RNA sequencing in organoids identified Akt-mTOR (mammalian target of rapamycin) pathway hyperactivation upon iDKO, and pharmacological Akt-mTOR axis inhibition rescued the iDKO defects. Our results demonstrate a requirement for Cbl/Cblb in the maintenance of ISCs by fine-tuning the Akt-mTOR axis to balance stem cell maintenance vs. commitment to differentiation.
Collapse
Affiliation(s)
- Neha Zutshi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bhopal C. Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pinaki Mondal
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin T. Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shuo Wang
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sicong Li
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew D. Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David F. Mercer
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adrian R. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sarah P. Thayer
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jennifer D. Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chi Lin
- Department of Radiation Oncology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Vimla Band
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
5
|
Alfhili MA, Alsughayyir J. Bufalin reprograms erythrocyte lifespan through p38 MAPK and Rac1 GTPase. Toxicon 2024; 240:107636. [PMID: 38316294 DOI: 10.1016/j.toxicon.2024.107636] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Ample evidence indicates that bufalin (BFN), a cardiotonic steroid in Bufo toad toxin, possesses a potent anticancer activity mainly by stimulating apoptosis in cancer cells. Human red blood cells (RBCs) undergo eryptosis which contributes to a plethora of pathological conditions. No reports, however, have examined the potential toxicity of BFN to RBCs. This study aims to characterize the biochemical mechanisms governing the influence of BFN on the physiology and lifespan of RBCs. Isolated RBCs from healthy volunteers were exposed to anticancer concentrations of commercially available BFN from the skin of Bufo gargarizans (10-200 μM) for 24 h at 37 °C. Photometric assays were used to estimate hemolysis and hemolytic markers, and flow cytometry was used to detect eryptotic markers. Phosphatidylserine externalization was captured by fluorescein isothiocyante-labeled annexin V, cellular dimensions by light scatter patterns, and intracellular Ca2+ and reactive oxygen species (ROS) by fluorogenic dyes Fluo4/AM and 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA), respectively. BFN caused Ca2+-independent hemolysis and release of LDH, AST, CK, and K+, and increased annexin V-bound cells, cytosolic Ca2+, cell shrinkage, and ROS levels. BFN also disrupted Na+ and Mg2+ trafficking, and was sensitive to PEG 8000, sucrose, SB203580, and NSC 23766. In whole blood, BFN depleted hemoglobin stores, increased fragmented RBCs, and was selectively toxic to reticulocytes, lymphocytes, and platelets. In conclusion, BFN elicits premature RBC death, subject to regulation by p38 MAPK and Rac1 GTPase, and is detrimental to other peripheral blood cells. Altogether, these novel findings prompt cautious consideration of the toxin in anticancer therapy.
Collapse
Affiliation(s)
- Mohammad A Alfhili
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 12372, Saudi Arabia.
| | - Jawaher Alsughayyir
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 12372, Saudi Arabia
| |
Collapse
|
6
|
Wang S, Argikar UA, Cheruzel L, Cho S, Crouch RD, Dhaware D, Heck CJS, Johnson KM, Kalgutkar AS, King L, Liu J, Ma B, Maw H, Miller GP, Seneviratne HK, Takahashi RH, Wei C, Khojasteh SC. Bioactivation and reactivity research advances - 2022 year in review‡. Drug Metab Rev 2023; 55:267-300. [PMID: 37608698 DOI: 10.1080/03602532.2023.2244193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/05/2023] [Indexed: 08/24/2023]
Abstract
With the 50th year mark since the launch of Drug Metabolism and Disposition journal, the field of drug metabolism and bioactivation has advanced exponentially in the past decades (Guengerich 2023).This has, in a major part, been due to the continued advances across the whole spectrum of applied technologies in hardware, software, machine learning (ML), and artificial intelligence (AI). LC-MS platforms continue to evolve to support key applications in the field, and automation is also improving the accuracy, precision, and throughput of these supporting assays. In addition, sample generation and processing is being aided by increased diversity and quality of reagents and bio-matrices so that what is being analyzed is more relevant and translatable. The application of in silico platforms (applied software, ML, and AI) is also making great strides, and in tandem with the more traditional approaches mentioned previously, is significantly advancing our understanding of bioactivation pathways and how these play a role in toxicity. All of this continues to allow the area of bioactivation to evolve in parallel with associated fields to help bring novel or improved medicines to patients with urgent or unmet needs.Shuai Wang and Cyrus Khojasteh, on behalf of the authors.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Lionel Cheruzel
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Sungjoon Cho
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Rachel D Crouch
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, TN, USA
| | | | - Carley J S Heck
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Kevin M Johnson
- Drug Metabolism and Pharmacokinetics, Inotiv, Maryland Heights, MO, USA
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Lloyd King
- Quantitative Drug Discovery, UCB Biopharma UK, Slough, UK
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Hlaing Maw
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Grover P Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, USA
| | | | - Cong Wei
- Drug Metabolism and Pharmacokinetics, Biogen Inc., Cambridge, MA, USA
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
7
|
Ye Q, Zhou X, Ren H, Han F, Lin R, Li J. An overview of the past decade of bufalin in the treatment of refractory and drug-resistant cancers: current status, challenges, and future perspectives. Front Pharmacol 2023; 14:1274336. [PMID: 37860119 PMCID: PMC10582727 DOI: 10.3389/fphar.2023.1274336] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
Profound progress has been made in cancer treatment in the past three decades. However, drug resistance remains prevalent and a critical challenge. Drug resistance can be attributed to oncogenes mutations, activated defensive mechanisms, ATP-bind cassette transporters overexpression, cancer stem cells, etc. Chinese traditional medicine toad venom has been used for centuries for different diseases, including resistant cancers. Bufalin is one of the bufadienolides in toad venom that has been extensively studied for its potential in refractory and drug-resistant cancer treatments in vitro and in vivo. In this work, we would like to critically review the progress made in the past decade (2013-2022) of bufalin in overcoming drug resistance in cancers. Generally, bufalin shows high potential in killing certain refractory and resistant cancer cells via multiple mechanisms. More importantly, bufalin can work as a chemo-sensitizer that enhances the sensitivity of certain conventional and targeted therapies at low concentrations. In addition, the development of bufalin derivatives was also briefly summarized and discussed. We also analyzed the obstacles and challenges and provided possible solutions for future perspectives. We hope that the collective information may help evoke more effort for more in-depth studies and evaluation of bufalin in both lab and possible clinical trials.
Collapse
Affiliation(s)
- Qingmei Ye
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, Hainan, China
| | - Xin Zhou
- The Fifth People’s Hospital of Hainan Province & Affiliated Dermatology Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Han Ren
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Fangxuan Han
- Hainan General Hospital & Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Rong Lin
- Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Juan Li
- Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| |
Collapse
|
8
|
Zutshi N, Mohapatra BC, Mondal P, An W, Goetz BT, Wang S, Li S, Storck MD, Mercer DF, Black AR, Thayer SP, Black JD, Lin C, Band V, Band H. Cbl and Cbl-b Ubiquitin Ligases are Essential for Intestinal Epithelial Stem Cell Maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541154. [PMID: 37292716 PMCID: PMC10245689 DOI: 10.1101/2023.05.17.541154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Among the signaling pathways that control the stem cell self-renewal and maintenance vs. acquisition of differentiated cell fates, those mediated by receptor tyrosine kinase (RTK) activation are well established as key players. CBL family ubiquitin ligases are negative regulators of RTKs but their physiological roles in regulating stem cell behaviors are unclear. While hematopoietic Cbl/Cblb knockout (KO) leads to a myeloproliferative disease due to expansion and reduced quiescence of hematopoietic stem cells, mammary epithelial KO led to stunted mammary gland development due to mammary stem cell depletion. Here, we examined the impact of inducible Cbl/Cblb double-KO (iDKO) selectively in the Lgr5-defined intestinal stem cell (ISC) compartment. Cbl/Cblb iDKO led to rapid loss of the Lgr5 Hi ISC pool with a concomitant transient expansion of the Lgr5 Lo transit amplifying population. LacZ reporter-based lineage tracing showed increased ISC commitment to differentiation, with propensity towards enterocyte and goblet cell fate at the expense of Paneth cells. Functionally, Cbl/Cblb iDKO impaired the recovery from radiation-induced intestinal epithelial injury. In vitro , Cbl/Cblb iDKO led to inability to maintain intestinal organoids. Single cell RNAseq analysis of organoids revealed Akt-mTOR pathway hyperactivation in iDKO ISCs and progeny cells, and pharmacological inhibition of the Akt-mTOR axis rescued the organoid maintenance and propagation defects. Our results demonstrate a requirement for Cbl/Cblb in the maintenance of ISCs by fine tuning the Akt-mTOR axis to balance stem cell maintenance vs. commitment to differentiation.
Collapse
|
9
|
Tu D, Ning J, Zou L, Wang P, Zhang Y, Tian X, Zhang F, Zheng J, Ge G. Unique Oxidative Metabolism of Bufalin Generates Two Reactive Metabolites That Strongly Inactivate Human Cytochrome P450 3A. J Med Chem 2022; 65:4018-4029. [DOI: 10.1021/acs.jmedchem.1c01875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Dongzhu Tu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Ning
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Liwei Zou
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yani Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangge Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550025, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
10
|
The Antagonistic Effect of Glutamine on Zearalenone-Induced Apoptosis via PI3K/Akt Signaling Pathway in IPEC-J2 Cells. Toxins (Basel) 2021; 13:toxins13120891. [PMID: 34941728 PMCID: PMC8704905 DOI: 10.3390/toxins13120891] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 11/29/2022] Open
Abstract
Zearalenone (ZEN) is a non-steroidal estrogen mycotoxin produced by Fusarium fungi, which inevitably exists in human and animal food or feed. Previous studies indicated that apoptosis seems to be a key determinant of ZEN-induced toxicity. This experiment aimed to investigate the protective effects of Glutamine (Gln) on ZEN-induced cytotoxicity in IPEC-J2 cells. The experimental results showed that Gln was able to alleviate the decline of cell viability and reduce the production of reactive oxygen species and calcium (Ca2+) induced by ZEN. Meanwhile, the mRNA expression of antioxidant enzymes such as glutathione reductase, glutathione peroxidase, and catalase was up-regulated after Gln addition. Subsequently, Gln supplementation resulted in the nuclear fission and Bad-fluorescence distribution of apoptotic cells were weakened, and the mRNA expression and protein expression of pro-apoptotic genes and apoptotic rates were significantly reduced. Moreover, ZEN reduced the phosphorylation Akt, decreased the expression of Bcl-2, and increased the expression of Bax. Gln alleviated the above changes induced by ZEN and the antagonistic effects of Gln were disturbed by PI3K inhibitor (LY294002). To conclude, this study revealed that Gln exhibited significant protective effects on ZEN-induced apoptosis, and this effect may be attributed to the PI3K/Akt signaling pathway.
Collapse
|
11
|
Nakamura K, Shiozaki A, Kosuga T, Shimizu H, Kudou M, Ohashi T, Arita T, Konishi H, Komatsu S, Kubota T, Fujiwara H, Okamoto K, Kishimoto M, Konishi E, Otsuji E. The expression of the alpha1 subunit of Na +/K +-ATPase is related to tumor development and clinical outcomes in gastric cancer. Gastric Cancer 2021; 24:1278-1292. [PMID: 34251542 DOI: 10.1007/s10120-021-01212-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND The Na+/K+-ATPase alpha1 subunit (ATP1A1) is a critical component of Na+/K+-ATPase (NKA), a membrane pump that maintains a low intracellular Na+/K+ ratio and retains cellular volume and osmolarity. ATP1A1 was recently implicated in tumor behavior. Therefore, the present study investigated the role of ATP1A1 in patients with gastric cancer (GC). METHODS Knockdown experiments were conducted on human GC cell lines using ATP1A1 siRNA, and its effects on proliferation, the cell cycle, apoptosis, and cellular movement were examined. Gene expression profiling was performed by a microarray analysis. Primary tumor samples from 192 GC patients who underwent gastrectomy were subjected to an immunohistochemical analysis. RESULTS High ATP1A1 expression levels were observed in NUGC4 and MKN74 cells. Cell proliferation was suppressed and apoptosis was induced by the siRNA-induced knockdown of ATP1A1. The microarray analysis showed that knockdown of ATP1A1 leads to the up-regulated expression of genes involved in the interferon (IFN) signaling pathway, such as STAT1, STAT2, IRF1, and IRF9. Furthermore, the depletion of ATP1A1 altered the phosphorylation of the MAPK pathway. The immunohistochemical analysis revealed that the expression of ATP1A1 was associated with the histological type, venous invasion, and the pathological T stage. Furthermore, the prognostic analysis showed a relationship between high ATP1A1 expression levels and poor postoperative survival. CONCLUSIONS ATP1A1 appears to regulate tumor progression by altering IFN signaling, and high ATP1A1 expression levels were associated with poor postoperative survival in GC patients. The present results provide novel insights into the function of ATP1A1 as a mediator and/or biomarker of GC.
Collapse
Affiliation(s)
- Kei Nakamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Michihiro Kudou
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Mitsuo Kishimoto
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.,Department of Pathology, Kyoto City Hospital, Kyoto, 604-8845, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
12
|
Zou D, Song J, Deng M, Ma Y, Yang C, Liu J, Wang S, Wen Z, Tang Y, Qu X, Zhang Y. Bufalin inhibits peritoneal dissemination of gastric cancer through endothelial nitric oxide synthase-mitogen-activated protein kinases signaling pathway. FASEB J 2021; 35:e21601. [PMID: 33913201 DOI: 10.1096/fj.202002780r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/19/2021] [Accepted: 04/01/2021] [Indexed: 01/11/2023]
Abstract
Peritoneal dissemination threatens the survival of patients with gastric cancer (GC). Bufalin is an extract of traditional Chinese medicine, which has been proved to have anticancer effect. The target of bufalin in suppressing gastric cancer peritoneal dissemination (GCPD) and the underlying mechanism are still unclear. In this research, GC cell line MGC-803 and high-potential peritoneal dissemination cell line MKN-45P were treated with bufalin or L-NAME. Malignant biological behavior and protein level of GC cell lines were detected with MTT, wound healing, transwell, adhesion, and western blotting. Bioinformatics analysis and patient tissues were used to verify the role of endothelial nitric oxide synthase (NOS3) in GC. Mice model was used to assess the effect of bufalin and role of NOS3 in vivo. We found that bufalin inhibited the proliferation, invasion, and migration in GC cell lines. NOS3, which was an independent prognostic factor of GC patients, was predicted to be a potential target of bufalin. Further experiments proved that bufalin reduced the phosphorylation of NOS3, thereby inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway, and ultimately suppressed GCPD by inhibiting EMT process. In conclusion, NOS3 was a potential therapeutic target and prognostic biomarker of GC. Bufalin could suppress GCPD through NOS3-MAPK signaling pathway, which provided more evidence support for intraperitoneal perfusion of bufalin to treat GCPD.
Collapse
Affiliation(s)
- Dan Zou
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Jincheng Song
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China.,Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mingming Deng
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yanju Ma
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, China
| | - Chunjiao Yang
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Jiaqing Liu
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Song Wang
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Zhenpeng Wen
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Yu Tang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Sheng X, Zhu P, Zhao Y, Zhang J, Li H, Zhao H, Qin J. Effect of PI3K/AKT/mTOR Signaling Pathway on Regulating and Controlling the Anti-Invasion and Metastasis of Hepatoma Cells by Bufalin. Recent Pat Anticancer Drug Discov 2021; 16:54-65. [PMID: 33530915 DOI: 10.2174/1574892816666210201120324] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND Autophagy plays a "double-edged sword" in the process of tumorigenesis, development and metastasis. OBJECTIVE In this study, we explored the effect of PI3K/AKT/mTOR autophagy-related signaling pathway on regulating and controlling the invasion and metastasis of liver cancer cells by Bufalin. METHODS The cell counting, migration, adhesion and invasion assay were used to evaluate the effect of Bufalin on cell proliferation, invasion and metastasis. The protein expression of PI3K/AKT/ mTOR signaling pathway were detected by the Western Blotting technique. RESULTS After inhibiting autophagy of HCC-LM3 cells, the inhibitory effect of Bufalin on adhesion, migration and invasion of HCC-LM3 cells was significantly enhanced. Synergistic inhibition was strongest when different autophagy inhibitors were combined with 3MA and CQ. After inhibiting autophagy, Bufalin significantly inhibited the protein expression of P-AKT, Cyclin D1, MMP- 2, MMP-9 and VEGF in HCC-LM3 cells. The protein expression of PTEN and E-Cadherin in HCC-LM3 cells was significantly increased. CONCLUSION The present study shows that the anti-tumor effect of Bufalin mainly inhibit proliferation, extracellular matrix degradation and angiogenesis of HCC by influencing autophagy. These findings confirm the capability of Bufalin in inhibiting metastasis of HCC and in parallel to current patents, could be applied as a novel therapeutic strategy in the prevention of metastasis of HCC.
Collapse
Affiliation(s)
- Xia Sheng
- Department of Pathology, Affiliated to Minhang Hospital, Fudan University, Xinsong Rd. 170, Minhang District, Shanghai 201100, China
| | - Pengfei Zhu
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Yi Zhao
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Jinwei Zhang
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Haijia Li
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Huan Zhao
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Jianmin Qin
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| |
Collapse
|
14
|
Li FJ, Hu JH, Ren X, Zhou CM, Liu Q, Zhang YQ. Toad venom: A comprehensive review of chemical constituents, anticancer activities, and mechanisms. Arch Pharm (Weinheim) 2021; 354:e2100060. [PMID: 33887066 DOI: 10.1002/ardp.202100060] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 12/31/2022]
Abstract
Toad venom, a traditional natural medicine, has been used for hundreds of years in China for treating different diseases. Many studies have been performed to elucidate the cardiotonic and analgesic activities of toad venom. Until the last decade, an increasing number of studies have documented that toad venom is a source of lead compound(s) for the development of potential cancer treatment drugs. Research has shown that toad venom contains 96 types of bufadienolide monomers and 23 types of indole alkaloids, such as bufalin, cinobufagin, arenobufagin, and resibufogenin, which exhibit a wide range of anticancer activities in vitro and, in particular, in vivo for a range of cancers. The main antitumor mechanisms are likely to be apoptosis or/and autophagy induction, cell cycle arrest, cell metastasis suppression, reversal of drug resistance, or growth inhibition of cancer cells. This review summarizes the chemical constituents of toad venom, analyzing their anticancer activities and molecular mechanisms for cancer treatments. We also outline the importance of further studies regarding the material basis and anticancer mechanisms of toad venom.
Collapse
Affiliation(s)
- Fang-Jie Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing-Hong Hu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan, China
| | - Xin Ren
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cheng-Mei Zhou
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan, China
| | - Yong-Qing Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
15
|
Xu Y, Tang L, Chen P, Chen M, Zheng M, Shi F, Wang Y. Tumor-Targeted Delivery of Bufalin-Loaded Modified Albumin-Polymer Hybrid for Enhanced Antitumor Therapy and Attenuated Hemolysis Toxicity and Cardiotoxicity. AAPS PharmSciTech 2021; 22:137. [PMID: 33880681 DOI: 10.1208/s12249-021-02000-2] [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: 11/05/2020] [Accepted: 03/22/2021] [Indexed: 12/19/2022] Open
Abstract
A novel albumin polymer hybrid with a core-shell structure was designed to target delivery of bufalin, which is an antineoplastic monomer with serious cardiotoxicity. The sheath layer was composed of ursodeoxycholic acid (UA)-modified bovine serum albumin (UA-BSA), while the stable core consisted of poly n-butyl cyanoacrylate (PBCA) nanoparticles. The UA-BSA was synthetized, and the substitution degree was characterized. The physical properties of bufalin-loaded UA-modified protein-PBCA nanocomplexes (BF-uPPNCs), such as morphology, particle size, and encapsulation efficiency, were evaluated. FTIR and DSC revealed the bufalin to be in an amorphous state. Furthermore, the in vitro release study indicated a sustained release profile of BF-uPPNCs. The MTT and cellular uptake study demonstrated that BF-uPPNCs significantly improved the inhibitory effect of the bufalin accompanied with an enhanced cell uptake capacity on HepG2 cells. In addition, in vivo research demonstrated that BF-uPPNCs had a better antitumor effect coupled with improved therapeutic effect, and reduced hemolysis, vascular irritation, and cardiotoxicity. This work therefore presented a novel albumin polymer hybrid with favorable stability, efficient tumor-targeted delivery potential, and side effect reduction ability, which can be a potential vehicle for an anticancer drug.
Collapse
|
16
|
Langat L, Langat MK, Wetschnig W, Knirsch W, Mulholland DA. Antiproliferative Bufadienolides from the Bulbs of Drimia altissima. JOURNAL OF NATURAL PRODUCTS 2021; 84:608-615. [PMID: 33478223 DOI: 10.1021/acs.jnatprod.0c01079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The bulbs of the South African Drimia altissima (Asparagaceae or Hyacinthaceae sensu APGII) have yielded a range of previously undescribed bufadienolides, drimianins A-G (1-7), the known bufadienolides bovogenin A (8), 3β-O-β-d-glucopyranosylbovogenin A (9), scillaren F (10), and altoside (11), the known homoisoflavonoid (3S)-3-(4'-methoxybenzyl)-5,6,7-trimethoxychroman-4-one (urgineanin C), the sesquiterpenoids 1β,6α-dihydroxy-4(15)-eudesmene and 6α-hydroxy-4(15)-eudesmen-1-one, polybotrin, adenosine, and 9R-hydroxy-(10E,12Z)-octadecadienoic acid ethyl ester. The bufadienolides isolated were tested at 10 μM in the NCI-60 cancer cell screen, and nine of these were selected for further screening at five concentrations. Drimianins C (3) and E (5) showed activity at the nanomolar level against a number of human cancer cell lines in the NCI-60 screen.
Collapse
Affiliation(s)
- Linda Langat
- Natural Products Research Group, Department of Chemistry, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Moses K Langat
- Natural Products Research Group, Department of Chemistry, University of Surrey, Guildford, GU2 7XH, United Kingdom
- Department of Chemistry, University of KwaZulu-Natal, Durban, 4041, South Africa
| | | | - Walter Knirsch
- Institute of Biology, NAWI Graz, University of Graz, 8010 Graz, Austria
| | - Dulcie A Mulholland
- Natural Products Research Group, Department of Chemistry, University of Surrey, Guildford, GU2 7XH, United Kingdom
- Department of Chemistry, University of KwaZulu-Natal, Durban, 4041, South Africa
| |
Collapse
|
17
|
Bawa S, Piccirillo R, Geisbrecht ER. TRIM32: A Multifunctional Protein Involved in Muscle Homeostasis, Glucose Metabolism, and Tumorigenesis. Biomolecules 2021; 11:biom11030408. [PMID: 33802079 PMCID: PMC7999776 DOI: 10.3390/biom11030408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/13/2022] Open
Abstract
Human tripartite motif family of proteins 32 (TRIM32) is a ubiquitous multifunctional protein that has demonstrated roles in differentiation, muscle physiology and regeneration, and tumor suppression. Mutations in TRIM32 result in two clinically diverse diseases. A mutation in the B-box domain gives rise to Bardet–Biedl syndrome (BBS), a disease whose clinical presentation shares no muscle pathology, while mutations in the NHL (NCL-1, HT2A, LIN-41) repeats of TRIM32 causes limb-girdle muscular dystrophy type 2H (LGMD2H). TRIM32 also functions as a tumor suppressor, but paradoxically is overexpressed in certain types of cancer. Recent evidence supports a role for TRIM32 in glycolytic-mediated cell growth, thus providing a possible mechanism for TRIM32 in the accumulation of cellular biomass during regeneration and tumorigenesis, including in vitro and in vivo approaches, to understand the broad spectrum of TRIM32 functions. A special emphasis is placed on the utility of the Drosophila model, a unique system to study glycolysis and anabolic pathways that contribute to the growth and homeostasis of both normal and tumor tissues.
Collapse
Affiliation(s)
- Simranjot Bawa
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA;
| | - Rosanna Piccirillo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy;
| | - Erika R. Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA;
- Correspondence: ; Tel.: +1-(785)-532-3105
| |
Collapse
|
18
|
Geraniin inhibits proliferation and induces apoptosis through inhibition of phosphatidylinositol 3-kinase/Akt pathway in human colorectal cancer in vitro and in vivo. Anticancer Drugs 2021; 31:575-582. [PMID: 32427739 DOI: 10.1097/cad.0000000000000929] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Geraniin, a polyphenolic component isolated from Phyllanthus amarus, has been reported to possess diverse biological activities, including antitumor, antiinflammatory, antihyperglycemic, antihypertensive, and antioxidant. However, the role and underlying mechanisms of geraniin in colorectal cancer still remain unclear. In the present study, we found that geraniin notably inhibited cell proliferation and clonogenic formation of colorectal cancer cell SW480 and HT-29 in a dose-dependent manner by Cell Counting Kit 8, EdU, and colony formation assays, respectively. Additionally, geraniin remarkably induced apoptosis of SW480 and HT-29 cells in a dose-dependent way by Hoechst 33342 staining, flow cytometric analysis, and TdT-mediated dUTP nick-end labeling assays and increased the expressions of Bax, caspase-3, and caspase-9, while decreased the level of Bcl-2. Besides, wound healing, transwell migration, and invasion assays demonstrated that geraniin obviously inhibited the migration and invasion of SW480 and HT-29 cells. Moreover, it also inhibited the levels of phospho (p)-phosphatidylinositol 3-kinase and p-Akt. Furthermore, in-vivo animal study revealed that geraniin had the significant inhibitory effects on tumor growth and promoted cancer cell apoptosis remarkably, which further confirmed the antitumor effect of geraniin. Taken together, the present study exhibited the positive role of geraniin in inhibiting proliferation and inducing apoptosis through suppression of phosphatidylinositol 3-kinase/Akt pathway in colorectal cancer cells in vitro and in vivo, which might provide new insights in searching for new drug candidates of anticolorectal cancer.
Collapse
|
19
|
Yao M, Zhang J, Li Z, Bai X, Ma J, Li Y. Liraglutide Protects Nucleus Pulposus Cells Against High-Glucose Induced Apoptosis by Activating PI3K/Akt/ mTOR/Caspase-3 and PI3K/Akt/GSK3β/Caspase-3 Signaling Pathways. Front Med (Lausanne) 2021; 8:630962. [PMID: 33681258 PMCID: PMC7933515 DOI: 10.3389/fmed.2021.630962] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Background and Objective: Diabetes mellitus (DM) is reportedly a significant risk factor for intervertebral disc degeneration (IDD). Incretin system and particularly glucagon-like peptide 1 (GLP-1) because of its glucose-lowering effects has become an important target in therapeutic strategies of type 2 diabetes (T2D). Liraglutide is a GLP-1 receptor (GLP-1R) agonist with glucoregulatory and insulinotropic functions as well as regulatory functions on cell proliferation, differentiation, and apoptosis. However, little is known on the roles and signaling pathways of apoptosis protecting effects of liraglutide in IDD. This study aimed to investigate the potential protective effects of liraglutide against high glucose-induced apoptosis of nucleus pulposus cells (NPCs) and the possible involved signaling pathways. Methods: The human NPCs were incubated with 100 nM liraglutide alone or in combination with LY294002 (PI3K inhibitor), rapamycin (mTOR inhibitor), and SB216763 (GSK3β inhibitor) in a high glucose culture for 48 h. The four groups were assessed further for apoptosis and genes expressions. The apoptotic effect was evaluated by flow cytometry and further confirmed by cell death detection enzyme-linked immunoassay plus (ELISAPLUS). The gene and protein expression levels were assessed by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting techniques. The results were comparatively assessed between the four groups. Results: The results confirmed the presence of GLP-1R in the NPCs indicating that liraglutide inhibited the high glucose-induced apoptosis, which was blocked by silencing GLP-1R with siRNA. Moreover, liraglutide stimulated the phosphorylation of Akt, mTOR and GSK3β. Treatment with LY294002 significantly increased the apoptosis of NPCs and reduced the levels of their downstream substrates (p-AKT, p-mTOR, and p-GSK3β). Further assessments revealed that activation of mTOR and GSK3β was almost completely inhibited by rapamycin and SB216763, respectively, which significantly increased the caspase-3 levels. Conclusion: Liraglutide could protect NPCs against high glucose-induced apoptosis by activating the PI3K/AKT/mTOR/caspase-3 and PI3K/AKT/GSK3β/caspase-3 signaling pathways.
Collapse
Affiliation(s)
- Mingyan Yao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Jing Zhang
- Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Zhihong Li
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Xiaoliang Bai
- Department of Orthopedics, Baoding No.1 Central Hospital, Baoding, China
| | - Jinhui Ma
- Department of Endocrinology, Affiliated Hospital of Hebei University, Baoding, China
| | - Yukun Li
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Agalakova NI, Kolodkin NI, Adair CD, Trashkov AP, Bagrov AY. Preeclampsia: Cardiotonic Steroids, Fibrosis, Fli1 and Hint to Carcinogenesis. Int J Mol Sci 2021; 22:ijms22041941. [PMID: 33669287 PMCID: PMC7920043 DOI: 10.3390/ijms22041941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/31/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
Despite prophylaxis and attempts to select a therapy, the frequency of preeclampsia does not decrease and it still takes the leading position in the structure of maternal mortality and morbidity worldwide. In this review, we present a new theory of the etiology and pathogenesis of preeclampsia that is based on the interaction of Na/K-ATPase and its endogenous ligands including marinobufagenin. The signaling pathway of marinobufagenin involves an inhibition of transcriptional factor Fli1, a negative regulator of collagen synthesis, followed by the deposition of collagen in the vascular tissues and altered vascular functions. Moreover, in vitro and in vivo neutralization of marinobufagenin is associated with the restoration of Fli1. The inverse relationship between marinobufagenin and Fli1 opens new possibilities in the treatment of cancer; as Fli1 is a proto-oncogene, a hypothesis on the suppression of Fli1 by cardiotonic steroids as a potential anti-tumor therapeutic strategy is discussed as well. We propose a novel therapy of preeclampsia that is based on immunoneutralization of the marinobufagenin by monoclonal antibodies, which is capable of impairing marinobufagenin-Na/K-ATPase interactions.
Collapse
Affiliation(s)
- Natalia I. Agalakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, 194223 St. Petersburg, Russia;
| | - Nikolai I. Kolodkin
- State Institute of Highly Pure Biopreparations and Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, 194223 St. Petersburg, Russia; or
| | - C. David Adair
- Department of Obstetrics and Gynecology, University of Tennessee, Chattanooga, TN 37402, USA; or
| | - Alexander P. Trashkov
- Konstantinov St. Petersburg Nuclear Physics Institute, National Research Centre Kurchatov Institute, 1 Orlova Roshcha, 188300 Gatchina, Russia;
| | - Alexei Y. Bagrov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, 44 Torez Prospect, 194223 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
21
|
Huang HC, Chang WT, Lee MS, Chen HY, Chen YH, Lin CC, Lin MK. Three bufadienolides induce cell death in the human lung cancer cell line CL1-5 mainly through autophagy. Bioorg Med Chem Lett 2021; 31:127715. [PMID: 33246109 DOI: 10.1016/j.bmcl.2020.127715] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/10/2020] [Accepted: 11/20/2020] [Indexed: 12/09/2022]
Abstract
The effects of 3 bufadienolides, namely kalantuboside B, kalantuboside A, and bryotoxin C, isolated from Kalanchoe tubiflora (Harvey) were evaluated and characterized in CL1-5 highly metastatic human lung cancer cells. In contrast to their apoptosis-promoting activity in other cancer cells, these bufadienolides only slight or did not induce apoptosis in CL1-5 cancer cells. Instead, they activated an autophagy pathway, as indicated by increased autophagosome formation. Autophagy induced by these bufadienolides was demonstrated to be linked to the down-regulation of p-mTOR and the up-regulation of LC3-II, ATG5, ATG7, and Beclin-1. Our findings revealed an autophagy as the alternative mechanism of drug action by bufadienolides in CL1-5 lung cancer cells and provided evidence that bufadienolides are a potential therapeutic strategy for highly metastatic human lung cancer.
Collapse
Affiliation(s)
- Hui-Chi Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Wen-Te Chang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Meng-Shiou Lee
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Hsing-Yu Chen
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Yu-Hua Chen
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| | - Chi-Chen Lin
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National ChungHsing University, 145 Xingda Rd., South Dist., Taichung 402, Taiwan
| | - Ming-Kuem Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan
| |
Collapse
|
22
|
Zhao Q, Peng C, Zheng C, He XH, Huang W, Han B. Recent Advances in Characterizing Natural Products that Regulate Autophagy. Anticancer Agents Med Chem 2020; 19:2177-2196. [PMID: 31749434 DOI: 10.2174/1871520619666191015104458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/16/2018] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
Autophagy, an intricate response to nutrient deprivation, pathogen infection, Endoplasmic Reticulum (ER)-stress and drugs, is crucial for the homeostatic maintenance in living cells. This highly regulated, multistep process has been involved in several diseases including cardiovascular and neurodegenerative diseases, especially in cancer. It can function as either a promoter or a suppressor in cancer, which underlines the potential utility as a therapeutic target. In recent years, increasing evidence has suggested that many natural products could modulate autophagy through diverse signaling pathways, either inducing or inhibiting. In this review, we briefly introduce autophagy and systematically describe several classes of natural products that implicated autophagy modulation. These compounds are of great interest for their potential activity against many types of cancer, such as ovarian, breast, cervical, pancreatic, and so on, hoping to provide valuable information for the development of cancer treatments based on autophagy.
Collapse
Affiliation(s)
- Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Chuan Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Xiang-Hong He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China.,The RNA Institute, University at Albany, State University of New York, Albany, NY 12222, United States
| |
Collapse
|
23
|
Hsu SK, Chang WT, Lin IL, Chen YF, Padalwar NB, Cheng KC, Teng YN, Wang CH, Chiu CC. The Role of Necroptosis in ROS-Mediated Cancer Therapies and Its Promising Applications. Cancers (Basel) 2020; 12:E2185. [PMID: 32764483 PMCID: PMC7465132 DOI: 10.3390/cancers12082185] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past decades, promising therapies targeting different signaling pathways have emerged. Among these pathways, apoptosis has been well investigated and targeted to design diverse chemotherapies. However, some patients are chemoresistant to these therapies due to compromised apoptotic cell death. Hence, exploring alternative treatments aimed at different mechanisms of cell death seems to be a potential strategy for bypassing impaired apoptotic cell death. Emerging evidence has shown that necroptosis, a caspase-independent form of cell death with features between apoptosis and necrosis, can overcome the predicament of drug resistance. Furthermore, previous studies have also indicated that there is a close correlation between necroptosis and reactive oxygen species (ROS); both necroptosis and ROS play significant roles both under human physiological conditions such as the regulation of inflammation and in cancer biology. Several small molecules used in experiments and clinical practice eliminate cancer cells via the modulation of ROS and necroptosis. The molecular mechanisms of these promising therapies are discussed in detail in this review.
Collapse
Affiliation(s)
- Sheng-Kai Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Wen-Tsan Chang
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Ling Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Nitin Balkrushna Padalwar
- Department of Chemistry, National Institute of Technology Tiruchirappalli, Tiruchirappalli 620015, Tamilnadu, India;
| | - Kai-Chun Cheng
- Department of Ophthalmology, Kaohsiung Municipal Hsiaokang Hospital, Kaohsiung 812, Taiwan;
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Chi-Huei Wang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
24
|
LingHu HR, Luo H, Gang L. Bufalin Induces Glioma Cell Death by Apoptosis or Necroptosis. Onco Targets Ther 2020; 13:4767-4778. [PMID: 32581545 PMCID: PMC7274536 DOI: 10.2147/ott.s242567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/04/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Bufalin is a component of Chinese traditional medicine, Chansu, which is reported to induce cell death among various kinds of tumors. Apoptosis evasion is a common problem of cancer treatment. Materials and Methods The proliferation of U-87 and U-373 treated by bufalin combined with or without apoptosis inhibitor was detected by MTT assay. The protein levels related to apoptosis and necroptosis were measured by Western blotting. Immunoprecipitation (IP) was applied for monitoring the formation of necrosome. The gene knockdown by CRISPR/Cas9 was applied to determine the roles of the proteins in apoptosis and necroptosis. Results In this study, we found that bufalin could induce apoptosis or necroptosis when U-87 and U-373 escaped from apoptosis. Bufalin triggered cell death by upregulating tumor necrosis factor (TNF) -α, TNF receptor 1 (TNFR1) and receptor-interacting protein 1 (RIPK1). Antagonizing cellular inhibitor of apoptosis 1 (cIAP1) and cIAP2 were also contributory. Caspase-8 activation led to apoptosis. When caspase-8 was functionally lost, necrosome consisted of RIPK1, receptor-interacting protein 3 (RIPK3) and mixed lineage kinase domain-like protein (MLKL) formed and necroptosis happened. The knockdown of above genes or the drug treatment confirmed the mechanism of bufalin-induced cell death. Cytotoxicity of bufalin to caspase-8 knockdown cell lines made control cell lines more sensitive to bufalin in their mixture. Discussion The cytotoxicity of bufalin to U-87 and U-373 was by inducing apoptosis or necroptosis when they were sensitive to apoptosis or not. The results indicated that seeking for treatments that could induce apoptosis and necroptosis was a good solution for the tumor evasion of apoptosis.
Collapse
Affiliation(s)
- Hai Rui LingHu
- Department of Neurosurgery and Neurocritical Care, Beijing Chaoyang Integrative Medicine Emergency Medical Center, Beijing 100022, People's Republic of China
| | - Hui Luo
- Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, People's Republic of China
| | - Lin Gang
- Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, People's Republic of China
| |
Collapse
|
25
|
Wei WL, An YL, Zhang YZ, Li ZW, Zhou Y, Lei M, Zhang JQ, Qu H, Da J, Wu WY, Guo DA. Quantitative analysis of fourteen bufadienolides in Venenum Bufonis crude drug and its Chinese patent medicines by ultra-high performance liquid chromatography coupled with tandem mass spectrometry. JOURNAL OF ETHNOPHARMACOLOGY 2020; 251:112490. [PMID: 31884035 DOI: 10.1016/j.jep.2019.112490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Venenum Bufonis, a product of the secretions of Bufo gargarizans Cantor or B. melanostictus Schneider, possessed an array of pharmacological activities, such as cardiotonic, anti-tumor, antinociceptive, anti-inflammatory, anesthetic and antimicrobial activities. However, there were few efficient methods for quality evaluation of Venenum Bufonis medicinal materials and its related Chinese patent medicines. AIM OF THE STUDY To establish an effective method for quality assessment of crude drugs and Chinese proprietary medicines of Venenum Bufonis, and explore the relationship of primary compounds - target - pathway - disease through a series of network databases. MATERIALS AND METHODS An ultra-high performance liquid chromatography coupled with tandem mass spectrometry (UHPLC-QqQ-MS/MS) method was developed and validated to simultaneously determine 14 bufadienolides for quantitative analysis of 71 batches of crude drugs and 20 kinds of Chinese patent medicines of Venenum Bufonis. Multiple reaction monitoring with good specificity and accuracy was applied to monitor the 14 bufadienolides in positive mode. RESULTS The methodology was validated with good specificity, precision, stability, repeatability and recovery. The low limits of quantification were in the range of 0.1-2.7 ng/mL. The relative standard deviation values for intra- and inter-day precisions ranged from 0.98% to 6.3% and from 2.39% to 6.76%, respectively. The recovery was varied from 87.78% to 110.57% for crude drugs and 88.32%-100.96% for Chinese proprietary medicine (Shexiang Baoxin Pill). The contents of 14 analytes in 71 batches of crude drugs and 20 sorts of Chinese proprietary medicines were procured, the results showed that the contents of crude drugs collected from the market exhibited great variations. Furthermore, 13 batches of crude drugs were identified as counterfeit with no bufadienolides detected. In addition, the total contents of bufadienolides in the same drug showed great difference among products from various manufacturers or brands. Subsequently, 9 bufadienolides with the higher contents were applied to screen the anti-tumor effect by network pharmacology, and 8 pathways which had prior correlation with bufadienolides were disclosed. CONCLUSION This method could be used for quality assessment of crude drugs and Chinese patent medicines of Venenum Bufonis, and the data could be served as the fundamental basis for drug research and development of Venenum Bufonis.
Collapse
Affiliation(s)
- Wen-Long Wei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Ya-Ling An
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Yan-Zhi Zhang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Zhen-Wei Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Yang Zhou
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Min Lei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Jian-Qing Zhang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Hua Qu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Juan Da
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Wan-Ying Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China.
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China.
| |
Collapse
|
26
|
Veschi S, Ronci M, Lanuti P, De Lellis L, Florio R, Bologna G, Scotti L, Carletti E, Brugnoli F, Di Bella MC, Bertagnolo V, Marchisio M, Cama A. Integrative proteomic and functional analyses provide novel insights into the action of the repurposed drug candidate nitroxoline in AsPC-1 cells. Sci Rep 2020; 10:2574. [PMID: 32054977 PMCID: PMC7018951 DOI: 10.1038/s41598-020-59492-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023] Open
Abstract
We recently identified nitroxoline as a repurposed drug candidate in pancreatic cancer (PC) showing a dose-dependent antiproliferative activity in different PC cell lines. This antibiotic is effective in several in vitro and animal cancer models. To date, the mechanisms of nitroxoline anticancer action are largely unknown. Using shotgun proteomics we identified 363 proteins affected by nitroxoline treatment in AsPC-1 pancreatic cancer cells, including 81 consistently deregulated at both 24- and 48-hour treatment. These proteins previously unknown to be affected by nitroxoline were mostly downregulated and interconnected in a single highly-enriched network of protein-protein interactions. Integrative proteomic and functional analyses revealed nitroxoline-induced downregulation of Na/K-ATPase pump and β-catenin, which associated with drastic impairment in cell growth, migration, invasion, increased ROS production and induction of DNA damage response. Remarkably, nitroxoline induced a previously unknown deregulation of molecules with a critical role in cell bioenergetics, which resulted in mitochondrial depolarization. Our study also suggests that deregulation of cytosolic iron homeostasis and of co-translational targeting to membrane contribute to nitroxoline anticancer action. This study broadens our understanding of the mechanisms of nitroxoline action, showing that the drug modulates multiple proteins crucial in cancer biology and previously unknown to be affected by nitroxoline.
Collapse
Affiliation(s)
- Serena Veschi
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Maurizio Ronci
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-Me.T), G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Paola Lanuti
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-Me.T), G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Laura De Lellis
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Rosalba Florio
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Giuseppina Bologna
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-Me.T), G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Luca Scotti
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Erminia Carletti
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-Me.T), G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Federica Brugnoli
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | | | - Valeria Bertagnolo
- Section of Anatomy and Histology, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Marco Marchisio
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-Me.T), G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy.
- Centre on Aging Sciences and Translational Medicine (Ce.S.I-Me.T), G. d'Annunzio University of Chieti-Pescara, Chieti, Italy.
| |
Collapse
|
27
|
Veklich TO, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Labyntseva RD, Shkrabak OA, Tsymbalyuk OV, Rodik RV, Kalchenko VI, Kosterin SO, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv;, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv;, Institute of High Technologies, Taras Shevchenko National University of Kyiv, Ukraine;, Institute of Organic Chemistry, National Academy of Sciences of Ukraine, Kyiv, Institute of Organic Chemistry, National Academy of Sciences of Ukraine, Kyiv, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv;. Inhibition of Na(+),K(+)-ATPase and activation of myosin ATPase by calix[4]arene C-107 cause stimulation of isolated smooth muscle contractile activity. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.01.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
28
|
TRIM32 Promotes the Growth of Gastric Cancer Cells through Enhancing AKT Activity and Glucose Transportation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4027627. [PMID: 32051827 PMCID: PMC6995489 DOI: 10.1155/2020/4027627] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/15/2019] [Accepted: 12/20/2019] [Indexed: 12/21/2022]
Abstract
Tripartite motif protein 32 (TRIM32), an E3 ubiquitin ligase, is a member of the TRIM protein family. However, the underlying function of TRIM32 in gastric cancer (GC) remains unclear. Here, we aimed to explore the function of TRIM32 in GC cells. TRIM32 was induced silencing and overexpression using RNA interference (RNAi) and lentiviral-mediate vector in GC cells, respectively. Moreover, the PI3K/AKT inhibitor LY294002 was used to examine the relationship between TRIM32 and AKT. Quantitative reverse-transcription PCR (qRT-PCR) and western blot were used to determine the mRNA and protein contents. The glucose analog 2-NBDG was used as a fluorescent probe for determining the activity of glucose transport. An annexin V-fluorescein isothiocyanate apoptosis detection kit was used to stain NCI-N87, MKN74, and MKN45 cells. Cell counting kit-8 (CCK-8) assay was used to examine cell proliferation. Our results indicated that TRIM32 was associated with poor overall survival of patients with GC. Moreover, TRIM32 was a proproliferation and antiapoptosis factor and involved in the AKT pathway in GC cells. Furthermore, TRIM32 possibly mediated the metabolism of glycolysis through targeting GLUT1 and HKII in GC cells. Importantly, TRIM32 silencing deeply suppressed the tumorigenicity of GC cells in vivo. Our findings not only enhanced the understanding of the function of TRIM32 but also indicated its potential value as a target in GC treatment.
Collapse
|
29
|
Xia D, Wu J, Xing M, Wang Y, Zhang H, Xia Y, Zhou P, Xu S. Iron overload threatens the growth of osteoblast cells via inhibiting the PI3K/AKT/FOXO3a/DUSP14 signaling pathway. J Cell Physiol 2019; 234:15668-15677. [PMID: 30693516 DOI: 10.1002/jcp.28217] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/16/2019] [Indexed: 01/24/2023]
Abstract
Iron overload is a common stress in the development of cells. Growing evidence has indicated that iron overload is associated with osteoporosis. Therefore, enhancing the understanding of iron overload would benefit the development of novel approaches to the treatment of osteoporosis. The purpose of the present study was to analyze the effect of iron overload on osteoblast cells, via the MC3T3-E1 cell line, and to explore its possible underlying molecular mechanisms. Ferric ammonium citrate (FAC) was utilized to simulate iron overload conditions in vitro. FAC-induced iron overload strongly suppressed proliferation of osteoblast cells and induced apoptosis. Moreover, iron overload strongly suppressed the expression of dual-specificity phosphatase 14 (DUSP14). Additionally, overexpression of DUSP14 protected osteoblast cells from the deleterious effects of iron overload, and this protective effect was mediated by FOXO3a. Additionally, matrine rescued the function of DUSP14 in osteoblast cells. Most importantly, our analysis demonstrated the essential role of the PI3K/AKT/FOXO3a/DUSP14 signaling pathway in the defense against iron overload in osteoblast cells. Overall, our results not only elucidate deleterious effects of iron overload, but also unveil its possible signaling pathway in osteoblast cells.
Collapse
Affiliation(s)
- Demeng Xia
- Department of Emergency, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jianghong Wu
- Department of Emergency, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Canada
| | - Yang Wang
- Department of Emergency, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hongyue Zhang
- Department of Emergency, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yan Xia
- Department of Emergency, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Panyu Zhou
- Department of Emergency, Changhai Hospital, Second Military Medical University, Shanghai, China.,Department of Orthopedics, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Shuogui Xu
- Department of Emergency, Changhai Hospital, Second Military Medical University, Shanghai, China.,Department of Orthopedics, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
30
|
Sun X, Ng TTH, Sham KWY, Zhang L, Chan MTV, Wu WKK, Cheng CHK. Bufalin, a Traditional Chinese Medicine Compound, Prevents Tumor Formation in Two Murine Models of Colorectal Cancer. Cancer Prev Res (Phila) 2019; 12:653-666. [PMID: 31431500 DOI: 10.1158/1940-6207.capr-19-0134] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/24/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
Chemoprevention is cost-effective for colorectal cancer when targeted at intermediate- or high-risk populations. Bufalin is a cardiac glycoside extracted from the traditional Chinese medicine (TCM) "Chan Su," which has been used as an anticancer agent. On the basis of the relative safety of bufalin, we investigated whether bufalin could act as a chemoprophylactic agent to prevent colon tumorigenesis in two murine models, namely colitis-associated colorectal cancer and Apc germline mutation-developed colorectal cancer. Our results revealed that long-term (12-16 weeks) administration of low-dose bufalin (0.5 mg/kg) effectively suppressed tumorigenesis in both colorectal cancer models, accompanied by attenuated epithelial cell proliferation (reduced bromodeoxyuridine incorporation, lower levels of cyclin A, cyclin D1, cyclin E, and cyclin-dependent kinases-2/4, and higher levels of p21 and p27) and promoted apoptosis (increased TUNEL positivity and caspase-3/9 cleavages, reduced levels of Bcl-2, Bcl-xL and survivin, and increased levels of Bax and Bak). Bufalin also suppressed the expression of proinflammatory mediators [reduced levels of cyclooxygenase-2, tumor TNFα, IL1β, IL6, C-X-C motif chemokine ligand (CXCL)-1, CXCL-2, and CXCL-5] in the colitis-associated colorectal cancer model. These effects were associated with the inhibition of oncogenic NF-κB and PI3K/Akt pathways. Our findings unveil a novel chemoprophylactic action of bufalin in colorectal cancer in vivo and provided efficacy data and mechanistic evidence for further clinical evaluation of this TCM compound for colorectal cancer chemoprevention in individuals at risk of colorectal cancer.
Collapse
Affiliation(s)
- Xiao Sun
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Tony T H Ng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Kathy W Y Sham
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Lin Zhang
- State Key Laboratory of Digestive Diseases, Institute of Digestive Diseases and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.,Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong
| | - William K K Wu
- State Key Laboratory of Digestive Diseases, Institute of Digestive Diseases and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong. .,Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong.,CUHK Shenzhen Research Institute, Shenzhen, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong. .,CUHK Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
31
|
Wang M, Tian Z, Zhu Y, Ding J, Li C, Zhou Y, Zhang Y. Sichong formula inhibits the proliferation and migration of human gastric cancer cells. Onco Targets Ther 2019; 12:5741-5750. [PMID: 31410020 PMCID: PMC6643054 DOI: 10.2147/ott.s199605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 06/15/2019] [Indexed: 12/30/2022] Open
Abstract
Purpose Traditional Chinese medicine (TCM) has gained increasing attention for the treatment of multiple chronic diseases, such as cancer. Here we aim to identify the antitumor activity of Sichong formula, a novel TCM, in human gastric cancer cells and investigate the underlying mechanisms. Methods The AGS and MKN45 gastric cancer cells were treated with Sichong formula at different concentrations. The proliferation rates were tested by CCK-8 and colony formation assays. Cell migration and invasion were tested by scratch and transwell assays. Gelatin zymography was used to detect the matrix metalloproteinase 9 (MMP9) activity in cell suspendents. Cell apoptosis was analyzed by Annexin V/PI staining and flow cytometry. The expression of interest proteins was tested by Western blot. Results Cell proliferation analysis indicated that Sichong formula inhibited cell viability of AGS and MKN45 cells in a dose- and time-dependent manner. The IC50 values were 240 μg/mL and 200 μg/mL for AGS and MKN45 cells, respectively. Furthermore, we found that Sichong formula could inhibit the invasion and migration of gastric cancer cells, which might be mediated by the downregulation of MMP9 activity. Flow cytometry results indicated that Sichong formula induced apoptosis in gastric cancer cells through upregulation of Bax/Bcl2 ratio and activation of caspase cascade. The results from Western blot indicated that Sichong formula resulted in cell autophagy and inactivation of AKT signaling pathway. Conclusion Our data suggest that Sichong formula inhibits the proliferation and migration and induces apoptosis in human gastric cancer cells. The inhibitory effect of Sichong formula was, at least partly, mediated by cell autophagy and AKT pathway.
Collapse
Affiliation(s)
- Meng Wang
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, People's Republic of China
| | - Zhaochun Tian
- Chinese Medicine Ophthalmology, Shandong Provincial Qianfoshan Hospital, Jinan 250014, People's Republic of China
| | - Yong Zhu
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, People's Republic of China
| | - Jian Ding
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, People's Republic of China
| | - Chen Li
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, People's Republic of China
| | - Yongkun Zhou
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, People's Republic of China
| | - Yunjie Zhang
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, People's Republic of China
| |
Collapse
|
32
|
An W, Lai H, Zhang Y, Liu M, Lin X, Cao S. Apoptotic Pathway as the Therapeutic Target for Anticancer Traditional Chinese Medicines. Front Pharmacol 2019; 10:758. [PMID: 31354479 PMCID: PMC6639427 DOI: 10.3389/fphar.2019.00758] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. Apoptosis is a process of programmed cell death and it plays a vital role in human development and tissue homeostasis. Mounting evidence indicates that apoptosis is closely related to the survival of cancer and it has emerged as a key target for the discovery and development of novel anticancer drugs. Various studies indicate that targeting the apoptotic signaling pathway by anticancer drugs is an important mechanism in cancer therapy. Therefore, numerous novel anticancer agents have been discovered and developed from traditional Chinese medicines (TCMs) by targeting the cellular apoptotic pathway of cancer cells and shown clinically beneficial effects in cancer therapy. This review aims to provide a comprehensive discussion for the role, pharmacology, related biology, and possible mechanism(s) of a number of important anticancer TCMs and their derivatives mainly targeting the cellular apoptotic pathway. It may have important clinical implications in cancer therapy.
Collapse
Affiliation(s)
- Weixiao An
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Nanchong Central Hospital, Nanchong, China
| | - Honglin Lai
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, China
| | - Yangyang Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Minghua Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
33
|
MicroRNA-32 targeting PTEN enhances M2 macrophage polarization in the glioma microenvironment and further promotes the progression of glioma. Mol Cell Biochem 2019; 460:67-79. [PMID: 31218569 DOI: 10.1007/s11010-019-03571-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/04/2019] [Indexed: 12/25/2022]
Abstract
This study was aimed to explore the molecular mechanism of macrophage polarization and its effect on glioma progression. THP1 cells were cocultured in conditioned medium from U87 human glioblastoma cells to simulate the glioma microenvironment. The expression of miR-32 and PTEN in THP1 cells was detected by real-time PCR. A luciferase reporter assay was conducted to confirm the target relation between miR-32 and PTEN. Western blot assays and ELISA were performed to detect PTEN, M2 macrophage-specific markers, PI3K/AKT signaling proteins, and apoptosis-related proteins. U87 cell proliferation was evaluated by CCK-8 and colony forming assays, and the migration ability of the cells was evaluated by Transwell and wound healing assays. The U87 culture supernatant promoted the M2 phenotype of THP1 cells. miR-32 was upregulated and PTEN was downregulated in THP1 cells with the M2 phenotype in the glioma microenvironment. Luciferase assays confirmed that PTEN expression was suppressed by miR-32 through interaction with the 3'UTR of PTEN. Overexpression of miR-32 suppressed PTEN expression in THP1 cells. Overexpression of miR-32 or downregulation of PTEN promoted the expression of M2 macrophage-specific markers, thereby enhancing M2 macrophage polarization. Additionally, miR-32 inhibited THP1 cell apoptosis via suppressing the PI3K/AKT signaling pathway. Most importantly, the proliferation and migration capacities of U87 cells treated with the THP1 culture supernatant after miR-32 overexpression were enhanced, and these effects could be reversed by cotransfection with pcDNA3.1-PTEN. miR-32 negatively modulates PTEN, thereby promoting M2 macrophage transformation through PI3K/AKT signaling, enhancing glioma proliferation and migration abilities.
Collapse
|
34
|
Wei WL, Hou JJ, Wang X, Yu Y, Li HJ, Li ZW, Feng ZJ, Qu H, Wu WY, Guo DA. Venenum bufonis: An overview of its traditional use, natural product chemistry, pharmacology, pharmacokinetics and toxicology. JOURNAL OF ETHNOPHARMACOLOGY 2019; 237:215-235. [PMID: 30905791 DOI: 10.1016/j.jep.2019.03.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/16/2019] [Accepted: 03/16/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The animal medicine of Venenum Bufonis (VB), a product of the secretions of Bufo gargarizans Cantor or B. melanostictus Schneider, has long been used as a traditional Chinese medicine (TCM) for the treatment of sunstroke and faint, acute filthy disease - abdominal pain or vomiting and diarrhea, etc. AIM OF THE REVIEW: This review is aimed at providing the comprehensive and up-to-date information of VB as regards its ethnopharmacological uses, constituents and their metabolism, pharmacokinetics, pharmacology and toxicology, all of which could be used as fundamental data for future research as well as development of new drugs. MATERIALS AND METHODS The information and data about the studies of VB were collected from scientific journals, material medica, historical documents, library, and electronic databases (PubMed, Google Scholar, Science Direct, Researchgate, Web of Science and CNKI). RESULTS To date, about 142 bufadienolides and 16 indole alkaloids have been isolated from VB in total. The extract and isolated compounds showed a wide range of in vitro and in vivo pharmacologic effects, such as cardiotonic, anti-tumor, antinociceptive, anti-inflammatory, anesthetic and antimicrobial activities. Especially, bufadienolides have been extensively studied due to its powerful anti-tumor activities against various cancer cells. Furthermore, their metabolites and metabolic pathways were concluded in detail, and the main metabolic pathways of bufadienolides were hydroxylation, 3-isomerization, 3-keto, 16-hydrolyzation, 3-O-sulfate and 3-O-glucuronide. CONCLUSIONS Although VB possesses significant anti-tumor effect against various cancer cell lines, the development of new drugs still remains to be a challenge due to its pharmacodynamic effects in vivo, druggability and toxicology. The main problem lies in its side effects in vivo, poor bioavailability, fast metabolism, cardiotoxicity and neurovirulence. Besides, studies on its metabolism and toxicology in vitro and in vivo, as well as clinical trials should be further conducted for the new drug development and the establishment of optimal dosage of consumption of its administration.
Collapse
Affiliation(s)
- Wen-Long Wei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Jin-Jun Hou
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Xia Wang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Yang Yu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Hao-Jv Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Zhen-Wei Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Zi-Jin Feng
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Hua Qu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Wan-Ying Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China.
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China.
| |
Collapse
|
35
|
Hong Y, Rao Y. Current status of nanoscale drug delivery systems for colorectal cancer liver metastasis. Biomed Pharmacother 2019; 114:108764. [DOI: 10.1016/j.biopha.2019.108764] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/25/2019] [Accepted: 03/06/2019] [Indexed: 12/24/2022] Open
|
36
|
Jiang YW, Cheng HY, Kuo CL, Way TD, Lien JC, Chueh FS, Lin YL, Chung JG. Tetrandrine inhibits human brain glioblastoma multiforme GBM 8401 cancer cell migration and invasion in vitro. ENVIRONMENTAL TOXICOLOGY 2019; 34:364-374. [PMID: 30549224 DOI: 10.1002/tox.22691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 11/19/2018] [Accepted: 11/24/2018] [Indexed: 06/09/2023]
Abstract
Tetrandrine (TET) has been reported to induce anti-cancer activity in many human cancer cells and also to inhibit cancer cell migration and invasion. However, there are no reports to show TET inhibits cell migration and invasion in human brain glioblastoma multiforme GBM 8401 cells. In this study, we investigated the anti-metastasis effects of TET on GBM 8401 cells in vitro. Under sub-lethal concentrations (from 1, 5 up to 10 μM), TET significantly inhibited cell mobility, migration and invasion of GBM 8401 cells that were assayed by wound healing and Transwell assays. Gelatin zymography assay showed that TET inhibited MMP-2 activity in GBM 8401 cells. Western blotting results indicated that TET inhibited several key metastasis-related proteins, such as p-EGFR(Tyr1068) , SOS-1, GRB2, Ras, p-AKT(Ser473) and p-AKT(Thr308) , NF-κB-p65, Snail, E-cadherin, N-cadherin, NF-κB, MMP-2 and MMP-9 that were significant reduction at 24 and 48 hours treatment by TET. TET reduced MAPK signaling associated proteins such as p-JNK1/2 and p-c-Jun in GBM 8401 cells. The electrophoretic mobility shift (EMSA) assay was used to investigate NF-κB and DNA binding was reduced by TET in a dose-dependently. Based on these findings, we suggested that TET could be used in anti-metastasis of human brain glioblastoma multiforme GBM 8401 cells in the future.
Collapse
Affiliation(s)
- Yi-Wen Jiang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Hsin-Yu Cheng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Tzong-Der Way
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Jin-Cherng Lien
- School of pharmacy, China Medical University, Taichung, Taiwan
| | - Fu-Shin Chueh
- Department of Food Nutrition and Health Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
37
|
Lan YL, Lou JC, Jiang XW, Wang X, Xing JS, Li S, Zhang B. A research update on the anticancer effects of bufalin and its derivatives. Oncol Lett 2019; 17:3635-3640. [PMID: 30915168 DOI: 10.3892/ol.2019.10062] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022] Open
Abstract
Bufalin (BF) is a cardiotonic steroid that has recently been found to have substantial anticancer activity; however, more efforts should be directed toward clarifying the detailed molecular mechanisms underlying this activity. BF could exert its anticancer effect by inducing apoptosis in various human cancer cells and thus triggering autophagic cancer cell death. The anti-inflammatory activities of BF are potentially important for its anticancer functions. Notably, some promising synthetic BF derivatives, including poly (ethylene glycol)-based polymeric prodrug of BF and BF211, have shown potent anticancer activity. Additionally, clinical trials regarding the use of BF-related agents in patients have supported the positive effect of BF as an anticancer treatment. Currently, large-scale randomized, double-blind, placebo or positive drug parallel controlled studies are required to confirm the anticancer potential of BF in various cancer types in the clinical setting. The present review will evaluate the potential mechanisms mediated by BF in intracellular signaling events in cancer cells and various promising BF derivatives that may have greater anticancer activity, thereby clarifying BF-mediated anticancer effects. The experimental and clinical results reviewed strongly emphasize the importance of this topic in future investigations.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China.,Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jia-Cheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Xue-Wen Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Xun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Jin-Shan Xing
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
38
|
Su EY, Chu YL, Chueh FS, Ma YS, Peng SF, Huang WW, Liao CL, Huang AC, Chung JG. Bufalin Induces Apoptotic Cell Death in Human Nasopharyngeal Carcinoma Cells through Mitochondrial ROS and TRAIL Pathways. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:237-257. [DOI: 10.1142/s0192415x19500125] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
The aim of this study was to investigate the effects of bufalin on human nasopharyngeal carcinoma NPC-TW 076 cells in vitro. Bufalin is a cardiotonic steroid and a key active ingredient of the Chinese medicine ChanSu. The extracts of Chansu are used for various cancer treatments in China. In the present study, bufalin induced cell morphological changes, decreased total cell viability and induced G2/M phase arrest of cell cycle in NPC-TW 076 cells. Results also indicated that bufalin induced chromatin condensation (cell apoptosis) and DNA damage by DAPI staining and comet assay, respectively. The induced apoptotic cell death was further confirmed by annexin-V/PI staining assay. In addition, bufalin also increased ROS and Ca[Formula: see text] production and decreased the levels of [Formula: see text]. Furthermore, the alterations of ROS, ER stress and apoptosis associated protein expressions were investigated by Western blotting. Results demonstrated that bufalin increased the expressions of ROS associated proteins, including SOD (Cu/Zn), SOD2 (Mn) and GST but decreased that of catalase. Bufalin increased ER stress associated proteins (GRP78, IRE-1[Formula: see text], IRE-1[Formula: see text], caspase-4, ATF-6[Formula: see text], Calpain 1, and GADD153). Bufalin increased the pro-apoptotic proteins Bax, and apoptotic associated proteins (cytochrome c, caspase-3, -8 and -9, AIF and Endo G) but reduced anti-apoptotic protein Bcl-2 in NPC-TW 076 cells. Furthermore, bufalin elevated the expressions of TRAIL-pathway associated proteins (TRAIL, DR4, DR5, and FADD). Based on these findings, we suggest bufalin induced apoptotic cell death via caspase-dependent, mitochondria-dependent and TRAIL pathways in human nasopharyngeal carcinoma NPC-TW 076 cells.
Collapse
Affiliation(s)
- En-Yun Su
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Yung-Lin Chu
- Department of Food Science, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Fu-Shin Chueh
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, Taiwan
- Department of Chinese Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Shu-Fen Peng
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wen-Wen Huang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Ching-Lung Liao
- College of Chinese Medicine, School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - An-Cheng Huang
- Department of Nursing, St. Mary’s Junior College of Medicine, Nursing and Management, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
39
|
Qi HY, Qu XJ, Liu J, Hou KZ, Fan YB, Che XF, Liu YP. Bufalin induces protective autophagy by Cbl-b regulating mTOR and ERK signaling pathways in gastric cancer cells. Cell Biol Int 2018; 43:33-43. [PMID: 30468278 DOI: 10.1002/cbin.11076] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/11/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Hai-Yan Qi
- Department of Medical Oncology; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province; The First Hospital of China Medical University; No. 155, North Nanjing Street Heping District Shenyang 110001 China
- Department of the First Medical Oncology; The Fourth Hospital of China Medical University; Shenyang 110032 China
| | - Xiu-Juan Qu
- Department of Medical Oncology; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province; The First Hospital of China Medical University; No. 155, North Nanjing Street Heping District Shenyang 110001 China
| | - Jing Liu
- Department of Medical Oncology; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province; The First Hospital of China Medical University; No. 155, North Nanjing Street Heping District Shenyang 110001 China
| | - Ke-Zuo Hou
- Department of Medical Oncology; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province; The First Hospital of China Medical University; No. 155, North Nanjing Street Heping District Shenyang 110001 China
| | - Yi-Bo Fan
- Department of Medical Oncology; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province; The First Hospital of China Medical University; No. 155, North Nanjing Street Heping District Shenyang 110001 China
| | - Xiao-Fang Che
- Department of Medical Oncology; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province; The First Hospital of China Medical University; No. 155, North Nanjing Street Heping District Shenyang 110001 China
| | - Yun-Peng Liu
- Department of Medical Oncology; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province; The First Hospital of China Medical University; No. 155, North Nanjing Street Heping District Shenyang 110001 China
| |
Collapse
|
40
|
Shih YL, Chou JS, Chen YL, Hsueh SC, Chung HY, Lee MH, Chen CP, Lee MZ, Hou HT, Lu HF, Chen KW, Chung JG. Bufalin Enhances Immune Responses in Leukemic Mice Through Enhancing Phagocytosis of Macrophage In Vivo. In Vivo 2018; 32:1129-1136. [PMID: 30150435 DOI: 10.21873/invivo.11355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIM Bufalin, bufadienolide present in Chan Su, has been shown to induce cancer cell apoptosis in many human cancer cells, including human leukemia cells, but its effects on immune responses are unknown. MATERIALS AND METHODS This study investigated whether bufalin affected immune responses of mice with WEHI-3 cell-generated leukemia in vivo. BALB/c mice were intraperitoneally injected with WEHI-3 cells to develop leukemia and then were treated with oral treatment with bufalin at different doses (0, 0.1, 0.2 and 0.4 mg/kg) for 2 weeks. At the end of treatment, all mice were weighted and blood was collected; liver and spleen tissues were collected for cell marker, phagocytosis, natural killer (NK) cell activity and T- and B-cell proliferation measurements by using flow cytometric assays. RESULTS When compared with the leukemia control group, bufalin increased the body weight, but reduced liver and spleen weights, and reduced CD3, CD16 and Mac-3 cell markers at 0.4 mg/kg treatment and increased CD11b marker at 0.1 and 0.2 mg/kg treatment. Furthermore, bufalin at 0.4 mg/kg increased phagocytosis by macrophages isolated from peripheral blood mononuclear cells and at 0.1 mg/kg by those from the peritoneal cavity. Bufalin (0.2 and 0.4 mg/kg) increased NK cell cytotoxic activity at effector:target ratio of 50:1. Bufalin increased B-cell proliferation at 0.1 and 0.2 mg/kg treatment but only increased T-cell proliferation at 0.1 mg/kg. Bufalin increased glutamate oxaloacetate transaminase level at all dose treatments, increased glutamic pyruvic transaminase level only at 0.1 mg/kg treatment, but reduced the level of lactate dehydrogenase at all dose levels in mice with WEHI-3 cell-induced leukemia in vivo. CONCLUSION Bufalin increased immune responses by enhancing phagocytosis in mice with leukemia mice.
Collapse
Affiliation(s)
- Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, R.O.C.,School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan, R.O.C.,School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan, R.O.C
| | - Jiann-Shang Chou
- Department of Anatomic Pathology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Yung-Liang Chen
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu, Taiwan, R.O.C
| | - Shu-Ching Hsueh
- Division of Hematology and Oncology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C.,Family Medicine and Community Medicine, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Hsueh-Yu Chung
- Jen-Teh Junior College of Medicine, Nursing and Management, Houlong, Taiwan, R.O.C
| | - Mei-Hui Lee
- Department of Genetic Counseling Center, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Chao-Ping Chen
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, R.O.C
| | - Ming-Zhe Lee
- Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Hsin-Tu Hou
- Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Hsu-Feng Lu
- Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Kuo-Wei Chen
- Division of Hematology and Oncology, Cheng Hsin General Hospital, Taipei, Taiwan, R.O.C.
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C. .,Department of Biotechnology, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
41
|
Bao F, Kang X, Xie Q, Wu J. HIF-α/PKM2 and PI3K-AKT pathways involved in the protection by dexmedetomidine against isoflurane or bupivacaine-induced apoptosis in hippocampal neuronal HT22 cells. Exp Ther Med 2018; 17:63-70. [PMID: 30651766 PMCID: PMC6307527 DOI: 10.3892/etm.2018.6956] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022] Open
Abstract
The present study investigated the mechanism underlying the protective effect of dexmedetomidine (Dex) on hippocampal neuronal HT22 cell apoptosis induced by the anesthetics isoflurane and bupivacaine. The cellular morphology was observed using a phase contrast microscope. The effects of anesthetics on cell proliferation were assayed using a Cell Counting Kit-8 (CCK-8). The levels of apoptosis were examined by flow cytometry utilizing Annexin V-fluorescein isothiocyanate/propidium iodide double staining, and the protein expression levels of cleaved caspase-3, phosphorylated phosphoinositide 3′-kinase (p-PI3K), p-protein kinase B (p-AKT), hypoxia inducible factor (HIF-α), pyruvate kinase M2 (PKM2), B-cell lymphoma (Bcl-2)-associated X protein (Bax), Bcl-2 and cytochrome c were detected by western blot analysis. In vitro treatment with anesthetics was identified to decrease cell proliferation (P<0.01), the effect of which was then markedly inhibited by treatment with Dex (P<0.01) or a PI3K/AKT agonist. Exposure to anesthetics induced apoptosis in HT22 cells (75.4%), which was significantly attenuated by co-treatment with Dex (26.2%) or the PI3K/AKT agonist (28.1%). Analysis of the protein expression levels revealed that exposure to anesthetics resulted in the activation of cleaved caspase-3, Bax, cytochrome c, HIF-α and PKM2 and decreased the expression levels of Bcl-2, p-PI3K and p-AKT. However, these changes were inhibited by treatment with Dex or the PI3K/AKT agonist. Dex protected hippocampal neuronal HT22 cells from anesthetic-induced apoptosis through the promotion of the PI3K/AKT pathway and inhibition of the HIF-α/PKM2 axis.
Collapse
Affiliation(s)
- Fangping Bao
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Qing Xie
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Jian Wu
- Department of Anesthesiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
42
|
Chen Q, Liu J. Transferrin and folic acid co-modified bufalin-loaded nanoliposomes: preparation, characterization, and application in anticancer activity. Int J Nanomedicine 2018; 13:6009-6018. [PMID: 30323588 PMCID: PMC6179727 DOI: 10.2147/ijn.s176012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim The aim of this study was to prepare transferrin (Tf) and folic acid (FA) co-modified bufalin (BF) liposomes for lung cancer treatment. Method In this study, (FA+Tf) BF-LPs were prepared using the high-pressure homogenization method. Results The EE% and DL% of prepared LPs were 82.3% and 10.7%, respectively, and the mean diameter was 120.4 nm from three batches. In vitro release showed that the release of BF from (FA+Tf) BF-LPs was slow with burst effects at an early stage. In vitro cytotoxicity assay showed that (FA+Tf) BF-LPs had a superior antiproliferative effect on A549 cells. An in vivo imaging study indicated that (FA+Tf) BF-LPs had obvious targeting characteristics on subcutaneous tumor, with the potential to actively deliver drugs to tumor tissues. In terms of the in vivo antitumor activity, (FA+Tf) BF-LPs treated mice showed a significantly suppressed tumor growth and no systemic toxicity in the body. Conclusion Through this study, it was found that the Tf and FA co-modified BF could be a very promising lung target preparation.
Collapse
Affiliation(s)
- Qiankun Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ji Liu
- Department of Anesthesia, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China,
| |
Collapse
|
43
|
Ma L, Meng Y, Tu C, Cao X, Wang H, Li Y, Man S, Zhou J, Li M, Liu Z, Su Y. A cardiac glycoside HTF-1 isolated from Helleborus thibetanus Franch displays potent in vitro anti-cancer activity via caspase-9, MAPK and PI3K-Akt-mTOR pathways. Eur J Med Chem 2018; 158:743-752. [DOI: 10.1016/j.ejmech.2018.09.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 11/25/2022]
|
44
|
Li Y, Angelova A, Liu J, Garamus VM, Li N, Drechsler M, Gong Y, Zou A. In situ phase transition of microemulsions for parenteral injection yielding lyotropic liquid crystalline carriers of the antitumor drug bufalin. Colloids Surf B Biointerfaces 2018; 173:217-225. [PMID: 30296646 DOI: 10.1016/j.colsurfb.2018.09.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 08/11/2018] [Accepted: 09/10/2018] [Indexed: 10/28/2022]
Abstract
In this work, we used the small angle X-ray scattering (SAXS) method for controlled preparation of in situ forming sustained-release carriers for the antitumor drug bufalin (BUF), which has very poor solubility and a considerable cardiotoxicity in a non-encapsulated state. To that aim, we exploited the pseudo-ternary phase diagram of an oil(O)/surfactant(S)/water(W) system containing medium chain capric/caprylic triglycerides (MCT) and a co-surfactant blend of Macrogol (15)-hydroxystearate (Solutol HS 15) and sorbitan monooleate (Span 80). Two compositions with different oil contents (sample B and C) were selected from the microemulsion region of the phase diagram in order to study the effect of the aqueous environment on their structural behavior. A phase transition from a microemulsion (ME) to a liquid crystalline phase (LC) was established by SAXS upon progressive dilution. The drug bufalin (BUF) was encapsulated in the microemulsions with low viscosity, whereas the release of the drug occurred from the in situ generated lamellar liquid crystalline structures. The formulations were characterized by SAXS, dynamic light scattering (DLS), cryogenic transmission electron microscopy (Cryo-TEM), rheology, drug loading and encapsulation efficiency, and in vitro release profiles. A correlation was suggested between the structures of the in situ phase-transition formed LCME formulations, the differences in their viscosities and drug release profiles. The performed cytotoxicity, cell apoptosis and pharmacokinetic experiments showed an enhanced bioavailability of BUF after encapsulation. These results suggest potential clinical applications for the obtained safe in situ phase-transition sustained-release formulations of BUF.
Collapse
Affiliation(s)
- Yawen Li
- Shanghai Key Laboratory of Functional Materials Chemistry, State Key Laboratory of Bioreactor Engineering and Institute of Applied Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Angelina Angelova
- Institut Galien Paris-Sud, CNRS UMR 8612, Univ. Paris-Sud, Université Paris-Saclay, LabEx LERMIT, F-92296, Châtenay-Malabry cedex, France
| | - Jianwen Liu
- Shanghai Key Laboratory of Functional Materials Chemistry, State Key Laboratory of Bioreactor Engineering and Institute of Applied Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Vasil M Garamus
- Helmholtz-Zentrum Geesthacht, Centre for Materials and Coastal Research, D-21502, Geesthacht, Germany
| | - Na Li
- National Center for Protein Science Shanghai and Shanghai Institute of Biochemistry and Cell Biology, Shanghai, 200237, PR China
| | - Markus Drechsler
- Keylab "Electron and Optical Microscopy", Bavarian Polymerinstitute (BPI), University of Bayreuth, D-95440, Bayreuth, Germany
| | - Yabin Gong
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 200437, PR China
| | - Aihua Zou
- Shanghai Key Laboratory of Functional Materials Chemistry, State Key Laboratory of Bioreactor Engineering and Institute of Applied Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, PR China.
| |
Collapse
|
45
|
Ma J, Ding Y. Geniposide suppresses growth, migration and invasion of MKN45 cells by down-regulation of lncRNA HULC. Exp Mol Pathol 2018; 105:252-259. [PMID: 30176223 DOI: 10.1016/j.yexmp.2018.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 08/23/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022]
Abstract
Gastric cancer (GC) is a serious disease with high incidence rate and high mortality. Geniposide (GEN) exhibits multiple biological properties including anti-tumor function. However, effect of GEN on GC is not well studied. Hence, the effects of GEN on GC were investigated in our study. HULC expression in GC tissue and GC cell lines (MKN45, SGC-7901, MKN28, AGS) was detected by qRT-PCR. Cell viability, colony formation, migration, invasion and cell apoptosis were examined by CCK-8 assay, survival fraction assay, modified two-chamber migration assay, Millicell Hanging Cell Culture and flow cytometry analysis, respectively. The expression of matrix metalloproteinase (MMP)-2/9 and vimentin was detected by qRT-PCR and western blot, respectively. The expression of PI3K/AKT and JNK was measured by western blot. The expression of HULC was up-regulated both in GC tissue and cell lines (P < .05, P < .01 or P < .001). GEN negatively regulated the expression of HULC in MKN45 cells (P < .05 or P < .01). GEN decreased cell viability (P < .05), colony formation (P < .01), migration (P < .05) and invasion (P < .05) while HULC overexpression led to the opposite results in GEN-treated cells. The expression of phosphatidylinositol 3' -kinase (PI3K)/ protein kinase B (AKT) and c-Jun N-terminal kinase (JNK) was down-regulated by GEN (all P < .05) while reversed by HULC overexpression. HULC was up-regulated in GC. GEN inhibited MNK45 cell viability, colony formation, migration and invasion while induced cell apoptosis by down-regulation of HULC in MKN45 cells. GEN inactivated PI3K/AKT and JNK signal pathways through down-regulation of HULC.
Collapse
Affiliation(s)
- Ji Ma
- Department of Endoscopy, Huaihe Hospital of Henan University, Kaifeng 475000, Henan, China
| | - Yong Ding
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
46
|
Jin X, Jiang R, Xiang Y, Fan Z, Wu Z, Yang B, Yang L, Wei S, Yang Y. Overexpression of retinoblastoma‑binding protein 4 contributes to the radiosensitivity of AGS gastric cancer cells via phosphoinositide3‑kinase/protein kinase B pathway suppression. Mol Med Rep 2018; 18:1571-1581. [PMID: 29901205 PMCID: PMC6072197 DOI: 10.3892/mmr.2018.9153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/04/2018] [Indexed: 12/16/2022] Open
Abstract
In the present study, the effects and underlying mechanism of RbAp48 on the radiosensitivity of AGS gastric cancer cells was investigated. Cell proliferation was determined with an MTT assay. Flow cytometry was performed to evaluate the cell cycle and apoptosis. Reverse transcription-quantitative polymerase chain reaction and western blot analysis were performed to detect mRNA and protein expression, respectively, including RbAp48, phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt). The results revealed that radiation enhanced the expression level of RbAp48 in AGS cells, and that RbAp48 combined with radiation reduced AGS cell proliferation. In addition, RbAp48 combined with radiation resulted in G2 phase arrest and induced apoptosis via regulation of the PI3K/Akt pathway. In conclusion, it was demonstrated that overexpression of RbAp48 enhanced the radiosensitivity of AGS gastric cancer cells via suppression of PI3K/Akt pathway activity, suggesting that RbAp48 may hold potential as a gene therapeutic strategy in the future, aiding in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Xiaoxi Jin
- Laboratory Department, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Rui Jiang
- Department of Hematology, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Yongsheng Xiang
- Department of Hematology, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Zhen Fan
- Department of Hematology, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Zhiwei Wu
- Department of Hematology, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Bo Yang
- Department of Hematology, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Lujun Yang
- Department of Hematology, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Shanshan Wei
- Department of Hematology, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Yan Yang
- Laboratory Department, Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
47
|
Inhibitor of growth 3 induces cell death by regulating cell proliferation, apoptosis and cell cycle arrest by blocking the PI3K/AKT pathway. Cancer Gene Ther 2018; 25:240-247. [DOI: 10.1038/s41417-018-0023-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/18/2018] [Accepted: 03/24/2018] [Indexed: 12/14/2022]
|
48
|
He Z, Dong W, Li Q, Qin C, Li Y. Sauchinone prevents TGF-β-induced EMT and metastasis in gastric cancer cells. Biomed Pharmacother 2018; 101:355-361. [DOI: 10.1016/j.biopha.2018.02.121] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/12/2018] [Accepted: 02/23/2018] [Indexed: 12/17/2022] Open
|
49
|
Nobiletin (NOB) suppresses autophagic degradation via over-expressing AKT pathway and enhances apoptosis in multidrug-resistant SKOV3/TAX ovarian cancer cells. Biomed Pharmacother 2018; 103:29-37. [PMID: 29635125 DOI: 10.1016/j.biopha.2018.03.126] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/21/2022] Open
Abstract
Chemotherapy could be used as an effective therapeutic treatment for ovarian cancer and subsequent peritoneal metastasis. However, the occurrence of drug resistance reduced the treatment effect originated from cancer chemotherapy. Accumulating evidences indicated the significant role of autophagy in tumor cell resistance to chemotherapy. Thus, inhibition of autophagy using natural compounds could be a promising candidate to overcome multidrug resistance in human ovarian cancer cells. Nobiletin (NOB), a polymethoxyflavonoid found in citrus fruits such as Citrus depressa and Citrus reticulate, exhibits a number of bioactivities. In the present study, NOB selectively suppressed the growth and proliferation of human SKOV3/TAX cells, inducing G0/G1 phase arrest and reducing G2/M phase, along with the increase of p53 and p21. In addition, NOB induced significant apoptosis in SKOV3/TAX cells through the intrinsic apoptosis pathway, as evidenced by the up-regulation of cleaved Caspase-9/-3 and PARP. Further, NOB impaired the autophagic degradation in SKOV3/TAX cells, resulting in autophagic flux inhibition. Moreover, the impaired autophagic flux enhanced NOB-induced apoptosis in SKOV3/TAX cells. Importantly, AKT signaling was activated by NOB, which was involved in autophagic degradation and apoptotic cell death. In conclusion, the findings here supplied the illustration that NOB could overcome multidrug resistance in human ovarian cancer cells through AKT-regulated suppression of autophagic degradation.
Collapse
|
50
|
Ding Y, Liu W, Wang X, Zhang L, Zhao M, Deng H, Liu Y. Bufalin induces apoptosis in human esophageal carcinoma ECA109 cells by inhibiting the activation of the mTOR/p70S6K pathway. Oncol Lett 2018; 15:9339-9346. [PMID: 29805658 DOI: 10.3892/ol.2018.8526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 03/07/2018] [Indexed: 01/30/2023] Open
Abstract
The present study examined whether bufalin could induce human esophageal carcinoma ECA109 cells apoptosis via inhibiting the activation of mechanistic target of rapamycin (mTOR)/p70 S6 kinase (p70S6K) pathway is discussed in this article. The present study used the esophageal squamous cell carcinoma ECA109 cell line to assess the apoptosis-inducing effects of bufalin via inhibition of the mTOR/p70S6K pathways. A plasmid containing the wild-type mTOR gene (wtmTOR) was transfected into ECA109 cells. The levels of p70S6K, phosphorylated (p)-p70S6K, cellular inhibitor of apoptosis-1 (cIAP-1) and Bcl-2-associated death promoter (BAD) in ECA109 cells were examined by western blot analysis, and apoptosis was detected by flow cytometry analysis and Giemsa staining. The results revealed that the expression of p-p70S6K was increased as the time progressed (at 0, 12 and 24 h), and then decreased at 30, 36, 42 and 48 h after transfection. The expression of cIAP-1 was significantly decreased as time progressed following the addition of bufalin, whereas that of BAD was increased. The levels of p-p70S6K and cIAP-1 were significantly higher in the wtmTOR-transfected group than in the control and empty vector-transfected groups, and then reduced following addition of bufalin; however, BAD expression was significantly lower in the wtmTOR-transfected group. The results of flow cytometry revealed the cell cycle of ECA109 was arrested at G2/M phase and the apoptotic rate was significantly lower in the wtmTOR-transfected group than in the control and empty vector-transfected groups, and then increased following addition of bufalin. In conclusion, the findings of the present study demonstrated that bufalin induced apoptosis in esophageal carcinoma cells via the inhibition of the mTOR/p70S6K pathway and indicated that treatment with bufalin could be combined with chemotherapy to overcome the resistance of esophageal carcinoma cells to chemotherapeutic-induced apoptosis.
Collapse
Affiliation(s)
- Yan Ding
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Wei Liu
- Department of Surgery, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaoling Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Lingling Zhang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Meng Zhao
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Huiyan Deng
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|