1
|
Yang S, Lan J, Li Z, Li M, Wu Y, Sun L, Zhang T, Ding Y. Bufonis venenum extract loaded novel cholesterol-free liposome for the treatment of hepatocellular carcinoma. Front Pharmacol 2024; 15:1486742. [PMID: 39654615 PMCID: PMC11625546 DOI: 10.3389/fphar.2024.1486742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Background This study aims to improve the solubility and the toxicity of Bufonis venenum, and finally enhance the therapeutic outcomes of hepatocellular carcinoma (HCC). Methods The cholesterol-free liposomes simultaneously encapsulate bufadienolides and indolealkylamines (Non-Cholesterol-Bufonis Venenum Extract-Liposome, Non-Chol-BVE-LP) was prepared by the thin-film evaporation technique. In vitro, the cytotoxicity, cell apoptosis study, cellular uptake and hemolysis studies were evaluated in HepG2 cells. In vivo, the biodistribution and anti-tumor activity studies were conducted in BALB/C mice with HepG2 cells. Results The liposomes showed good size distribution, encapsulation efficiency drug loading capacity and slower drug release. Non-Chol-BVE-LP had higher cytotoxicity on HepG2 cells and induced more apoptosis on HepG2 Cells compared with BVE. In addition, the liposomes could accumulate in tumor by passive targeting, thus facilitating the anti-tumor effects. In vivo, Non-Chol-BVE-LP showed equivalent anti-tumor efficacy to the first-line anti-HCC drug sorafenib. Conclusion The study provided new ideas for the development and clinical application of Bufonis venenum related formulation and offered new drug for the treatment of HCC.
Collapse
Affiliation(s)
- Siqi Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ya Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liyan Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- National Innovation Platform for medical industry-education integration, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Kocjan J, Rydel M, Adamek M. Hepatocellular Carcinoma (HCC) Metastasis to the Diaphragm Muscle: A Systematic Review and Meta-Analysis of Case Reports. Cancers (Basel) 2024; 16:3076. [PMID: 39272934 PMCID: PMC11394088 DOI: 10.3390/cancers16173076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
The purpose of this study was to conduct a systematic review and meta-analysis of case reports presenting HCC spread to the diaphragm muscle and to determine possible risk factors for this condition. An extensive literature search was performed using the following electronic databases: MEDLINE, CINAHL, ScienceDirect, Google Scholar, and DOAJ. A total of 18 articles describing 27 hepatocellular carcinoma patients were included in this review. The presence of HCC cells in the superior liver segment is strongly associated with metastases to the diaphragm. Among the two types of diaphragm involvement by HCC cells, diaphragm infiltration occurs much more frequently than diaphragm adhesion. However, an HCC nodule in the 8th liver segment and a higher number of liver segments involved by HCC cells predispose patients to diaphragm adhesion. Hepatitis B is a risk factor for diaphragm metastases in recurrent HCC. The tumor diameter is not associated with HCC spread to the diaphragm muscle. We did not find specific symptoms reported by patients that could indicate HCC metastasis to the diaphragm muscle. The presence of hepatitis B and the localization of HCC cells in superior liver segments, especially in the 8th liver segment, should be take into consideration in the diagnostic process.
Collapse
Affiliation(s)
- Janusz Kocjan
- Diaphragm Concept Academy, Private Clinic Centre Specializing in Treating of Diaphragm Disorders, 32-300 Olkusz, Poland
| | - Mateusz Rydel
- Department of Thoracic Surgery, Faculty of Medicine with Dentistry Division, Medical University of Silesia, 40-055 Katowice, Poland
| | - Mariusz Adamek
- Department of Thoracic Surgery, Faculty of Medicine with Dentistry Division, Medical University of Silesia, 40-055 Katowice, Poland
- Faculty of Health Sciences with Institute of Maritime and Tropical Medicine, Department of Radiology, Medical University of Gdansk, 80-210 Gdansk, Poland
| |
Collapse
|
3
|
Cui Y, Lan L, Lv J, Zhao B, Kong J, Lai Y. Chalcomoracin promotes apoptosis and endoplasmic reticulum stress in hepatocellular carcinoma cells. J Antibiot (Tokyo) 2024; 77:428-435. [PMID: 38724630 DOI: 10.1038/s41429-024-00732-4] [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: 09/21/2023] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 06/28/2024]
Abstract
Chalcomoracin (CMR), a Diels-Alder adduct obtained from mulberry leaves, demonstrated wide-spectrum anti-cancer activity. Herein, we aimed to explore the function of CMR and how it works in hepatocellular carcinoma (HCC). Human HCC cell lines Hep3B and SNU-387 were cultured and treated with various concentrations of CMR (1.5, 3, and 6 µM). Subsequently, the effects of CMR on cell viability, colony formation, apoptosis, migration, and invasion abilities were studied in vitro. Furthermore, the levels of endoplasmic reticulum (ER) stress-related proteins and mitogen-activated protein kinase (MAPK) pathway-related proteins in cells under CMR exposure were detected using western blot. Experiments in vivo were conducted to examine the effects of CMR on tumor growth in HCC. CMR administration inhibited the viability and clonogenic, migration, and invasion abilities, as well as promoted cell apoptosis and ER stress in Hep3B and SNU-387 cells. In addition, CMR treatment reduced the phosphorylation levels of ERK, P38, and JNK in the MAPK pathway. Moreover, an in vivo study showed that CMR administration could inhibit tumorigenesis and MAPK pathway activity in HCC. Our data indicate that CMR has the potential to inhibit the development of HCC, potentially through the inhibition of the MAPK pathway. These findings suggest that CMR may have promising applications as an anticancer agent in future therapeutics for HCC.
Collapse
Affiliation(s)
- Yongliang Cui
- Department of Hepatobiliary Pancreatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China
| | - Liqin Lan
- Department of Intensive Care Unit, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China
| | - Jiahui Lv
- Department of Hepatobiliary Pancreatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China
| | - Bixing Zhao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China
| | - Jinfeng Kong
- Department of Liver Disease, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China.
| | - Yongping Lai
- Department of Hepatobiliary Pancreatic Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, 350028, China.
| |
Collapse
|
4
|
Li X, Zhou M, Chen W, Sun J, Zhao Y, Wang G, Wang B, Pan Y, Zhang J, Xu J. Integrating network pharmacology, bioinformatics, and experimental validation to unveil the molecular targets and mechanisms of galangin for treating hepatocellular carcinoma. BMC Complement Med Ther 2024; 24:208. [PMID: 38816744 PMCID: PMC11137903 DOI: 10.1186/s12906-024-04518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/23/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Galangin, a flavonoid compound, is derived from Alpinia officinarum Hance. Previous studies have shown that galangin can inhibit the proliferation of hepatocellular carcinoma (HCC), but its mechanism is still unclear. This study aims to investigate the potential targets and molecular mechanisms of galangin on HCC through network pharmacology, bioinformatics, molecular docking, and experimental in vitro validation. METHODS In this study, network pharmacology was used to investigate the targets and mechanisms of galangin in the treatment of HCC. AutoDockTools software was used to simulate and calculate the binding of galangin to its core targets. GO and KEGG enrichment analyses were conducted in the DAVID database to explore the main biological functions and signaling pathways impacted by galangin intervention. In addition, bioinformatics was applied to examine the correlation between the differential expressions of the anti-HCC core targets of galangin and the survival of patients with HCC. Finally, the findings obtained from network pharmacology and bioinformatics were verified in cell experiments. RESULTS A total of 67 overlapping target genes of galangin and HCC were identified. Through the analysis of the protein-protein interaction (PPI) network, 10 hub genes with the highest degree of freedom were identified, including SRC, ESR1, MMP9, CDK4, CCNB1, MMP2, CDK2, CDK1, CHK1, and PLK1. These genes were found to be closely related to the degradation of the extracellular matrix, signal transduction, and the cell cycle. GO and KEGG enrichment analyses revealed that galangin exerts an anti-HCC role by affecting various signaling pathways, including the cell cycle, pathways in cancer, and the PI3K-Akt signaling pathway. The results of molecular docking indicated a significant interaction between galangin and CCNB1, CDK4, CDK1, and PLK1. Bioinformatics analysis revealed that CCNB1, CDK4, CDK1, and PLK1 were upregulated in the liver of patients with HCC at both the mRNA and protein levels. Flow cytometry analysis showed that galangin induced G0/G1 phase arrest and cell apoptosis in HepG2 and Huh7 cells. Additionally, galangin suppressed the expression of key proteins and mRNAs involved in the cell cycle pathway. CONCLUSIONS These results suggest that galangin inhibits the growth of HCC cells by arresting the cell cycle at the G0/G1 phase.
Collapse
Affiliation(s)
- Xiaoliang Li
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education & International Joint Research Center of Human-Machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province & Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs & Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Haikou, 571199, Hainan Province, China
| | - Mingyan Zhou
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China
| | - Weijia Chen
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China
| | - Jiangbo Sun
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China
| | - Yihang Zhao
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China
| | - Gaoan Wang
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China
| | - Bingshu Wang
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China
| | - Yipeng Pan
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China.
| | - Junqing Zhang
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China.
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education & International Joint Research Center of Human-Machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province & Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs & Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, No. 3 Xueyuan Road, Haikou, 571199, Hainan Province, China.
| | - Jian Xu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, No. 368 Yehai Road, Haikou, Hainan Province, 570311, China.
| |
Collapse
|
5
|
Deng ZT, Liang SF, Huang GK, Wang YQ, Tu XY, Zhang YN, Li S, Liu T, Cheng BB. Autophagy plays a pro-apoptotic role in arsenic trioxide-induced cell death of liver cancer. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:295-302. [PMID: 38599914 DOI: 10.1016/j.joim.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/18/2023] [Indexed: 04/12/2024]
Abstract
OBJECTIVE The effects of arsenic trioxide (As2O3) on hepatocellular carcinoma have been documented widely. Autophagy plays dual roles in the survival and death of cancer cells. Therefore, we investigated the exact role of autophagy in As2O3-induced apoptosis in liver cancer cells. METHODS The viability of hepatoma cells was determined using the MTT assay with or without fetal bovine serum. The rate of apoptosis in liver cancer cells treated with As2O3 was evaluated using flow cytometry, Hoechst 33258 staining, and TUNEL assays. The rate of autophagy among liver cancer cells treated with As2O3 was detected using immunofluorescence, Western blot assay and transmission electron microscopy. RESULTS Upon treatment with As2O3, the viability of HepG2 and SMMC-7721 cells was decreased in a time- and dose-dependent manner. The apoptosis rates of both liver cancer cell lines increased with the concentration of As2O3, as shown by flow cytometry. Apoptosis in liver cancer cells treated with As2O3 was also shown by the activation of the caspase cascade and the regulation of Bcl-2/Bax expression. Furthermore, As2O3 treatment induced autophagy in liver cancer cells; this finding was supported by Western blot, immunofluorescence of LC3-II and beclin 1, and transmission electron microscopy. In liver cancer cells, As2O3 inhibited the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signal pathway that plays a vital role in both apoptosis and autophagy. The PI3K activator SC-79 partially reversed As2O3-induced autophagy and apoptosis. Furthermore, inhibiting autophagy with 3-methyladenine partially reversed the negative effects of As2O3 on cell viability. Serum starvation increased autophagy and amplified the effect of As2O3 on cell death. CONCLUSION As2O3 induces apoptosis and autophagy in liver cancer cells. Autophagy induced by As2O3 may have a proapoptotic effect that helps to reduce the viability of liver cancer cells. This study provides novel insights into the effects of As2O3 against liver cancer. Please cite this article as: Deng ZT, Liang SF, Huang GK, Wang YQ, Tu XY, Zhang YN, Li S, Liu T, Cheng BB. Autophagy plays a pro-apoptotic role in arsenic trioxide-induced cell death of liver cancer. J Integr Med. 2024; 22(3): 295-302.
Collapse
Affiliation(s)
- Zheng-Ting Deng
- Department of Respiration, Taizhou Affiliated Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China; Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Department of Febrile Diseases, School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Shu-Fang Liang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Guo-Kai Huang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Yu-Qian Wang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Xiao-Yu Tu
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Ya-Ni Zhang
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Tao Liu
- Department of Febrile Diseases, School of Traditional Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Bin-Bin Cheng
- Oncology Department of Traditional Chinese Medicine, the First Affiliated Hospital of Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
6
|
Wu M, Sun T, Xing L. Circ_0004913 Inhibits Cell Growth, Metastasis, and Glycolysis by Absorbing miR-184 to Regulate HAMP in Hepatocellular Carcinoma. Cancer Biother Radiopharm 2023; 38:708-719. [PMID: 33021399 DOI: 10.1089/cbr.2020.3779] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Circular RNA (circRNA) can regulate the progression of hepatocellular carcinoma (HCC). However, the role and potential mechanism of circ_0004913 in HCC are not explored. Methods: Circ_0004913 was identified from two GSE datasets (GSE94508 and GSE97322) as a differentially expressed circRNA between HCC and normal tissues. Levels of circ_0004913, microRNA-184 (miR-184), and hepcidin (HAMP) were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell proliferation, migration, and invasion were estimated by methyl thiazolyl tetrazolium, colony formation, and Transwell assays, respectively. Levels of all proteins were examined by Western blot. Glucose consumption and lactate and ATP production were analyzed by the glucose, lactate, and ATP assay kits. Dual-luciferase reporter, RNA immunoprecipitation (RIP), and RNA pull-down assays were performed to verify the interactions among miR-184 and circ_0004913 or HAMP. The mice xenograft models were established to assess the effect of circ_0004913 on tumor growth in vivo. Results: Circ_0004913 was downregulated in HCC, and its expression impeded cell proliferation, migration, and invasion, EMT, and glycolysis in HCC cells. miR-184 was identified as a target miRNA of circ_0004913, and their expression levels were negatively correlated. miR-184 overexpression could reverse the inhibitory effect of circ_0004913 on HCC cell progression. Moreover, as a target gene of miR-184, HAMP expression was positively correlated with circ_0004913 expression in HCC tissues, and repression of miR-184 could inhibit the progression of HCC cells by increasing HAMP expression. Circ_0004913 could inhibit JAK2/STAT3/AKT signaling pathway and tumor growth in vivo by regulating the miR-184/HAMP axis. Conclusion: Circ_0004913 inhibited the tumorigenesis of HCC by sponging miR-184 to regulate HAMP expression in vitro and in vivo.
Collapse
Affiliation(s)
- Mingyuan Wu
- Department of Gastroenterology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tanlezi Sun
- Basic Medical College, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lianjun Xing
- Department of Gastroenterology, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Luo Y, Xiang J, Tang S, Huang S, Zhou Y, Shen H. Ursolic acid induces apoptosis and pyroptosis in Reh cells by upregulating of the JNK signalling pathway based on network pharmacology and experimental validation. Heliyon 2023; 9:e23079. [PMID: 38144346 PMCID: PMC10746475 DOI: 10.1016/j.heliyon.2023.e23079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Objective To explore the mechanism of ursolic acid (UA) against acute B lymphoblastic leukaemia (B-ALL) based on network pharmacological analysis, molecular docking and experimental verification. Methods The core targets, functional processes, and biological pathways of UA in B-ALL were predicted by network pharmacology and molecular docking. The efficacy and mechanism of UA against B-ALL were verified through in vitro experiments such as cell viability assays, CCK-8 assays, LDH assays, AO/EB staining, flow cytometry, and Western blot assays. Results Network pharmacology analysis of the core targets indicated that the effects of UA on B-ALL were related to programmed cell death (apoptosis and pyroptosis). Molecular docking results showed that FOS, CASP8, MAPK8, IL-1β and JUN were the key targets of UA against B-ALL. The MTS assay showed that UA decreased the viability of Reh cells in a concentration- and time-dependent manner. Cellular and Western blot experiments found that UA induced Reh cell apoptosis and pyroptosis by upregulating the JNK signalling pathway. Conclusions Our findings demonstrated that UA could induce Reh cell apoptosis and pyroptosis by activating the JNK signalling pathway to exert anti-B-ALL effects. This indicates that UA may become a potential drug for the effective treatment of B-ALL.
Collapse
Affiliation(s)
- Ying Luo
- Institute of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Critical Human Diseases Prevention of the Education Department of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Jing Xiang
- Institute of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Critical Human Diseases Prevention of the Education Department of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Shuangyang Tang
- Institute of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Critical Human Diseases Prevention of the Education Department of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Shiting Huang
- Institute of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Critical Human Diseases Prevention of the Education Department of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Yishan Zhou
- Institute of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Critical Human Diseases Prevention of the Education Department of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Haiyan Shen
- Institute of Biochemistry and Molecular Biology, The Key Laboratory of Environment and Critical Human Diseases Prevention of the Education Department of Hunan Province, Hengyang Medical College, University of South China, Hengyang 421001, China
| |
Collapse
|
8
|
Zhang Y, Liu X, Dang W, Liu L. Thymoquinone inhibits lung cancer stem cell properties via triggering YAP degradation. Carcinogenesis 2023; 44:426-435. [PMID: 37105709 DOI: 10.1093/carcin/bgad026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 03/26/2023] [Accepted: 04/27/2023] [Indexed: 04/29/2023] Open
Abstract
Due to the characteristics of high recurrence and metastasis, it is still difficult to cure lung cancer. Cancer stem cells (CSCs) are a group of tumor cells with self-renewal ability and differentiation potential, which are responsible for lung cancer recurrence. Therefore, targeting CSCs may provide a new strategy for lung cancer treatment. Thymoquinone (TQ), the main active ingredient isolated from black seed oil, has shown significant anti-cancer effects in various cancers. However, the effect of TQ on lung cancer stem cells (LCSCs) has never been clarified. In the present study, we successfully separated and enriched lung cancer tumorsphere cells. Our data showed that TQ significantly inhibited the stem-like properties of LCSCs. In addition, we found TQ promoted Yes-associated protein (YAP) phosphorylation and ubiquitination, and the inhibitory effects of TQ on LCSCs could be enhanced by silencing YAP. Taken together, these results suggest that TQ, functions by targeting YAP, may be a potential therapeutic agent against lung cancer.
Collapse
Affiliation(s)
- Yujiao Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xizhi Liu
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wenhui Dang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, Xi'an Aerospace General Hospital, Xi'an, China
| |
Collapse
|
9
|
Wang CD, Ma BQ, Yi BS. Resveratrol reverses drug resistance of esophageal cancer cell line Eca109/DDP via regulating ferroptosis. Shijie Huaren Xiaohua Zazhi 2023; 31:477-484. [DOI: 10.11569/wcjd.v31.i12.477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/28/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Resveratrol not only has anti-tumor effects, but also can enhance the chemosensitivity of tumor cells to a variety of chemotherapeutic agents. However, its effect on cisplatin sensitivity of drug-resistant esophageal cancer cells remains unclear.
AIM To investigate whether resveratrol reverses the drug resistance of esophageal cancer Eca109/DDP cells and to explore the potential mechanism involved from the perspective of ferroptosis.
METHODS The optimal treatment time and concentration of resveratrol and cisplatin were determined by MTT assay. Cell proliferation and the intracellular levels of malon-dialdehyde (MDA), glutathione (GSH), reactive oxygen species (ROS), and ferrous iron were detected. The protein expression of the ferroptosis-related molecules acyl coenzyme A synthetase long chain family member 4 (ACSL4), ferritin heavy chain (FTH), glutathione peroxidase 4 (GPX4), and tumor protein 53 (P53) was detected by Western blot assay.
RESULTS MTT assay showed that compared with cisplatin alone, resveratrol combined with cisplatin significantly inhibited the growth of Eca109/DDP cells (P < 0.05). The combination of resveratrol and cisplatin not only reduced the number of colonies formed (P < 0.05), but also increased the levels of ferrous iron, MDA, and ROS (P < 0.05) and decreased the level of GSH (P < 0.05) in Eca109/DDP cells. The inhibitory effect of resveratrol on Eca109/DDP cell proliferation was partially reversed by ferrostatin-1 (Fer-1) and deferoxamine (DFO) (P < 0.05). Western blot analysis showed that compared with other groups, the protein expression of FTH and GPX4 in the resveratrol and cisplatin combination group was significantly decreased (P < 0.05), and the protein expression of ACSL4 and P53 was significantly increased (P < 0.05).
CONCLUSION Resveratrol can inhibit cell proliferation and reverse cisplatin resistance by regulating ferroptosis in Eca109/DDP cells.
Collapse
Affiliation(s)
- Chen-Deng Wang
- Department of Gastrointestinal, Hernia, Bariatric metabolic, and Trauma Surgery, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Bai-Qiang Ma
- Department of Gastrointestinal, Hernia, Bariatric metabolic, and Trauma Surgery, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Bi-Shun Yi
- Department of Gastrointestinal, Hernia, Bariatric metabolic, and Trauma Surgery, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
10
|
Wang G, Zhou Y, Yi B, Long Y, Ma B, Zhang Y. Comprehensive analysis of the prognostic value and biological function of TDG in hepatocellular carcinoma. Cell Cycle 2023; 22:1478-1495. [PMID: 37224078 PMCID: PMC10281473 DOI: 10.1080/15384101.2023.2216501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/30/2023] [Accepted: 04/27/2023] [Indexed: 05/26/2023] Open
Abstract
Epigenetics plays an important role in the malignant progression of tumors, in which DNA methylation can alter genetic performance without altering the DNA sequence. As a key regulator demethylation, thymine-DNA glycosylase (TDG) has been reported to participate in malignant progression of multiple tumors. In this study, we demonstrate that TDG is highly expressed in hepatocellular carcinoma (HCC) and its high expression is closely related to the poor prognosis of patients. Decreasing TDG expression can significantly inhibit the malignant biological behavior of HCC cells. ABL proto-oncogene 1(ABL1) was identified as a downstream gene regulated by TDG demethylation. In addition, TDG can affect the Hippo signaling pathway through ABL1 to regulate HCC cell proliferation, apoptosis, invasion and migration. Overall, our study demonstrated that TDG reduces DNA methylation of ABL1, increases ABL1 protein expression, and affects the Hippo signaling pathway to regulate the malignant progression of HCC.
Collapse
Affiliation(s)
- Guoliang Wang
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yinwen Zhou
- Department of Surgery, Zunyi Medical University, Zunyi, Guizhou, China
| | - Bin Yi
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yanli Long
- Department of Pathology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Bo Ma
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yi Zhang
- Department of Hepatobiliary Surgery, Department of Organ Transplantation, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
11
|
Kuok C, Wang Q, Fong P, Qin Y, Meng L. Inhibitory Effect of Hernandezine on the Proliferation of Hepatocellular Carcinoma. Biol Pharm Bull 2023; 46:245-256. [PMID: 36724952 DOI: 10.1248/bpb.b22-00612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Hepatocellular carcinoma (HCC) causes 830000 deaths every year and is becoming the third malignant tumor worldwide. One of the primary reasons is the lack of effective drugs. Hernandezine (HER), a bisbenzylisoquinoline alkaloid of Thalictrum simplex, has been confirmed to have antitumor activity. But there are few reports about its effect on HCC and the underlying mechanisms still remain unclear. Therefore, the antitumor effects and mechanisms of HER on HCC were evaluated in HepG2 and Hep3B cells. The in vitro experiments demonstrated that HER significantly induced G0/G1 phase arrest, inhibited the proliferation and promoted cell apoptosis in liver cancer cell lines. In the mechanisms, the antitumor effects of HER on liver cancer cells were mediated by phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) pathway and reactive oxygen species (ROS), simultaneously. In one way, HER inhibited the activities of PI3K-AKT pathway, which interrupt the dimer formation of cyclin-dependent kinase 4 (CDK4) and cyclin D1 (CCND1) and result to G0/G1 phase arrest. In another way, after HER treatment, ROS accumulated in liver cancer cells and caused mitochondria injury which further influenced the expression of apoptosis-related proteins and eventually resulted to HepG2 and Hep3B cell apoptosis. In addition, HER showed a tumor restrain function in HepG2 and Hep3B bearing nude mice. Overall, these findings indicated that HER is a promising antitumor drug, which may provide a new direction for clinical HCC treatment.
Collapse
Affiliation(s)
- Chiufai Kuok
- Faculty of Health Sciences and Sports, Macao Polytechnic University
| | - Qi Wang
- School of Traditional Chinese Medicine, China Pharmaceutical University
| | - Pedro Fong
- Faculty of Health Sciences and Sports, Macao Polytechnic University
| | - Yong Qin
- School of Traditional Chinese Medicine, China Pharmaceutical University
| | - Lirong Meng
- Faculty of Health Sciences and Sports, Macao Polytechnic University
| |
Collapse
|
12
|
Mo J, Tong Y, Ma J, Wang K, Feng Y, Wang L, Jiang H, Jin C, Li J. The mechanism of flavonoids from Cyclocarya paliurus on inhibiting liver cancer based on in vitro experiments and network pharmacology. Front Pharmacol 2023; 14:1049953. [PMID: 36817123 PMCID: PMC9936097 DOI: 10.3389/fphar.2023.1049953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: Cyclocarya paliurus (Batal.) Iljinsk., a subtropical tree belonging to the family Juglandaceae, is rich in polysaccharides, flavonoids, and terpenoids. It has important pharmacological effects such as lowering blood lipids, blood sugar, and blood pressure. However, little has been discerned regarding anti tumor effects and their potential mechanisms. Method: In vitro cell culture experiments were used to test the effect of C. paliurus total flavonoids (CTFs) extract on apoptosis mechanisms in HepG2 cells. Network pharmacology was applied to further explore the effects of CTFs on liver cancer as well as the mechanisms through which these effects might be achieved. Both 3 hydroxyflavone and luteolin were randomly selected to verify the effect on inducing apoptosis and inhibiting the proliferation of HepG2 cells. Results and Discussion: Network pharmacological analysis was applied to these 62 compounds and their targets, and 13 flavonoids were further screened for their potential anti liver cancer activity. These 13 flavonoids included: tangeretin, baicalein, 7,3'-dihydroxyflavone, velutin, 3-hydroxyflavone, chrysin, kumatakenin, tricin, luteolin, chrysoeriol, apigenin, pinocembrin, and butin. Together, these flavonoids were predicted to interact with AKT1, MAPK3, PIK3CA, EGFR, MAP2K1, SRC, IGF1R, IKBKB, MET, and MAPK14. It was predicted that the inhibitory effect on hepatocellular carcinoma would be accomplished by regulation of core proteins relating to such KEGG pathways as cancer, PI3K-Akt, proteoglycans in cancer, microRNAs in cancer, and endocrine resistance via core target proteins. Both 3-hydroxyflavone and luteolin were demonstrated to induce apoptosis and inhibit the proliferation of HepG2 cells. Our study provides scientific evidence supporting the use of CTFs for the treatment of liver cancer.
Collapse
Affiliation(s)
- Jinggang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Yingpeng Tong
- School of Advanced Study, Taizhou University, Taizhou, China
| | - Junxia Ma
- Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China
| | - Kunpeng Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Yifu Feng
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Liezhi Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Hao Jiang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China
| | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang, China,*Correspondence: Chong Jin, ; Junmin Li,
| | - Junmin Li
- School of Advanced Study, Taizhou University, Taizhou, China,Zhejiang Provincial Key Laboratory of Evolutionary Ecology and Conservation, Taizhou University, Taizhou, China,*Correspondence: Chong Jin, ; Junmin Li,
| |
Collapse
|
13
|
Wang C, Yang L, Xu S, Guo H, Guan H, Wang Q, Jiang X, Fei M, Zhang J. Systematic analysis of the role and significance of target genes of active ingredients of traditional Chinese medicine injections in the progression and immune microenvironment of hepatocellular carcinoma. Front Pharmacol 2023; 13:1095965. [PMID: 36686660 PMCID: PMC9852871 DOI: 10.3389/fphar.2022.1095965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
Background: Traditional Chinese medicine in China is an important adjuvant therapy for the treatment of hepatocellular carcinoma (HCC) and traditional Chinese medicines injections have a wide range of clinical applications. The purpose of this study was to identify the active ingredients and related genes of traditional Chinese medicine injections that can treat hepatocellular carcinoma. Methods: Effective small molecule components were extracted from 14 types of traditional Chinese medicines from 8 injections and the main gene targets were identified. The 968 patients with HCC were classified based on the target gene set, and the characteristics of patients with different subtypes were analyzed. Patients with two subtypes of HCC were compared with normal tissues and cirrhosis to identify important gene targets related to traditional Chinese medicines in HCC progression. Results: In this study, 138 important genes associated with traditional Chinese medicines were identified and two HCC subtypes were identified. By analyzing the differences between the two subtypes, 25 related genes were associated with HCC subtypes. Through clinical and pharmacological analysis, this study identified quercetin as an important traditional Chinese medicines small molecule and secreted phosphoprotein 1 (SPP1) as an important oncogene in HCC. Conclusion: Traditional Chinese medicines injection is an important adjuvant treatment modality for HCC. SPP1 is an important oncogene in HCC.
Collapse
Affiliation(s)
- Chao Wang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Lili Yang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shaoheng Xu
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hui Guo
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hewen Guan
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qiannan Wang
- Department of Plastic Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xueyan Jiang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mingyang Fei
- Department of Neurosurgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jinbao Zhang
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China,*Correspondence: Jinbao Zhang,
| |
Collapse
|
14
|
Guo Y, Hu HT, Xu SJ, Xia WL, Zhao Y, Zhao XH, Zhu WB, Li FT, Li HL. Proteoglycan-4 predicts good prognosis in patients with hepatocellular carcinoma receiving transcatheter arterial chemoembolization and inhibits cancer cell migration in vitro. Front Oncol 2022; 12:1023801. [PMID: 36439456 PMCID: PMC9691762 DOI: 10.3389/fonc.2022.1023801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/24/2022] [Indexed: 08/26/2023] Open
Abstract
PURPOSE To search for adaptive response molecules that affect the efficacy of transcatheter arterial chemoembolization (TACE), analyze their clinical correlation with and prognostic value for hepatocellular carcinoma (HCC), and explore their impact on cell biological behavior and their mechanisms of action. METHODS HCC tissue gene sequencing was used to identify differentially expressed genes. The expression of proteoglycan 4 (PRG4) in the serum of 117 patients with HCC who received TACE was detected by enzyme-linked immunosorbent assay. Serum-free medium mimicked TACE-induced nutrient deprivation. Cells with stable knockdown of PRG4 (shPRG4) were constructed to verify the effect and mechanism of PRG4 on the biological behavior of HCC cells in vitro. RESULTS The expression of PRG4 was significantly elevated under TACE-induced starvation conditions. Low PRG4 expression was associated with worse response to TACE treatment, shorter survival time, and stronger HCC migration ability. Furthermore, in vitro experiments showed that knockdown of PRG4 promoted HCC cell migration by enhancing epithelial-mesenchymal transition (EMT) while did not affect proliferation. When PRG4 expression was low, starvation treatment impaired the migratory ability of HCC cells and reduced the chemosensitivity of HCC cells to epirubicin. CONCLUSIONS PRG4 expression predicts survival and TACE treatment response in patients with HCC. Furthermore, knockdown of PRG4 enhanced EMT, leading to HCC cell migration. PRG4 may serve as a biomarker for HCC patients receiving TACE.
Collapse
Affiliation(s)
- Yuan Guo
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hong Tao Hu
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Shi Jun Xu
- Department of Radiology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wei Li Xia
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yan Zhao
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xiao Hui Zhao
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wen Bo Zhu
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Fang Ting Li
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Hai Liang Li
- Department of Minimal Invasive Intervention, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
15
|
Wang S, Cui Q, Chen X, Zhu X, Lin K, Zheng Q, Wang Y, Li D. Ailanthone Inhibits Cell Proliferation in Tongue Squamous Cell Carcinoma via PI3K/AKT Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3859489. [PMID: 36387351 PMCID: PMC9643058 DOI: 10.1155/2022/3859489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 10/03/2022] [Accepted: 10/21/2022] [Indexed: 07/22/2023]
Abstract
Tongue squamous cell carcinoma (TSCC) is the most widespread and invasive subtype of oral cancer with high recurrence rates. Ailanthone (AIL) is an active ingredient in the plant extracts of Ailanthus altissima (Mill.) Swingle. Here, we showed that AIL inhibited the proliferation of human TSCC, the cell viability of Cal-27 and Tca8113 was significantly decreased after AIL treatment for 24 h. Hoechst 33258 staining demonstrated apoptotic characteristics (such as chromatin aggregation) after AIL treatment. The ratio of early- and late-apoptotic cells in AIL-treated Cal-27 and TCA8113 cells increased remarkably when compared with the control group. Bcl-2/Bax ratio and the levels of PARP1, caspase-9, and caspase-3 decreased after AIL treatment, accompanied by significant increase of cleaved PARP1, cleaved caspase-9, and caspase-3 in Cal-27 and TCA8113 cells. Meanwhile, AIL led to Cal-27 cell cycle arrest at G2/M phase. Western blot implied decreased levels of CDK1 and cyclin B1 after AIL treatment. The level of phospho-PI3K p55 subunit and p-Akt were significantly downregulated by AIL in both Cal-27 and TCA8113 cells. These findings implied the potential applications of AIL in the treatment of human TSCC.
Collapse
Affiliation(s)
- Shuhan Wang
- Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, China
- College of Stomatology, Binzhou Medical University, Yantai 264003, Shandong, China
- College of Stomatology, Qilu Medical University, Zibo 255300, Shandong, China
| | - Qixiao Cui
- College of Stomatology, Binzhou Medical University, Yantai 264003, Shandong, China
| | - Xiaoyu Chen
- Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, China
| | - Xuejie Zhu
- Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, China
| | - Kehao Lin
- College of Stomatology, Binzhou Medical University, Yantai 264003, Shandong, China
| | - Qiusheng Zheng
- Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, China
| | - Yuliang Wang
- College of Stomatology, Binzhou Medical University, Yantai 264003, Shandong, China
- Department of Oral and Maxillofacial Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai 264100, Shandong, China
| | - Defang Li
- Collaborative Innovation Platform for Modernization and Industrialization of Regional Characteristic Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai 264003, Shandong, China
| |
Collapse
|
16
|
Mohapatra P, Chandrasekaran N. Wnt/β-catenin targeting in liver carcinoma through nanotechnology-based drug repurposing: A review. Biomed Pharmacother 2022; 155:113713. [PMID: 36126453 DOI: 10.1016/j.biopha.2022.113713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/08/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Liver cancer is the fifth most widespread in the world, with a high fatality rate and poor prognosis.However,surgicalresction,thermal/radiofrequencyablation,chemo/radioembolization and pathway targeting to the cancer cells are all possible options for treating Liver Carcinoma. Unfortunately, once the tumour has developed and spread, diagnosis often occurs too late. The targeted therapy has demonstrated notable, albeit modest, efficacy in some patients with advanced HCC. This demonstrates the necessity of creating additional focused treatments and, in pursuit of this end, the need to find ever-more pathways as prospective targets. Despite the critical need, there are currently no Wnt signalling directed therapy on the research field, only a few methods have progressed beyond the early stage of clinical studies. In the present study, we report that repurposing of drug previously licensed for other diseases is one possible strategy inhibit malignant cell proliferation and renewal by removing individuals protein expression in the Wnt/β-catenin pathway. Particularly β-catenin complex is present in Liver cancer, where tumour necrosis factor is indispensable for the complex formation and β-catenin interactions are disrupted upon drug in nano-carrier through nanotechnology. This study findings not only highlight that repurposing drug could improve liver cancer treatment outcomes but also focused to character traits and functions of the Wnt signalling cascade's molecular targets and how they could be used to get anti-tumour results method to targeting Wnt/β-catenin in liver carcinoma.
Collapse
|
17
|
Wei L, Wang Z, Jing N, Lu Y, Yang J, Xiao H, Guo H, Sun S, Li M, Zhao D, Li X, Qi W, Zhang Y. Frontier progress of the combination of modern medicine and traditional Chinese medicine in the treatment of hepatocellular carcinoma. Chin Med 2022; 17:90. [PMID: 35907976 PMCID: PMC9338659 DOI: 10.1186/s13020-022-00645-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/20/2022] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC, accounting for 90% of primary liver cancer) was the sixth most common cancer in the world and the third leading cause of cancer death in 2020. The number of new HCC patients in China accounted for nearly half of that in the world. HCC was of occult and complex onset, with poor prognosis. Clinically, at least 15% of patients with HCC had strong side effects of interventional therapy (IT) and have poor sensitivity to chemotherapy and targeted therapy. Traditional Chinese medicine (TCM), as a multi-target adjuvant therapy, had been shown to play an active anti-tumor role in many previous studies. This review systematically summarized the role of TCM combined with clinically commonly used drugs for the treatment of HCC (including mitomycin C, cyclophosphamide, doxorubicin, 5-fluorouracil, sorafenib, etc.) in the past basic research, and summarized the efficacy of TCM combined with surgery, IT and conventional therapy (CT) in clinical research. It was found that TCM, as an adjuvant treatment, played many roles in the treatment of HCC, including enhancing the tumor inhibition, reducing toxic and side effects, improving chemosensitivity and prolonging survival time of patients. This review summarized the advantages of integrated traditional Chinese and modern medicine in the treatment of HCC and provides a theoretical basis for clinical research.
Collapse
Affiliation(s)
- Lai Wei
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Zeyu Wang
- Department of Scientific Research, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Niancai Jing
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Yi Lu
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Jili Yang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Hongyu Xiao
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Huanyu Guo
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Shoukun Sun
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Mingjing Li
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Yue Zhang
- Department of Integrated Chinese and Western Medicine, Jilin Cancer Hospital, Changchun, 130000, Jilin, China.
| |
Collapse
|
18
|
Yang X, Sun J, Wen B, Wang Y, Zhang M, Chen W, Zhao W, He C, Zhong X, Liu Y, Li T, Sun H, He S. Biejiajian Pill Promotes the Infiltration of CD8 + T Cells in Hepatocellular Carcinoma by Regulating the Expression of CCL5. Front Pharmacol 2021; 12:771046. [PMID: 34899325 PMCID: PMC8661106 DOI: 10.3389/fphar.2021.771046] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 10/29/2021] [Indexed: 12/17/2022] Open
Abstract
Tumor-infiltrating CD8+T lymphocytes are mostly associated with a favorable prognosis in numerous cancers, including hepatocellular carcinoma (HCC). Biejiajian Pill (BJJP) is a common type of traditional Chinese medicine that is widely used in the treatment of HCC in China. Previous studies showed that BJJP suppressed the growth of HCC cells both in vivo and in vitro, by exerting direct cytotoxic effects on tumor cells. The present study demonstrated that in addition to direct cytotoxicity, BJJP inhibits the growth of tumor cells by promoting the infiltration of CD8+T cells into the tumor in H22-bearing mice. Mechanistically, chemokine ligand 5 (CCL5) was identified as one of the most highly expressed chemokines by tumor cells in vivo after treatment with BJJP. Additionally, CCL5 was knocked down in H22 cells and the results showed that knockdown of the gene significantly impaired the infiltration of CD8+T cells in vivo. Furthermore, the effects of BJJP on human HCC cell lines were assessed in vitro. Similarly, cells treated with BJJP had higher expression of CCL5 mRNA, which was consistent with increased levels of CCL5 protein in human tumor cells. These findings provide new insights into the anticancer effects of BJJP, which regulated the expression of CCL5 and the infiltration of CD8+T cells. The results, therefore, suggest that BJJP has great potential application in clinical practice.
Collapse
Affiliation(s)
- Xuemei Yang
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jialing Sun
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Bin Wen
- Department of Traditional Chinese Medicine, Hospital of PLA, Guangzhou, China
| | - Yu Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingjia Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Weicong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wenting Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chunyu He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaodan Zhong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Tong Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Songqi He
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Jia W, Liang S, Cheng B, Ling C. The Role of Cancer-Associated Fibroblasts in Hepatocellular Carcinoma and the Value of Traditional Chinese Medicine Treatment. Front Oncol 2021; 11:763519. [PMID: 34868982 PMCID: PMC8636329 DOI: 10.3389/fonc.2021.763519] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/28/2021] [Indexed: 01/10/2023] Open
Abstract
Invasion and metastasis are the main reasons for the high mortality of liver cancer, which involve the interaction of tumor stromal cells and malignant cells. Cancer-associated fibroblasts (CAFs) are one of the major constituents of tumor stromal cells affecting tumor growth, invasion, and metastasis. The heterogeneous properties and sources of CAFs make both tumor-supporting and tumor-suppression effects possible. The mechanisms for CAFs in supporting hepatocellular carcinoma (HCC) progression can be categorized into upregulated aggressiveness and stemness, transformed metabolism toward glycolysis and glutamine reductive carboxylation, polarized tumor immunity toward immune escape of HCC cells, and increased angiogenesis. The tumor-suppressive effect of fibroblasts highlights the functional heterogenicity of CAF populations and provides new insights into tumor–stromal interplay mechanisms. In this review, we introduced several key inflammatory signaling pathways in the transformation of CAFs from normal stromal cells and the heterogeneous biofunctions of activated CAFs. In view of the pleiotropic regulation properties of traditional Chinese medicine (TCM) and heterogeneous effects of CAFs, we also introduced the application and values of TCM in the treatment of HCC through targeting CAFs.
Collapse
Affiliation(s)
- Wentao Jia
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Shufang Liang
- Department of Traditional Chinese Medicine, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Binbin Cheng
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| | - Changquan Ling
- School of Traditional Chinese Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
20
|
Zhu H, Wang SY, Zhu JH, Liu H, Kong M, Mao Q, Zhang W, Li SL. Efficacy and safety of transcatheter arterial chemoembolization combined with ginsenosides in hepatocellular carcinoma treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153700. [PMID: 34425474 DOI: 10.1016/j.phymed.2021.153700] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/30/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Transcatheter arterial chemoembolization (TACE) is a standard therapy to treat hepatocellular carcinoma (HCC), but often limited for its complications. Ginsenosides, including total ginsenosides (GS), Rg3, Rh2 and CK, have been clinically used as adjuvants of TACE in HCC therapy. However, partial clinical observations concerning the efficacy and safety of the combinational treatment were contradictory. PURPOSE To investigate the efficacy and safety of TACE and ginsenosides combination for HCC therapy. METHODS Randomized controlled trials (RCTs) regarding TACE and ginsenosides for HCC up to May 2021 were screened from six databases (PubMed, Embase, Cochrane Library, China National Knowledge Infrastructure, Chinese VIP Information and Web of Science). The outcomes of tumor response, adverse reactions (ADRs), quality of life (QOL), survival rates (OS) and liver function were extracted and evaluated by meta-analysis, respectively. RESULTS A total of 18 RCTs with 1308 HCC patients were enrolled, and most of the eligible studies had unclear bias risk. Compared with TACE, combining ginsenosides improved objective response rate [ORR, risk ratio (RR) 1.39, 95% confidence intervals (CI) 1.20∼1.61], disease control rate (DCR, RR 1.21, 95% CI 1.12∼1.30), QOL (RR 1.54, 95% CI 1.25∼1.90), one- (RR 1.37, 95% CI 1.16∼1.62) and two- (RR 1.43, 95% CI 1.06∼1.95) year OS, and A level of Child-pugh, as well as reduced the risks of nausea and vomiting, pyrexia, ache, hyperbilirubinemia, anorexia, fatigue, leukopenia, thrombocytopenia and myelosuppression. Subgroup analyses showed that both short- and long- treatment durations of ginsenosides enhanced the A level of Child-pugh, and reduced nausea and vomiting, ache and hyperbilirubinemia. Besides, combining Rg3 benefited DCR, ORR and QOL, and alleviated nausea and vomiting, hyperbilirubinemia, leukopenia, myelosuppression, thrombocytopenia and α-fetoprotein, while combining GS alleviated nausea and vomiting, ache and hyperbilirubinemia, combining Rh2 alleviated thrombocytopenia, and combining CK alleviated nausea and vomiting, pyrexia, ache and leukopenia, respectively. CONCLUSION The results suggested that combining ginsenosides could continuously benefit the efficacy and safety of TACE in HCC treatment, and Rg3 is the prior selection during the combination.
Collapse
Affiliation(s)
- He Zhu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| | - Si-Yu Wang
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Jin-Hao Zhu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Hui Liu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Ming Kong
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Qian Mao
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Wei Zhang
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China.
| |
Collapse
|
21
|
Chen F, Lao Z, Zhang H, Wang J, Wang S. Knockdown of circ_0001883 may inhibit epithelial-mesenchymal transition in laryngeal squamous cell carcinoma via the miR-125-5p/PI3K/AKT axis. Exp Ther Med 2021; 22:1007. [PMID: 34345289 PMCID: PMC8311254 DOI: 10.3892/etm.2021.10440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 06/09/2021] [Indexed: 12/27/2022] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a malignant tumor with increasing incidence and poor prognosis. Circular RNAs (circRNAs) are known to modulate tumorigenesis and cancer development that may function through microRNAs (miRs). The aim of the present study was to investigate the functional roles of circ_0001883 in LSCC and the underlying molecular mechanism. The expression of circ_0001883 was upregulated and measured using reverse transcription-quantitative PCR (RT-qPCR) and RNase R. miR-125b-5p expression was downregulated in LSCC tissues and cells as determined using RT-qPCR. Subsequently, knockdown of circ_0001883 inhibited LSCC cell migration, invasion and epithelial-mesenchymal transition (EMT), which were tested by wound healing assays, Transwell assays and western blotting, respectively. Bioinformatics analysis predicted that circ_0001883 was a sponge of miR-125b-5p, which was verified using a dual-luciferase reporter assay. Knockdown of circ_0001883 played a functional role by sponging miR-125b-5p. Additionally, circ_0001883 and miR-125b-5p influenced phosphorylation of PI3K and AKT, detected via western blotting. In an in vivo study, knockdown of circ_0001883 reduced tumor volume and weight in mice, along with enhanced miR-125b-5p and E-cadherin expression levels, and decreased N-cadherin, phosphorylated (p)-PI3K/PI3K and p-AKT/AKT ratios. In conclusion, knockdown of circ_0001883 inhibited cell migration, invasion and EMT of LSCC by sponging miR-125b-5p. This is hypothesized to be via the PI3K/AKT signaling pathway, which suggested that circ_0001883 has potential for LSCC therapy.
Collapse
Affiliation(s)
- Fu Chen
- Department of Radiation Oncology, Eye and ENT Hospital of Fudan University, Shanghai 200031, P.R. China
| | - Zheng Lao
- Radiotherapy Division, Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Haiyan Zhang
- Department of Radiation Oncology, Eye and ENT Hospital of Fudan University, Shanghai 200031, P.R. China
| | - Jie Wang
- Department of Radiation Oncology, Eye and ENT Hospital of Fudan University, Shanghai 200031, P.R. China
| | - Shengzi Wang
- Department of Radiation Oncology, Eye and ENT Hospital of Fudan University, Shanghai 200031, P.R. China
| |
Collapse
|
22
|
Genetic Toxicology and Safety Pharmacological Evaluation of Forsythin. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6610793. [PMID: 34239584 PMCID: PMC8233079 DOI: 10.1155/2021/6610793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/21/2021] [Accepted: 06/08/2021] [Indexed: 11/18/2022]
Abstract
Introduction Forsythin is the main ingredient of Forsythia suspensa and is widely used in treatment of fever, viral cold, gonorrhea, and ulcers clinically. This study aimed to evaluate the potential genetic toxicity of forsythin and its safety for human use. Methods Based on the Good Laboratory Practice regulations and test guidelines, the genetic toxicity of forsythin was assessed by the Ames test, chromosome aberration (CA) test, and bone marrow micronucleus (MN) test in vivo. In the Ames test, five strains of Salmonella typhimurium were exposed to different concentrations of forsythin in the presence or absence of the S9 mixture, and then, the number of His + revertant colonies was counted. In the CA test, Chinese hamster lung (CHL) fibroblast cells were treated with different concentrations of forsythin, mitomycin C, or cyclophosphamide in the presence or absence of the S9 mixture, and the chromosomal aberrations were determined. In the MN test, bone marrow was isolated from the mice with different treatments, and the ratios of polychromatic erythrocytes (PCE) and erythrocytes (PCE/(PCE + NCE)) were measured. Finally, beagle dogs were divided into four groups (negative control, low dose, medium dose, and high dose groups), and then, a telemetry system was used to evaluate the safe use of forsythin. Results Ames test results showed that the number of colonies in all test strains with different treatments showed no significantly dose-dependent increase in the presence or absence of the S9 mixture (p > 0.05). In the CA test, the number of cells with aberrations in the CHL fibroblast cells treated with low, medium, and high doses of forsythin for 24/48 h in the absence of the S9 mixture was, respectively, 5.0/2.5, 4.5/1.5, and 5.0/5.0, and in the presence of the S9 mixture, the number was, respectively, 5.0, 5.0, and 4.5. These results showed that there was no significant difference compared to the negative control group either in the presence (2.0) or in the absence (4.0/2.5 for 24/48 h) of the S9 mixture (p > 0.05). The MN test showed that the values of PCE/(PCE + NCE) in the negative, positive controls, and forsythin treatment groups were all more than 20%, which indicated that forsythin had no cytotoxicity. Additionally, no significant toxicological effects of forsythin on blood pressure, respiration, temperature, electrocardiogram, and other physiological indicators in the conscious beagle dogs of different groups were observed by the telemetry method. Conclusion Our findings showed that forsythin has low probability of genetic toxicity and no significant toxicological effects, which implied that forsythin is suitable for further development and potential application.
Collapse
|
23
|
Chao WW, Liou YJ, Ma HT, Chen YH, Chou ST. Phytochemical composition and bioactive effects of ethyl acetate fraction extract (EAFE) of Glechoma hederacea L. J Food Biochem 2021; 45:e13815. [PMID: 34121206 DOI: 10.1111/jfbc.13815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/26/2021] [Accepted: 05/20/2021] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is a highly malignant cancer that exists worldwide. Herbal medicine plays an important role in the management and treatment of various diseases worldwide. The herbal medicine Glechoma hederacea L. has a variety of biological activities and belongs to the Labiatae family. The current study investigated the in vitro effects of ethyl acetate fraction extract (EAFE) of G. hederacea on HepG2 cells and its possible mechanism. The phytochemical composition of EAFE was analyzed by high performance liquid chromatography (HPLC). Bioactive effects of the EAFE were assessed using the MTT assay, annexin V-FITC/PI staining, PhiphiLux-G1 D2 kit, DAPI and comet assay, flow cytometry, western blotting. In this study, we found that rosmarinic acid (RA), caffeic acid (CA), and ferulic acid (FA) were the abundant polyphenols in EAFE of G. hederacea. This fraction extract could significantly inhibit HepG2 cell proliferation, make cells apoptosis, and cause S phase arrest. The apoptogenic activity of EAFE involved reactive oxygen species (ROS) induction, Ca2+ accumulation, mitochondrial membrane potential (MMP, ΔΨm) destruction, regulate the Bax/Bcl-2 ratio and caspases 3, 9 cascade. We propose that the EAFE can inhibit the proliferation of HepG2 cell via intracellular ROS mediated apoptosis. EAFE could be developed as a possible anti-HCC agent or pharmaceutical industries. PRACTICAL APPLICATIONS: 1. The rosmarinic acid, caffeic acid, and ferulic acid were the main polyphenolic components in the ethyl acetate fraction extract (EAFE) of Glechoma hederacea. 2. The EAFE treatment exerted cytotoxicity by inducing S arrest and apoptosis in HepG2 cells. 3. Antitumor effect of EAFE through the mitochondria-mediated pathway and ROS-mediated ER stress.
Collapse
Affiliation(s)
- Wen Wan Chao
- Department of Nutrition and Health Sciences, Kainan University, Taoyuan, Taiwan
| | - Yu Jhen Liou
- Department of Food and Nutrition, Providence University, Taichung, Taiwan.,Department of Nutrition, Show Chwan Memorial Hospital, Changhua City, Taiwan
| | - Hao Ting Ma
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
| | - Yi Hua Chen
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
| | - Su-Tze Chou
- Department of Food and Nutrition, Providence University, Taichung, Taiwan
| |
Collapse
|
24
|
Design of dual targeting immunomicelles loaded with bufalin and study of their anti-tumor effect on liver cancer. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2021; 19:408-417. [PMID: 34130942 DOI: 10.1016/j.joim.2021.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/07/2020] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Bufalin is an effective drug for the treatment of liver cancer. But its high toxicity, poor water-solubility, fast metabolism and short elimination half-life limit its use in tumor treatment. How to make the drug accumulate in the tumor and reduce side effects while maintaining its efficacy are urgent problems to be solved. The goal of this study is to solve these problems. METHODS A copolymer with tunable poly-N-isopropylacrylamide and polylactic acid was designed and synthesized. The corresponding dual targeting immunomicelles (DTIs) loaded with bufalin (DTIs-BF) were synthesized by copolymer self-assembly in an aqueous solution. The size and structure of DTIs-BF were determined by ZetaSizer Nano-ZS and transmission electron microscopy. Then, its temperature sensitivity, serum stability, critical micelle concentration (CMC), entrapment efficiency (EE), drug release and non-cytotoxicity of blank block copolymer micelles (BCMs) were evaluated. Next, the effects of DTIs-BF on cellular uptake, cytotoxicity, and tumor cell inhibition were evaluated. Finally, the accumulation of DTIs-fluorescein isothiocyanate (FITC) and the in vivo anti-tumor effect were observed using an interactive video information system. RESULTS DTIs-BF had a small size, spherical shape, good temperature sensitivity, high serum stability, low CMC, high EE, and slow drug release. The blank BCMs had very low cytotoxicity. Compared with free bufalin, the in vitro cellular internalization and cytotoxicity of DTIs-BF against SMMC-7721 cells were significantly enhanced, and the effects were obviously better at 40 °C than 37 °C. In addition, the therapeutic effect on SMMC-7721 cells was further enhanced by the programmed cell death specifically caused by bufalin. When DTIs-FITC were injected intravenously in BALB/c nude mice bearing liver cancer, the accumulation of FITC was significantly increased in tumors. CONCLUSION DTIs-BF is a potentially effective nano-formulation and has broad prospects in the clinical treatment of liver cancer.
Collapse
|
25
|
Feng Y, Jin H, Guo K, Xiang Y, Zhang Y, Du W, Shen M, Ruan S. Results from a Meta-analysis of Combination of PD-1/PD-L1 and CTLA-4 Inhibitors in Malignant Cancer Patients: Does PD-L1 Matter? Front Pharmacol 2021; 12:572845. [PMID: 33716732 PMCID: PMC7949479 DOI: 10.3389/fphar.2021.572845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 01/25/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Combination therapy with immune checkpoint inhibitors (ICIs) has been widely used for clinical treatment in recent years, which has a better survival benefit. However, not all patients can derive clinical benefit from combination immunotherapy. Therefore, it is necessary to explore the biomarkers of combination immunotherapy. Methods: We retrieved articles from electronic databases including PubMed, EMBASE and Cochrane. The statistical analysis was performed using RevMan software. Progression free survival (PFS), overall survival (OS) and objective response rate (ORR) were the outcome indicators. In the unselect population, we compared combination therapy with other treatments. In addition, we also conducted subgroup analysis on PFS, OS and ORR according to PD-L1 status. Results: Seven studies were included in the analysis for a total of 3,515 cases. In the unselected population, we found that combination therapy has longer PFS, OS, and better ORR than other treatments for cancer patients. The longer PFS was showed in PD-L1 ≥ 5% cases (HR = 0.64, 95% CI: 0.56–0.76; p < 0.001) than PD-L1 ≥ 1% cases (HR = 0.72, 95% CI: 0.66–0.79; p < 0.001), while ORR and OS have not related to the status of PD-L1. Conclusion: This study supported the efficacy of combination therapy with immune checkpoint inhibitors (ICIs), and also showed that PFS in patients with malignant tumors is positively correlated with PD-L1 expression. Due to the limited number of trials included, more high-quality clinical randomized controlled trials should be conducted to confirm the review findings.
Collapse
Affiliation(s)
- Yuqian Feng
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Huimin Jin
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Kaibo Guo
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuying Xiang
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiting Zhang
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Wurong Du
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Minhe Shen
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shanming Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
26
|
Wang Y, Tan J, Hu P, Pei Q, Wen Y, Ma W, Shi K, Li Z, Li H, Cheng F, Gu X, Yao X, Man Y, Zhao R, Feng S, Ding X, Yang T. Traditional Chinese medicine compound, Bu Sheng Hui Yang Fang, promotes the proliferation of lymphocytes in the immunosuppressed mice potentially by upregulating IL-4 signaling. Biomed Pharmacother 2021; 134:111107. [PMID: 33341059 DOI: 10.1016/j.biopha.2020.111107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 01/04/2023] Open
Abstract
The immune system plays a pivotal role in defending against infection and cancer immunosurveillance during the onset and procession of malignant disease. Cancer patients are frequently immunocompromised and subject to refractory infection and relapse of leukemia, due to the cytotoxic agents and immunosuppressive glucocorticoids in the chemotherapy regimens. Bu Shen Hui Yang Fang (BSHY), a traditional Chinese compound, was widely used in China to enhance the immune system of leukemia patients combined with chemotherapy and effectively lowered their risk of infection, with specific mechanism unknown yet. Thus, we investigated the effects of BSHY on the immune system using immunosuppressive mouse models. By analyzing the immune system of immunosuppressed BALB/C mice induced by hydrocortisone, we found an increase of CD4+ and CD8+ lymphocytes in the spleens of mice after BSHY treatment. Furthermore, we found the enhanced immune system in BSHY treated group was due to increased proliferation and decreased apoptosis of lymphocytes. Cytokine array analysis revealed that interleukin 4 (IL-4) was reduced in the plasma of immunosuppressed mice but returned to a normal level after BSHY treatment. Moreover, we found IL-4 was an adverse prognostic factor in acute myeloid leukemia patients and part of them could be elevated by BSHY. Mechanistically, we found BSHY enhances the proliferation of lymphocytes in a Stat6-dependent manner. In summary, our current study demonstrates that BSHY enhances the proliferation of lymphocytes in the immunosuppressed mice via upregulating IL-4 signaling.
Collapse
Affiliation(s)
- Yajie Wang
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Jiabin Tan
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Peng Hu
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Qiang Pei
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Yan Wen
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Wenqing Ma
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Keqian Shi
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Zengzheng Li
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Huiyuan Li
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Fengyu Cheng
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Xuezhong Gu
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Xiangmei Yao
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Yan Man
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Renbin Zhao
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Shuai Feng
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China
| | - Xiao Ding
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Tonghua Yang
- Yunnan Blood Disease Clinical Medical Center, Yunnan Blood Disease Hospital, National Key Clinical Specialty of Hematology, Department of Hematology, The First People's Hospital of Yunnan Province, Kunming, China; Kunming University of Science and Technology, Kunming, China.
| |
Collapse
|
27
|
Wu J, Luo D, Li S. Network Pharmacology-Oriented Identification of Key Proteins and Signaling Pathways Targeted by Xihuang Pill in the Treatment of Breast Cancer. BREAST CANCER-TARGETS AND THERAPY 2020; 12:267-277. [PMID: 33324095 PMCID: PMC7733446 DOI: 10.2147/bctt.s284076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022]
Abstract
Purpose The compound traditional Chinese medicine Xihuang pill (XHP) has been adopted to treat breast cancer (BC) for centuries, but its specific mechanism of action is unclear. Materials and Methods The active ingredients and related targets of XHP were screened using the TCMSP and TCMID databases. GSE139038 was downloaded from the GEO database, and differentially expressed genes (DEGs) were analyzed. The intersection of targets and DEGs were chosen to build an ingredients–target genes network. Protein–protein interaction network construction and functional enrichment analysis of target genes were conducted. Results A PPI network of 37 targets was constructed, and seven core nodes (FOS, MYC, JUN, PPARG, MMP9, PTGS2, SERPINE1) were identified. Functional enrichment analysis revealed that the aforementioned targets were mainly enriched in the IL-17, toll-like receptor, and tumor necrosis factor signaling pathways, which are deeply involved in the inflammatory microenvironment of tumors. Conclusion This network pharmacology study indicated that XHP can inhibit the development of BC by targeting a variety of proteins and signaling pathways involved in the inflammatory microenvironment.
Collapse
Affiliation(s)
- Jiafa Wu
- School of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China.,Henan Engineering Research Center of Food Microbiology, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China
| | - Dongping Luo
- The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China
| | - Shengnan Li
- School of Medicine, Henan Polytechnic University, Jiaozuo, Henan, People's Republic of China
| |
Collapse
|
28
|
Zhang S, Zhang M, Chen J, Zhao J, Su J, Zhang X. Ginsenoside Compound K Regulates HIF-1α-Mediated Glycolysis Through Bclaf1 to Inhibit the Proliferation of Human Liver Cancer Cells. Front Pharmacol 2020; 11:583334. [PMID: 33363466 PMCID: PMC7753211 DOI: 10.3389/fphar.2020.583334] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/10/2020] [Indexed: 11/13/2022] Open
Abstract
This study aimed to demonstrate that ginsenoside compound K (20 (S)-ginsenoside CK; CK) downregulates Bcl-2-associated transcription factor 1 (Bclaf1), which inhibits the hypoxia-inducible factor-1α (HIF-1α)-mediated glycolysis pathway to inhibit the proliferation of liver cancer cells. Treatment of hepatoma cells (Bel-7404 and Huh7) under hypoxic conditions with different concentrations of CK showed that CK inhibited the proliferation of hepatoma cells in a time- and concentration-dependent manner; furthermore, the ability of the cells to form colonies was reduced, and cell growth was blocked in the G0/G1 phase. CK promoted the degradation of HIF-1α ubiquitination in liver cancer cells by regulating the expression of HIF-1α and related ubiquitination proteins; moreover, it reduced the activity of key enzymes involved in glycolysis, the pressure of cellular glycolysis, and the rate of real-time ATP production, thereby inhibiting the glycolysis pathway. It also decreased the expression of Bclaf1 in hypoxic liver cancer cells and thus reduced the ability of Bclaf1 to bind to HIF-1α. CK treatment of Bel-7404 and Huh7 cells with CRISPR/Cas9-engineered knock out of Bclaf1 gene under hypoxic conditions further suppressed the expression of HIF-1α, promoted HIF-1α ubiquitination, and inhibited the glycolysis pathway. In a rat model of primary liver cancer induced by diethylnitrosamine, positron emission tomography and computed tomography scans showed that after CK administration, tumor tissue volumes were reduced and glucose uptake capacity decreased. Increased Bclaf1 and HIF-1α expression promoted the ubiquitination of HIF-1α and inhibited the glycolysis pathway, thereby inhibiting the proliferation of liver cancer cells. In summary, this study confirmed by in vitro and in vivo experiments that in hypoxic liver cancer cells CK downregulates the expression of Bclaf1, inhibits the HIF-1α-mediated glycolysis pathway, and inhibits cell proliferation, suggesting that the CK-mediated effects on Bclaf1 may represent a novel therapeutic approach for the treatment of liver cancer patients.
Collapse
Affiliation(s)
- Silin Zhang
- College of Medicine, Yanbian University, Yanji, China
| | | | - Jiaxin Chen
- College of Medicine, Yanbian University, Yanji, China
| | - Jiaqi Zhao
- College of Medicine, Yanbian University, Yanji, China
| | - Jielin Su
- College of Medicine, Yanbian University, Yanji, China
| | - Xuewu Zhang
- College of Medicine, Yanbian University, Yanji, China
| |
Collapse
|
29
|
Uncovering the Mechanism of the Effects of Pien-Tze-Huang on Liver Cancer Using Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4863015. [PMID: 32963562 PMCID: PMC7492898 DOI: 10.1155/2020/4863015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 01/25/2023]
Abstract
Pien-Tze-Huang (PTH) has a long history in the treatment of liver cancer. However, its molecular mechanism of action remains unclear. TCMSP and TCM were used to collect the active ingredients. Bioactive compounds targets were predicted by reverse pharmacophore models. The antiliver cancer targets of PTH were selected by gene comparison of liver cancer in the GEO database. Molecular docking was used to verify the binding activity of the targets and the active ingredients. The DAVID was used to analyze the gene function and signal pathway. A model was built with Cytoscape. The core genes were obtained by PPI network. We screened the 4 main medicinal ingredients of PTH to obtain 16 active ingredient, 190 potential targets, and 6 core genes. We found that active small molecules exert anticancer effects by multiple pathways. The core genes were involved in multiple biological processes. We also found that eight chemical components play a greater role in inhibiting liver cancer. PTH achieves the effect of inhibiting liver cancer through the synergistic effect of multiple components, multiple targets, and multiple pathways. This study provides a potential scientific basis for further elucidating the molecular mechanism of action of PTH against liver cancer.
Collapse
|
30
|
Zeng C, Zhao C, Ge F, Li Y, Cao J, Ying M, Lu J, He Q, Yang B, Dai X, Zhu H. Machado-Joseph Deubiquitinases: From Cellular Functions to Potential Therapy Targets. Front Pharmacol 2020; 11:1311. [PMID: 32982735 PMCID: PMC7479174 DOI: 10.3389/fphar.2020.01311] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/07/2020] [Indexed: 12/13/2022] Open
Abstract
Ubiquitination is known as important post-translational modification in cancer-related pathways. Human deubiquitinases (DUBs), with functions of modulating the ubiquitination process, are a family with about 100 proteins. They mainly function by cutting ubiquitin chains of the substrates. The Machado-Joseph domain-containing proteases (MJDs) is one of the sub-families of DUBs, consisting of four members, namely, Ataxin-3, Ataxin-3L, JOSD1, and JOSD2. Recent studies have provided new insights into biological functions of MJDs in the progression of Machado-Joseph disease or cancer diseases. In this review, we summarized the cellular functions and regulatory mechanisms of MJDs in Machado-Joseph disease and cancer pathways. Furthermore, we summarized MJDs genetic alterations in different human cancers by exploring the public databases (cBioportal). The aim of this review is to provide a comprehensive account based on our current knowledge about emerging insights into MJDs in physiology and disease, which might shed light on fundamental biological questions and promise to provide a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Chenming Zeng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chenxi Zhao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fujing Ge
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuekang Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macau
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoyang Dai
- Center for Drug Safety Evaluation and Research, Zhejiang University, Hangzhou, China
| | - Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
31
|
Chen CY, Fang JY, Chen CC, Chuang WY, Leu YL, Ueng SH, Wei LS, Cheng SF, Hsueh C, Wang TH. 2-O-Methylmagnolol, a Magnolol Derivative, Suppresses Hepatocellular Carcinoma Progression via Inhibiting Class I Histone Deacetylase Expression. Front Oncol 2020; 10:1319. [PMID: 32850418 PMCID: PMC7431949 DOI: 10.3389/fonc.2020.01319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022] Open
Abstract
Magnolia officinalis is widely used in Southeast Asian countries for the treatment of fever, headache, diarrhea, and stroke. Magnolol is a phenolic compound extracted from M. officinalis, with proven antibacterial, antioxidant, anti-inflammatory, and anticancer activities. In this study, we modified magnolol to synthesize a methoxylated derivative, 2-O-methylmagnolol (MM1), and investigated the use of MM1, and magnolol in the treatment of liver cancer. We found that both magnolol and MM1 exhibited inhibitory effects on the growth, migration, and invasion of hepatocellular carcinoma (HCC) cell lines and halted the cell cycle at the G1 phase. MM1 also demonstrated a substantially better tumor-suppressive effect than magnolol. Further analysis suggested that by inhibiting class I histone deacetylase expression in HCC cell lines, magnolol and MM1 induced p21 expression and p53 activation, thereby causing cell cycle arrest and inhibiting HCC cell growth, migration, and invasion. Subsequently, we verified the significant tumor-suppressive effects of magnolol and MM1 in an animal model. Collectively, these findings demonstrate the anti-HCC activities of magnolol and MM1 and their potential for clinical use.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan
| | - Jia-You Fang
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Chin-Chuan Chen
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan
| | - Wen-Yu Chuang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan.,Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shir-Hwa Ueng
- Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Li-Shan Wei
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Shu-Fang Cheng
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Taoyuan City, Taiwan
| | - Chuen Hsueh
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Department of Anatomic Pathology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan City, Taiwan
| | - Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Taoyuan City, Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan City, Taiwan.,Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| |
Collapse
|
32
|
Ran G, Chen X, Xie Y, Zheng Q, Xie J, Yu C, Pittman N, Qi S, Yu FX, Agbandje-McKenna M, Srivastava A, Ling C. Site-Directed Mutagenesis Improves the Transduction Efficiency of Capsid Library-Derived Recombinant AAV Vectors. Mol Ther Methods Clin Dev 2020; 17:545-555. [PMID: 32258217 PMCID: PMC7114622 DOI: 10.1016/j.omtm.2020.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/26/2022]
Abstract
Recombinant adeno-associated virus (rAAV) vectors selected from capsid libraries present enormous advantages in high selectivity of tissue tropism and their potential use in human gene therapy applications. For example, rAAV-LK03, was used in a gene therapy trial for hemophilia A (ClinicalTrials.gov: NCT03003533). However, high doses in patients resulted in severe adverse events and subsequent loss of factor VIII (FVIII) expression. Thus, additional strategies are needed to enhance the transduction efficiency of capsid library-derived rAAV vectors such that improved clinical efficacy can be achieved at low vector doses. In this study, we characterized two commonly used library-derived rAAV vectors, rAAV-DJ and rAAV-LK03. It was concluded that rAAV-DJ shared similar transport pathways (e.g., cell surface binding, endocytosis-dependent internalization, and cytoplasmic trafficking) with rAAV serotype 2, while rAAV-LK03 and rAAV serotype 3 shared similar transport pathways. We then performed site-directed mutagenesis of surface-exposed tyrosine (Y), serine (S), aspartic acid (D), and tryptophan (W) residues on rAAV-DJ and rAAV-LK03 capsids. Our results demonstrated that rAAV-DJ-S269T and rAAV-LK03-Y705+731F variants had significantly enhanced transduction efficiency compared to wild-type counterparts. Our studies suggest that the strategy of site-directed mutagenesis should be applicable to other non-natural AAV variants for their optimal use in human gene therapy.
Collapse
Affiliation(s)
- Gai Ran
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Xiao Chen
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yilin Xie
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Qingyun Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Jinyan Xie
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Chenghui Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Nikea Pittman
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Sixian Qi
- Institute of Pediatrics, Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Arun Srivastava
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Genetics Institute, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA
- Department of Molecular Genetics and Microbiology
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Chen Ling
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32611, USA
| |
Collapse
|
33
|
Using Traditional Chinese Medicine to Treat Hepatocellular Carcinoma by Targeting Tumor Immunity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9843486. [PMID: 32595757 PMCID: PMC7305542 DOI: 10.1155/2020/9843486] [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: 02/28/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
As the leading cause of cancer-related death, hepatocellular carcinoma (HCC) threatens human health and limited treatments are available to cure the disease efficiently and effectively. The particularly immunotolerant environment of the liver lowers the efficacy of current therapies in patients with advanced HCC. Traditional Chinese medicine (TCM) is gathering increasing interest due to the immunoregulatory properties of certain compounds. In advanced HCC, TCM can restore immunosurveillance to promote antitumor effects in several ways, including the upregulation of immunostimulatory factors and the downregulation of immunosuppressive factors. The characteristic multitarget regulation of TCM compounds may provide new insights regarding effective HCC immunotherapies. Here, we review the immunoregulatory potency of TCMs for treating HCC and explain how individual TCM drugs and complex formulas remodel the immune environment in various cell- and cytokine-dependent manners.
Collapse
|