1
|
Wu C, Gao Y, Jin Z, Huang Z, Wang H, Lu S, Guo S, Zhang F, Zhang J, Huang J, Tao X, Liu X, Zhang X, You L, Li Q, Wu J. PTPRG-AS1 regulates the KITLG/KIT pathway through the ceRNA axis to promote the malignant progression of gastric cancer and the intervention effect of Compound Kushen injection on it. Pharmacol Res 2025; 215:107743. [PMID: 40250508 DOI: 10.1016/j.phrs.2025.107743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Gastric cancer (GC) is a common malignant tumor with high mortality, recurrence, and metastasis rates. Compound Kushen injection (CKI) combination chemotherapy has been clinically used for the treatment of GC in China for many years, but its underlying mechanisms of action remain unclear. Recent reports have highlighted the important role of the competing endogenous RNA (ceRNA) mechanism of noncoding RNA (ncRNA) and messenger RNA (mRNA) formation in GC and other tumors. This study aimed to investigate the effects of CKI on GC from the ceRNA perspective. We confirmed the inhibitory effect of CKI on GC in mouse models and cell lines. By examining the GC cell lines sensitive to CKI treatment, we developed the CNScore method to analyze the ceRNA network, revealing that the CKI-GC ceRNA network promotes GC proliferation and metastasis through the PTPRG-AS1/hsa-miR-421/KITLG axis. Finally, we constructed GC cell models with PTPRG-AS1 overexpression or knockdown and GC liver metastasis models and found that PTPRG-AS1 can sponge hsa-miR-421, releasing KITLG and promoting GC proliferation and metastasis through the KITLG/KIT pathway. Taken together, CKI can suppress these malignant phenotypes by regulating the PTPRG-AS1/hsa-miR-421/KITLG axis.
Collapse
Affiliation(s)
- Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yifei Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhengsen Jin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Haojia Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fanqin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaqi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoyu Tao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinkui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Leiming You
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Qinglin Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang Province 310022, China.
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
2
|
Xu T, Chakraborty S, Wei D, Tran M, Rhea R, Wei B, Nguyen P, Gagea M, Xie X, Wu L, Cohen L, Liao Z, Yang P. Evaluation of the protective effect of Compound Kushen Injection against radiation‑induced lung injury in mice. Mol Med Rep 2025; 31:88. [PMID: 39917996 PMCID: PMC11831882 DOI: 10.3892/mmr.2025.13453] [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: 02/16/2024] [Accepted: 11/07/2024] [Indexed: 02/19/2025] Open
Abstract
Radiation‑induced lung injury (RILI) is a prevalent complication following thoracic radiation, and currently there is a lack of effective intervention options. The present study investigated the potential of Compound Kushen Injection (CKI), a botanical drug, to mitigate inflammatory responses in mice with RILI, along with its underlying mechanisms of action. C3H mice underwent total lung irradiation (TLI) and intraperitoneal injection of CKI (2, 4 or 8 ml/kg) once daily for 8 weeks. Pre‑radiation treatment with 4 or 8 ml/kg CKI starting 2 weeks before TLI or concurrent treatment of 8 ml/kg CKI with TLI led to a significantly longer overall survival compared with the TLI vehicle‑treated group. Micro‑computed tomography evaluations showed that concurrent treatment with 8 ml/kg CKI was associated with a significantly lower incidence of RILI. Histological evaluations revealed that concurrent CKI (4 and 8 ml/kg) treatment significantly reduced grades of lung inflammation. Following radiation at 72 h, TLI plus vehicle‑treated mice had significantly elevated serum IL6, IL17A, and transforming growth factor β (TGF‑β) levels compared with non‑irradiated normal mice. Conversely, mice that received TLI plus CKI displayed lower cytokine levels than those in the TLI plus vehicle‑treated mice. Immunohistochemistry staining showed a reduction of TGF‑β positive cells in the lung tissues of TLI mice after CKI treatment. The concurrent TLI CKI‑treated mice had a significantly reduced cyclooxygenase 2 (COX‑2) activity and COX‑2 metabolites compared with TLI vehicle‑treated mice. These data highlight that CKI substantially reduced radiation‑induced lung inflammation, mitigated RILI incidence, and prolonged overall survival.
Collapse
Affiliation(s)
- Ting Xu
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sharmistha Chakraborty
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daoyan Wei
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Megan Tran
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robyn Rhea
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bo Wei
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Phuong Nguyen
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mihai Gagea
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoxue Xie
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lirong Wu
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lorenzo Cohen
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhongxing Liao
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Peiying Yang
- Department of Palliative, Rehabilitation and Integrative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
3
|
Gao K, Liu L, Lei S, Li Z, Huo P, Wang Z, Dong L, Deng W, Bu D, Zeng X, Li C, Zhao Y, Zhang W, Wang W, Wu Y. HERB 2.0: an updated database integrating clinical and experimental evidence for traditional Chinese medicine. Nucleic Acids Res 2025; 53:D1404-D1414. [PMID: 39558177 PMCID: PMC11701625 DOI: 10.1093/nar/gkae1037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
Clinical trials and meta-analyses are considered high-level medical evidence with solid credibility. However, such clinical evidence for traditional Chinese medicine (TCM) is scattered, requiring a unified entrance to navigate all available evaluations on TCM therapies under modern standards. Besides, novel experimental evidence has continuously accumulated for TCM since the publication of HERB 1.0. Therefore, we updated the HERB database to integrate four types of evidence for TCM: (i) we curated 8558 clinical trials and 8032 meta-analyses information for TCM and extracted clear clinical conclusions for 1941 clinical trials and 593 meta-analyses with companion supporting papers. (ii) we updated experimental evidence for TCM, increased the number of high-throughput experiments to 2231, and curated references to 6 644. We newly added high-throughput experiments for 376 diseases and evaluated all pairwise similarities among TCM herbs/ingredients/formulae, modern drugs and diseases. (iii) we provide an automatic analyzing interface for users to upload their gene expression profiles and map them to our curated datasets. (iv) we built knowledge graph representations of HERB entities and relationships to retrieve TCM knowledge better. In summary, HERB 2.0 represents rich data type, content, utilization, and visualization improvements to support TCM research and guide modern drug discovery. It is accessible through http://herb.ac.cn/v2 or http://47.92.70.12.
Collapse
Affiliation(s)
- Kai Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo 315010, China
| | - Liu Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Shuangshuang Lei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Zhinong Li
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Peipei Huo
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhihao Wang
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Lei Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Wenxin Deng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
| | - Dechao Bu
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Chun Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yi Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing 100029, China
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Wei Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yang Wu
- Key Laboratory of Intelligent Information Processing, Research Center for Ubiquitous Computing Systems, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
4
|
Lv M, Feng Y, Zeng S, Zhang Y, Shen W, Guan W, E X, Zeng H, Zhao R, Yu J. Network pharmacology in combination with bibliometrics analysis on the mechanism of compound Kushen injection in the treatment of radiation pneumonia and lung cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9789-9809. [PMID: 38918234 DOI: 10.1007/s00210-024-03238-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024]
Abstract
Radiation pneumonia is a common adverse reaction during radiotherapy in lung cancer patients, which negatively impacts the quality of life and survival of patients. Recent studies have shown that compound Kushen injection (CKI), a traditional Chinese medicine (TCM), has great anti-inflammatory and anticancer potential, but the mechanism is still unclear. We used CiteSpace, the R package "bibliometrix," and VOSviewers to perform a bibliometrics analysis of 162 articles included from the Web of Science core collection. A network pharmacology-based approach was used to screen effective compounds, screen and predict target genes, analyze biological functions and pathways, and construct regulatory networks and protein interaction networks. Molecular docking experiments were used to identify the affinity of key compounds and core target. The literature metrology analysis revealed that over 90% of the CKI-related studies were conducted by Chinese scholars and institutions, with a predominant focus on tumors, while research on radiation pneumonia remained limited. Our investigation identified 60 active ingredients of CKI, 292 genes associated with radiation pneumonia, 533 genes linked to lung cancer, and 37 common targets of CKI in the treatment of both radiation pneumonia and lung cancer. These core potential targets were found to be significantly associated with the OS of lung cancer patients, and the key compounds exhibited a good docking affinity with these targets. Additionally, GO and KEGG enrichment analysis highlighted that the bioinformatics annotation of these common genes mainly involved ubiquitin protein ligase binding, cytokine receptor binding, and the PI3K/Akt signaling pathway. Our study revealed that the main active components of CKI, primarily quercetin, luteolin, and naringin, might act on major core targets, including AKT1, PTGS2, and PPARG, and further regulated key signaling pathways such as the PI3K/Akt pathway, thereby playing a crucial role in the treatment of radiation pneumonia and lung cancer. Moreover, this study had a certain promotional effect on further clinical application and provided a theoretical basis for subsequent experimental research.
Collapse
Affiliation(s)
- Minghe Lv
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Yue Feng
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Su Zeng
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Yang Zhang
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Wenhao Shen
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Wenhui Guan
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Xiangyu E
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China
| | - Hongwei Zeng
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China.
| | - Ruping Zhao
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China.
| | - Jingping Yu
- Department of Radiotherapy, Shuguang Hospital, Affiliated to Shanghai University of Chinese Traditional Medicine, Zhang Heng Road, Pudong New Area, Shanghai, 201203, China.
| |
Collapse
|
5
|
Li Y, Dong J, Qin JJ. Small molecule inhibitors targeting heat shock protein 90: An updated review. Eur J Med Chem 2024; 275:116562. [PMID: 38865742 DOI: 10.1016/j.ejmech.2024.116562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/10/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024]
Abstract
As a molecular chaperone, heat shock protein 90 (HSP90) plays important roles in the folding, stabilization, activation, and degradation of over 500 client proteins, and is extensively involved in cell signaling, proliferation, and survival. Thus, it has emerged as an important target in a variety of diseases, including cancer, neurodegenerative diseases, and viral infections. Therefore, targeted inhibition of HSP90 provides a valuable and promising therapeutic strategy for the treatment of HSP90-related diseases. This review aims to systematically summarize the progress of research on HSP90 inhibitors in the last five years, focusing on their structural features, design strategies, and biological activities. It will refer to the natural products and their derivatives (including novobiocin derivatives, deguelin derivatives, quinone derivatives, and terpenoid derivatives), and to synthetic small molecules (including resorcinol derivatives, pyrazoles derivatives, triazole derivatives, pyrimidine derivatives, benzamide derivatives, benzothiazole derivatives, and benzofuran derivatives). In addition, the major HSP90 small-molecule inhibitors that have moved into clinical trials to date are also presented here.
Collapse
Affiliation(s)
- Yulong Li
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinyun Dong
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Jiang-Jiang Qin
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
6
|
Cui B, Cheng X, Zhang X, Chen L, Pang W, Liu Y, Yang Z, Li H, He X, Li X, Bi X. Anti-cancer activity and mechanism of flurbiprofen organoselenium compound RY-1-92 in non-small cell lung cancer. RSC Med Chem 2024; 15:1737-1745. [PMID: 38784458 PMCID: PMC11110739 DOI: 10.1039/d4md00058g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/15/2024] [Indexed: 05/25/2024] Open
Abstract
Lung cancer is one of the malignancies with the highest incidence and mortality rates worldwide, and non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer types. In this study, the anti-cancer activities of a novel flurbiprofen organic selenium compound, RY-1-92, on NSCLC cells and a mouse model and the underlying molecular mechanisms were explored. We found that compound RY-1-92 can significantly inhibit the viability, colony formation and migration of A549, NCI-H460 lung cancer cells. Flow cytometry analysis showed that RY-1-92 also can lead to G2/M cell cycle arrest and apoptosis induced in lung cancer cells. Further, RY-1-92 can decrease the tumor size in the Lewis lung cancer tumor-bearing mouse model. The protein levels of cell cycle-related proteins CDK1/cyclinB1 were decreased, while the apoptosis-related protein BAX was increased dramatically after RY-1-92 treatment in vitro and in vivo. Impressively, it was found that TRPV1 might act as a potential molecular target of RY-1-92 using the SEA search server. Furthermore, down-regulation on TRPV1 and its downstream associated factors including p-AKT protein and MAPK signaling pathway-related proteins after RY-1-92 treatment was observed in A549, NCI-H460 lung cancer cells. Taken together, our findings shed light on the potential of RY-1-92 as a novel small molecular drug for NSCLC, and it is of great significance for its further in-depth research and development.
Collapse
Affiliation(s)
- Bo Cui
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Xianda Cheng
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Xin Zhang
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Lili Chen
- College of Life Science, Liaoning University Shenyang 110036 China
- Shenyang Key Laboratory of Chronic Disease Occurrence and Nutrition Intervention, College of Life Sciences, Liaoning University Shenyang 110036 China
- College of Mathematics and Statistics, Liaoning University Shenyang 110036 China
| | - Wenqian Pang
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Yue Liu
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Zhe Yang
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Hui Li
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Xianran He
- Institute for Interdisciplinary Research, Jianghan University Wuhan Economic and Technological Development Zone Wuhan 430056 China
| | - Xiaolong Li
- Shenzhen Fushan Biological Technology Co., Ltd Kexing Science Park A1 1005, Nanshan Zone Shenzhen 518057 China
| | - Xiuli Bi
- College of Life Science, Liaoning University Shenyang 110036 China
- Shenyang Key Laboratory of Chronic Disease Occurrence and Nutrition Intervention, College of Life Sciences, Liaoning University Shenyang 110036 China
| |
Collapse
|
7
|
Qiu G, Li F, Kowah JAH, Xie J, Long Q, Wang L, Liu X. Novel chiral matrine derivatives as potential antitumor agents: Design, synthesis and biological evaluation. Bioorg Chem 2024; 146:107276. [PMID: 38479132 DOI: 10.1016/j.bioorg.2024.107276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024]
Abstract
Since the thalidomide incident, research on chiral drugs has escalated immensely. Differences in drug configuration can lead to significant variations in therapeutic efficacy. Matrine, a natural product esteemed for its low toxicity and high water solubility, has garnered significant attention in research endeavors. Nonetheless, its precise target has proven elusive. In this study, we designed and synthesized a novel chiral matrine derivative. Their cytotoxicity against three types of tumor cells was assessed. Comparing the newly synthesized derivatives to the parent matrine, most compounds exhibited significantly enhanced inhibitory effects on cancer cells. Among them, Q12 exhibited the highest activity, with IC50 values of 8.31 μM against rat glioma cells C6, 6.3 μM against human liver cancer cells HepG2 and 7.14 μM against human gastric cancer cells HGC-27, meanwhile showing low toxicity. Based on IC50 values, we constructed a preliminary structure-activity relationship (SAR). Compound Q12 significantly suppressed the cloning and migration of HepG2 cells. Further mechanistic studies indicated that Q12 inhibited Topo I in HepG2 cells, leading to DNA damage, induction of G0/G1 cell cycle arrest and ultimately causing apoptosis. The molecular docking experiments provided a rational binding mode of Q12 with the Topo I-DNA complex. In vivo, experiments demonstrated that Q12 exhibited a higher tumor growth inhibition rate (TGI) compared to the positive control drug Lenvatinib, while maintaining good safety. In summary, it suggests that Topo I might be a potential target for matrine and Q12 represents a promising candidate for cancer treatment.
Collapse
Affiliation(s)
- Gan Qiu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Fan Li
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Jamal A H Kowah
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Junwei Xie
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Qingfeng Long
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Lisheng Wang
- School of Medicine, Guangxi University, Nanning 530004, China.
| | - Xu Liu
- School of Medicine, Guangxi University, Nanning 530004, China.
| |
Collapse
|
8
|
Qin L, Zhong Y, Li Y, Yang Y. TCM targets ferroptosis: potential treatments for cancer. Front Pharmacol 2024; 15:1360030. [PMID: 38738174 PMCID: PMC11082647 DOI: 10.3389/fphar.2024.1360030] [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: 12/22/2023] [Accepted: 04/08/2024] [Indexed: 05/14/2024] Open
Abstract
Ferroptosis is caused by the accumulation of cellular reactive oxygen species that exceed the antioxidant load that glutathione (GSH) and phospholipid hydroperoxidases with GSH-based substrates can carry When the antioxidant capacity of cells is reduced, lipid reactive oxygen species accumulate, which can cause oxidative death. Ferroptosis, an iron-dependent regulatory necrosis pathway, has emerged as a new modality of cell death that is strongly associated with cancer. Surgery, chemotherapy and radiotherapy are the main methods of cancer treatment. However, resistance to these mainstream anticancer drugs and strong toxic side effects have forced the development of alternative treatments with high efficiency and low toxicity. In recent years, an increasing number of studies have shown that traditional Chinese medicines (TCMs), especially herbs or herbal extracts, can inhibit tumor cell growth and metastasis by inducing ferroptosis, suggesting that they could be promising agents for cancer treatment. This article reviews the current research progress on the antitumor effects of TCMs through the induction of ferroptosis. The aim of these studies was to elucidate the potential mechanisms of targeting ferroptosis in cancer, and the findings could lead to new directions and reference values for developing better cancer treatment strategies.
Collapse
Affiliation(s)
- Liwen Qin
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Yuhan Zhong
- Laboratory of Liver Transplantation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Precision Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yongfeng Yang
- Department of Respiratory and Critical Care Medicine, Institute of Respiratory Health, Center of Precision Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Samuels N, Ben-Arye E. Integrative Medicine for Cancer-Related Pain: A Narrative Review. Healthcare (Basel) 2024; 12:403. [PMID: 38338288 PMCID: PMC10855884 DOI: 10.3390/healthcare12030403] [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: 11/13/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Cancer-related pain (C-RP) is a prevalent and debilitating concern among patients with cancer, with conventional treatments limited in their ability to provide adequate relief, and by the adverse effects associated with their use. Complementary and integrative medicine (CIM) modalities have been shown to be potentially effective and safe for the treatment of pain and related symptoms, when used in conjunction with conventional medications and under medical supervision. An increasing number of oncology centers provide CIM within their conventional supportive and palliative care service, in an "Integrative Oncology" (IO) setting. A large body of clinical research, including systematic reviews and guidelines such as those published in 2022 by the Society for Integrative Oncology (SIO), in collaboration with the American Society for Clinical Oncology (ASCO), support the use of some CIM modalities for C-RP and related concerns. These include acupuncture for general and peri-operative/procedural pain, as well as aromatase inhibitor-associated arthralgia (AIA); reflexology or acupressure for pain during systemic therapy for cancer; hypnosis for procedural pain or pain due to diagnostic workup; and massage for pain experienced by patients during palliative and hospice care. Further research is needed, within both randomized control trials and pragmatic non-controlled studies which are more reflective of the real-life IO setting. This review summarizes the evidence supporting the use of CIM for C-RP; the analgesic mechanism of the modalities presented; and the challenges facing IO researchers, as well as the implementation of the 2022 SIO-ASCO guideline recommendations.
Collapse
Affiliation(s)
- Noah Samuels
- Center for Integrative Complementary Medicine, Shaare Zedek Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| | - Eran Ben-Arye
- Integrative Oncology Program, The Oncology Service, Lin Carmel, and Zebulun Medical Centers, Clalit Health Services, Haifa 3535152, Israel;
- Department of Family Medicine, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
10
|
Xu T, Chakraborty S, Wei D, Tran M, Rhea R, Wei B, Nguyen P, Gagea M, Cohen L, Liao Z, Yang P. Evaluation of the Protective Effect of Compound Kushen Injection Against Radiation- induced Pneumonitis in Mice. RESEARCH SQUARE 2024:rs.3.rs-3880937. [PMID: 38352564 PMCID: PMC10862984 DOI: 10.21203/rs.3.rs-3880937/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Background Radiation-induced lung injury (RILI) via inflammation is a common adverse effect of thoracic radiation that negatively impacts patient quality of life and survival. Compound kushen injection (CKI), a botanical drug treatment, was examined for its ability to reduce RILI, and inflammatory responses and improve survival in mice exposed total lung irradiation (TLI). CKI's specific mechanisms of action were also evaluated. Methods C3H mice underwent TLI and were treated with CKI (2, 4, or 8 mL/kg) intraperitoneally once a day for 8 weeks. The effects of CKI on survival were estimated by Kaplan-Meier survival analysis and compared by log-rank test. RILI damage was evaluated by histopathology and micro-computed tomography (CT). Inflammatory cytokines and cyclooxygenase metabolites were examined by IHC staining, western blot, and ELISA. Results Pre-irradiation treatment with 4 or 8 mL/kg CKI starting 2 weeks before TLI or concurrent treatment with 8 mL/kg CKI were associated with a significantly longer survival compared with TLI vehicle-treated group ( P < 0.05). Micro-CT images evaluations showed that concurrent treatment with 8 mL/kg CKI was associated with significantly lower incidence of RILI ( P < 0.05). Histological evaluations revealed that concurrent TLI treatment of CKI (4 and 8 mL/kg) significantly reduced lung inflammation (p < 0.05). Mechanistic investigation showed that at 72 hours after radiation, TLI plus vehicle mice had significantly elevated serum IL6, IL17A, and TGF-β levels compared with non-irradiated, age-matched normal mice; in contrast, levels of these cytokines in mice that received TLI plus CKI treatment were lower than those in the TLI plus vehicle-treated mice ( P < 0.05) and similar to the nonirradiated mice. IHC staining showed that the CKI treatment led to a reduction of TGF-β positive cells in the lung tissues of TLI mice (P < 0.01). The concurrent CKI with TLI treatment group had a significant reduction in COX-2 activity and COX-2 metabolites compared with the TLI vehicle-treated group ( P < 0.05). Conclusions These data suggest that CKI treatment was associated with reduced radiation-induced inflammation in lung tissues, reduced RILI, and improved survival. Further investigation of CKI in human clinical trials as a potential radioprotector against RILI to improve patients' quality of life and survival is warranted.
Collapse
|
11
|
Lu T, Kong B, Wang Y, Yu J, Pan Y, Chen D, Li H, Chen X, Yuan Z, Yang Z, Zhang J, Ding T, Zhang G, Fan Q, Wang X. Compound Kushen injection combined with transarterial chemoembolization for hepatocellular carcinoma: An evidence map and overview of systematic reviews. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117267. [PMID: 37838291 DOI: 10.1016/j.jep.2023.117267] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE For the treatment of hepatocellular carcinoma (HCC), compound Kushen injection (CKi) is commonly used in combination with transarterial chemoembolization (TACE). AIMS OF THE STUDY Our objective was to evaluate the reporting quality, methodological quality, risk of bias, and certainty of evidence for CKi combined with TACE for the treatment of patients with HCC by conducting systematic reviews (SRs). The purpose of this study was to improve the clinical application of CKis, strengthen clinical decision-making regarding CKis, and inform future research. MATERIALS AND METHODS We used eight databases to systematically search SRs of CKi combined with TACE for HCC through February 21, 2023. The quality of reporting of SRs was evaluated using the 2009 Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, methodological quality using A MeaSurement Tool to Assess systematic Reviews 2, risk of bias using the Risk of Bias in Systematic Review, and certainty of evidence using the Grading of Recommendations Assessment. Finally, the assessment results were visualized by the evidence mapping method. This overview has been registered on PROSPERO with the registration title "Compound Kushen injection for hepatocellular carcinoma: An overview of systematic reviews" and registration number CRD42022369120. RESULTS A total of 12 SRs meeting the inclusion criteria were included. In terms of reporting quality, 42% of SRs reported relatively complete reports and 58% had certain deficiencies. The methodological quality of all SRs was " critically low". The risk of bias was evaluated as low in 33% of SRs and high in 67% of SRs. The results of the evidence synthesis showed that, in the "moderate" level of evidence, CKi combined with TACE resulted in a 12.7%-21.5% benefit for one-year survival rate, 11.7%-17.2% benefit for objective response rate (ORR), 20.5%-27.1% benefit for quality of life, 22.2% benefit for nausea and vomiting, and 24.7%-27.4% benefit for leukopenia in HCC patients. CONCLUSION In conclusion, CKi combined with TACE improved survival, ORR and quality of life in patients with HCC, and reduced adverse events. The results should be interpreted with caution due to the low methodological quality of the included SRs. The clinical efficacy of CKis must be confirmed in a large number of randomized controlled trials.
Collapse
Affiliation(s)
- Taicheng Lu
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Bingtan Kong
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Yue Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Jingwen Yu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yuancan Pan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Dong Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Haiming Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xing Chen
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Zichun Yuan
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Zhengzheng Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Jiahui Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Tongjing Ding
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Ganlin Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| | - Qingsheng Fan
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| | - Xiaomin Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| |
Collapse
|
12
|
Li Y, Lu Y, Nian M, Sheng Q, Zhang C, Han C, Dou X, Ding Y. Therapeutic potential and mechanism of Chinese herbal medicines in treating fibrotic liver disease. Chin J Nat Med 2023; 21:643-657. [PMID: 37777315 DOI: 10.1016/s1875-5364(23)60443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Indexed: 10/02/2023]
Abstract
Liver fibrosis is a pathological condition characterized by replacement of normal liver tissue with scar tissue, and also the leading cause of liver-related death worldwide. During the treatment of liver fibrosis, in addition to antiviral therapy or removal of inducers, there remains a lack of specific and effective treatment strategies. For thousands of years, Chinese herbal medicines (CHMs) have been widely used to treat liver fibrosis in clinical setting. CHMs are effective for liver fibrosis, though its mechanisms of action are unclear. In recent years, many studies have attempted to determine the possible mechanisms of action of CHMs in treating liver fibrosis. There have been substantial improvements in the experimental investigation of CHMs which have greatly promoted the understanding of anti-liver fibrosis mechanisms. In this review, the role of CHMs in the treatment of liver fibrosis is described, based on studies over the past decade, which has addressed the various mechanisms and signaling pathways that mediate therapeutic efficacy. Among them, inhibition of stellate cell activation is identified as the most common mechanism. This article provides insights into the research direction of CHMs, in order to expand its clinical application range and improve its effectiveness.
Collapse
Affiliation(s)
- Yanwei Li
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yunrui Lu
- Liaoning University of Traditional Chinese Medicine, Shenyang 110000, China
| | - Mozuo Nian
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Qiuju Sheng
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Chong Zhang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Chao Han
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Xiaoguang Dou
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
13
|
Qu Z, Wang W, Adelson DL. Chromosomal level genome assembly of medicinal plant Sophora flavescens. Sci Data 2023; 10:572. [PMID: 37644152 PMCID: PMC10465603 DOI: 10.1038/s41597-023-02490-8] [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: 06/02/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Sophora flavescens is a medicinal plant in the genus Sophora of the Fabaceae family. The root of S. flavescens is known in China as Kushen and has a long history of wide use in multiple formulations of Traditional Chinese Medicine (TCM). In this study, we used third-generation Nanopore long-read sequencing technology combined with Hi-C scaffolding technology to de novo assemble the S. flavescens genome. We obtained a chromosomal level high-quality S. flavescens draft genome. The draft genome size is approximately 2.08 Gb, with more than 80% annotated as Transposable Elements (TEs), which have recently and rapidly proliferated. This genome size is ~5x larger than its closest sequenced relative Lupinus albus L. . We annotated 60,485 genes and examined their expression profiles in leaf, stem and root tissues, and also characterised the genes and pathways involved in the biosynthesis of major bioactive compounds, including alkaloids, flavonoids and isoflavonoids. The assembled genome highlights the very different evolutionary trajectories that have occurred in recently diverged Fabaceae, leading to smaller duplicated genomes.
Collapse
Affiliation(s)
- Zhipeng Qu
- Zhendong Center, Department of Molecular and Biomedical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
| | - Wei Wang
- Beijing Zhendong Research Institute, Shanxi Zhendong Pharmaceutical Co Ltd, Beijing, 10587, China
- Shanxi Provincial Key Laboratory of Functional Food with Homology of Medicine and Food, Department of Pharmacy, Changzhi Medical College, Changzhi, 046012, China
| | - David L Adelson
- Zhendong Center, Department of Molecular and Biomedical Sciences, The University of Adelaide, Adelaide, 5005, Australia.
- South Australian Museum, Adelaide, 5000, Australia.
| |
Collapse
|
14
|
Shang W, Ye A, Tong YK. Sub-Cellular Dynamic Analysis of BGC823 Cells after Treatment with the Multi-Component Drug CKI Using Raman Spectroscopy. Int J Mol Sci 2023; 24:12750. [PMID: 37628931 PMCID: PMC10454546 DOI: 10.3390/ijms241612750] [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: 06/26/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Multi-component drugs (MCDs) can induce various cellular changes covering multiple levels, from molecular and subcellular structure to cell morphology. A "non-invasive" method for comprehensively detecting the dynamic changes of cellular fine structure and chemical components on the subcellular level is highly desirable for MCD studies. In this study, the subcellular dynamic processes of gastric cancer BGC823 cells after treatment with a multi-component drug, Compound Kushen Injection (CKI), were investigated using a homemade, high-resolution, confocal Raman spectroscopy (RS) device combined with bright-field imaging. The Raman spectra of the nucleus, cytoplasm and intracellular vesicles (0.4-1 μm) were collected simultaneously for each cell treated with CKI at different times and doses. The RS measurements showed that CKI decreased the DNA signatures, which the drug is known to inhibit. Meanwhile, the CKI-induced subcellular dynamic changes in the appearance of numerous intracellular vesicles and the deconstruction of cytoplasm components were observed and discussed. The results demonstrated that high-resolution subcellular micro-Raman spectroscopy has potential for detecting fine cellular dynamic variation induced by drugs and the screening of MCDs in cancer therapy.
Collapse
Affiliation(s)
- Wenhao Shang
- Key Laboratory for the Physics and Chemistry of Nanodevices, School of Electronics, Peking University, Beijing 100871, China
- Biomed-X Center, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Anpei Ye
- Key Laboratory for the Physics and Chemistry of Nanodevices, School of Electronics, Peking University, Beijing 100871, China
- Biomed-X Center, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yu-Kai Tong
- Key Laboratory for the Physics and Chemistry of Nanodevices, School of Electronics, Peking University, Beijing 100871, China
| |
Collapse
|
15
|
Huang Z, Wu C, Zhou W, Lu S, Tan Y, Wu Z, You R, Stalin A, Guo F, Zhang J, Liu P, Wang W, Duan X, You L, Wu J. Compound Kushen Injection inhibits epithelial-mesenchymal transition of gastric carcinoma by regulating VCAM1 induced by the TNF signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 118:154984. [PMID: 37487253 DOI: 10.1016/j.phymed.2023.154984] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/04/2023] [Accepted: 07/15/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Gastric carcinoma (GC) treatment needs to be developed rapidly. Compound Kushen Injection (CKI), a formula from traditional Chinese medicine, has been used clinically in combination with chemotherapy to treat GC with satisfactory results. However, the molecular mechanism by which CKI acts to cure GC is still unclear. METHODS In the present study, in vivo and in vitro experiments were used to assess the efficacy of CKI. Using ceRNA microarray and TMT technologies, the molecular mechanism of CKI was further investigated at the transcriptional and protein levels, and a bioinformatics approach was employed to investigate and functionally validate key CKI targets in GC. RESULTS When combined with cisplatin (DDP), CKI significantly increased its efficacy in preventing the proliferation and metastasis of GC cells and malignant-looking tumors in mice. High-throughput sequencing data and bioinformatics analysis showed that CKI regulated the TNF signaling pathway, epithelial-mesenchymal transition (EMT), with VCAM1 as a key target. The transcription factors CEBPB, JUN, RELA, NFKB1, the EMT mesenchymal-like cell markers N-cadherin and vimentin, as well as the expression of VCAM1 and its upstream signaling driver TNF, were all downregulated by CKI. In contrast, the expression of the EMT epithelial-like cell marker E-cadherin was upregulated. CONCLUSION CKI can effectively inhibit GC growth and metastasis, improve body's immunity, and protect normal tissues from damage. The molecular mechanism by which CKI inhibits metastasis of GC is by regulating VCAM1 induced by the TNF signaling pathway to inhibit EMT of GC. Our results provide an important clue to clarify precisely the multi-scale molecular mechanism of CKI in the treatment of GC.
Collapse
Affiliation(s)
- Zhihong Huang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chao Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Zhou
- Department of pharmacy, China-Japan Friendship Hospital, Beijing 100029, China
| | - Shan Lu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yingying Tan
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhishan Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Rongli You
- Shanxi Zhendong Pharmaceutical Co., Ltd., Shanxi 47100, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Fengying Guo
- School of Management, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingyuan Zhang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Pengyun Liu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- Shanxi Zhendong Pharmaceutical Co., Ltd., Shanxi 47100, China
| | - Xiaoxia Duan
- Beijing Zestbridge Medical Technology Co., Ltd., Beijing, 100176, China.
| | - Leiming You
- Department of Immunology and Microbiology, School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Jiarui Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
16
|
Wang W, Liu D, Yang L, Chen L, Miao M, Liu Y, Yin Y, Wei M, Liu G, An Y, Zheng M. Compound Kushen injection attenuates angiotensin II‑mediated heart failure by inhibiting the PI3K/Akt pathway. Int J Mol Med 2023; 51:23. [PMID: 36734284 PMCID: PMC9943540 DOI: 10.3892/ijmm.2023.5226] [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: 11/03/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Compound Kushen injection (CKI) is a type of traditional Chinese medicine that has previously been studied for the treatment of various types of cancer. Previous studies have reported that CKI regulates cell apoptosis by downregulating the PI3K/Akt pathway. The present study aimed to determine whether CKI alleviates heart failure (HF) by attenuating cardiomyocyte apoptosis via the inhibition of the PI3K/Akt pathway. Angiotensin II (Ang II) was used to elicit HF, and osmotic minipumps with either Ang II (2 µg/kg/day) or phosphate‑buffered saline (PBS; 200 µl) were subcutaneously implanted into 6‑week‑old male C57BL/6 mice for 3 weeks. In addition, PBS or CKI (25 mg/kg/day) were subcutaneously infused once a day for 3 weeks. Echocardiography was used to examine hemodynamics. The myocardial injury biomarkers, cardiac troponin I and N‑terminal (NT)‑pro hormone B‑type natriuretic peptide, were assessed using enzyme‑linked immunosorbent assay. Transmission electron microscopy was used to determine the morphology of the myocardium. The rate of apoptosis was detected using TUNEL staining and flow cytometry (FCM), and the expression levels of apoptosis‑related proteins were measured using western blot (WB) analysis. Moreover, H9C2 cells were treated with CKI (2 mg/ml) or LY294002 (an inhibitor of the PI3K/Akt pathway; 25 µmol/l) in combination with Ang II (1 µmol/l) for 48 h. Cell Counting Kit‑8 assay, FCM and WB analysis were performed in the H9C2 cells to examine cell viability, cell cycle distribution and representative signaling proteins. It was found that CKI promoted healthy cardiac function, reduced myocardial structural damage and reduced the rate of cardiomyocyte apoptosis. CKI markedly attenuated the expression of apoptosis‑related proteins in the PI3K/Akt pathway. The results of the in vitro experiments indicated that CKI promoted cardiomyocyte proliferation and inhibited apoptosis, similar to LY294002. On the whole, the present study demonstrates that CKI reduces cardiomyocyte apoptosis, promotes healthy cardiac function and attenuates Ang II‑mediated HF. These ameliorative effects may be associated with the inhibition of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Wei Wang
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Da Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Liyun Yang
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lixia Chen
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Mengdan Miao
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Yongsheng Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Mei Wei
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Yonghui An
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China,Professor Yonghui An, Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Yuhua, Shijiazhuang, Hebei 050031, P.R. China, E-mail:
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China,Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, Hebei 050000, P.R. China,Correspondence to: Professor Mingqi Zheng, Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Yuhua, Shijiazhuang, Hebei 050031, P.R. China, E-mail:
| |
Collapse
|
17
|
Yu HB, Hu JQ, Han BJ, Cao HJ, Chen ST, Chen X, Xiong HT, Gao J, Du YY, Zheng HG. Evaluation of efficacy and safety for compound kushen injection combined with intraperitoneal chemotherapy for patients with malignant ascites: A systematic review and meta-analysis. Front Pharmacol 2023; 14:1036043. [PMID: 36937874 PMCID: PMC10020185 DOI: 10.3389/fphar.2023.1036043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 02/23/2023] [Indexed: 03/06/2023] Open
Abstract
Objectives: Compound Kushen injection (CKI) combined with intraperitoneal chemotherapy (IPC) is widely used in the treatment of malignant ascites (MA). However, evidence about its efficacy and safety remains limited. This review aimed to evaluate the efficacy and safety of CKI combined with IPC for the treatment of MA. Methods: Protocol of this review was registered in PROSPERO (CRD42022304259). Randomized controlled trials (RCTs) on the efficacy and safety of IPC with CKI for the treatment of patients with MA were searched through 12 electronic databases and 2 clinical trials registration platforms from inception until 20 January 2023. The Cochrane risk-of-bias tool was used to assess the quality of the included trials through the risk of bias assessment. We included RCTs that compared IPC single used or CKI combined with IPC for patients with MA schedule to start IPC. The primary outcome was identified as an objective response rate (ORR), while the secondary outcomes were identified as the quality of life (QoL), survival time, immune functions, and adverse drug reactions (ADRs). The Revman5.4 and Stata17 software were used to calculate the risk ratio (RR) at 95% confidence intervals (CI) for binary outcomes and the mean difference (MD) at 95% CI for continuous outcomes. The certainty of the evidence was assessed according to the GRADE criteria. Results: A total of 17 RCTs were assessed, which included 1200 patients. The risk of bias assessment of the Cochrane risk-of-bias tool revealed that one study was rated high risk and the remaining as unclear or low risk. Meta-analysis revealed that CKI combined with IPC had an advantage in increasing ORR (RR = 1.31, 95% CI 1.20 to 1.43, p < 0.00001) and QoL (RR = 1.50, 95% CI 1.23 to 1.83, p < 0.0001) when compared with IPC alone. Moreover, the combined treatment group showed a lower incidence of myelosuppression (RR = 0.51, 95%CI 0.40-0.64, p < 0.00001), liver dysfunction (RR = 0.33, 95%CI 0.16 to 0.70, p = 0.004), renal dysfunction (RR = 0.39, 95%CI 0.17 to 0.89, p = 0.02), and fever (RR = 0.51, 95%CI 0.35 to 0.75, p = 0.0007) compared to those of the control group. The quality of evidence assessment through GRADE criteria showed that ORR, myelosuppression, and fever were rated moderate, renal dysfunction and liver dysfunction were rated low, and QoL and abdominal pain were rated very low. Conclusion: The efficacy and safety of CKI combined with IPC were superior to that with IPC alone for the treatment of MA, which indicates the potentiality of the treatment. However, more high-quality RCTs are required to validate this conclusion. Systematic Review Registration: [https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022304259], identifier [PROSPERO 2022 CRD42022304259].
Collapse
Affiliation(s)
- Hui-Bo Yu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jia-Qi Hu
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Bao-Jin Han
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Hui-Juan Cao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shun-Tai Chen
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Chen
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hong-Tai Xiong
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jin Gao
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yan-Yuan Du
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hong-Gang Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Hong-Gang Zheng,
| |
Collapse
|
18
|
Wang X, Zhang S, Han K, Wang L, Liu X. Induction of Apoptosis by Matrine Derivative ZS17 in Human Hepatocellular Carcinoma BEL-7402 and HepG2 Cells through ROS-JNK-P53 Signalling Pathway Activation. Int J Mol Sci 2022; 23:ijms232415991. [PMID: 36555631 PMCID: PMC9783520 DOI: 10.3390/ijms232415991] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies and ranks third among cancer-related deaths worldwide. Using matrine as a lead compound, 12 matrine derivatives were designed and synthesised, and their antiproliferative activities were evaluated in four cancer cell lines. Eight of the twelve compounds showed strong antiproliferative activity, with an IC50 of <10 μM. The compound ZS17 exhibited strong antiproliferative activity in hepatocellular carcinoma cell lines with IC50 values in the range of 3.014−3.388 μM, which was much lower than that of matrine. Furthermore, we explored the role of ZS17 in inducing apoptosis in HCC cells in vitro and in vivo, as well as possible mechanisms involved. ZS17 inhibited the proliferation of BEL-7402 and HepG2 cells in time- and dose-dependent manners. In addition, we found that ZS17 significantly induced apoptosis and ROS (reactive oxygen species) production, promoted JNK phosphorylation, activated p53, and activated the caspase signalling pathway. Furthermore, the antioxidant NAC, JNK inhibitor SP600125, and Si-JNK increased cell viability, re-established cell metastasis, and inhibited ZS17-induced apoptosis. An in vivo antitumour assay demonstrated that ZS17 significantly reduced the number of migrating HepG2 cells in zebrafish embryos and suppressed the growth of HepG2 xenografts in nude mice without any obvious side effects. Our study demonstrated that the ROS-JNK-P53 pathway plays an important role in the destruction of liver tumour cells by ZS17. Thus, ZS17 may represent a promising chemotherapeutic agent for the treatment of HCC patients.
Collapse
Affiliation(s)
| | | | | | | | - Xu Liu
- Correspondence: (L.W.); (X.L.)
| |
Collapse
|
19
|
Liu X, Bai M, Li H, Ye P, Duan X, Wu C, Huang Z, Lu S, Zhang J, Zhao Z, Guo F, You R, Qin W, Wang W, Han A, Shen L, Wang Y, Zhao Z, Luo H, Wu J. Single-cell RNA-sequencing uncovers compound kushen injection synergistically improves the efficacy of chemotherapy by modulating the tumor environment of breast cancer. Front Immunol 2022; 13:965342. [DOI: 10.3389/fimmu.2022.965342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundDue to lack of enough specific targets and the immunosuppressive tumor microenvironment (TME) of triple-negative breast cancer (TNBC), TNBC patients often cannot benefit from a single treatment option. This study aims to explore the regulatory effects of Compound kushen injection (CKI) plus chemotherapy on the TME of TNBC from a single cell level.MethodsA mouse TNBC model in BALB/c mice was established to evaluate the antitumor efficacy and toxicity of CKI combined with chemotherapy. Flow cytometry was used to observe the influence of CKI on the lymphocyte populations in the tumor bearing mice. Both bulk RNA sequencing (RNA-seq) and single-cell RNA-seq (scRNA-seq) were applied to portray the modulation of CKI combined with chemotherapy on the TME of TNBC mice.ResultsCKI significantly enhanced the anticancer activity of chemotherapy in vivo with no obvious side effects. Flow cytometry results revealed a significantly higher activation of CD8+ T lymphocytes in the spleens and tumors of the mice with combination therapy. Bulk RNA-seq indicated that CKI could promote the cytotoxic immune cell infiltrating into tumor tissues. Meanwhile, scRNA-seq further revealed that CKI combined with chemotherapy could enhance the percentage of tumor-infiltrating CD8+ T cells, inhibit tumor-promoting signaling pathways, and promote T cell activation and positive regulation of immune response. In addition, CKI showed obvious anticancer activity against MDA-MB-231 breast tumor cells in vitro.ConclusionsThe combination of CKI and chemotherapy might provide a higher efficiency and lower toxicity strategy than a single chemotherapy drug for TNBC. CKI potentiates the anti-TNBC effects of chemotherapy by activating anti-tumor immune response in mice.
Collapse
|
20
|
Wang J, Wang Q, Zhang P, Zhang R, He J. Efficacy and safety of traditional Chinese medicine for the treatment of pancreatic cancer: An overview of systematic reviews and meta-analyses. Front Pharmacol 2022; 13:896017. [PMID: 36120323 PMCID: PMC9475193 DOI: 10.3389/fphar.2022.896017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/02/2022] [Indexed: 12/09/2022] Open
Abstract
Pancreatic cancer is a highly malignant tumor with poor prognosis. Currently available Western medical management strategies are unable to prolong the survival time and reduce the mortality of patients with pancreatic cancer. Traditional Chinese medicine has achieved promising results in many clinical studies. This systematic review and meta-analysis (SR/MA) aimed to explore the benefits and evaluate the quality of evidence of traditional Chinese medicine-based interventions for preventing and treating pancreatic cancer. A systematic search of eight databases for SRs/MAs of randomized controlled trials on traditional Chinese medicine treatment for pancreatic cancer was conducted (from inception to April 2022). The methodological quality of the SRs/MAs was assessed using AMSTAR 2.0, and the quality of evidence was evaluated using the GRADE guide. Nine SRs/MAs, including 145 randomized controlled trials, were considered eligible for this study. The literature were published between 2014 and 2022. The sample size of randomized controlled trials in the MAs ranged from 336 to 1,989. The methodological quality of the nine studies was critically low. Among the 59 outcome indicators of the nine SRs/MAs, seven, 33, and 19 had moderate-, low-, and critically low-quality evidence, respectively, while high-quality evidence was not identified. The results for the long-term indicators, short-term indicators, and adverse reactions in the SRs/MAs displayed consistencies and differences. In conclusion, the methodological and evidence quality of the current evidence is generally low, highlighting the need for additional focus on implementation processes. Some evidence with moderate quality validated that several specific traditional Chinese medicine were optimum for improving the short-term clinical efficacy. However, more objective and high-quality investigations are warranted to verify the efficacy of traditional Chinese medicine for pancreatic cancer.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuyuan Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Orthopaedics, China-Japan Friendship Hospital, Beijing, China
| | - Peitong Zhang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Peitong Zhang,
| | - Ruoqi Zhang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jie He
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
21
|
Harata-Lee Y, Qu Z, Bateman E, Xiao X, Keller MD, Bowen J, Wang W, Adelson DL. Compound Kushen injection reduces severity of radiation-induced gastrointestinal mucositis in rats. Front Oncol 2022; 12:929735. [PMID: 36033515 PMCID: PMC9403047 DOI: 10.3389/fonc.2022.929735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/18/2022] [Indexed: 12/04/2022] Open
Abstract
Mucositis, or damage/injury to mucous membranes of the alimentary, respiratory, or genitourinary tract, is the major side effect associated with anticancer radiotherapies. Because there is no effective treatment for mucositis at present, this is a particular issue as it limits the dose of therapy in cancer patients and significantly affects their quality of life. Gastrointestinal mucositis (GIM) occurs in patients receiving radiotherapies to treat cancers of the stomach, abdomen, and pelvis. It involves inflammation and ulceration of the gastrointestinal (GI) tract causing diarrhea, nausea and vomiting, abdominal pain, and bloating. However, there is currently no effective treatment for this debilitating condition. In this study, we investigated the potential of a type of traditional Chinese medicine (TCM), compound Kushen injection (CKI), as a treatment for GIM. It has previously been shown that major groups of chemical compounds found in CKI have anti-inflammatory effects and are capable of inhibiting the expression of pro-inflammatory cytokines. Intraperitoneal administration of CKI to Sprague Dawley (SD) rats that concurrently received abdominal irradiation over five fractions resulted in reduced severity of GIM symptoms compared to rats administered a vehicle control. Histological examination of the intestinal tissues revealed significantly less damaged villus epithelium in CKI-administered rats that had reduced numbers of apoptotic cells in the crypts. Furthermore, it was also found that CKI treatment led to decreased levels of inflammatory factors including lower levels of interleukin (IL)-1β and IL-6 as well as myeloperoxidase (MPO)-producing cells in the intestinal mucosa. Together, our data indicate a novel effect of CKI to reduce the symptoms of radiation-induced GIM by inhibiting inflammation in the mucosa and apoptosis of epithelial cells.
Collapse
Affiliation(s)
- Yuka Harata-Lee
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Zhipeng Qu
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Emma Bateman
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Xi Xiao
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Marianne D. Keller
- Preclinical, Imaging and Research Laboratories (PIRL), South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Joanne Bowen
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Wei Wang
- Zhendong Research Institute, Zhendong Pharmaceutical, Beijing, China
| | - David L. Adelson
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: David L. Adelson,
| |
Collapse
|
22
|
Xu Y, Zeng P, Wang H, Han K, Qiu G, Wei Y, Chen R, Wang L, Liu X. Novel matrinic acid derivatives bearing 2-anilinothiazole structure for non-small cell lung cancer treatment with improved Hsp90 targeting effect. Drug Dev Res 2022; 83:1434-1454. [PMID: 35841121 DOI: 10.1002/ddr.21974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/03/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
Involved in mediating the folding and maturation of more than 300 client proteins, many of which are oncoproteins, Hsp90 has emerged as a promising drug target for cancer therapy. In particular, inhibiting Hsp90 plays a vital role in the treatment of non-small cell lung cancer. Owing to undesirable outcomes of Hsp90 inhibitors in clinical trials, a series of matrinic acid compounds bearing 2-anilinothiazole moiety were designed based on the structural features allocation shared among Hsp90 inhibitors within the ATP-binding pocket. Most of the compounds showed potent anticancer activities validated by MTT assay. Among them, the most potent compound C4 (IC50 < 10 μM against four cell lines) was chosen for further mechanism study. Notably, C4 showed a better safety profile than 17AAG with a higher SI value. Thermal shift assay data indicated C4 exhibited a strong binding affinity with Hsp90 (-18.85 ± 0.56°C) comparable to radicicol. Mechanism studies verified that C4 significantly inhibited proliferation and migration activities of A549 cells. Besides, C4 can induce a prolonged G1-phase and cell apoptosis. Western blot analysis results indicated C4 could moderately suppress Hsp90 and upregulate Hsp70 expression. Furthermore, the downregulated trend of the client proteins of Hsp90, such as β-Catenin and Bcl-2, were consistent with the cellular effect of C4, suggesting that C4 could exert anticancer activity via targeting Hsp90. In the xenograft model in vivo, C4 effectively inhibited lung cancer growth without obvious side effects. Collectively, C4 could be a promising therapeutic agent for lung cancer and the novel scaffold provided new insights into the design of Hsp90 inhibitors.
Collapse
Affiliation(s)
- Yiming Xu
- Medical College, Guangxi University, Nanning, China.,Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Panke Zeng
- Medical College, Guangxi University, Nanning, China
| | - Haodong Wang
- Medical College, Guangxi University, Nanning, China
| | - Keyan Han
- Medical College, Guangxi University, Nanning, China
| | - Gan Qiu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, China
| | - Yongquan Wei
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, China
| | - Rui Chen
- Guangxi University of Chinese Medicine, Nanning, China
| | - Lisheng Wang
- Medical College, Guangxi University, Nanning, China
| | - Xu Liu
- Medical College, Guangxi University, Nanning, China
| |
Collapse
|
23
|
Kirdeeva Y, Fedorova O, Daks A, Barlev N, Shuvalov O. How Should the Worldwide Knowledge of Traditional Cancer Healing Be Integrated with Herbs and Mushrooms into Modern Molecular Pharmacology? Pharmaceuticals (Basel) 2022; 15:868. [PMID: 35890166 PMCID: PMC9320176 DOI: 10.3390/ph15070868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Traditional herbal medicine (THM) is a "core" from which modern medicine has evolved over time. Besides this, one third of people worldwide have no access to modern medicine and rely only on traditional medicine. To date, drugs of plant origin, or their derivates (paclitaxel, vinblastine, vincristine, vinorelbine, etoposide, camptothecin, topotecan, irinotecan, and omacetaxine), are very important in the therapy of malignancies and they are included in most chemotherapeutic regimes. To date, 391,000 plant and 14,000 mushroom species exist. Their medical and biochemical capabilities have not been studied in detail. In this review, we systematized the information about plants and mushrooms, as well as their active compounds with antitumor properties. Plants and mushrooms are divided based on the regions where they are used in ethnomedicine to treat malignancies. The majority of their active compounds with antineoplastic properties and mechanisms of action are described. Furthermore, on the basis of the available information, we divided them into two priority groups for research and for their potential of use in antitumor therapy. As there are many prerequisites and some examples how THM helps and strengthens modern medicine, finally, we discuss the positive points of THM and the management required to transform and integrate THM into the modern medicine practice.
Collapse
Affiliation(s)
- Yulia Kirdeeva
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
| | - Olga Fedorova
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
| | - Alexandra Daks
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
| | - Nikolai Barlev
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
- Orekhovich Institute of Biomedical Chemistry, 119435 Moscow, Russia
| | - Oleg Shuvalov
- Institute of Cytology, Russian Academy of Sciences, 194064 St. Petersburg, Russia; (Y.K.); (O.F.); (A.D.)
- Orekhovich Institute of Biomedical Chemistry, 119435 Moscow, Russia
| |
Collapse
|
24
|
Xu PL, Cheng CS, Jiao JY, Chen H, Chen Z, Li P. Matrine injection inhibits pancreatic cancer growth via modulating carbonic anhydrases- a network pharmacology-based study with in vitro validation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 287:114691. [PMID: 34597654 DOI: 10.1016/j.jep.2021.114691] [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: 08/08/2021] [Revised: 09/14/2021] [Accepted: 09/25/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Matrine injection is a complex mixture of plant bioactive substances extracted from Sophora flavescens Aiton and Smilax glabra Roxb. Since its approval by the Chinese Food and Drug Administration (CFDA) in 1995, Matrine injection has been clinically used as a complementary and alternative treatment for various cancers; however, the underlying mechanism of pancreatic cancer treatment is yet to be elucidated. AIM OF THE STUDY The present study explores the potential mechanism of matrine injection on pancreatic cancer through network pharmacology technique and in vitro experimental validation. MATERIALS AND METHODS Genes differentially expressed in pancreatic cancer were obtained from the Gene Expression Omnibus (GEO) database (GSE101448). The potential active components of matrine injection were selected following a literature search, and target prediction was performed by the SwissTarget Prediction database. Overlapping genes associated with survival were screened by the Gene Expression Profiling Interactive Analysis (GEPIA) database. In vitro experimental validation was performed with cell counting kit-8 (CCK-8) assay, apoptosis detection, cell cycle analysis, immunoblotting, and co-immunoprecipitation of the identified proteins. RESULTS One thousand seven hundred genes differentially expressed among pancreatic tumor and non-tumor tissues were screened out. Sixteen active components and 226 predicted target genes were identified in matrine injection. A total of 25 potential target genes of matrine injection for the treatment of pancreatic cancer were obtained. Among them, the prognostic target genes carbonic anhydrase 9 (CA9) and carbonic anhydrase 12 (CA12) based on the GEPIA database are differently expressed in tumors compared to adjacent normal tissue. In vitro experiments, the results of CCK-8 assay, apoptosis and cell cycle analysis, immunoblotting, and co-immunoprecipitation showed that matrine injection inhibited Capan-1 and Mia paca-2 proliferation, arrested the cell cycle at the S phase, and induced apoptosis through up-regulated CA12 and down-regulated CA9. CONCLUSIONS In this study, bioinformatics and network pharmacology were applied to explore the treatment mechanism on pancreatic cancer with matrine injection. This study demonstrated that matrine injection inhibited proliferation, arrested the cell cycle, and induced apoptosis of pancreatic cancer cells. The mechanism may be related to the induction of CA12 over-expression, and CA9 reduced expression. As novel targets for pancreatic cancer treatment, Carbonic anhydrases require further study.
Collapse
Affiliation(s)
- Pan-Ling Xu
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| | - Chien-Shan Cheng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Ju-Ying Jiao
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Hao Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhen Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Ping Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| |
Collapse
|
25
|
Yu ZY, Peng RY, Han M, Grant S, Yang GY, Liu JP, Cao HJ. Adjunctive effect of compound Kushen Injection to chemotherapy for non-small cell lung cancer: An evidence map and overview of systematic reviews. JOURNAL OF ETHNOPHARMACOLOGY 2021; 281:114538. [PMID: 34418510 DOI: 10.1016/j.jep.2021.114538] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 08/08/2021] [Accepted: 08/17/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Extract from Kushen (Sophora flavescens Aiton) and Baituling (Heterosmilax japonica Kunth), Compound Kushen Injection has a long history for cancer treatment in China. As a common adjunctive drug in chemotherapy of non-small cell lung cancer (NSCLC), the evidence of effectiveness and safety of Compound Kushen Injection needs to be synthesized. AIM OF THE STUDY The objective of this overview is to synthesize recent evidence and assess the methodological quality of systematic reviews (SRs) of Compound Kushen Injection, in treating NSCLC as an adjunctive treatment of chemotherapy. MATERIALS AND METHODS We searched PubMed, PubMed Central, China National Knowledge Infrastructure, Wanfang Data Knowledge Service Platform and VIP China Science and Technology Journal Database from inception to April 22, 2020. We included SRs of the efficacy and safety of Compound Kushen Injection combined with chemotherapy in the treatment of NSCLC. Two authors assessed eligibility and extracted data. The quality of SRs was assessed using AMSTAR-2. A meta-analysis was conducted for the original trials where good homogeneity was present. Evidence maps using bubble plots illustrated overall results. The quality of the evidence was graded by the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach. RESULTS Twelve SRs were included, with 91 randomized controlled trials and 7466 participants. Overall, the quality of the included SRs was low. Only six SRs reported specific chemotherapy regimens, three using vinorelbine combined with cisplatin (NP), one using paclitaxel combined with cisplatin (TP), one using gemcitabine combined with cisplatin (GP), and one discussed the three regimens in subgroups. Meta-analysis showed that CKI used as an adjuvant to chemotherapy was superior to chemotherapy alone in promoting tumor complete response rate, tumor partial response rate, Kamofsky score, and relieving some chemotherapy related side effects. Evidence mapping showed that Kushen Injection combined with NP and GP had better effect. The evidence quality of increasing Kamofsky score and decreasing the incidence of leukopenia were moderate, others were low or very low. CONCLUSIONS There are some favorable effects in improving short-term effectiveness, quality of life and alleviating some side effects of chemotherapy in patients with NSCLC. Results are more promising when Compound Kushen injection is used as an adjunctive to NP and GP. Promising results are however, compromised by the poor quality overall of the clinical trials.
Collapse
Affiliation(s)
- Ze-Yu Yu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Rong-Yan Peng
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mei Han
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | | | | | - Jian-Ping Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hui-Juan Cao
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
26
|
Wu C, Huang ZH, Meng ZQ, Fan XT, Lu S, Tan YY, You LM, Huang JQ, Stalin A, Ye PZ, Wu ZS, Zhang JY, Liu XK, Zhou W, Zhang XM, Wu JR. A network pharmacology approach to reveal the pharmacological targets and biological mechanism of compound kushen injection for treating pancreatic cancer based on WGCNA and in vitro experiment validation. Chin Med 2021; 16:121. [PMID: 34809653 PMCID: PMC8607619 DOI: 10.1186/s13020-021-00534-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Compound kushen injection (CKI), a Chinese patent drug, is widely used in the treatment of various cancers, especially neoplasms of the digestive system. However, the underlying mechanism of CKI in pancreatic cancer (PC) treatment has not been totally elucidated. METHODS Here, to overcome the limitation of conventional network pharmacology methods with a weak combination with clinical information, this study proposes a network pharmacology approach of integrated bioinformatics that applies a weighted gene co-expression network analysis (WGCNA) to conventional network pharmacology, and then integrates molecular docking technology and biological experiments to verify the results of this network pharmacology analysis. RESULTS The WGCNA analysis revealed 2 gene modules closely associated with classification, staging and survival status of PC. Further CytoHubba analysis revealed 10 hub genes (NCAPG, BUB1, CDK1, TPX2, DLGAP5, INAVA, MST1R, TMPRSS4, TMEM92 and SFN) associated with the development of PC, and survival analysis found 5 genes (TSPOAP1, ADGRG6, GPR87, FAM111B and MMP28) associated with the prognosis and survival of PC. By integrating these results into the conventional network pharmacology study of CKI treating PC, we found that the mechanism of CKI for PC treatment was related to cell cycle, JAK-STAT, ErbB, PI3K-Akt and mTOR signalling pathways. Finally, we found that CDK1, JAK1, EGFR, MAPK1 and MAPK3 served as core genes regulated by CKI in PC treatment, and were further verified by molecular docking, cell proliferation assay, RT-qPCR and western blot analysis. CONCLUSIONS Overall, this study suggests that the optimized network pharmacology approach is suitable to explore the molecular mechanism of CKI in the treatment of PC, which provides a reference for further investigating biomarkers for diagnosis and prognosis of PC and even the clinical rational application of CKI.
Collapse
Affiliation(s)
- Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhi-Hong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zi-Qi Meng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiao-Tian Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Ying-Ying Tan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Lei-Ming You
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jia-Qi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Antony Stalin
- State Key Laboratory of Subtropical Silviculture, Department of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou, 311300, China
| | - Pei-Zhi Ye
- National Cancer Center/National Clinical Research Center for Cancer/Chinese Medicine Department of the Caner Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Shan Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jing-Yuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xin-Kui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Wei Zhou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
- China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiao-Meng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jia-Rui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
27
|
Zhou W, Wu C, Zhao C, Huang Z, Lu S, Fan X, Tan Y, Stalin A, You R, Liu X, Zhang J, Wu Z, Wu J. An Advanced Systems Pharmacology Strategy Reveals AKR1B1, MMP2, PTGER3 as Key Genes in the Competing Endogenous RNA Network of Compound Kushen Injection Treating Gastric Carcinoma by Integrated Bioinformatics and Experimental Verification. Front Cell Dev Biol 2021; 9:742421. [PMID: 34646828 PMCID: PMC8502965 DOI: 10.3389/fcell.2021.742421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric carcinoma (GC) is a severe tumor of the digestive tract with high morbidity and mortality and poor prognosis, for which novel treatment options are urgently needed. Compound Kushen injection (CKI), a classical injection of Chinese medicine, has been widely used to treat various tumors in clinical practice for decades. In recent years, a growing number of studies have confirmed that CKI has a beneficial therapeutic effect on GC, However, there are few reports on the potential molecular mechanism of action. Here, using systems pharmacology combined with proteomics analysis as a core concept, we identified the ceRNA network, key targets and signaling pathways regulated by CKI in the treatment of GC. To further explore the role of these key targets in the development of GC, we performed a meta-analysis to compare the expression differences between GC and normal gastric mucosa tissues. Functional enrichment analysis was further used to understand the biological pathways significantly regulated by the key genes. In addition, we determined the significance of the key genes in the prognosis of GC by survival analysis and immune infiltration analysis. Finally, molecular docking simulation was performed to verify the combination of CKI components and key targets. The anti-gastric cancer effect of CKI and its key targets was verified by in vivo and in vitro experiments. The analysis of ceRNA network of CKI on GC revealed that the potential molecular mechanism of CKI can regulate PI3K/AKT and Toll-like receptor signaling pathways by interfering with hub genes such as AKR1B1, MMP2 and PTGERR3. In conclusion, this study not only partially highlighted the molecular mechanism of CKI in GC therapy but also provided a novel and advanced systems pharmacology strategy to explore the mechanisms of traditional Chinese medicine formulations.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,China-Japan Friendship Hospital, Beijing, China
| | - Chao Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chongjun Zhao
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Huang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaotian Fan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- State Key Laboratory of Subtropical Silviculture, Department of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou, China
| | - Rongli You
- Shanxi Zhendong Pharmaceutical Co., Ltd., Shanxi, China
| | - Xinkui Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhishan Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
28
|
Liu X, Wu Y, Zhang Y, Bu D, Wu C, Lu S, Huang Z, Song Y, Zhao Y, Guo F, Ye P, Fu C, Shen L, Zhang J, Wang H, Duan X, Wu J. High Throughput Transcriptome Data Analysis and Computational Verification Reveal Immunotherapy Biomarkers of Compound Kushen Injection for Treating Triple-Negative Breast Cancer. Front Oncol 2021; 11:747300. [PMID: 34604090 PMCID: PMC8484800 DOI: 10.3389/fonc.2021.747300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022] Open
Abstract
Background Although notable therapeutic and prognostic benefits of compound kushen injection (CKI) have been found when it was used alone or in combination with chemotherapy or radiotherapy for triple-negative breast cancer (TNBC) treatment, the effects of CKI on TNBC microenvironment remain largely unclear. This study aims to construct and validate a predictive immunotherapy signature of CKI on TNBC. Methods The UPLC-Q-TOF-MS technology was firstly used to investigate major constituents of CKI. RNA sequencing data of CKI-perturbed TNBC cells were analyzed to detect differential expression genes (DEGs), and the GSVA algorithm was applied to explore significantly changed pathways regulated by CKI. Additionally, the ssGSEA algorithm was used to quantify immune cell abundance in TNBC patients, and these patients were classified into distinct immune infiltration subgroups by unsupervised clustering. Then, prognosis-related genes were screened from DEGs among these subgroups and were further overlapped with the DEGs regulated by CKI. Finally, a predictive immunotherapy signature of CKI on TNBC was constructed based on the LASSO regression algorithm to predict mortality risks of TNBC patients, and the signature was also validated in another TNBC cohort. Results Twenty-three chemical components in CKI were identified by UPLC-Q-TOF-MS analysis. A total of 3692 DEGs were detected in CKI-treated versus control groups, and CKI significantly activated biological processes associated with activation of T, natural killer and natural killer T cells. Three immune cell infiltration subgroups with 1593 DEGs were identified in TNBC patients. Then, two genes that can be down-regulated by CKI with hazard ratio (HR) > 1 and 26 genes that can be up-regulated by CKI with HR < 1 were selected as key immune- and prognosis-related genes regulated by CKI. Lastly, a five-gene prognostic signature comprising two risky genes (MARVELD2 and DYNC2I2) that can be down-regulated by CKI and three protective genes (RASSF2, FERMT3 and RASSF5) that can be up-regulated by CKI was developed, and it showed a good performance in both training and test sets. Conclusions This study proposes a predictive immunotherapy signature of CKI on TNBC, which would provide more evidence for survival prediction and treatment guidance in TNBC as well as a paradigm for exploring immunotherapy biomarkers in compound medicines.
Collapse
Affiliation(s)
- Xinkui Liu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Wu
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Yingying Zhang
- Department of Vascular Neurosurgery, New Era Stroke Care and Research Institute, The People's Liberation Army (PLA) Rocket Force Characteristic Medical Center, Beijing, China
| | - Dechao Bu
- Pervasive Computing Research Center, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Chao Wu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Shan Lu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Huang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Yurong Song
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhao
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fengying Guo
- School of Management, Beijing University of Chinese Medicine, Beijing, China
| | - Peizhi Ye
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changgeng Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liangliang Shen
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Jingyuan Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Haojia Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Xianchun Duan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Jiarui Wu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
29
|
Yang Y, Sun M, Li W, Liu C, Jiang Z, Gu P, Li J, Wang W, You R, Ba Q, Li X, Wang H. Rebalancing TGF-β/Smad7 signaling via Compound kushen injection in hepatic stellate cells protects against liver fibrosis and hepatocarcinogenesis. Clin Transl Med 2021; 11:e410. [PMID: 34323416 PMCID: PMC8255064 DOI: 10.1002/ctm2.410] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Liver fibrosis and fibrosis-related hepatocarcinogenesis are a rising cause for morbidity and death worldwide. Although transforming growth factor-β (TGF-β) is a critical mediator of chronic liver fibrosis, targeting TGF-β isoforms and receptors lead to unacceptable side effect. This study was designed to explore the antifibrotic effect of Compound kushen injection (CKI), an approved traditional Chinese medicine formula, via a therapeutic strategy of rebalancing TGF-β/Smad7 signaling. METHODS A meta-analysis was performed to evaluate CKI intervention on viral hepatitis-induced fibrosis or cirrhosis in clinical randomized controlled trials (RCTs). Mice were given carbon tetrachloride (CCl4 ) injection or methionine-choline deficient (MCD) diet to induce liver fibrosis, followed by CKI treatment. We examined the expression of TGF-β/Smad signaling and typical fibrosis-related genes in hepatic stellate cells (HSCs) and fibrotic liver tissues by qRT-PCR, Western blotting, RNA-seq, immunofluorescence, and immunohistochemistry. RESULTS Based on meta-analysis results, CKI improved the liver function and relieved liver fibrosis among patients. In our preclinical studies by using two mouse models, CKI treatment demonstrated promising antifibrotic effects and postponed hepatocarcinogenesis with improved liver function and histopathologic features. Mechanistically, we found that CKI inhibited HSCs activation by stabilizing the interaction of Smad7/TGF-βR1 to rebalance Smad2/Smad3 signaling, and subsequently decreased the extracellular matrix formation. Importantly, Smad7 depletion abolished the antifibrotic effect of CKI in vivo and in vitro. Moreover, matrine, oxymatrine, sophocarpine, and oxysophocarpine were identified as material basis responsible for the antifibrosis effect of CKI. CONCLUSIONS Our results unveil the approach of CKI in rebalancing TGF-β/Smad7 signaling in HSCs to protect against hepatic fibrosis and hepatocarcinogenesis in both preclinical and clinical studies. Our study suggests that CKI can be a candidate for treatment of hepatic fibrosis and related oncogenesis.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Mayu Sun
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Weida Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chaobao Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zheshun Jiang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Pengfei Gu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jingquan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Wang
- Beijing Zhendong Pharmaceutical Research Institute Co., Ltd.BeijingChina
| | - Rongli You
- Beijing Zhendong Pharmaceutical Research Institute Co., Ltd.BeijingChina
| | - Qian Ba
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaoguang Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- CAS Key Laboratory of Nutrition, Metabolism and Food SafetyShanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
30
|
Wang KX, Chen YP, Lu AP, Du GH, Qin XM, Guan DG, Gao L. A metabolic data-driven systems pharmacology strategy for decoding and validating the mechanism of Compound Kushen Injection against HCC. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114043. [PMID: 33753143 DOI: 10.1016/j.jep.2021.114043] [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: 10/19/2020] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Compound Kushen Injection (CKI) is a widely used TCM formula for treatment of carcinomatous pain and tumors of digestive system including hepatocellular carcinoma (HCC). However, the potential mechanisms of CKI for treatment of HCC have not been systematically and deeply studied. AIM OF STUDY A metabolic data-driven systems pharmacology approach was utilized to investigate the potential mechanisms of CKI for treatment of HCC. MATERIALS AND METHODS Based on phenotypic data generated by metabolomics and genotypic data of drug targets, a propagation model based on Dijkstra program was proposed to decode the effective network of key genotype-phenotype of CKI in treating HCC. The pivotal pathway was predicted by target propagation mode of our proposed model, and was validated in SMMC-7721 cells and diethylnitrosamine-induced rats. RESULTS Metabolomics results indicated that 12 differential metabolites, and 5 metabolic pathways might be involved in the anti-HCC effect of CKI. A total of 86 metabolic related genes that affected by CKI were obtained. The results calculated by propagation model showed that 6475 shortest distance chains might be involved in the anti-HCC effect of CKI. According to the results of propagation mode, EGFR was identified as the core target of CKI for the anti-HCC effect. Finally, EGFR and its related pathway EGFR-STAT3 signaling pathway were validated in vivo and in vitro. CONCLUSION The proposed method provides a methodological reference for explaining the underlying mechanism of TCM in treating HCC.
Collapse
Affiliation(s)
- Ke-Xin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China.
| | - Yu-Peng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Ai-Ping Lu
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong, China.
| | - Guan-Hua Du
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China; Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China.
| | - Dao-Gang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China.
| | - Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, China.
| |
Collapse
|
31
|
Yang Y, Sun M, Yao W, Wang F, Li X, Wang W, Li J, Gao Z, Qiu L, You R, Yang C, Ba Q, Wang H. Compound kushen injection relieves tumor-associated macrophage-mediated immunosuppression through TNFR1 and sensitizes hepatocellular carcinoma to sorafenib. J Immunother Cancer 2021; 8:jitc-2019-000317. [PMID: 32179631 PMCID: PMC7073790 DOI: 10.1136/jitc-2019-000317] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background There is an urgent need for effective treatments for hepatocellular carcinoma (HCC). Immunotherapy is promising especially when combined with traditional therapies. This study aimed to investigate the immunomodulatory function of an approved Chinese medicine formula, compound kushen injection (CKI), and its anti-HCC efficiency in combination with low-dose sorafenib. Methods Growth of two murine HCC cells was evaluated in an orthotopic model, a subcutaneous model, two postsurgical recurrence model, and a tumor rechallenge model with CKI and low-dose sorafenib combination treatment. In vivo macrophage or CD8+ T cell depletion and in vitro primary cell coculture models were used to determine the regulation of CKI on macrophages and CD8+ T cells. Results CKI significantly enhanced the anticancer activity of sorafenib at a subclinical dose with no obvious side effects. CKI and sorafenib combination treatment prevented the postsurgical recurrence and rechallenged tumor growth. Further, we showed that CKI activated proinflammatory responses and relieved immunosuppression of tumor-associated macrophages in the HCC microenvironment by triggering tumor necrosis factor receptor superfamily member 1 (TNFR1)-mediated NF-κB and p38 MAPK signaling cascades. CKI-primed macrophages significantly promoted the proliferation and the cytotoxic ability of CD8+ T cells and decreased the exhaustion, which subsequently resulted in apoptosis of HCC cells. Conclusions CKI acts on macrophages and CD8+ T cells to reshape the immune microenvironment of HCC, which improves the therapeutic outcomes of low-dose sorafenib and avoids adverse chemotherapy effects. Our study shows that traditional Chinese medicines with immunomodulatory properties can potentiate chemotherapeutic drugs and provide a promising approach for HCC treatment.
Collapse
Affiliation(s)
- Yang Yang
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mayu Sun
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenbo Yao
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Feng Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoguang Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Pharmacology, Beijing Zhendong Pharmaceutical Research Institute Co, Ltd, Beijing, China
| | - Jingquan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihu Gao
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lin Qiu
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rongli You
- Department of Pharmacology, Beijing Zhendong Pharmaceutical Research Institute Co, Ltd, Beijing, China
| | - Chenghua Yang
- CAS Key Laboratory of Nutrition, Metabolism and Food safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qian Ba
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Xiao Z, Chen Z, Han R, Lu L, Li Z, Lin J, Hu L, Huang X, Lin L. Comprehensive TCM treatments combined with chemotherapy for advanced non-small cell lung cancer: A randomized, controlled trial. Medicine (Baltimore) 2021; 100:e25690. [PMID: 33950949 PMCID: PMC8104195 DOI: 10.1097/md.0000000000025690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/08/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE We conducted this study to evaluate the efficacy and safety of traditional Chinese medicine (TCM) in advanced non-small cell lung cancer (NSCLC) patients who underwent chemotherapy. DESIGN This was a prospective, open-label, randomized controlled trial. NSCLC patients at stage IIIA, IIIB, or IV were randomly assigned to either TCM plus chemotherapy or chemotherapy alone. The comprehensive TCM treatment consisted of Kang Ai injection, herbal decoction, and Zhenqifuzheng capsules. The primary endpoint was quality of life (QOL) measured by the Functional Assessment of Cancer Therapy-Lung version 4.0. The secondary endpoints were chemotherapy completion rate, tumor response, and adverse events. All assessments were done at baseline, the third week, and the sixth week. RESULTS Thirty-nine participants were randomly assigned to the treatment group and 36 to the control group. The QOL scores were significantly improved in the treatment group compared with those of the control group in social well-being (cycle 1, P = .048; cycle 2, P = .015), emotional well-being (cycle 1, P = .047; cycle 2, P = 4.29E-05), and functional well-being (cycle 1, P = .030; cycle 2, P = .003), while the QOL scores in the above 3 domains declined in the control group (P < .05). Both groups had a decline in the physical well-being score (cycle 1, P = .042; cycle 2, P = .017) and lung cancer symptom score (cycle 1, P = .001; cycle 2, P = .001) after 2 courses of intervention. The deterioration in physical well-being and lung cancer symptoms was noticeably smaller in the treatment group (P < .05). There were significant differences between the 2 groups in social well-being, emotional well-being, functional well-being, lung cancer symptom domain, and the total score (P < .05). Patients in the treatment group had a significantly lower incidence of platelet reduction than the control group (P = .028) after 2 cycles of treatment. No significant difference in nonhematological adverse events (AEs) was observed. CONCLUSION This study illustrated that comprehensive TCM treatment could promote the QOL of NSCLC patients, alleviate symptoms, and reduce the AEs caused by chemotherapy, verifying the synergistic and attenuating effects of TCM in NSCLC patients undergoing chemotherapy. TRIAL REGISTRATION Chinese Clinical Trial Registry (www.chictr.org.cn): ChiCTR-TRC-13003637.
Collapse
Affiliation(s)
- Zhiwei Xiao
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | | | - Rui Han
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Liming Lu
- Clinical Research Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zeyun Li
- Guangzhou University of Chinese Medicine
| | - Jietao Lin
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Leihao Hu
- Guangzhou University of Chinese Medicine
| | - Xuewu Huang
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Lizhu Lin
- Oncology Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| |
Collapse
|
33
|
Jian T, Chen J, Ding X, Lv H, Li J, Wu Y, Ren B, Tong B, Zuo Y, Su K, Li W. Flavonoids isolated from loquat (Eriobotrya japonica) leaves inhibit oxidative stress and inflammation induced by cigarette smoke in COPD mice: the role of TRPV1 signaling pathways. Food Funct 2020; 11:3516-3526. [PMID: 32253400 DOI: 10.1039/c9fo02921d] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic, progressive lung disease with few successful treatments, and is strongly associated with cigarette smoking (CS). Since the novel coronavirus has spread worldwide seriously, there is growing concern that patients who have chronic respiratory conditions like COPD can easily be infected and are more prone to having severe illness and even mortality because of lung dysfunction. Loquat leaves have long been used as an important material for both pharmaceutical and functional applications in the treatment of lung disease in Asia, especially in China and Japan. Total flavonoids (TF), the main active components derived from loquat leaves, showed remarkable anti-inflammatory and antioxidant activities. However, their protective activity against CS-induced COPD airway inflammation and oxidative stress and its underlying mechanism still remain not well-understood. The present study uses a CS-induced mouse model to estimate the morphological changes in lung tissue. The results demonstrated that TF suppressed the histological changes in the lungs of CS-challenged mice, as evidenced by reduced generation of pro-inflammatory cytokines including interleukin 6 (IL-6), IL-1β, tumor necrosis factor α (TNF-α), nitric oxide (NO), and inducible nitric oxide synthase (iNOS) and diminished the protein expression of transient receptor potential vanilloid 1 (TRPV1). Moreover, TF also inhibited phosphorylation of IKK, IκB and NFκB and increased p-Akt. Interestingly, TF could inhibit CS-induced oxidative stress in the lungs of COPD mice. TF treatment significantly inhibited the level of malondialdehyde (MDA) and increased the activity of superoxide dismutase (SOD). In addition, TF markedly downregulated TRPV1 and cytochrome P450 2E1 (CYP2E1) and upregulated the expression of SOD-2, while the p-JNK level was observed to be inhibited in COPD mice. Taken together, our findings showed that the protective effect and putative mechanism of the action of TF resulted in the inhibition of inflammation and oxidative stress through the regulation of TRPV1 and the related signal pathway in lung tissues. It suggested that TF derived from loquat leaves could be considered to be an alternative or a new functional material and used for the treatment of CS-induced COPD.
Collapse
Affiliation(s)
- Tunyu Jian
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Jian Chen
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China. and Department of Food Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaoqin Ding
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Han Lv
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Jiawei Li
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Yuexian Wu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Bingru Ren
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Bei Tong
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Yuanyuan Zuo
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Kelei Su
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210000, China and Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Weilin Li
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China. and Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
34
|
Cui J, Qu Z, Harata-Lee Y, Shen H, Aung TN, Wang W, Kortschak RD, Adelson DL. The effect of compound kushen injection on cancer cells: Integrated identification of candidate molecular mechanisms. PLoS One 2020; 15:e0236395. [PMID: 32730293 PMCID: PMC7392229 DOI: 10.1371/journal.pone.0236395] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 07/05/2020] [Indexed: 12/22/2022] Open
Abstract
Traditional Chinese Medicine (TCM) preparations are often extracts of single or multiple herbs containing hundreds of compounds, and hence it has been difficult to study their mechanisms of action. Compound Kushen Injection (CKI) is a complex mixture of compounds extracted from two medicinal plants and has been used in Chinese hospitals to treat cancer for over twenty years. To demonstrate that a systematic analysis of molecular changes resulting from complex mixtures of bioactives from TCM can identify a core set of differentially expressed (DE) genes and a reproducible set of candidate pathways. We used in vitro cancer models to measure the effect of CKI on cell cycle phases and apoptosis, and correlated those phenotypes with CKI induced changes in gene expression. We treated two cancer cell lines with or without CKI and assessed the resulting phenotypes by employing cell viability and proliferation assays. Based on these results, we carried out high-throughput transcriptome data analysis to identify genes and candidate pathways perturbed by CKI. We integrated these differential gene expression results with previously reported results and carried out validation of selected differentially expressed genes. CKI induced cell-cycle arrest and apoptosis in the cancer cell lines tested. In these cells CKI also altered the expression of 363 core candidate genes associated with cell cycle, apoptosis, DNA replication/repair, and various cancer pathways. Of these, 7 are clinically relevant to cancer diagnosis or therapy, 14 are cell cycle regulators, and most of these 21 candidates are downregulated by CKI. Comparison of our core candidate genes to a database of plant medicinal compounds and their effects on gene expression identified one-to-one, one-to-many and many-to-many regulatory relationships between compounds in CKI and DE genes. By identifying genes and promising candidate pathways associated with CKI treatment based on our transcriptome-based analysis, we have shown that this approach is useful for the systematic analysis of molecular changes resulting from complex mixtures of bioactives.
Collapse
Affiliation(s)
- Jian Cui
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Zhipeng Qu
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Yuka Harata-Lee
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Hanyuan Shen
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Thazin Nwe Aung
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Wei Wang
- Zhendong Research Institute, Shanxi-Zhendong Pharmaceutical Co Ltd, Beijing, China
| | - R. Daniel Kortschak
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - David L. Adelson
- Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
- * E-mail:
| |
Collapse
|
35
|
Zhou W, Wu J, Zhang J, Liu X, Guo S, Jia S, Zhang X, Zhu Y, Wang M. Integrated bioinformatics analysis to decipher molecular mechanism of compound Kushen injection for esophageal cancer by combining WGCNA with network pharmacology. Sci Rep 2020; 10:12745. [PMID: 32728182 PMCID: PMC7391752 DOI: 10.1038/s41598-020-69708-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Compound Kushen injection (CKI), a medicine in widespread clinical use in China, has proven therapeutic effects on cancer. However, few molecular mechanism analyses have been carried out. To address this problem, bioinformatics approaches combining weighted gene co-expression network analysis with network pharmacology methods were undertaken to elucidate the underlying molecular mechanisms of CKI in the treatment of esophageal cancer (ESCA). First, the key gene modules related to the clinical traits of ESCA were analysed by WCGNA. Based on the results, the hub genes related to CKI treatment for ESCA were explored through network pharmacology. Molecular docking simulation was performed to recognize the binding activity of hub genes with CKI compounds. The results showed that the potential hub targets, including EGFR, ErbB2, CCND1 and IGF1R, are therapeutic targets of CKI for the treatment of ESCA. Moreover, these targets were significantly enriched in many pathways related to cancer and signalling pathways, such as the PI3K-Akt signalling pathway and ErbB signalling pathway. In conclusion, this research partially highlighted the molecular mechanism of CKI in the treatment of ESCA, offering great potential in the identification of the effective compounds in CKI and biomarkers for ESCA treatment.
Collapse
MESH Headings
- Algorithms
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Computational Biology/methods
- Cyclin D1/chemistry
- Cyclin D1/metabolism
- Databases, Genetic
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- ErbB Receptors/chemistry
- ErbB Receptors/metabolism
- Esophageal Neoplasms/drug therapy
- Esophageal Neoplasms/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Regulatory Networks/drug effects
- Humans
- Kaplan-Meier Estimate
- Models, Molecular
- Molecular Docking Simulation
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/metabolism
- Receptor, IGF Type 1/chemistry
- Receptor, IGF Type 1/metabolism
- Sequence Analysis, RNA
Collapse
Affiliation(s)
- Wei Zhou
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Jiarui Wu
- Beijing University of Chinese Medicine, Beijing, 100102, China.
| | - Jingyuan Zhang
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Xinkui Liu
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Siyu Guo
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - ShanShan Jia
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Xiaomeng Zhang
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Yingli Zhu
- Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Miaomiao Wang
- Beijing University of Chinese Medicine, Beijing, 100102, China
| |
Collapse
|
36
|
Huh JW, Lee JH, Jeon E, Ryu HW, Oh SR, Ahn KS, Jun HS, Ha UH. Maackiain, a compound derived from Sophora flavescens, increases IL-1β production by amplifying nigericin-mediated inflammasome activation. FEBS Open Bio 2020; 10:1482-1491. [PMID: 32428336 PMCID: PMC7396426 DOI: 10.1002/2211-5463.12899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/08/2020] [Accepted: 05/15/2020] [Indexed: 11/09/2022] Open
Abstract
Sophora flavescens is used as a traditional herbal medicine to modulate inflammatory responses. However, little is known about the impact of (-)-maackiain, a compound derived from S. flavescens, on the activation of inflammasome/caspase-1, a key factor in interleukin-1β (IL-1β) processing. Here, we report that (-)-maackiain potently amplified caspase-1 cleavage in macrophages in response to nigericin (Nig). In macrophages primed with either lipopolysaccharide or monophosphoryl lipid A, Nig-mediated caspase-1 cleavage was also markedly promoted by (-)-maackiain. Notably, (-)-maackiain induced the production of vimentin, an essential mediator for the activation of the NOD-, LRR-, and pyrin domain-containing protein 3 inflammasome, thereby contributing to promotion of the formation of the inflammasome complex to activate caspase-1. Taken together, our data suggest that (-)-maackiain exerts an immunostimulatory effect by promoting IL-1β production via activation of the inflammasome/caspase-1 pathway. Thus, the potent inflammasome-activating effect of (-)-maackiain may be clinically useful as an acute immune-stimulating agent.
Collapse
Affiliation(s)
- Jin-Won Huh
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Jung-Hoon Lee
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Eekhyoung Jeon
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk, Korea
| | - Hyun Sik Jun
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Un-Hwan Ha
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| |
Collapse
|
37
|
Zhou W, Wu J, Zhu Y, Meng Z, Liu X, Liu S, Ni M, Jia S, Zhang J, Guo S. Study on the mechanisms of compound Kushen injection for the treatment of gastric cancer based on network pharmacology. BMC Complement Med Ther 2020; 20:6. [PMID: 32020871 PMCID: PMC7076865 DOI: 10.1186/s12906-019-2787-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022] Open
Abstract
Background As an effective prescription for gastric cancer (GC), Compound Kushen Injection (CKI) has been widely used even though few molecular mechanism analyses have been carried out. Methods In this study, we identified 16 active ingredients and 60 GC target proteins. Then, we established a compound-predicted target network and a GC target protein-protein interaction (PPI) network by Cytoscape 3.5.1 and systematically analyzed the potential targets of CKI for the treatment of GC. Finally, molecular docking was applied to verify the key targets. In addition, we analyzed the mechanism of action of the predicted targets by Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses. Results The results showed that the potential targets, including CCND1, PIK3CA, AKT1, MAPK1, ERBB2, and MMP2, are the therapeutic targets of CKI for the treatment of GC. Functional enrichment analysis indicated that CKI has a therapeutic effect on GC by synergistically regulating some biological pathways, such as the cell cycle, pathways in cancer, the PI3K-AKT signaling pathway, the mTOR signaling pathway, and the FoxO signaling pathway. Moreover, molecular docking simulation indicated that the compounds had good binding activity to PIK3CA, AKT1, MAPK1, ERBB2, and MMP2 in vivo. Conclusion This research partially highlighted the molecular mechanism of CKI for the treatment of GC, which has great potential in the identification of the effective compounds in CKI and biomarkers to treat GC.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China.
| | - Yingli Zhu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Ziqi Meng
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Xinkui Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Shuyu Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Mengwei Ni
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Shanshan Jia
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| | - Siyu Guo
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 of North Three-ring East Road, Chao Yang District, Beijing, China
| |
Collapse
|
38
|
Xu Y, Jing D, Zhao D, Wu Y, Xing L, Ur Rashid H, Wang H, Wang L, Cao H. New modification strategy of matrine as Hsp90 inhibitors based on its specific L conformation for cancer treatment. Bioorg Med Chem 2020; 28:115305. [PMID: 31928863 DOI: 10.1016/j.bmc.2020.115305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/22/2019] [Accepted: 01/02/2020] [Indexed: 10/25/2022]
Abstract
The similarity of spatial structure between radicicol and matrine urged us to perform conformation modification of matrine, followed by L-shaped matrine derivatives, 6, 12, 21a-h and 22a-h were originally designed, synthesized and evaluated for Hsp90N inhibitors as anticancer agents. TSA (Thermal Shift Assay) results indicated that 21e, 22a-c and 22e-g exhibited strong binding force against Hsp90N with∣ΔTm∣ > 3, meanwhile, MTT assay also revealed these compounds displayed potent anticancer activity with IC50 values below 25 μM against HepG2, HeLa and MDA-MB-231 cells lines. Then, compound 22g with a high ΔTm = 10.92 was chosen as a representative to perform further mechanism study. It can induce cell apoptosis, arrest the cell cycle at the S phase and decrease the expression level of Hsp90 in Hela cell. These results originally provided targeted modification strategy for matrine derivatives to serve as Hsp90 inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Yiming Xu
- Medicinal College, Guangxi University, Nanning 530004, China; School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Dewang Jing
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Dong Zhao
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Lu Xing
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an 710021, China
| | - Haroon Ur Rashid
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China; Department of Chemistry, Sarhad University of Science & Information Technology, Peshawar, Khyber Pakhtunkhwa 25120, Pakistan
| | - Haodong Wang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Lisheng Wang
- Medicinal College, Guangxi University, Nanning 530004, China.
| | - Huiling Cao
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an 710021, China.
| |
Collapse
|
39
|
Compound Kushen Injection as an Adjunctive Therapy for the Treatment of Non-Small-Cell Lung Cancer: A Meta-Analysis of Randomized Controlled Trials. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7241927. [PMID: 31781277 PMCID: PMC6875363 DOI: 10.1155/2019/7241927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 08/22/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
Objectives To evaluate the efficacy and safety of compound Kushen injection (CKI) combined with chemo treatment (chemo) for non-small-cell lung cancer (NSCLC). Methods We systematically searched the literature published in seven databases, including Embase, PubMed, central, MEDLINE, CNKI, Wanfang, and VIP, from their inception to April 2019 for all randomized controlled trials (RCTs) comparing CKI plus chemo with chemo alone in patients with NSCLC. Our main end point was clinical efficiency and the secondary outcomes were Karnofsky performance score (KPS), immune function, and adverse events. The Cochrane risk of bias tool was applied for quality assessment. Results 10 studies involving 1019 participants were included. The clinical response rate (relative risk (RR) = 1.21, 95% confidence interval (CI): 1.06 to 1.37; P=0.003), KPS (RR = 2.18, 95% CI: 1.49 to 3.17; P < 0.0001), immune function (mean differences (MD) = 0.82, 95% CI: 0.12 to 1.52; P=0.02) and adverse effects (RR = 0.67, 95% CI: 0.60 to 0.74; P < 0.00001) in the CKI plus chemo group showed significant differences when compared with chemo alone. Conclusions CKI combined with chemo can improve clinical efficiency, KPS, and immune function and reduce adverse reactions in patients with NSCLC when compared with chemo alone. However, more rigorously designed RCTs are needed to validate this benefit, as some of the included RCTs are of low methodological quality.
Collapse
|
40
|
Shen H, Qu Z, Harata-Lee Y, Cui J, Aung TN, Wang W, Kortschak RD, Adelson DL. A New Strategy for Identifying Mechanisms of Drug-drug Interaction Using Transcriptome Analysis: Compound Kushen Injection as a Proof of Principle. Sci Rep 2019; 9:15889. [PMID: 31685921 PMCID: PMC6828681 DOI: 10.1038/s41598-019-52375-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/24/2019] [Indexed: 01/08/2023] Open
Abstract
Drug-drug interactions (DDIs), especially with herbal medicines, are complex, making it difficult to identify potential molecular mechanisms and targets. We introduce a workflow to carry out DDI research using transcriptome analysis and interactions of a complex herbal mixture, Compound Kushen Injection (CKI), with cancer chemotherapy drugs, as a proof of principle. Using CKI combined with doxorubicin or 5-Fu on cancer cells as a model, we found that CKI enhanced the cytotoxic effects of doxorubicin on A431 cells while protecting MDA-MB-231 cells treated with 5-Fu. We generated and analysed transcriptome data from cells treated with single treatments or combined treatments and our analysis showed that opposite directions of regulation for pathways related to DNA synthesis and metabolism which appeared to be the main reason for different effects of CKI when used in combination with chemotherapy drugs. We also found that pathways related to organic biosynthetic and metabolic processes might be potential targets for CKI when interacting with doxorubicin and 5-Fu. Through co-expression analysis correlated with phenotype results, we selected the MYD88 gene as a candidate major regulator for validation as a proof of concept for our approach. Inhibition of MYD88 reduced antagonistic cytotoxic effects between CKI and 5-Fu, indicating that MYD88 is an important gene in the DDI mechanism between CKI and chemotherapy drugs. These findings demonstrate that our pipeline is effective for the application of transcriptome analysis to the study of DDIs in order to identify candidate mechanisms and potential targets.
Collapse
Affiliation(s)
- Hanyuan Shen
- Zhendong Australia - China Centre for Molecular Chinese Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Zhipeng Qu
- Zhendong Australia - China Centre for Molecular Chinese Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Yuka Harata-Lee
- Zhendong Australia - China Centre for Molecular Chinese Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Jian Cui
- Zhendong Australia - China Centre for Molecular Chinese Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Thazin Nwe Aung
- Zhendong Australia - China Centre for Molecular Chinese Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Wei Wang
- Zhendong Research Institute, Shanxi-Zhendong Pharmaceutical Co Ltd, Beijing, China
| | - R Daniel Kortschak
- Zhendong Australia - China Centre for Molecular Chinese Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - David L Adelson
- Zhendong Australia - China Centre for Molecular Chinese Medicine, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, 5005, Australia.
| |
Collapse
|
41
|
Zhang J, Qu Z, Yao H, Sun L, Harata-Lee Y, Cui J, Aung TN, Liu X, You R, Wang W, Hai L, Adelson DL, Lin L. An effective drug sensitizing agent increases gefitinib treatment by down regulating PI3K/Akt/mTOR pathway and up regulating autophagy in non-small cell lung cancer. Biomed Pharmacother 2019; 118:109169. [PMID: 31310954 DOI: 10.1016/j.biopha.2019.109169] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023] Open
Abstract
Gefitinib is one of commonly used first-line treatment options for patients with positive EGFR mutation in non-small cell lung cancer (NSCLC). However, most patients with gefitinib treatment relapse over time due to the loss of drug sensitivity. Compound Kushen injection (CKI) has been used to treat lung cancer, including EGFR-mutated NSCLC. In this report, we examined the anti-cancer and drug sensitivity increased activities of CKI in gefitinib less sensitive NSCLC cell lines H1650 and H1975. Bioinformatics analysis was applied to uncover gene regulation and molecular mechanisms of CKI. Our results indicated that when associating with gefitinib in a dose-dependent fashion, CKI demonstrated the ability to inhibit the proliferation and to increase the sensitivity to gefitinib treatment in gefitinib less sensitive cell lines. This could be the results of down regulation of the PI3K/Akt/mTOR pathway and up regulation of autophagy, which were identified as the potential primary targets of CKI to increase gefitinib treatment effect.
Collapse
Affiliation(s)
- Jue Zhang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Zhipeng Qu
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Hong Yao
- Foshan hospital of TCM, Guangzhou University of Chinese Medicine, Foshan, Guangdong Province, PR China
| | - Lingling Sun
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Yuka Harata-Lee
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jian Cui
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Thazin Nwe Aung
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Xiaomin Liu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Rongli You
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, PR China
| | - Wei Wang
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lina Hai
- Zhendong Pharmaceutical Research Institute Co., Ltd., Beijing, PR China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, PR China
| | - David L Adelson
- Department of Genetics and Evolution, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Lizhu Lin
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China.
| |
Collapse
|
42
|
Jiang X, Hou D, Wei Z, Zheng S, Zhang Y, Li J. Extracellular and intracellular microRNAs in pancreatic cancer: from early diagnosis to reducing chemoresistance. ACTA ACUST UNITED AC 2019. [DOI: 10.1186/s41544-019-0014-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
43
|
Cui J, Qu Z, Harata-Lee Y, Nwe Aung T, Shen H, Wang W, Adelson DL. Cell cycle, energy metabolism and DNA repair pathways in cancer cells are suppressed by Compound Kushen Injection. BMC Cancer 2019; 19:103. [PMID: 30678652 PMCID: PMC6345000 DOI: 10.1186/s12885-018-5230-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In this report we examine candidate pathways perturbed by Compound Kushen Injection (CKI), a Traditional Chinese Medicine (TCM) that we have previously shown to alter the gene expression patterns of multiple pathways and induce apoptosis in cancer cells. METHODS We have measured protein levels in Hep G2 and MDA-MB-231 cells for genes in the cell cycle pathway, DNA repair pathway and DNA double strand breaks (DSBs) previously shown to have altered expression by CKI. We have also examined energy metabolism by measuring [ADP]/[ATP] ratio (cell energy charge), lactate production and glucose consumption. Our results demonstrate that CKI can suppress protein levels for cell cycle regulatory proteins and DNA repair while increasing the level of DSBs. We also show that energy metabolism is reduced based on reduced glucose consumption and reduced cellular energy charge. RESULTS Our results validate these pathways as important targets for CKI. We also examined the effect of the major alkaloid component of CKI, oxymatrine and determined that it had no effect on DSBs, a small effect on the cell cycle and increased the cell energy charge. CONCLUSIONS Our results indicate that CKI likely acts through the effect of multiple compounds on multiple targets where the observed phenotype is the integration of these effects and synergistic interactions.
Collapse
Affiliation(s)
- Jian Cui
- Department of Molecular and Biomedical Science, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
- Zhendong Australia - China Centre for Molecular Chinese Medicine, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
| | - Zhipeng Qu
- Department of Molecular and Biomedical Science, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
- Zhendong Australia - China Centre for Molecular Chinese Medicine, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
| | - Yuka Harata-Lee
- Department of Molecular and Biomedical Science, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
- Zhendong Australia - China Centre for Molecular Chinese Medicine, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
| | - Thazin Nwe Aung
- Department of Molecular and Biomedical Science, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
- Zhendong Australia - China Centre for Molecular Chinese Medicine, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
| | - Hanyuan Shen
- Department of Molecular and Biomedical Science, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
- Zhendong Australia - China Centre for Molecular Chinese Medicine, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
| | - Wei Wang
- Zhendong Australia - China Centre for Molecular Chinese Medicine, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
- Zhendong Research Institute, Shanxi-Zhendong Pharmaceutical Co Ltd, Beijing, China
| | - David L. Adelson
- Department of Molecular and Biomedical Science, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
- Zhendong Australia - China Centre for Molecular Chinese Medicine, The University of Adelaide, North Terrace, Adelaide, 5005 South Australia Australia
| |
Collapse
|
44
|
Zhang D, Wu J, Duan X, Wang K, Ni M, Liu S, Zhang X, Zhang B, Zhao Y. Network Meta-Analysis of Chinese Herbal Injections Plus the FOLFOX Regimen for the Treatment of Colorectal Cancer in China. Integr Cancer Ther 2019; 18:1534735419827098. [PMID: 30791732 PMCID: PMC7242776 DOI: 10.1177/1534735419827098] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/30/2018] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE The aim of the present network meta-analysis (NMA) was to explore the comparative effectiveness and safety of different Chinese herbal injections (CHIs) combined with the FOLFOX regimen versus FOLFOX alone for colorectal cancer (CRC). METHODS A comprehensive search for randomized controlled trials (RCTs) was performed with regard to different CHIs for treating CRC in several electronic databases up to July 2016. The quality assessment of the included RCTs was conducted according to the Cochrane risk of bias tool. Standard pair-wise and Bayesian NMA were designed to compare the effectiveness and safety of different CHIs combined with the FOLFOX regimen by utilizing WinBUGS 1.4.3 and Stata 13.1 software, simultaneously. RESULTS Initially, a total of 820 citations were retrieved through comprehensive searching, and 60 eligible articles involving 4849 participants and 14 CHIs were ultimately included. The results of the current evidence indicated that the FOLFOX regimen combined with Delisheng, Kanglaite, Shenqifuzheng, or Aidi injections were associated with the most favorable clinical efficacy compared with the FOLFOX regimen alone. Additionally, the FOLFOX regimen combined with Delisheng, Xiaoaiping, Lentinan, Kangai, Shenqifuzheng, or Aidi injections improved performance status among patients with CRC. CONCLUSIONS The results of cluster analysis demonstrated that the combination of Compound matrine injection and FOLFOX regimen was associated with more preferable and beneficial outcomes than other CHIs groups. Nevertheless, the additional results from multicenter trials and high-quality studies will be pivotal for supporting our findings.
Collapse
Affiliation(s)
- Dan Zhang
- Beijing University of Chinese Medicine,
Beijing, China
| | - Jiarui Wu
- Beijing University of Chinese Medicine,
Beijing, China
| | - Xiaojiao Duan
- Beijing University of Chinese Medicine,
Beijing, China
| | - Kaihuan Wang
- Beijing University of Chinese Medicine,
Beijing, China
| | - Mengwei Ni
- Beijing University of Chinese Medicine,
Beijing, China
| | - Shuyu Liu
- Beijing University of Chinese Medicine,
Beijing, China
| | | | - Bing Zhang
- Beijing University of Chinese Medicine,
Beijing, China
| | - Yi Zhao
- Beijing University of Chinese Medicine,
Beijing, China
| |
Collapse
|
45
|
Wang H, Hu H, Rong H, Zhao X. Effects of compound Kushen injection on pathology and angiogenesis of tumor tissues. Oncol Lett 2018; 17:2278-2282. [PMID: 30719109 PMCID: PMC6351733 DOI: 10.3892/ol.2018.9861] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/07/2018] [Indexed: 12/17/2022] Open
Abstract
Effects of compound Kushen injection on pathology and angiogenesis of tumor tissues were investigated. Forty nude mice were used to establish the liver cancer model of nude mice, and were divided into model group (n=10), low-dose compound Kushen injection group (n=10), medium-dose compound Kushen injection group (n=10) and high-dose compound Kushen injection group (n=10). When the tumor volume reached 0.5 cm3, 200, 400 and 600 µl of compound Kushen were injected into the mice of the low-, medium- and high-dose compound Kushen injection groups, respectively, for 3 consecutive days; while 400 µl normal saline were injected into the mice of the model group. At 9 days after treatment, the mice were sacrificed, and the tumor was taken and weighed. The tumor inhibition rate was calculated, α-smooth muscle actin (α-SMA) and cluster of differentiation 31 (CD31) were detected via immunohistochemistry, and the vascular maturity index (VMI) and microvessel density (MVD) were also detected. With the increase of compound Kushen injection dose, the tumor mass was decreased significantly (P<0.05), and the tumor inhibition rate was obviously increased (P<0.05). In the model group, the nuclei were large and deeply stained, and there were many mitotic figures, and more small blood vessels could be seen. In the three Kushen injection groups, the number of mitotic figures was slightly decreased, and the vascular distribution was reduced. With the increase of compound Kushen injection dose, MVD of transplanted tumor was decreased significantly, but VMI was increased significantly (P<0.05). Compound Kushen injection can reduce the angiogenesis in tumor tissues and play a role in inhibiting tumor growth. Therefore, anti-angiogenesis may be one of the important mechanisms of compound Kushen injection in inhibiting tumor growth.
Collapse
Affiliation(s)
- Hongyan Wang
- Pharmacy of Traditional Chinese Medicine, The First Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| | - Haiyan Hu
- Department of Gastroenterology, The First Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| | - Hua Rong
- Institute of Pharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| | - Xuwei Zhao
- Institute of Pharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang 161000, P.R. China
| |
Collapse
|
46
|
Jin Y, Yang Q, Liang L, Ding L, Liang Y, Zhang D, Wu B, Yang T, Liu H, Huang T, Shen H, Tu H, Pan Y, Wei Y, Yang Y, Zhou F. Compound kushen injection suppresses human acute myeloid leukaemia by regulating the Prdxs/ROS/Trx1 signalling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:277. [PMID: 30454068 PMCID: PMC6245615 DOI: 10.1186/s13046-018-0948-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 10/29/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND The increase in the levels of reactive oxygen species (ROS) in acute myeloid leukemia (AML) patients has been previously described; thus, it is important to regulate ROS levels in AML. METHODS Flow cytometry were used to assess the in vitro effect of compound kushen injection (CKI). Quantitative proteomics were used to analyse the mechanism. The AML patient-derived xenograft (PDX) model were used to evaluate the in vivo effect of CKI. RESULTS We found that intracellular ROS levels in AML cells were decreased, the antioxidant capacity were increased when treated with CKI. CKI inhibited the proliferation of AML cells and enhanced the cytotoxicity of AML cells, which has few toxic effects on haematopoietic stem cells (HSCs) and T cells. At the single-cell level, individual AML cells died gradually by CKI treatment on optofluidic chips. CKI promoted apoptosis and arrested cell cycle at G1/G0 phase in U937 cells. Furthermore, higher peroxiredoxin-3 (Prdx3) expression levels were identified in CKI-treated U937 cells through quantitative proteomics detection. Mechanically, the expression of Prdx3 and peroxiredoxin-2 (Prdx2) was up-regulated in CKI-treated AML cells, while thioredoxin 1 (Trx1) was reduced. Laser confocal microscopy showed that the proteins Prdx2 could be Interacted with Trx1 by CKI treatment. In vivo, the survival was longer and the disease was partially alleviated by decreased CD45+ immunophenotyping in peripheral blood in the CKI-treated group in the AML PDX model. CONCLUSIONS Antioxidant CKI possess better clinical application against AML through the Prdxs/ROS/Trx1 signalling pathway.
Collapse
Affiliation(s)
- Yanxia Jin
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Qian Yang
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Li Liang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, Hubei, China
| | - Lu Ding
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Yuxing Liang
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Dongdong Zhang
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Balu Wu
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Tian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Hailing Liu
- Department of Clinical Haematology, Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Tingting Huang
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Hui Shen
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Honglei Tu
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yi Yang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, Hubei, China
| | - Fuling Zhou
- Department of Haematology, Zhongnan Hospital, Wuhan University, No. 169 Donghu Road, Wuchang District, Wuhan, 430071, Hubei Province, China.
| |
Collapse
|
47
|
Ma H, Huang Q, Qu W, Li L, Wang M, Li S, Chu F. In vivo and in vitro anti-inflammatory effects of Sophora flavescens residues. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:497-503. [PMID: 29913301 DOI: 10.1016/j.jep.2018.06.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/04/2018] [Accepted: 06/14/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The dried roots of Sophora flavescens Ait. (Leguminosae) is traditionally used as antipyretic medicine to reduce inflammation. It is well known that alkaloids and flavonoids are the main constituents of S. flavescens. However, the clinical researches and applications of S. flavescens is mainly based on its water-extracted alkaloids, its flavonoids may still remain in residues and have been underused. With development and manufacturing of S. flavescens in recent years, more herb residues are being produced. Since they are typically treated as waste and dumped openly in landfill sites, which can cause pollution, there is a great need to explore these wastes as recyclable resources and increase their added value. To date, whether other bioactive components would be found in the residues of S. flavescens is still unknown. If the extraction method of these active ingredients was established, the residues of S. flavescens could be turned from the harm to a benefit and make great sense of the comprehensive utilization of S. flavescens resources. This study aimed to establish an extraction method of the residues of S. flavescens and investigate the anti-inflammatory effect of it both in vivo and in vitro. MATERIALS AND METHODS Dried S. flavescens were decocted with distilled water firstly, then the residues were powdered and extracted with ethyl acetate by using ultrasonic wave. HPLC was utilized to analyze the chemical constituents of the water extracts of S. flavescens (WSF) and the ethyl acetate extracts of residues of S. flavescens (RSF). In vivo, the anti-inflammatory effect of WSF and RSF were evaluated using the xylene-induced auricle edema, acetic acid-induced peritoneal permeability and carrageenan-induced hind paw edema methods. In vitro, the inhibitory activities of WSF and RSF on NO, TNF-α, IL-6 and MCP-1 production of LPS-treated RAW264.7 cells were measured. RESULTS The major ingredients of RSF were flavonoids, while WSF almost had no flavonoids. In vivo, WSF and RSF (200 mg/kg) could significantly inhibit the edema in the xylene-induced mice auricle edema and carrageenan-induced hind paw edema as well as the peritoneal permeability increased by acetic acid. They can also lower production levels of PGE2 in inflamed paw tissues. In vitro experimental results showed that RSF (25-100 μg/mL) could significantly inhibit the release of pro-inflammatory cytokines NO, TNF-α, IL-6 and MCP-1 on LPS-induced RAW264.7 cells. The in vitro suppress effect of WSF had no dose-response relationship. CONCLUSIONS The residues of S. flavescens had obvious flavonoids with anti-inflammatory activity. This study provided evidence for the reuse of residues from S. flavescens in the food additive, medicine and cosmetic industries.
Collapse
Affiliation(s)
- Hongyan Ma
- Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials/School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou 510006, PR China.
| | - Qi Huang
- Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials/School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou 510006, PR China.
| | - Wenshan Qu
- Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials/School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou 510006, PR China.
| | - Linyuan Li
- Key Laboratory of State Administration of Traditional Chinese Medicine for Production & Development of Cantonese Medicinal Materials/School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou 510006, PR China.
| | - Min Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances/School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou 510006, PR China.
| | - Shao Li
- Bioinformatics Division, TNLIST and Department of Automation, Tsinghua University, Beijing 100084, PR China.
| | - Fujiang Chu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances/School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, 280 Wai Huan Dong Road, Guangzhou 510006, PR China.
| |
Collapse
|
48
|
Cinobufacini Injection Improves the Efficacy of Chemotherapy on Advanced Stage Gastric Cancer: A Systemic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:7362340. [PMID: 30254688 PMCID: PMC6142757 DOI: 10.1155/2018/7362340] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022]
Abstract
Gastric cancer has a high morbidity and mortality. Chemotherapy regimens are routine advanced stage gastric cancer (AGC) treatment protocols, but most of these drugs have side-effects such as myelosuppression and gastrointestinal disorders. Cinobufacini, an extractive from TCM, could suppress cell proliferation and inhibit gastric cancer. In this study, we comprehensively reviewed the literature on the efficacy comparison between Cinobufacini injection combined with chemotherapy and chemotherapy solely used in AGC treatment. We extracted data for from six electronic databases to evaluate the efficacy of Cinobufacini injection on AGC patients. Twelve studies with a total of 853 patients were finally included in our study. The results indicated that Cinobufacini injection could increase response rate and disease control rate of chemotherapy on AGC, improve the life quality of AGC patients, increase leukocytes, improve anemia, improve hand-foot syndrome induced by chemotherapy, and relieve cancer pain. This study has its own limitations that prevented us from drawing a definite conclusion and more well-designed clinical trials of TCM are needed.
Collapse
|
49
|
Jia L, Lin H, Oppenheim J, Howard OMZ, Li J, Fan H, Zhao Z, Farrar W, Zhang Y, Colburn N, Young MR, Li W, Newman D, O'Keefe BR, Beutler J, Liu J, Hao X, Yang X, Ji T, White JD. US National Cancer Institute-China Collaborative Studies on Chinese Medicine and Cancer. J Natl Cancer Inst Monogr 2018; 2017:4617822. [PMID: 29140488 DOI: 10.1093/jncimonographs/lgx007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/08/2017] [Indexed: 12/13/2022] Open
Abstract
Since 2007, the US National Cancer Institute (NCI) Office of Cancer Complementary and Alternative Medicine (OCCAM), together with the Cancer Institute of the China Academy of Chinese Medical Sciences (CICACMS), institutes at China Academy of Sciences and Chinese Academy of Medical Sciences, have engaged in collaborations on Chinese medicine (CM) and cancer research. Through these collaborations, CM drugs and compounds have been studied at NCI labs. This paper summarizes the discoveries and progress on these research projects, exploring the aspects of cancer prevention, botanical drug mechanisms of action and component analysis/quality control (QC), and anticancer activity screening. These and other related projects have been presented in various jointly convened workshops and have provided the backdrop for establishing a new organization, the International Consortium for CM and Cancer, to promote international collaborations in this field.
Collapse
Affiliation(s)
- Libin Jia
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongsheng Lin
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Joost Oppenheim
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - O M Zack Howard
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Li
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Huiting Fan
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhizheng Zhao
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - William Farrar
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Zhang
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Nancy Colburn
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Matthew R Young
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Weidong Li
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - David Newman
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Barry R O'Keefe
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - John Beutler
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Jikai Liu
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaojiang Hao
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaosheng Yang
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Tengfei Ji
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| | - Jeffrey D White
- Office of Cancer Complementary and Alternative Medicine, Division of Cancer Treatment and Diagnosis, and Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD; Cancer Institute, China Academy of Chinese Medical Sciences, Beijing, China; Laboratory of Molecular Immunoregulation, Laboratory of Cancer Prevention, Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD; Natural Products Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, MD; Kunming Institute of Botany, China Academy of Sciences, Kunming, China; Key Laboratory of Chemistry for Natural Products in Guizhou Province, China Academy of Sciences, Gui Yang, China; Institute of Materia Medica, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
50
|
Identification of candidate anti-cancer molecular mechanisms of Compound Kushen Injection using functional genomics. Oncotarget 2018; 7:66003-66019. [PMID: 27602759 PMCID: PMC5323210 DOI: 10.18632/oncotarget.11788] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/24/2016] [Indexed: 12/31/2022] Open
Abstract
Compound Kushen Injection (CKI) has been clinically used in China for over 15 years to treat various types of solid tumours. However, because such Traditional Chinese Medicine (TCM) preparations are complex mixtures of plant secondary metabolites, it is essential to explore their underlying molecular mechanisms in a systematic fashion. We have used the MCF-7 human breast cancer cell line as an initial in vitro model to identify CKI induced changes in gene expression. Cells were treated with CKI for 24 and 48 hours at two concentrations (1 and 2 mg/mL total alkaloids), and the effect of CKI on cell proliferation and apoptosis were measured using XTT and Annexin V/Propidium Iodide staining assays respectively. Transcriptome data of cells treated with CKI or 5-Fluorouracil (5-FU) for 24 and 48 hours were subsequently acquired using high-throughput Illumina RNA-seq technology. In this report we show that CKI inhibited MCF-7 cell proliferation and induced apoptosis in a dose-dependent fashion. We integrated and applied a series of transcriptome analysis methods, including gene differential expression analysis, pathway over-representation analysis, de novo identification of long non-coding RNAs (lncRNA) as well as co-expression network reconstruction, to identify candidate anti-cancer molecular mechanisms of CKI. Multiple pathways were perturbed and the cell cycle was identified as the potential primary target pathway of CKI in MCF-7 cells. CKI may also induce apoptosis in MCF-7 cells via a p53 independent mechanism. In addition, we identified novel lncRNAs and showed that many of them might be expressed as a response to CKI treatment.
Collapse
|