1
|
Wu Q, Zhu C, Zhao T, Liu T, Da M. Downregulation of LncRNA CCAT1 Enhances Chemosensitivity in Cisplatin-Resistant Gastric Cancer Cells. Drug Dev Res 2025; 86:e70048. [PMID: 39829433 DOI: 10.1002/ddr.70048] [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: 10/02/2024] [Revised: 12/05/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Chemotherapy is an effective treatment for gastric cancer. However, many patients develop resistance to chemotherapeutic agents during clinical treatment. LncRNA CCAT1 has recently been shown to influence cellular resistance to specific chemotherapeutic drugs, but its role in gastric cancer remains underexplored. This study aims to investigate the role of LncRNA CCAT1 in cisplatin resistance in gastric cancer cells and its potential underlying mechanisms. Gastric cancer cell lines with acquired resistance were established. The expression of CCAT1 was assessed in both cisplatin-sensitive and cisplatin-resistant AGS cell lines. CCAT1 expression was knocked down in AGS/DDP cells, and the changes in IC50 values were measured using the Cell Counting Kit-8 (CCK-8) assay. Apoptosis in gastric cancer cells was evaluated by flow cytometry. Additionally, Western blotting was employed to measure the expression levels of PI3K/AKT/mTOR signaling pathway proteins and apoptosis-related proteins in both interference and control groups. RT-qPCR results indicated that CCAT1 expression was significantly elevated in cisplatin-resistant gastric cancer cells compared to non-resistant cells. Similarly, CCK-8 assay results demonstrated that knocking down CCAT1 in resistant cells increased their sensitivity to cisplatin treatment. Flow cytometry and Western blot results further confirmed that silencing CCAT1 promoted apoptosis in these cells. Additionally, the expression of PI3K/AKT/mTOR signaling pathway proteins was higher in resistant cells compared to their sensitive counterparts, and silencing CCAT1 in AGS/DDP cells resulted in reduced expression of these proteins. In conclusion, the above studies demonstrated that LncRNA CCAT1 induced cisplatin resistance in gastric cancer cells.
Collapse
Affiliation(s)
- Qiong Wu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Chenglou Zhu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Tiantian Zhao
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Tianxiang Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China
| | - Mingxu Da
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
- The First School of Clinical Medicine, Gansu University of Chinese Medicine, Lanzhou, China
- Department of Surgical Oncology, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
2
|
Cui M, Zhang Y, Tang Y, Fan Q, Chen X, Li J, Qiao C, Chen X, Lin R, Yu X, Zhao C. Hepatotoxicity of Phytolacca acinosa Roxb mediated by phytolaccagenin via ferroptosis/PPAR/P53/arachidonic acid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156433. [PMID: 39892312 DOI: 10.1016/j.phymed.2025.156433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/18/2025] [Accepted: 01/25/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND The traditional Chinese medicine Phytolacca acinosa Roxb (PAR), known as Shanglu, possesses recognized therapeutic benefits against many diseases. PAR is also hepatotoxic, making it a major public health problem. However, the specific toxic substances and molecular mechanisms of PAR remain unclear. Therefore, appropriate animal models and methods are essential to confirm the toxic components and related mechanisms of PAR. METHODS L-02 cells and zebrafish larvae at 4 days post-fertilization (4 dpf) were used as models and treated with various concentrations of phytolaccagenin (Phy), esculentoside A (EsA), and esculentoside H (EsH). The hepatotoxicity of three samples was assessed based on liver phenotype, pathological assessments, and biochemical index in zebrafish and proliferative activity, apoptosis level, and biochemical index in L02 cells. The transcriptomic technique was used to explore the related signaling pathways and potential mechanisms in vitro and in zebrafish , and the findings were validated by RT-PCR. RESULTS The results of acute toxicity tests indicated that Phy exhibited substantially more severe hepatotoxicity than EsA, while EsH did not lead to any obvious toxic effects. Especially, under sublethal exposure ( CONCLUSION This study identified Phy as a key hepatotoxic component of PAR. Furthermore, using transcriptomic techniques, we preliminarily investigated the hepatotoxic mechanisms of Phy in vitro and in vivo. The results of the present study showed that Phy affects several signaling pathways, including those involved in lipid metabolism, oxidative stress, and apoptosis, finally leading to hepatotoxicity. These findings provide invaluable insights into the safe use of PAR in clinical settings.
Collapse
Affiliation(s)
- Muyao Cui
- Beijing University of Chinese Medicine, Beijing 100029, China; Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yao Zhang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Tang
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qiqi Fan
- Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaolu Chen
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China
| | - Jiaqi Li
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China
| | - Chuanqi Qiao
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China
| | - Xue Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ruichao Lin
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China.
| | - Xue Yu
- Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Chongjun Zhao
- Beijing University of Chinese Medicine, Beijing 100029, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing 100102, China; Traditional Chinese Medicine Processing Technology Inheritance Base of National Administration of Traditional Chinese Medicine, China.
| |
Collapse
|
3
|
Chen Z, Yu T, Wang Y, Li J, Zhang B, Zhou L. Mechanistic insights into the role of traditional Chinese medicine in treating gastric cancer. Front Oncol 2025; 14:1443686. [PMID: 39906672 PMCID: PMC11790455 DOI: 10.3389/fonc.2024.1443686] [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: 06/04/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Gastric cancer remains a leading cause of cancer-related mortality worldwide, with advanced stages presenting significant challenges due to metastasis and drug resistance. Traditional Chinese Medicine (TCM) offers a promising complementary approach characterized by holistic treatment principles and minimal side effects. This review comprehensively explores the multifaceted mechanisms by which TCM addresses gastric cancer. Specifically, we detail how TCM inhibits aerobic glycolysis by downregulating key glycolytic enzymes and metabolic pathways, thereby reducing the energy supply essential for cancer cell proliferation. We examine how TCM suppresses angiogenesis by targeting the vascular endothelial growth factor (VEGF) and cyclooxygenase-2 (COX-2) pathways, effectively starving tumors of nutrients and oxygen required for growth and metastasis. Furthermore, TCM modulates the immune microenvironment by enhancing the activity of effector immune cells such as CD4+ and CD8+ T cells and natural killer (NK) cells while reducing immunosuppressive cells like regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). These actions collectively contribute to slowing tumor progression, inhibiting metastasis, and enhancing the body's antitumor response. The insights presented underscore the significant potential of TCM as an integral component of comprehensive gastric cancer treatment strategies, highlighting avenues for future research and clinical application to improve patient outcomes.
Collapse
Affiliation(s)
- Ziqiang Chen
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ting Yu
- Department of Rheumatism, Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yunhe Wang
- Department of Endocrinology, Metabolism and Gastroenterology, Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jiaxin Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Bo Zhang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liya Zhou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
4
|
Cheng W, Feng W, Tian G, Liu J, Ba Z, Yu M, Yan R, Liu L, He Y, Li X, Zhang J. Study of Serum Metabolic Biomarkers and Prediction Models of Cantharidin-Induced Nephrotoxicity in Rats Based on Dynamic Metabolomics. J Appl Toxicol 2024. [PMID: 39676217 DOI: 10.1002/jat.4743] [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: 08/05/2024] [Revised: 11/10/2024] [Accepted: 11/24/2024] [Indexed: 12/17/2024]
Abstract
The clinical application of cantharidin (CTD) is seriously limited due to its nephrotoxicity. Therefore, this study aims to investigate sensitive biomarkers for the evaluation and prediction of nephrotoxicity induced by CTD in rat. A total of 80 rats were randomly divided into four groups: control group and three doses of CTD groups. After 0, 1, 5, 15, and 28 days of intragastric administration, rat serum and urine were collected for biochemical indexes, then serum was used for metabolomic analyses, and rat kidney was collected for pathological and ultrastructural observation. The levels of serum crea (Scr), blood urea nitrogen (BUN), urea, urine crea (Ucrea), and urinary microalbumin (UmALB) were significantly increased after administration of different doses of CTD (p < 0.05). Additionally, histopathology and cell ultrastructure observation of kidney showed significant cell inflammatory infiltration and glomerular edema. Seven metabolic biomarkers including 6-hydroxymelatonin were significantly disturbed by CTD. The CatBoost Classifier prediction model was used to establish the CTD nephrotoxicity prediction model, and the prediction accuracy and precision were 0.645 and 0.640, respectively. Moreover, 6-hydroxymelatonin was found to be most useful biomarkers for evaluating the CTD nephrotoxicity. Finally, the seven metabolic biomarkers were found mainly involved in pyruvate metabolism, pantothenate and CoA biosynthesis.
Collapse
Affiliation(s)
- Weina Cheng
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Wenzhong Feng
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Guanghuan Tian
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jingxian Liu
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Zhixun Ba
- Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ming Yu
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Rong Yan
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Liu Liu
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Yanmei He
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Xiaofei Li
- School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Jianyong Zhang
- School of Pharmacy, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry Education and Joint International Research Laboratory of Ethnomedicine Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
5
|
Liu Y, Yin Q, Liu B, Lu Z, Liu M, Meng L, He C, Chang J. Fisetin reduces ovalbumin-triggered airway remodeling by preventing phenotypic switching of airway smooth muscle cells. Respir Res 2024; 25:370. [PMID: 39402516 PMCID: PMC11479573 DOI: 10.1186/s12931-024-03005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND The transformation of airway smooth muscle cells (ASMCs) from a quiescent phenotype to a hypersecretory and hypercontractile phenotype is a defining feature of asthmatic airway remodeling. Fisetin, a flavonoid compound, possesses anti-inflammatory characteristics in asthma; yet, its impact on airway remodeling and ASMCs phenotype transition has not been investigated. OBJECTIVES This research seeked to assess the impact of fisetin on ovalbumin (OVA) induced asthmatic airway remodeling and ASMCs phenotype transition, and clarify the mechanisms through network pharmacology predictions as well as in vivo and in vitro validation. METHODS First, a fisetin-asthma-ASMCs network was constructed to identify potential targets. Subsequently, cellular and animal studies were carried out to examine the inhibitory effects of fisetin on airway remodeling in asthmatic mice, and to detemine how fisetin impacts the phenotypic transition of ASMCs. RESULTS Network analysis indicated that fisetin might affect asthma via mediating the phosphatidylinositol 3-kinase (PI3K)/ protein kinase B (AKT) pathway. Intraperitoneal administration of fisetin in vivo reduced airway inflammation and remodeling, as shown by reduced inflammatory cells, decreased T helper type 2 (Th2) cytokine release, diminished collagen accumulation, mitigated airway smooth muscle thickening, and decreased expression of osteopontin (OPN), collagen-I and α-smooth muscle actin (α-SMA). Moreover, fisetin suppressed the PI3K/AKT pathway in asthmatic lung tissue. According to the in vitro data, fisetin downregulated the expression of the synthetic phenotypic proteins OPN and collagen-I, contractile protein α-SMA, and inhibited cellular migration, potentially through the PI3K/AKT pathway. CONCLUSION These results suggest that fisetin inhibits airway remodeling in asthma by regulating ASMCs phenotypic shift, emphasizing that fisetin is a promising candidate for the treatment of airway smooth muscle remodeling.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Qiling Yin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Bin Liu
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Zheng Lu
- Tai'an Tumour Prevention and Treatment Hospital, Tai'an, Shandong, 271000, China
| | - Meijun Liu
- Shandong First Medical University, Tai'an, Shandong, 271000, China
| | - Ling Meng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China.
| | - Chao He
- Department of Gastrointestinal Surgery, The Affiliated Taian City Central Hospital of Qingdao University, Tai'an, Shandong, 271000, China.
| | - Jin Chang
- Department of Oncology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong, 271000, China.
| |
Collapse
|
6
|
Jiao W, Li H, Wu Y, Wen Q, Wang W, Tian J, Ren Y, Ma J, Zhao D, Zhao J, Zhang Y, Han G. Dual Targeted Nanoparticles Encapsulating Cantharidin for Treatment of Hepatocellular Carcinoma and Lymphatic Metastasis. ACS APPLIED NANO MATERIALS 2024; 7:20609-20625. [DOI: 10.1021/acsanm.4c03587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- Wenwen Jiao
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Hao Li
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Yingjie Wu
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Qing Wen
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Wenzhen Wang
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Jia Tian
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Yulong Ren
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Jinyuan Ma
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Danxiang Zhao
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Junli Zhao
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Yu Zhang
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| | - Guang Han
- Henan Province Engineering Research Center of High Value Utilization to Natural Medical Resource in Yellow River Basin, School of Pharmacy, Henan University, Kaifeng 475004, China
- State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, China
- Kaifeng Key Lab for Application of Local Chrysanthemum Morifolium in Food & Drug, Kaifeng 475004, China
| |
Collapse
|
7
|
Li P, Ma X, Gu X. The essential roles of lncRNAs/PI3K/AKT axis in gastrointestinal tumors. Front Cell Dev Biol 2024; 12:1442193. [PMID: 39161590 PMCID: PMC11330846 DOI: 10.3389/fcell.2024.1442193] [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: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
The role of long noncoding RNA (lncRNA) in tumors, particularly in gastrointestinal tumors, has gained significant attention. Accumulating evidence underscores the interaction between various lncRNAs and diverse molecular pathways involved in cancer progression. One such pivotal pathway is the PI3K/AKT pathway, which serves as a crucial intracellular mechanism maintaining the balance among various cellular physiological processes for normal cell growth and survival. Frequent dysregulation of the PI3K/AKT pathway in cancer, along with aberrant activation, plays a critical role in driving tumorigenesis. LncRNAs modulate the PI3K/AKT signaling pathway through diverse mechanisms, primarily by acting as competing endogenous RNA to regulate miRNA expression and associated genes. This interaction significantly influences fundamental biological behaviors such as cell proliferation, metastasis, and drug resistance. Abnormal expression of numerous lncRNAs in gastrointestinal tumors often correlates with clinical outcomes and pathological features in patients with cancer. Additionally, these lncRNAs influence the sensitivity of tumor cells to chemotherapy in multiple types of gastrointestinal tumors through the abnormal activation of the PI3K/AKT pathway. These findings provide valuable insights into the mechanisms underlying gastrointestinal tumors and potential therapeutic targets. However, gastrointestinal tumors remain a significant global health concern, with increasing incidence and mortality rates of gastrointestinal tumors over recent decades. This review provides a comprehensive summary of the latest research on the interactions of lncRNA and the PI3K/AKT pathway in gastrointestinal tumor development. Additionally, it focuses on the functions of lncRNAs and the PI3K/AKT pathway in carcinogenesis, exploring expression profiles, clinicopathological characteristics, interaction mechanisms with the PI3K/AKT pathway, and potential clinical applications.
Collapse
Affiliation(s)
- Penghui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiao Ma
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
8
|
Safenraiter ME, Soldini MPC, Del Río MG. Cantharidin: A Multiporpuse Beetlejuice. NEOTROPICAL ENTOMOLOGY 2024; 53:964-971. [PMID: 38750300 DOI: 10.1007/s13744-024-01164-3] [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: 12/06/2023] [Accepted: 05/08/2024] [Indexed: 07/18/2024]
Abstract
Cantharidin is produced by beetles of two families, Meloidae (true blister beetles) and Oedemeridae (false blister beetles). Nevertheless, it is mainly members of the meloid family that have been widely studied in the traditional medicines and pharmacology of different cultures and countries. The meloids cantharidin's role is going to be reviewed in this paper, including the cantharidin discovery, its adaptative function, and worldwide uses. Finally, we recovered information on the implementation of this compound in South American civilizations in different therapeutic treatments as well as sexual stimulants and aphrodisiacs.
Collapse
Affiliation(s)
- Melania Edith Safenraiter
- Laboratorio de Entomología, CICYTTP-CONICET/UADER/Gob. Entre Ríos, Materi y España, Diamante, Entre Ríos, Argentina.
| | - María Paula Campos Soldini
- Laboratorio de Entomología, CICYTTP-CONICET/UADER/Gob. Entre Ríos, Materi y España, Diamante, Entre Ríos, Argentina
| | - María Guadalupe Del Río
- División Entomología, Museo de La Plata, Univ Nacional de La Plata, CONICET, La Plata, Buenos Aires, Argentina
| |
Collapse
|
9
|
Liu R, Yang C, Yang X, Yu J, Tang W. Network toxicology, molecular docking technology, and experimental verification revealed the mechanism of cantharidin-induced testicular injury in mice. Toxicol Appl Pharmacol 2024; 486:116921. [PMID: 38582374 DOI: 10.1016/j.taap.2024.116921] [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: 11/13/2023] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
As a protein kinase inhibitor, cantharidin (CTD) exhibits antitumor activities. However, CTD is highly toxic, thereby limiting clinical applications. Moreover, relatively few studies have investigated CTD-induced reproductive toxicity, thus the underlying mechanism remains unclear. In this study, the toxic effects of CTD on mouse testis were confirmed in vivo and the potential mechanism was predicted by network toxicology (NT) and molecular docking technology. Proteins involved in the signaling pathways and core targets were verified. The results showed that different concentrations of CTD induced weight loss increased the testicular coefficient, and caused obvious pathological damage to testicular cells. The NT results showed that the main targets of CTD-induced testicular injury (TI) included AKT1, Caspase 3, Bcl-2, and Bax. The results of pathway enrichment analysis showed that CTD-induced TI was closely related to apoptosis and the PI3K/AKT and HIF-1 signaling pathways. Molecular docking methods confirmed high affinity between CTD and key targets. Western blot analysis showed that CTD inhibited expression of PI3K, AKT, and the anti-apoptotic protein Bcl-2, while promoting expression of the pro-apoptotic proteins Bax and Caspase 3. These results suggest that CTD-induced TI involves multiple targets and pathways, and the underlying mechanism was associated with inhibition of the apoptosis-related PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Ruxia Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Changfu Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xin Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Jia Yu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Wenchao Tang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
10
|
Wu Y, Liu Z, He Z, Yi J, Qiao X, Tan C, Xing Y, Zeng Y, Yang D, Yin J, Fan B, Zeng G. Cantharidin analogue alleviates dextran sulfate sodium-induced colitis in mice by inhibiting the activation of NF-κB signaling. Eur J Med Chem 2023; 260:115731. [PMID: 37643546 DOI: 10.1016/j.ejmech.2023.115731] [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: 04/23/2023] [Revised: 07/28/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
Ulcerative colitis is a chronic inflammatory disease with a remitting-relapsing clinical course, it has evolved into a global burden given its high incidence worldwide. Cantharidin (CTD) derivatives are a class of compounds whose structures characterized with a 7-oxabicyclo [2.2.1]heptane core. Though potent cytotoxicity CTD and its derivatives showed, their clinical usage as anti-cancer drugs was limited by the toxicity in organs. In order to find new CTD analogues with good activity and lower toxicity, 21 CTD analogues with or without alkynyl substitution at C5 position of 7-oxabicyclo [2.2.1]heptane core were synthesized, some compounds showed better in vitro anti-inflammatory activity compared to CTD and norcantharidin (NCTD). Based on the structure-activity relationship results of in vitro experiment, analogue 3i was chosen for further study. Results from the acute toxicity in mice showed that 3i was hypotoxic with the single-dose MTD (maximum tolerated dose) for oral administration is over 1852 mg/kg, at least 35-fold lower than that of NCTD. Mechanism study indicated that 3i could potently inhibit TNF-α induced activation of NF-κB signaling by down-regulation the expression levels of phosphor- IKK, IκBα, and NF-κB p65, and alleviated dextran sulfate sodium-induced colitis in mice. This study indicated that CTD analogues with alkynyl substitution at C5 position of 7-oxabicyclo [2.2.1]heptane core is a kind of new compounds with good anti-inflammatory activity and lower toxicity in vivo, and might be used as therapeutic agents for inflammatory diseases.
Collapse
Affiliation(s)
- Yihang Wu
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650504, China
| | - Zixiu Liu
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650504, China
| | - Zhenxiu He
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650504, China
| | - Jumei Yi
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650504, China
| | - Xingfang Qiao
- Chongqing Key Laboratory of Traditional Chinese Medicine Health, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Chunbin Tan
- Chongqing Key Laboratory of Traditional Chinese Medicine Health, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Yajing Xing
- Chongqing Key Laboratory of Traditional Chinese Medicine Health, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Yaobo Zeng
- Chongqing Key Laboratory of Traditional Chinese Medicine Health, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China
| | - Dajian Yang
- Chongqing Key Laboratory of Traditional Chinese Medicine Health, Chongqing Academy of Chinese Materia Medica, Chongqing 400065, China.
| | - Junlin Yin
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650504, China
| | - Baomin Fan
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650504, China.
| | - Guangzhi Zeng
- Key Laboratory of Chemistry in Ethnic Medicinal Resources, State Ethnic Affairs Commission and Ministry of Education, Yunnan Minzu University, Kunming 650504, China.
| |
Collapse
|
11
|
Liao Y, Gui Y, Li Q, An J, Wang D. The signaling pathways and targets of natural products from traditional Chinese medicine treating gastric cancer provide new candidate therapeutic strategies. Biochim Biophys Acta Rev Cancer 2023; 1878:188998. [PMID: 37858623 DOI: 10.1016/j.bbcan.2023.188998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/26/2023] [Accepted: 10/08/2023] [Indexed: 10/21/2023]
Abstract
Gastric cancer (GC) is one of the severe malignancies with high incidence and mortality, especially in Eastern Asian countries. Significant advancements have been made in diagnosing and treating GC over the past few decades, resulting in tremendous improvements in patient survival. In recent years, traditional Chinese medicine (TCM) has garnered considerable attention as an alternative therapeutic approach for GC due to its multicomponent and multitarget characteristics. Consequently, natural products found in TCM have attracted researchers' attention, as growing evidence suggests that these natural products can impede GC progression by regulating various biological processes. Nevertheless, their molecular mechanisms are not systematically uncovered. Here, we review the major signaling pathways involved in GC development. Additionally, clinical GC samples were analyzed. Moreover, the anti-GC effects of natural products, their underlying mechanisms and potential targets were summarized. These summaries are intended to facilitate further relevant research, and accelerate the clinical applications of natural products in GC treatment.
Collapse
Affiliation(s)
- Yile Liao
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Gui
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Qingzhou Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jun An
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dong Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
12
|
Deng Z, Gao Y, Nguyen T, Chai J, Wu J, Li J, Abdel-Rahman MA, Xu X, Chen X. The Potent Antitumor Activity of Smp43 against Non-Small-Cell Lung Cancer A549 Cells via Inducing Membranolysis and Mitochondrial Dysfunction. Toxins (Basel) 2023; 15:toxins15050347. [PMID: 37235381 DOI: 10.3390/toxins15050347] [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: 04/10/2023] [Revised: 05/06/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Research has been conducted to investigate the potential application of scorpion venom-derived peptides in cancer therapy. Smp43, a cationic antimicrobial peptide from Scorpio maurus palmatus venom, has been found to exhibit suppressive activity against the proliferation of multiple cancer cell lines. However, its impact on non-small-cell lung cancer (NSCLC) cell lines has not been previously investigated. This study aimed to determine the cytotoxicity of Smp43 towards various NSCLC cell lines, particularly A549 cells with an IC50 value of 2.58 μM. The results indicated that Smp43 was internalized into A549 cells through membranolysis and endocytosis, which caused cytoskeleton disorganization, a loss of mitochondrial membrane potential, an accumulation of reactive oxygen species (ROS), and abnormal apoptosis, cell cycle distribution, and autophagy due to mitochondrial dysfunction. Additionally, the study explored the in vivo protective effect of Smp43 in xenograft mice. The findings suggest that Smp43 has potential anticarcinoma properties exerted via the inducement of cellular processes related to cell membrane disruption and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Ze Deng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yahua Gao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tienthanh Nguyen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiali Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | | | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
13
|
Yan J, Deng XL, Ma SQ, Hui Li Y, Gao YM, Shi GT, Wang HS. Cantharidin suppresses hepatocellular carcinoma development by regulating EZH2/H3K27me3-dependent cell cycle progression and antitumour immune response. BMC Complement Med Ther 2023; 23:160. [PMID: 37202806 DOI: 10.1186/s12906-023-03975-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/25/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Cantharidin (CTD) is a major ingredient of cantharis (Mylabris phalerata Pallas) and has been used extensively in traditional Chinese medicines. It has been shown to exhibit anticancer activity in multiple types of cancer, especially hepatocellular carcinoma (HCC). However, there is no systematic study on the relationships among the regulatory networks of its targets in HCC therapy. We focused on histone epigenetic regulation and the influence of CTD on the immune response in HCC. METHODS We performed a comprehensive analysis of novel CTD targets in HCC based on network pharmacology and RNA-seq approaches. The mRNA levels of target genes were analyzed by qRT-PCR, and the corresponding protein levels were confirmed using enzyme-linked immunosorbent assay (ELISA) and immunohistochemical staining (IHC). ChIP-seq data were visualized by IGV software. The associations of gene transcript levels with the cancer immune score and infiltration level were investigated using TIMER. In vivo, the H22 mouse model of hepatocellular carcinoma was established by treatment with CTD and 5-Fu. The immune cell proportions in the blood were elevated in model mice, as shown by flow cytometry. RESULTS We identified 58 targets of CTD, which were involved in various pathways in cancer, including apoptosis, the cell cycle, EMT and immune pathways. Moreover, we found that 100 EMT-related genes were differentially expressed after CTD treatment in HCC cells. Interestingly, our results confirmed that the EZH2/H3K27me3 -related cell cycle pathway is a therapeutic target of CTD in antitumour. In addition, we evaluated the influence of CTD on the immune response. Our data showed that the significantly enriched gene sets were positively correlated with the chemokine biosynthetic and chemokine metabolic modules. The proportions of CD4+/CD8 + T cells and B cells were increased, but the proportion of Tregs was decreased after treatment with CTD in vivo. Moreover, we found that the expression of the inflammatory factor and immune checkpoint genes PD-1/PD-L1 was significantly reduced in the mouse model. CONCLUSION We performed a novel integrated analysis of the potential role of CTD in HCC treatment. Our results provide innovative insight into the mechanism by which cantharidin exerts antitumour effects by regulating target genes expression to mediate apoptosis, EMT, cell cycle progression and the immune response in HCC. Based on the effect of CTD on the immune response, it can be used as a potential effective drug to activate antitumour immunity for the treatment of liver cancer.
Collapse
Affiliation(s)
- Jia Yan
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiu Ling Deng
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Shi Qi Ma
- Inner Mongolia Hospital of Traditional Chinese Medicine, Hohhot, Inner Mongolia, China
| | - Yu Hui Li
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yu Min Gao
- School of Public health, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Gui Tao Shi
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| | - Hai Sheng Wang
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
14
|
Hu J, Cheng M, Li Y, Shi B, He S, Yao Z, Jiang J, Yu H, He Z, Zhao Y, Zheng H, Hua B, Liu R. Ginseng-containing traditional medicine preparations in combination with fluoropyrimidine-based chemotherapy for advanced gastric cancer: A systematic review and meta-analysis. PLoS One 2023; 18:e0284398. [PMID: 37068063 PMCID: PMC10109524 DOI: 10.1371/journal.pone.0284398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
BACKGROUND Ginseng-containing traditional medicine preparations (G-TMPs) in combination with fluoropyrimidine-based chemotherapy (FBC) are well-known treatments for advanced gastric cancer (AGC), with a superior efficacy to FBC alone. However, evidence regarding their efficacy remains limited. The purpose of this meta-analysis is to evaluate the efficacy and safety of G-TMPs in combination with FBC for the treatment of AGC. METHODS Eight electronic databases were searched for randomized controlled trials (RCTs) using G-TMPs with FBC for the treatment of AGC. The primary outcome included the tumor response, while the secondary outcomes included the quality of life (QoL), proportions of peripheral blood lymphocytes, adverse drug reactions (ADRs), and levels of cancer biomarkers. The quality of evidence for each outcome was assessed using GRADE profilers. RESULTS A total of 1,960 participants were involved in the 26 RCTs included. Patients treated with FBC plus G-TMPs had better objective response (risk ratio [RR] = 1.23, 95% confidence interval [CI]: 1.13 to 1.35, p < 0.00001) and disease control (RR = 1.13, 95% CI: 1.08 to 1.19, p < 0.00001) rates than those treated with FBC alone. Additionally, the combination group had a better QoL, higher proportions of CD3+ T cells, CD4+ T cells, and natural killer cells, as well as a higher CD4+/CD8+ T-cell ratio. Furthermore, lower levels of CA19-9, CA72-4, and CEA were confirmed in the combination treatment group. In addition, G-TMPs reduced the incidence of ADRs during chemotherapy. CONCLUSION In combination with FBC, G-TMPs can potentially enhance efficacy, reduce ADRs, and improve prognosis for patients with AGC. However, high-quality randomized studies remain warranted. SYSTEMATIC REVIEW REGISTRATION PROSPERO Number: CRD42021264938.
Collapse
Affiliation(s)
- Jiaqi Hu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Mengqi Cheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yue Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Bolun Shi
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shulin He
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ziang Yao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Juling Jiang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huibo Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongning He
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuwei Zhao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Zhai BT, Sun J, Shi YJ, Zhang XF, Zou JB, Cheng JX, Fan Y, Guo DY, Tian H. Review targeted drug delivery systems for norcantharidin in cancer therapy. J Nanobiotechnology 2022; 20:509. [DOI: 10.1186/s12951-022-01703-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022] Open
Abstract
AbstractNorcantharidin (NCTD) is a demethylated derivative of cantharidin (CTD), the main anticancer active ingredient isolated from traditional Chinese medicine Mylabris. NCTD has been approved by the State Food and Drug Administration for the treatment of various solid tumors, especially liver cancer. Although NCTD greatly reduces the toxicity of CTD, there is still a certain degree of urinary toxicity and organ toxicity, and the poor solubility, short half-life, fast metabolism, as well as high venous irritation and weak tumor targeting ability limit its widespread application in the clinic. To reduce its toxicity and improve its efficacy, design of targeted drug delivery systems based on biomaterials and nanomaterials is one of the most feasible strategies. Therefore, this review focused on the studies of targeted drug delivery systems combined with NCTD in recent years, including passive and active targeted drug delivery systems, and physicochemical targeted drug delivery systems for improving drug bioavailability and enhancing its efficacy, as well as increasing drug targeting ability and reducing its adverse effects.
Graphical Abstract
Collapse
|
16
|
Xu Y, Wang M, Ning S, Yang Z, Zhou L, Xia X. Development of Glycyrrhetinic Acid and Folate Modified Cantharidin Loaded Solid Lipid Nanoparticles for Targeting Hepatocellular Carcinoma. Molecules 2022; 27:molecules27206786. [PMID: 36296377 PMCID: PMC9610810 DOI: 10.3390/molecules27206786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/01/2022] [Accepted: 10/09/2022] [Indexed: 11/22/2022] Open
Abstract
Cantharidin (CTD) is the major component of anticancer drugs obtained from Mylabris Cichorii and has a good inhibitory effect on several cancers, including hepatocellular carcinoma (HCC) and breast cancer. However, due to its toxicity, oral administration can cause various adverse reactions, limiting its clinical application. The aim of this work was to design glycyrrhetinic acid (GA)- and/or folate (FA)-modified solid lipid nanoparticles (SLNs) for the encapsulation of CTD to target HCC. Four CTD-loaded SLNs (cantharidin solid lipid nanoparticles (CSLNs), glycyrrhetinic acid-modified cantharidin solid lipid nanoparticles (GA-CSLNs), folate-modified cantharidin solid lipid nanoparticles (FA-CSLNs), and glycyrrhetinic acid and folate-modified cantharidin solid lipid nanoparticles (GA-FA-CSLNs)) were prepared by the emulsion ultrasonic dispersion method, and their physicochemical parameters were determined (particle size and distribution, morphology, zeta-potential, entrapment efficiency, drug loading, and hemolysis). Additionally, the antitumor activities of the four SLNs were evaluated comprehensively by tests for cytotoxicity, cell migration, cell cycle, apoptosis, cellular uptake, competition suppression assay, and in vivo tumor suppression assay. Four SLNs showed spherical shapes and mean diameters in the range of 75–110 nm with size dispersion (PDI) within the range of 0.19–0.50 and zeta-potential approximately –10 mV. The entrapment efficiency of CTD in SLNs was higher than 95% for all tested formulations, and no hemolysis was observed. Compared to GA-CSLNs or CSLNs, GA-FA-CSLNs and FA-CSLNs showed stronger cytotoxicity on hepatocellular carcinoma cells (HepG2), and the cytotoxicity of GA-FA-CSLNs on hepatocyte cells (L-02) was remarkably reduced compared with other formulations. GA-FA-CSLNs and FA-CSLNs also increased the inhibition of HepG2 cell migration, and FA-CSLNs had the highest apoptosis rate. The cell cycle results indicated that HepG2 cells were arrested mainly in the S phase and G2/M phase. Analysis of competition inhibition experiments showed that GA and FA ligands had targeted effects on HepG2 cells. The in vivo tumor inhibition experiment showed that GA-FA-CSLNs and FA-CSLNs had excellent tumor inhibition ability—their tumor inhibition rates were 96.46% and 89.92%, respectively. Our results indicate that GA-FA-CSLNs and FA-CSLNs have a promising future in the therapeutic intervention of HCC.
Collapse
Affiliation(s)
- Yilin Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Min Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- The Second Hospital of Hunan University of Chinese Medicine, Changsha 410005, China
| | - Shuangcheng Ning
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhonglan Yang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Correspondence: (L.Z.); (X.X.); Tel.: +86-138-7596-5134 (L.Z.); +86-139-7313-1320 (X.X.)
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Correspondence: (L.Z.); (X.X.); Tel.: +86-138-7596-5134 (L.Z.); +86-139-7313-1320 (X.X.)
| |
Collapse
|
17
|
Yu C, Li Y, Chen G, Wu C, Wang X, Zhang Y. Bioactive constituents of animal-derived traditional Chinese medicinal materials for breast cancer: opportunities and challenges. J Zhejiang Univ Sci B 2022; 23:547-563. [PMID: 35794685 PMCID: PMC9264107 DOI: 10.1631/jzus.b2101019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/20/2022] [Indexed: 11/11/2022]
Abstract
Breast cancer is globally the most common invasive cancer in women and remains one of the leading causes of cancer-related deaths. Surgery, radiotherapy, chemotherapy, immunotherapy, and endocrine therapy are currently the main treatments for this cancer type. However, some breast cancer patients are prone to drug resistance related to chemotherapy or immunotherapy, resulting in limited treatment efficacy. Consequently, traditional Chinese medicinal materials (TCMMs) as natural products have become an attractive source of novel drugs. In this review, we summarized the current knowledge on the active components of animal-derived TCMMs, including Ophiocordycepssinensis-derived cordycepin, the aqueous and ethanolic extracts of O.sinensis, norcantharidin (NCTD), Chansu, bee venom, deer antlers, Ostreagigas, and scorpion venom, with reference to marked anti-breast cancer effects due to regulating cell cycle arrest, proliferation, apoptosis, metastasis, and drug resistance. In future studies, the underlying mechanisms for the antitumor effects of these components need to be further investigated by utilizing multi-omics technologies. Furthermore, large-scale clinical trials are necessary to validate the efficacy of bioactive constituents alone or in combination with chemotherapeutic drugs for breast cancer treatment.
Collapse
Affiliation(s)
- Chaochao Yu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yi Li
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Guopeng Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Chaoyan Wu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xiuping Wang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yingwen Zhang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
18
|
Impact of Amarogentin on Gastric Carcinoma Cell Multiplication, Apoptosis and Migration via circKIF4A/miR-152-3p. J Immunol Res 2022; 2022:2156204. [PMID: 35747689 PMCID: PMC9213178 DOI: 10.1155/2022/2156204] [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: 04/15/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 11/18/2022] Open
Abstract
Objective The active ingredients extracted from natural plants have anti-GC actions and can slow down gastric carcinoma (GC) progression. To investigate the impact of Amarogentin (AG) on GC cell multiplication, apoptosis and migration and the possible mechanisms. Methods qRT-PCR quantification of circKIF4A and miR-152-3p in GC tissues and normal counterparts as well as HGC-27 (human GC cell strain) and GES-1 (human gastric mucosal epithelial cell strain) was performed. HGC-27 cells were intervened by AG of various concentrations. si-NC, si-circKIF4A were further transfected into HGC-27 cells. Besides, pcDNA and pcDNA-circKIF4A were transfected into HGC-27 cells, after which 60 mmol/L AG was added for intervention. Cell multiplication, clone formation, as well as apoptosis and migration measurements were made by MTT, plate clone formation, flow cytometry and Transwell assays, respectively; Double luciferase reporter assay was performed for targeting relationship identification between circKIF4A and miR-152-3p; Western blots were carried out to measure Bax and Bcl-2 protein levels. Results circKIF4A increased (P <0.05) and miR-152-3p decreased (P <0.05) in GC tissues and cell strains. Concentration-dependently, AG intervention contributed to enhanced cell multiplication inhibitory rate, apoptosis rate, miR-152-3p expression and Bax protein level (P <0.05), together with declined number of cell clones formed, migrating cells, circKIF4A expression and Bcl-2 protein level (P <0.05). After transfection of si-circKIF4A, cell multiplication inhibition rate, apoptosis rate and Bax protein level enhanced (P <0.05), while cell clones formed and migrating cells as well as Bcl-2 protein level reduced (P <0.05). miR-152-3p can be controlled by circKIF4A; pcDNA-circKIF4A transfection antagonized AG's effects on HGC-27 cell multiplication, clone formation, apoptosis and migration. Conclusion AG can decrease GC multiplication, clone formation and migration and induce apoptosis via modulating circKIF4A/miR-152-3p expression.
Collapse
|
19
|
Wang H, Luo Y, Chu Z, Ni T, Ou S, Dai X, Zhang X, Liu Y. Poria Acid, Triterpenoids Extracted from Poria cocos, Inhibits the Invasion and Metastasis of Gastric Cancer Cells. Molecules 2022; 27:molecules27113629. [PMID: 35684565 PMCID: PMC9182142 DOI: 10.3390/molecules27113629] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Poria cocos (P. cocos) is an important medicinal fungus in traditional Chinese medicine. Poria acid (PA), a triterpenoid compound, is an effective component of traditional Chinese medicine P. cocos. This experiment investigated the anti-gastric cancer biological activity of PA in vitro. Methods: The effect of PA on the viability of gastric cancer cells was detected by the thiazolyl blue (MTT) assay. Cell adhesion assays were used to detect changes in the adhesion of cells treated after PA (0, 20, 40, and 80 µmol/L). The ability of cell invasion and migration were detected by Transwell assays and wound healing assays. A high-content imaging system was used to dynamically record the motility of the gastric cancer cells after PA (0, 20, 40, and 80 µmol/L) treatment. Western blotting was used to detect the expression of epithelial–mesenchymal transformation (EMT), invasion and migration related proteins. Results: The MTT assay showed that the proliferation of gastric cancer cells was significantly inhibited after PA treatment. Cell adhesion experiments showed that the adhesion of gastric cancer cells was significantly decreased after PA treatment. Compared with the control group, the wound healing area of the gastric cancer cells treated with different concentrations of PA decreased. The Transwell assay showed that the number of gastric cancer cells passing through the cell membrane were significantly reduced after PA treatment. In addition, after PA treatment, the cells’ movement distance and average movement speed were significantly lower than those of the control group. Finally, PA can significantly alter the expression of EMT-related proteins E-cadherin, N-cadherin, and Vimentin and decreased the expressions of metastasis-related proteins matrix metalloproteinase (MMP) 2, MMP-9 and tissue inhibition of matrix metalloproteinase (TIMP)1 in the gastric cancer cells. Conclusions: Triterpenoids from P. cocos have significant biological activity against gastric cancer, and the mechanism may be involved in the process of epithelial–mesenchymal transformation.
Collapse
Affiliation(s)
- Haibo Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Y.L.); (Z.C.); (T.N.); (S.O.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China;
- Correspondence: (H.W.); (X.Z.); (Y.L.)
| | - Yuanyuan Luo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Y.L.); (Z.C.); (T.N.); (S.O.)
| | - Zewen Chu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Y.L.); (Z.C.); (T.N.); (S.O.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China;
| | - Tengyang Ni
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Y.L.); (Z.C.); (T.N.); (S.O.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China;
| | - Shiya Ou
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Y.L.); (Z.C.); (T.N.); (S.O.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China;
| | - Xiaojun Dai
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China;
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225001, China
| | - Xiaochun Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Y.L.); (Z.C.); (T.N.); (S.O.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China;
- Yangzhou Hospital of Traditional Chinese Medicine, Yangzhou 225001, China
- Correspondence: (H.W.); (X.Z.); (Y.L.)
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, China; (Y.L.); (Z.C.); (T.N.); (S.O.)
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou 225001, China;
- Correspondence: (H.W.); (X.Z.); (Y.L.)
| |
Collapse
|
20
|
Oxidative Stress and AKT-Associated Angiogenesis in a Zebrafish Model and Its Potential Application for Withanolides. Cells 2022; 11:cells11060961. [PMID: 35326412 PMCID: PMC8946239 DOI: 10.3390/cells11060961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress and the AKT serine/threonine kinase (AKT) signaling pathway are essential regulators in cellular migration, metastasis, and angiogenesis. More than 300 withanolides were discovered from the plant family Solanaceae, exhibiting diverse functions. Notably, the relationship between oxidative stress, AKT signaling, and angiogenesis in withanolide treatments lacks comprehensive understanding. Here, we summarize connecting evidence related to oxidative stress, AKT signaling, and angiogenesis in the zebrafish model. A convenient vertebrate model monitored the in vivo effects of developmental and tumor xenograft angiogenesis using zebrafish embryos. The oxidative stress and AKT-signaling-modulating abilities of withanolides were highlighted in cancer treatments, which indicated that further assessments of their angiogenesis-modulating potential are necessary in the future. Moreover, targeting AKT for inhibiting AKT and its AKT signaling shows the potential for anti-migration and anti-angiogenesis purposes for future application to withanolides. This particularly holds for investigating the anti-angiogenetic effects mediated by the oxidative stress and AKT signaling pathways in withanolide-based cancer therapy in the future.
Collapse
|
21
|
Liu F, Zhu XT, Li Y, Wang CJ, Fu JL, Hui J, Xiao Y, Liu L, Yan R, Li XF, Liu Y. Magnesium demethylcantharidate inhibits hepatocellular carcinoma cell invasion and metastasis via activation transcription factor FOXO1. Eur J Pharmacol 2021; 911:174558. [PMID: 34634308 DOI: 10.1016/j.ejphar.2021.174558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 11/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world, develops rapidly and has a high mortality rate. Relapsed metastasis is the most important factor affecting prognosis and is also the main cause of death for patients with HCC. Cantharidin is a kind of folk medicine for malignant tumors in China. Because of its cytotoxicity, the application of cantharidin is very limited. Magnesium demethylcantharidate (MDC) is a derivative of cantharidin independently developed by our laboratory. Our results show that MDC has anticancer activity and exhibited lower toxicity than cantharidin. However, whether MDC affects the invasion and metastasis of HCC cells and the underlying molecular mechanisms remain obscure. Transwell and Matrigel assays showed that MDC could effectively inhibit the invasion and metastasis of the HCC cell lines SMMC-7721 and SK-Hep1 in a dose-dependent manner. Moreover, MDC significantly inhibited the expression of invasion and metastasis related proteins MMP-2 and MMP-9. In addition, our study found that MDC inhibited the invasion and metastasis of HCC cell lines SMMC-7721 and SK-Hep1 by activating transcription factor FOXO1. Interestingly, the combination of MDC and sorafenib significantly inhibited the invasion and metastasis of HCC cell lines SMMC-7721 and SK-Hep1 compared with the single drug treatment via the activated transcription factor FOXO1. Our work revealed that MDC obviously inhibited the invasion and metastasis of HCC cells, and suggested that MDC could be a potential candidate molecule against the invasion and metastasis of HCC.
Collapse
Affiliation(s)
- Fang Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xin-Ting Zhu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China; Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China
| | - Yi Li
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Chen-Jing Wang
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Jia-Li Fu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Jing Hui
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China; Life Sciences Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China; Life Sciences Institute, Zunyi Medical University, Zunyi, 563000, China
| | - Liu Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Rong Yan
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xiao-Fei Li
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Yun Liu
- College of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, 563000, China; Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, 563000, China; Life Sciences Institute, Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
22
|
Long Noncoding RNAs Regulate the Radioresistance of Breast Cancer. Anal Cell Pathol (Amst) 2021; 2021:9005073. [PMID: 34595090 PMCID: PMC8478560 DOI: 10.1155/2021/9005073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BRCA) has severely threatened women's health worldwide. Radiotherapy is a treatment for BRCA, which applies high doses of ionizing radiation to induce cancer cell death and reduce disease recurrence. Radioresistance is one of the most important elements that affect the therapeutic efficacy of radiotherapy. Long noncoding RNAs (lncRNAs) are suggested to dominate crucial roles in regulating the biological behavior of BRCA. Currently, some studies indicate that overexpression or inhibition of lncRNAs can greatly alter the radioresistance of BRCA. In this review, we summarized the knowledge on the classification and function of lncRNAs and the molecular mechanism of BRCA radioresistance, listed lncRNAs related to the BRCA radioresistance, highlighted their underlying mechanisms, and discussed the potential application of these lncRNAs in regulating BRCA radioresistance.
Collapse
|
23
|
Hu S, Chang J, Ruan H, Zhi W, Wang X, Zhao F, Ma X, Sun X, Liang Q, Xu H, Wang Y, Yang Y. Cantharidin inhibits osteosarcoma proliferation and metastasis by directly targeting miR-214-3p/DKK3 axis to inactivate β-catenin nuclear translocation and LEF1 translation. Int J Biol Sci 2021; 17:2504-2522. [PMID: 34326690 PMCID: PMC8315017 DOI: 10.7150/ijbs.51638] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Background: As the leading primary bone cancer in adolescents and children, osteosarcoma patients with metastasis show a five-year-survival-rate of 20-30%, without improvement over the past 30 years. Wnt/β-catenin is important in promoting osteosarcoma development. DKK3 is a Wnt/β-catenin antagonist and predicted to have the specific binding site in 3′-UTR with miR-214-3p. Methods: miR-214-3p and DKK3 levels were investigated in human osteosarcoma tissues and cells by RT-qPCR; the prognostic importance of DKK3 level in osteosarcoma patients was determined with Log-rank test; direct binding between DKK3 with miR-214-3p was identified with targetscan; anti-osteosarcoma mechanism of cantharidin was investigated by miR-214-3p silence/over-expression with or without cantharidin treatment, and nuclear/cytoplasmic protein assay in osteosarcoma cells. Results: Down-regulated DKK3 indicated poor prognosis of osteosarcoma patients. Up-regulated miR-214-3p promoted proliferation and migration, while suppressed apoptosis of osteosarcoma cells by increasing β-catenin nuclear translocation and LEF1 translation via degradation of DKK3. Cantharidin suppressed viabilities, migration and invasion, while promoted cell cycle arrest and apoptosis in 143B and U-2 OS cells via down-regulating miR-214-3p to up-regulate DKK3, thus inhibited p-GSK-3β expression, β-catenin nuclear translocation and LEF1 translation. Meanwhile, cantharidin inhibited tumor growth in xenograft-bearing mice with 143B cell injection in tibia. Conclusion: miR-214-3p mediated Wnt/β-catenin/LEF1 signaling activation by targeting DKK3 to promote oncogenesis of osteosarcoma; cantharidin inhibited proliferation and metastasis of osteosarcoma cells via down-regulating miR-214-3p to up-regulate DKK3 and decrease β-catenin nuclear translocation, indicating that cantharidin may be a prospective candidate for osteosarcoma treatment by targeting miR-214-3p/DKK3/β-catenin signaling.
Collapse
Affiliation(s)
- Shaopu Hu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Junli Chang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Hongfeng Ruan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Wenlan Zhi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Xiaobo Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Fulai Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Xiaoping Ma
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Xingyuan Sun
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| | - Yanping Yang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.,Key laboratory of theory and therapy of muscles and bones, Ministry of Education, Shanghai, 200032, China
| |
Collapse
|
24
|
Effective Material Basis and Mechanism Analysis of Compound Banmao Capsule against Tumors Using Integrative Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6653460. [PMID: 34055017 PMCID: PMC8112962 DOI: 10.1155/2021/6653460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 01/05/2023]
Abstract
Purpose Compound banmao capsule (CBC), a well-known traditional Chinese medical material, is known to inhibit various tumors. However, its material basis and pharmacological mechanisms remain to be elucidated. This study aimed to investigate the effective material basis and mechanisms of action of CBC against tumors. Methods Active compounds of CBC were identified using public database and reports to build a network. The corresponding targets of active compounds were retrieved from online databases, and the antitumor targets were identified by GeneCards database. The antitumor hub targets were generated via protein-protein interaction analysis using String, and key compounds and targets from the integrative network were detected by molecular docking and ADMET. Top targets in hepatocellular carcinoma were confirmed by quantitative real-time PCR (qPCR). Finally, the multivariate biological network was built to identify the integrating mechanisms of action of CBC against tumor cells. Results A total of 128 compounds and 436 targets of CBC were identified successfully. Based on the generated multivariate biological network analysis, 25 key compounds, nine hub targets, and two pathways were further explored. Effective material bases of cantharidin, baicalein, scutellarin, sesamin, and quercetin were verified by integrative network analysis. PTGS2, ESR1, and TP53 were identified as hub targets via multivariate biological network analysis and confirmed using qPCR. Furthermore, VEGF and estrogen signaling pathways seem to play a role in the antitumor activity of CBC. Thus, breast cancer may be a potential clinical indication of CBC. Conclusion This study successfully identified the material basis of CBC and its synergistic mechanisms of action against tumor cells.
Collapse
|
25
|
Ling J, Chang A, Ye H, Zhao H, Zhuo X. TXNIP, CXCL1, and AREG as key genes in formaldehyde-induced head and neck carcinoma: an in silico analysis. Inhal Toxicol 2021; 33:113-120. [PMID: 33821754 DOI: 10.1080/08958378.2021.1908461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Reports have shown that formaldehyde (FA) can induce malignant transformation in cells via complicated mechanisms. Therefore, we aimed to investigate the possible molecules, pathways, and therapeutic agents for FA-induced head and neck cancer (HNC) by using bioinformatics approaches. METHODS High throughput data were analyzed to screen the differentially expressed genes (DEGs) between FA-treated nasal epithelium cells and controls. Then, the functions of the DEGs were annotated and the hub genes, as well as the key genes, were further screened out. Afterwards, potential drugs were predicted by using the connectivity map (CMAP) tool. RESULTS The information of a microarray-based dataset GSE21477 was extracted and analyzed. A total of 210 upregulated and 83 downregulated DEGs were generated, which might be enriched in various pathways, such as Cytokine-cytokine receptor interaction, Jak-STAT signaling pathway, and Toll-like receptor signaling pathway. Among these DEGs, three hub genes including TXNIP, CXCL1, and AREG, were identified as the key genes because they might affect the prognosis of HNC. Finally, a major active ingredient of blister beetles, Cantharidin, was predicted to be one of the potential drugs reversing FA-induced malignant transformation in head and neck epithelium cells. CONCLUSION The present analysis gave us a novel insight into the mechanisms of FA-induced malignant transformation in head and neck epithelium cells, and predicted several small agents for the prevention or treatment of HNC. Future experiment studies are warranted to validate the findings.
Collapse
Affiliation(s)
- Junjun Ling
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Aoshuang Chang
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Huiping Ye
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Houyu Zhao
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xianlu Zhuo
- Department of Otorhinolaryngology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
26
|
Kittiwattanokhun A, Samosorn S, Innajak S, Watanapokasin R. Inhibitory effects on chondrosarcoma cell metastasis by Senna alata extract. Biomed Pharmacother 2021; 137:111337. [PMID: 33582453 DOI: 10.1016/j.biopha.2021.111337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/27/2021] [Accepted: 01/27/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Senna alata L. Roxb or candle bush is a traditional medicinal plant with a wide range of biological activities including anti-inflammatory, antimicrobial and antifungal. Leaf extract of S. alata showed the anti-tumor activity in various cancer cell lines. In this study, we focused on the inhibitory mechanism of S. alata extract (SAE) on cancer metastasis including cell migration, cell invasion and signaling pathways in chondrosarcoma SW1353 cells. PURPOSE This study aimed to evaluate the anti-metastatic mechanisms of Senna alata extract on chondrosarcoma SW1353 cells. METHODS Screening for phytochemicals in biologically active fraction of SAE was analysed by 1H NMR spectroscopy. Cell viability and cytoxicity were determined by using MTT assay. Cell migration was observed by scratch wound healing and transwell migration assay. Cell invasion and cell adhesion assay were examined by Matrigel coated transwell chambers or plates. The expression of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs), MAPKs and PI3K/Akt signaling pathways and NF-κB were detected by Western blot analysis. RESULTS The SAE treatment at the sub-cytoxic and non-cytotoxic concentrations significantly inhibited cell migration, cell invasion and cell adhesion of SW1353 cells in a dose-dependent manner. The results from Western blot analysis showed decreased MMP-2 and MMP-9 expression, while increased TIMP-1 and TIMP-2 expression in SAE treated cells. Moreover, SAE suppressed phosphorylation of ERK1/2, p38 and Akt but decreased NF-κB transcription factor expression in SW1353 cells. CONCLUSION These results revealed that SAE could reduce MMP-2 and MMP-9 expression by downregulation of NF-κB which is downstream of MAPKs and PI3K/Akt signaling pathway in SW1353 cells resulting in reduced cancer cell migration and invasion. Therefore, SAE may have the potential use as an alternative treatment of chondrosarcoma metastasis.
Collapse
Affiliation(s)
- Athicha Kittiwattanokhun
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, 114 Sukhumvit 23, Bangkok 10110, Thailand
| | - Siritron Samosorn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Srinakharinwirot University, 114 Sukhumvit 23, Bangkok 10110, Thailand
| | - Sukanda Innajak
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, 114 Sukhumvit 23, Bangkok 10110, Thailand
| | - Ramida Watanapokasin
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, 114 Sukhumvit 23, Bangkok 10110, Thailand.
| |
Collapse
|
27
|
Antitumor potential of the protein phosphatase inhibitor, cantharidin, and selected derivatives. Bioorg Med Chem 2021; 32:116012. [PMID: 33454654 DOI: 10.1016/j.bmc.2021.116012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 12/21/2022]
Abstract
Cantharidin is a potent natural protein phosphatase monoterpene anhydride inhibitor secreted by several species of blister beetle, with its demethylated anhydride analogue, (S)-palasonin, occurring as a constituent of the higher plant Butea frondosa. Cantharidin shows both potent protein phosphatase inhibitory and cancer cell cytotoxic activities, but possible preclinical development of this anhydride has been limited thus far by its toxicity. Thus, several synthetic derivatives of cantharidin have been prepared, of which some compounds exhibit improved antitumor potential and may have use as lead compounds. In the present review, the potential antitumor activity, structure-activity relationships, and development of cantharidin-based anticancer drug conjugates are summarized, with protein phosphatase-related and other types of mechanisms of action discussed. Protein phosphatases play a key role in the tumor microenvironment, and thus described herein is also the potential for developing new tumor microenvironment-targeted cancer chemotherapeutic agents, based on cantharidin and its naturally occurring analogues and synthetic derivatives.
Collapse
|
28
|
Anticancer activities of TCM and their active components against tumor metastasis. Biomed Pharmacother 2020; 133:111044. [PMID: 33378952 DOI: 10.1016/j.biopha.2020.111044] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Traditional Chinese Medicine (TCM) has the characteristics of multiple targets, slight side effects and good therapeutic effects. Good anti-tumor effects are shown by Traditional Chinese Medicine prescription, Chinese patent medicine, single Traditional Chinese Medicine and Traditional Chinese medicine monomer compound. Clinically, TCM prolonged the survival time of patients and improved the life quality of patients, due to less side effects. Cancer metastasis is a complex process involving numerous steps, multiple genes and their products. During the process of tumor metastasis, firstly, cancer cell increases its proliferative capacity by reducing autophagy and apoptosis, and then the cancer cell capacity is stimulated by increasing the ability of tumors to absorb nutrients from the outside through angiogenesis. Both of the two steps can increase tumor migration and invasion. Finally, the purpose of tumor metastasis is achieved. By inhibiting autophagy and apoptosis of tumor cells, angiogenesis and EMT outside the tumor can inhibit the invasion and migration of cancer, and consequently achieve the purpose of inhibiting tumor metastasis. This review explores the research achievements of Traditional Chinese Medicine on breast cancer, lung cancer, hepatic carcinoma, colorectal cancer, gastric cancer and other cancer metastasis in the past five years, summarizes the development direction of TCM on cancer metastasis research in the past five years and makes a prospect for the future.
Collapse
|
29
|
Lv X, Li P, Wang J, Gao H, Hei Y, Zhang J, Li S. hsa_circ_0000520 influences herceptin resistance in gastric cancer cells through PI3K-Akt signaling pathway. J Clin Lab Anal 2020; 34:e23449. [PMID: 32701211 PMCID: PMC7595902 DOI: 10.1002/jcla.23449] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/22/2022] Open
Abstract
Background To investigate whether hsa_circ_0000520 affects Herceptin resistance in gastric cancer by regulating the PI3K‐AKT signaling. Methods The expression of hsa_circ_0000520 was detected by qRT‐PCR in gastric cancer tissues and cell lines. A Herceptin‐resistant gastric cancer cell was established. PcDNA and pcDNA‐hsa_circ_0000520 were transfected into NCI‐N87R cells and treated with Herceptin at a concentration of 10 μg/mL for 24 hours. MTT tested cell proliferation, and apoptosis was measured by flow cytometry. IGF‐1 treatment was used to activate PI3K‐Akt signaling. The expression levels of related proteins were detected. Results The expression of hsa_circ_0000520 was reduced in gastric cancer tissues and cell lines, and hsa_circ_0000520 in NCI‐N87R cells was significantly lower than that of NCI‐N87 cells. Compared with the CON group, the cell viability of the Herceptin group was significantly reduced, the apoptosis rate was significantly increased, the level of Bax protein was significantly increased, and the levels of Bcl‐2, p‐PI3K, and p‐Akt protein were significantly reduced. Compared with the Herceptin + pcDNA group, the cell viability of the Herceptin + hsa_circ_0000520 group was significantly reduced, the apoptosis rate was significantly increased, the level of Bax protein was significantly increased, and the levels of p‐PI3K and p‐Akt proteins were significantly reduced. After IGF‐1 treatment, the cell viability was significantly increased, the apoptosis rate was significantly reduced, the level of Bax protein was significantly reduced, and the level of Bcl‐2 protein was significantly increased. Conclusion Hsa_circ_0000520 overexpression may reverse the Herceptin resistance of gastric cancer cells by inhibiting the PI3K‐Akt signaling pathway.
Collapse
Affiliation(s)
- Xukun Lv
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Peizhe Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Jinkai Wang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Hengling Gao
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Yingrui Hei
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Jianxian Zhang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| | - Shuliang Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Linqing, China
| |
Collapse
|
30
|
Naz F, Wu Y, Zhang N, Yang Z, Yu C. Anticancer Attributes of Cantharidin: Involved Molecular Mechanisms and Pathways. Molecules 2020; 25:E3279. [PMID: 32707651 PMCID: PMC7397086 DOI: 10.3390/molecules25143279] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is a preeminent threat to the human race, causing millions of deaths each year on the Earth. Traditionally, natural compounds are deemed promising agents for cancer treatment. Cantharidin (CTD)-a terpenoid isolated from blister beetles-has been used extensively in traditional Chinese medicines for healing various maladies and cancer. CTD has been proven to be protein phosphatase 2A (PP2A) and heat shock transcription factor 1 (HSF-1) inhibitor, which can be potential targets for its anticancer activity. Albeit, it harbors some toxicities, its immense anticancer potential cannot be overlooked, as the cancer-specific delivery of CTD could help to rescue its lethal effects. Furthermore, several derivatives have been designed to weaken its toxicity. In light of extensive research, the antitumor activity of CTD is evident in both in vitro as well as in vivo cancer models. CTD has also proven efficacious in combination with chemotherapy and radiotherapy and it can also target some drug-resistant cancer cells. This mini-review endeavors to interpret and summarize recent information about CTD anticancer potential and underlying molecular mechanisms. The pertinent anticancer strength of CTD could be employed to develop an effective anticarcinogenic drug.
Collapse
Affiliation(s)
| | | | | | - Zhao Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (F.N.); (Y.W.); (N.Z.)
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (F.N.); (Y.W.); (N.Z.)
| |
Collapse
|
31
|
Tian S, Song X, Wang Y, Wang X, Mou Y, Chen Q, Zhao H, Ma K, Wu Z, Yu H, Han X, Wang H, Wang S, Ji X, Zhang Y. Chinese herbal medicine Baoyuan Jiedu decoction inhibits the accumulation of myeloid derived suppressor cells in pre-metastatic niche of lung via TGF-β/CCL9 pathway. Biomed Pharmacother 2020; 129:110380. [PMID: 32554250 DOI: 10.1016/j.biopha.2020.110380] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/24/2020] [Accepted: 06/07/2020] [Indexed: 12/14/2022] Open
Abstract
Baoyuan Jiedu (BYJD for short) decoction, a traditional Chinese medicine formula, is composed of Astragalus, Ginseng, Aconite root, Honeysuckle, Angelica, Licorice, which has the functions of nourishing qi and blood, enhancing immune function, improving quality of life and prolonging survival time of tumor patients. The present study aimed to investigate the effect and mechanism of BYJD decoction on reversing the pre-metastatic niche. We showed that BYJD decoction could prolong the survival time of 4T1 tumor-bearing mice. Moreover, we found that the BYJD decoction inhibited the formation of lung pre-metastatic niche and inhibited recruitment of myeloid derived suppressor cells (MDSCs) in the lung. Mechanistically, we showed that the proteins and genes expression of TGF-β, Smad2, Smad3, p-Smad2/3, Smad4, CCL9 in the TGF-β/CCL9 signaling pathway were suppressed by BYJD decoction. In line with the above findings, our results confirm that BYJD decoction inhibits the accumulation of MDSC in pre-metastatic niche of lung via TGF-β/CCL9 pathway.
Collapse
Affiliation(s)
- Sheng Tian
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - XiaoTong Song
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - XiaoYan Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China
| | - Yue Mou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qian Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - HaiJun Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ke Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - ZhiChun Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - HuaYun Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - XiaoChun Han
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - HuaXin Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - ShiJun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - XuMing Ji
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; College of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China.
| | - YaNan Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China; Shandong Co-Innovation Center of Classic TCM Formula, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
32
|
Yang S, Sun S, Xu W, Yu B, Wang G, Wang H. Astragalus polysaccharide inhibits breast cancer cell migration and invasion by regulating epithelial‑mesenchymal transition via the Wnt/β‑catenin signaling pathway. Mol Med Rep 2020; 21:1819-1832. [PMID: 32319619 PMCID: PMC7057808 DOI: 10.3892/mmr.2020.10983] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) serves an important role in tumor migration and invasion. Astragalus polysaccharide (APS), which is the main component of the traditional Chinese medicine Astragalus membranaceus, has been identified to display an antitumor effect. However, the effects and mechanisms of APS during breast cancer migration and invasion are not completely understood. The present study investigated whether APS inhibited breast cancer migration and invasion by modulating the EMT pathway. An MTT assay and a Ki67 immunofluorescence staining assay demonstrated that APS inhibited the proliferation of breast cancer cells. The results of the wound healing and Transwell Matrigel invasion assays suggested that APS decreased the migration and invasion of breast cancer cells. The western blotting and immunofluorescence analyses further demonstrated that APS had a regulatory effect on EMT-related molecules. APS decreased the expression levels of Snail and vimentin, but increased E-cadherin expression. APS also downregulated Wnt1, β-catenin and downstream target expression. Additionally, the present results suggested that APS decreased the proliferation, and EMT-mediated migration and invasion of cells by inhibiting the Wnt/β-catenin signaling pathway. The present study suggested that APS may serve as a promising therapeutic agent for breast cancer.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Shuqin Sun
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Wanqun Xu
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Bangxu Yu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Guimei Wang
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Haibo Wang
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
33
|
Construction of a circRNA-miRNA-mRNA network based on competitive endogenous RNA reveals the function of circRNAs in osteosarcoma. Cancer Cell Int 2020; 20:48. [PMID: 32063749 PMCID: PMC7011443 DOI: 10.1186/s12935-020-1134-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
Background Osteosarcoma (OS) is a common primary malignant bone tumour. Growing evidence suggests that circular RNAs (circRNAs) are closely related to the development of tumours. However, the function of circRNAs in OS remains unknown. Here, we aimed to determine the regulatory mechanisms of circRNAs in OS. Methods The expression profiles of OS circRNA (GSE96964), microRNA (GSE65071) and mRNA (GSE33382) were downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed circRNAs, miRNAs and mRNAs in OS. A ceRNA network was constructed based on circRNA-miRNA pairs and miRNA-mRNA pairs. MRNAs with significant prognostic differences were identified by the TARGET database in the network. Functional and pathway enrichment analyses were performed, and interactions between proteins were predicted using Cytoscape. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to elucidate the possible functions of these differentially expressed circRNAs. Results A total of 15 downregulated circRNAs, 136 upregulated miRNAs and 52 downregulated mRNAs were identified in OS. Finally, a circRNA-miRNA-mRNA network was constructed in OS based on 14 circRNAs, 24 miRNAs, and 52 mRNAs. GO and KEGG pathway analyses suggested that the mRNAs in the network may be involved in the pathogenesis and progression of OS. Four mRNAs identified by the TARGET database were significantly associated with OS survival prognosis. A circRNA-miRNA-mRNA subnetwork was constructed based on these four mRNAs. Conclusion Our results provide a deeper understanding of the regulatory mechanisms by which circRNAs compete for endogenous RNAs in OS.
Collapse
|