1
|
Li J, Liang H, Liu L, Gao X, Liu Y, Zhang M, Yuan X, Ren S, Zhang W. Structural diversity and biological activities of terpenoids derived from Tripterygium wilfordii Hook. f. RSC Adv 2025; 15:12594-12608. [PMID: 40264893 PMCID: PMC12012611 DOI: 10.1039/d4ra09048a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 04/05/2025] [Indexed: 04/24/2025] Open
Abstract
Terpenoids, a heterogeneous group of natural products, have garnered considerable attention in the field of drug discovery. This is attributed to their vast diversity, intricate structural features, and extensive biological activities. Tripterygium wilfordii Hook. f., a traditional medicinal plant with widespread application in East Asia, is particularly enriched in terpenoids, which can be classified into sesquiterpenoids, diterpenoids, and triterpenoids. The present review provides a comprehensive elaboration of the chemical structures and biological activities of 217 terpenoids isolated from T. wilfordii. The purpose is to shed light on their potential in pharmacological research and to stimulate innovative drug discovery as well as clinical applications. These terpenoids display a broad spectrum of biological activities, such as antitumor, anti - inflammatory, immunosuppressive, and other therapeutic effects. Nevertheless, their clinical application is impeded by issues related to toxicity and poor bioavailability. Future research efforts should be concentrated on exploring effective strategies to alleviate toxicity and enhance drug delivery systems. In addition, in - depth investigation into the structure-activity relationships and the identification of new active constituents are crucial for the development of more potent and safer drugs. This review serves as an exhaustive reference for the discovery and development of novel drugs based on the natural active products of T. wilfordii, providing valuable insights and guidance for researchers in the relevant field.
Collapse
Affiliation(s)
- Jiping Li
- School of Public Health, Qiqihar Medical University Qiqihar 161006 China
| | - Hong Liang
- School of Public Health, Qiqihar Medical University Qiqihar 161006 China
| | - Likun Liu
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Xiuli Gao
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Yang Liu
- Office of Academic Research, Qiqihar Medical University Qiqihar 161006 China
| | - Meng Zhang
- Office of Academic Research, Qiqihar Medical University Qiqihar 161006 China
| | - Xiaoan Yuan
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Shan Ren
- Research Institute of Medicine of Pharmacy, Qiqihar Medical University Qiqihar 161006 China
| | - Wei Zhang
- Office of Academic Research, Qiqihar Medical University Qiqihar 161006 China
| |
Collapse
|
2
|
Huang Y, Wang G, Zhang N, Zeng X. MAP3K4 kinase action and dual role in cancer. Discov Oncol 2024; 15:99. [PMID: 38568424 PMCID: PMC10992237 DOI: 10.1007/s12672-024-00961-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
It is commonly known that the MAPK pathway is involved in translating environmental inputs, regulating downstream reactions, and maintaining the intrinsic dynamic balance. Numerous essential elements and regulatory processes are included in this pathway, which are essential to its functionality. Among these, MAP3K4, a member of the serine/threonine kinases family, plays vital roles throughout the organism's life cycle, including the regulation of apoptosis and autophagy. Moreover, MAP3K4 can interact with key partners like GADD45, which affects organism's growth and development. Notably, MAP3K4 functions as both a tumor promotor and suppressor, being activated by a variety of factors and triggering diverse downstream pathways that differently influence cancer progression. The aim of this study is to provide a brief overview of physiological functions of MAP3K4 and shed light on its contradictory roles in tumorigenesis.
Collapse
Affiliation(s)
- Yuxin Huang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Guanwen Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Ningning Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China.
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, China.
| |
Collapse
|
3
|
Geng S, Chen L, Lin W, Wan F, Le Z, Hu W, Chen H, Liu X, Huang Q, Zhang H, Lu JJ, Kong L. Exploring the Therapeutic Potential of Triptonide in Salivary Adenoid Cystic Carcinoma: A Comprehensive Approach Involving Network Pharmacology and Experimental Validation. Curr Pharm Des 2024; 30:2276-2289. [PMID: 38910414 DOI: 10.2174/0113816128315277240610052453] [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: 03/23/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Salivary Adenoid Cystic Carcinoma (ACC) is characterized by a highly invasive and slow-growing pattern, and its etiology remains unidentified. Triptonide (TN) has demonstrated efficacy as a pharmacotherapeutic agent against ACC. Nonetheless, the specific targets and mechanism of molecular action underlying the effectiveness of TN in treating ACC have not been elucidated. OBJECTIVES By integrating network pharmacology within laboratory experiments, this research delves into the prospective targets and molecular mechanisms associated with the application of TN in treating ACC. METHODS Initially, pertinent targets associated with TN against ACC were acquired from public databases. Subsequently, a combination of network pharmacology and bioinformatics analysis was utilized to screen the top 10 hub targets and key signal pathways of TN-treating ACC. Finally, in vitro experiments involving various molecular assays were conducted to evaluate the biological phenotypes of cells following TN treatment, encompassing assessments of apoptosis levels, plate migration, and other parameters, thereby validating pivotal genes and pathways. RESULTS A total of 23 pertinent targets for TN in relation to ACC were identified, with the top 10 hub genes being MAPK8, PTGS2, RELA, MAPK14, NR3C1, HDAC1, PPARG, NFKBIA, AR, and PGR. There was a significant correlation between the TNF signaling pathway and the treatment of ACC with TN. In vitro experiments demonstrated that TN treatment elevated RELA phosphorylation while concurrently reducing MAPK14 phosphorylation and inducing G2/M arrest. TN exhibited the ability to enhance the apoptosis rate through increased caspase-3 activity, elevated levels of Reactive Oxygen Species (ROS), mitochondrial dysfunction, and inhibition of cell migration. CONCLUSION There is a potential therapeutic role for TN in the treatment of ACC through the activation of the TNF signaling pathway. Among the identified candidates, MAPK8, HDAC1, PTGS2, RELA, NR3C1, PPARG, NFKBIA, AR, and PGR emerge as the most pertinent therapeutic targets for TN in the context of ACC treatment.
Collapse
Affiliation(s)
- Shikai Geng
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Li Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Wanzun Lin
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Fangzhu Wan
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Ziyu Le
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Wei Hu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Huaiyuan Chen
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Xingyu Liu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, China
| | - Qingting Huang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Haojiong Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jiade J Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| |
Collapse
|
4
|
Chaudhary MR, Chaudhary S, Sharma Y, Singh TA, Mishra AK, Sharma S, Mehdi MM. Aging, oxidative stress and degenerative diseases: mechanisms, complications and emerging therapeutic strategies. Biogerontology 2023; 24:609-662. [PMID: 37516673 DOI: 10.1007/s10522-023-10050-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/28/2023] [Indexed: 07/31/2023]
Abstract
Aging accompanied by several age-related complications, is a multifaceted inevitable biological progression involving various genetic, environmental, and lifestyle factors. The major factor in this process is oxidative stress, caused by an abundance of reactive oxygen species (ROS) generated in the mitochondria and endoplasmic reticulum (ER). ROS and RNS pose a threat by disrupting signaling mechanisms and causing oxidative damage to cellular components. This oxidative stress affects both the ER and mitochondria, causing proteopathies (abnormal protein aggregation), initiation of unfolded protein response, mitochondrial dysfunction, abnormal cellular senescence, ultimately leading to inflammaging (chronic inflammation associated with aging) and, in rare cases, metastasis. RONS during oxidative stress dysregulate multiple metabolic pathways like NF-κB, MAPK, Nrf-2/Keap-1/ARE and PI3K/Akt which may lead to inappropriate cell death through apoptosis and necrosis. Inflammaging contributes to the development of inflammatory and degenerative diseases such as neurodegenerative diseases, diabetes, cardiovascular disease, chronic kidney disease, and retinopathy. The body's antioxidant systems, sirtuins, autophagy, apoptosis, and biogenesis play a role in maintaining homeostasis, but they have limitations and cannot achieve an ideal state of balance. Certain interventions, such as calorie restriction, intermittent fasting, dietary habits, and regular exercise, have shown beneficial effects in counteracting the aging process. In addition, interventions like senotherapy (targeting senescent cells) and sirtuin-activating compounds (STACs) enhance autophagy and apoptosis for efficient removal of damaged oxidative products and organelles. Further, STACs enhance biogenesis for the regeneration of required organelles to maintain homeostasis. This review article explores the various aspects of oxidative damage, the associated complications, and potential strategies to mitigate these effects.
Collapse
Affiliation(s)
- Mani Raj Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Yogita Sharma
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Thokchom Arjun Singh
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Shweta Sharma
- Chitkara School of Health Sciences, Chitkara University, Chandigarh, Punjab, 140401, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
5
|
Kumar S, Singh SK, Srivastava P, Suresh S, Rana B, Rana A. Interplay between MAP kinases and tumor microenvironment: Opportunity for immunotherapy in pancreatic cancer. Adv Cancer Res 2023. [PMID: 37268394 DOI: 10.1016/bs.acr.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC), commonly called pancreatic cancer, is aggressive cancer usually detected at a late stage, limiting treatment options with modest clinical responses. It is projected that by 2030, PDAC will be the second most common cause of cancer-related mortality in the United States. Drug resistance in PDAC is common and significantly affects patients' overall survival (OS). Oncogenic KRAS mutations are nearly uniform in PDAC, affecting over 90% of patients. However, effective drugs directed to target prevalent KRAS mutants in pancreatic cancer are not in clinical practice. Accordingly, efforts are continued on identifying alternative druggable target(s) or approaches to improve patient outcomes with PDAC. In most PDAC cases, the KRAS mutations turn-on the RAF-MEK-MAPK pathways, leading to pancreatic tumorigenesis. The MAPK signaling cascade (MAP4K→MAP3K→MAP2K→MAPK) plays a central role in the pancreatic cancer tumor microenvironment (TME) and chemotherapy resistance. The immunosuppressive pancreatic cancer TME is another unfavorable factor affecting the therapeutic efficacy of chemotherapy and immunotherapy. The immune checkpoint proteins (ICPs), including CTLA-4, PD-1, PD-L1, and PD-L2, are critical players in T cell dysfunction and pancreatic tumor cell growth. Here, we review the activation of MAPKs, a molecular trait of KRAS mutations and their impact on pancreatic cancer TME, chemoresistance, and expression of ICPs that could influence the clinical outcomes in PDAC patients. Therefore, understanding the interplay between MAPK pathways and TME could help to design rational therapy combining immunotherapy and MAPK inhibitors for pancreatic cancer treatment.
Collapse
|
6
|
Zhang M, Meng M, Liu Y, Qi J, Zhao Z, Qiao Y, Hu Y, Lu W, Zhou Z, Xu P, Zhou Q. Triptonide effectively inhibits triple-negative breast cancer metastasis through concurrent degradation of Twist1 and Notch1 oncoproteins. Breast Cancer Res 2021; 23:116. [PMID: 34922602 PMCID: PMC8684143 DOI: 10.1186/s13058-021-01488-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/13/2021] [Indexed: 01/19/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is highly metastatic and lethal. Due to a lack of druggable targets for this disease, there are no effective therapies in the clinic. Methods We used TNBC cells and xenografted mice as models to explore triptonide-mediated inhibition of TNBC metastasis and tumor growth. Colony formation assay was used to quantify the tumorigenesis of TNBC cells. Wound-healing and cell trans-well assays were utilized to measure cell migration and invasion. Tube formation assay was applied to access tumor cell-mediated vasculogenic mimicry. Western blot, quantitative-PCR, immunofluorescence imaging, and immunohistochemical staining were used to measure the expression levels of various tumorigenic genes in TNBC cells. Results Here, we showed that triptonide, a small molecule from the traditional Chinese medicinal herb Tripterygium wilfordii Hook F, potently inhibited TNBC cell migration, invasion, and vasculogenic mimicry, and effectively suppressed TNBC tumor growth and lung metastasis in xenografted mice with no observable toxicity. Molecular mechanistic studies revealed that triptonide strongly triggered the degradation of master epithelial-mesenchymal transition (EMT)-inducing protein Twist1 through the lysosomal system and reduced Notch1 expression and NF-κB phosphorylation, which consequently diminished the expression of pro-metastatic and angiogenic genes N-cadherin, VE-cadherin, and vascular endothelial cell growth factor receptor 2 (VEGFR2). Conclusions Triptonide effectively suppressed TNBC cell tumorigenesis, vasculogenic mimicry, and strongly inhibited the metastasis of TNBC via degradation of Twist1 and Notch1 oncoproteins, downregulation of metastatic and angiogenic gene expression, and reduction of NF-κB signaling pathway. Our findings provide a new strategy for treating highly lethal TNBC and offer a potential new drug candidate for combatting this aggressive disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01488-7.
Collapse
Affiliation(s)
- Mengli Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yuxi Liu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Jindan Qi
- School of Nursing, Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China
| | - Zhe Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yingnan Qiao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yanxing Hu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Wei Lu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Zhou Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Peng Xu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China.
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, 199 Ren Ai Road, Suzhou Industrial Park, Suzhou, 215123, Jiangsu, People's Republic of China. .,State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, People's Republic of China.
| |
Collapse
|
7
|
Lim YX, Lin H, Seah SH, Lim YP. Reciprocal Regulation of Hippo and WBP2 Signalling-Implications in Cancer Therapy. Cells 2021; 10:cells10113130. [PMID: 34831354 PMCID: PMC8625973 DOI: 10.3390/cells10113130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/08/2021] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Cancer is a global health problem. The delineation of molecular mechanisms pertinent to cancer initiation and development has spurred cancer therapy in the form of precision medicine. The Hippo signalling pathway is a tumour suppressor pathway implicated in a multitude of cancers. Elucidation of the Hippo pathway has revealed an increasing number of regulators that are implicated, some being potential therapeutic targets for cancer interventions. WW domain-binding protein 2 (WBP2) is an oncogenic transcriptional co-factor that interacts, amongst others, with two other transcriptional co-activators, YAP and TAZ, in the Hippo pathway. WBP2 was recently discovered to modulate the upstream Hippo signalling components by associating with LATS2 and WWC3. Exacerbating the complexity of the WBP2/Hippo network, WBP2 itself is reciprocally regulated by Hippo-mediated microRNA biogenesis, contributing to a positive feedback loop that further drives carcinogenesis. Here, we summarise the biological mechanisms of WBP2/Hippo reciprocal regulation and propose therapeutic strategies to overcome Hippo defects in cancers through targeting WBP2.
Collapse
Affiliation(s)
- Yvonne Xinyi Lim
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 119077, Singapore; (Y.X.L.); (H.L.); (S.H.S.)
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
| | - Hexian Lin
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 119077, Singapore; (Y.X.L.); (H.L.); (S.H.S.)
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
| | - Sock Hong Seah
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 119077, Singapore; (Y.X.L.); (H.L.); (S.H.S.)
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Yoon Pin Lim
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore
- Correspondence:
| |
Collapse
|
8
|
Traub B, Roth A, Kornmann M, Knippschild U, Bischof J. Stress-activated kinases as therapeutic targets in pancreatic cancer. World J Gastroenterol 2021; 27:4963-4984. [PMID: 34497429 PMCID: PMC8384741 DOI: 10.3748/wjg.v27.i30.4963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/17/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a dismal disease with high incidence and poor survival rates. With the aim to improve overall survival of pancreatic cancer patients, new therapeutic approaches are urgently needed. Protein kinases are key regulatory players in basically all stages of development, maintaining physiologic functions but also being involved in pathogenic processes. c-Jun N-terminal kinases (JNK) and p38 kinases, representatives of the mitogen-activated protein kinases, as well as the casein kinase 1 (CK1) family of protein kinases are important mediators of adequate response to cellular stress following inflammatory and metabolic stressors, DNA damage, and others. In their physiologic roles, they are responsible for the regulation of cell cycle progression, cell proliferation and differentiation, and apoptosis. Dysregulation of the underlying pathways consequently has been identified in various cancer types, including pancreatic cancer. Pharmacological targeting of those pathways has been the field of interest for several years. While success in earlier studies was limited due to lacking specificity and off-target effects, more recent improvements in small molecule inhibitor design against stress-activated protein kinases and their use in combination therapies have shown promising in vitro results. Consequently, targeting of JNK, p38, and CK1 protein kinase family members may actually be of particular interest in the field of precision medicine in patients with highly deregulated kinase pathways related to these kinases. However, further studies are warranted, especially involving in vivo investigation and clinical trials, in order to advance inhibition of stress-activated kinases to the field of translational medicine.
Collapse
Affiliation(s)
- Benno Traub
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Aileen Roth
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Marko Kornmann
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| | - Joachim Bischof
- Department of General and Visceral Surgery, Ulm University Hospital, Ulm 89081, Germany
| |
Collapse
|
9
|
Tan S, Zhao Z, Qiao Y, Zhang B, Zhang T, Zhang M, Qi J, Wang X, Meng M, Zhou Q. Activation of the tumor suppressive Hippo pathway by triptonide as a new strategy to potently inhibit aggressive melanoma cell metastasis. Biochem Pharmacol 2021; 185:114423. [PMID: 33476574 DOI: 10.1016/j.bcp.2021.114423] [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: 10/20/2020] [Revised: 01/02/2021] [Accepted: 01/13/2021] [Indexed: 02/08/2023]
Abstract
Metastatic melanoma has a very high mortality rate despite the availability of chemotherapy, radiotherapy, and immunotherapy; therefore, more effective therapeutics are needed. The Hippo pathway plays an inhibitory role in melanoma progression, but the tumor suppressors Salvador homolog-1 (SAV1) and large tumor suppressor 1 (LATS1) in this pathway are down-regulated in melanoma. As a result, the downstream oncogenic Yes-associated protein (YAP) is active, resulting in uncontrolled melanoma growth and metastasis. Therapeutics for remedying SAV1 and LATS1 deficiency in melanoma have not yet been reported in the literature. Here, we show that the small molecule triptonide (MW 358 Da) robustly suppressed melanoma cell tumorigenicity, migration, and invasion. Furthermore, triptonide markedly reduced tumor growth and melanoma lung metastasis in tumor-bearing mice with low toxicity. Molecular mechanistic studies revealed that triptonide promoted SAV1 and LATS1 expression, strongly activated the tumor-suppressive Hippo pathway, degraded oncogenic YAP via the lysosomal pathway, and reduced levels of tumorigenic microphthalmia-associated transcription factor (MITF) in melanoma cells. Triptonide also strongly inhibited activation of AKT, a SAV1-binding signaling protein. Collectively, our results conceptually demonstrate that induction of SAV1 and LATS1 expression and activation of the tumor-suppressive Hippo pathway by triptonide potently inhibits aggressive melanoma cell growth and metastasis. These findings suggest a new strategy for developing therapeutics to treat metastatic melanoma and highlight a novel drug candidate against aggressive melanoma.
Collapse
Affiliation(s)
- Shijie Tan
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Zhe Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu 215123, PR China
| | - Yingnan Qiao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, PR China; Suzhou Institute of Systems Medicine, Suzhou 215123, PR China
| | - Tong Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Mengli Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Jindan Qi
- School of Nursing, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Xiaohua Wang
- School of Nursing, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China.
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123, PR China; National Clinical Research Center for Hematology Diseases, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
10
|
Ren Y, Kinghorn AD. Development of Potential Antitumor Agents from the Scaffolds of Plant-Derived Terpenoid Lactones. J Med Chem 2020; 63:15410-15448. [PMID: 33289552 PMCID: PMC7812702 DOI: 10.1021/acs.jmedchem.0c01449] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Naturally occurring terpenoid lactones and their synthetic derivatives have attracted increasing interest for their promising antitumor activity and potential utilization in the discovery and design of new antitumor agents. In the present perspective article, selected plant-derived five-membered γ-lactones and six-membered δ-lactones that occur with terpenoid scaffolds are reviewed, with their structures, cancer cell line cytotoxicity and in vivo antitumor activity, structure-activity relationships, mechanism of action, and the potential for developing cancer chemotherapeutic agents discussed in each case. The compounds presented include artemisinin (ART, 1), parthenolide (PTL, 2), thapsigargin (TPG, 3), andrographolide (AGL, 4), ginkgolide B (GKL B, 5), jolkinolide B (JKL B, 6), nagilactone E (NGL E, 7), triptolide (TPL, 8), bruceantin (BRC, 9), dichapetalin A (DCT A, 10), and limonin (LMN, 11), and their naturally occurring analogues and synthetic derivatives. It is hoped that this contribution will be supportive of the future development of additional efficacious anticancer agents derived from natural products.
Collapse
Affiliation(s)
- Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A. Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
11
|
Ling YJ, Ding TY, Dong FL, Gao YJ, Jiang BC. Intravenous Administration of Triptonide Attenuates CFA-Induced Pain Hypersensitivity by Inhibiting DRG AKT Signaling Pathway in Mice. J Pain Res 2020; 13:3195-3206. [PMID: 33293856 PMCID: PMC7718987 DOI: 10.2147/jpr.s275320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/19/2020] [Indexed: 01/11/2023] Open
Abstract
Background Currently, medical treatment of inflammatory pain is limited by a lack of safe and effective therapies. Triptonide (TPN), a major component of Tripterygium wilfordii Hook.f. with low toxicity, has been shown to have good anti-inflammatory and neuroprotective effects. The present study aims to investigate the effects of TPN on chronic inflammatory pain. Materials and Methods Inflammatory pain was induced by intraplantar injection of complete Freund’s adjuvant (CFA). TPN’s three different doses were intravenously administered to compare the analgesic efficacy: 0.1 mg/kg, 0.5 mg/kg, and 2.0 mg/kg. The foot swelling was quantitated by measuring paw volume. Mechanical allodynia and thermal hyperalgesia were assessed with von Frey filament testing and Hargreaves’ test, respectively. Western blots, qRT–PCR and immunofluorescence tests were used to analyze the expression of pAKT, tumor necrosis factor-α (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6). Two AKT inhibitors, AKT inhibitor Ⅳ and MK-2206, were used to examine AKT’s effects on pain behavior and cytokines expression. Results Intravenous treatment with TPN attenuated CFA-induced paw edema, mechanical allodynia, and thermal hyperalgesia. Western blotting and immunofluorescence results showed that CFA induced AKT activation in the dorsal root ganglion (DRG) neurons. However, these effects were suppressed by treatment with TPN. Furthermore, TPN treatment inhibited CFA-induced increase of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6. Consistent with the in vivo data, TPN inhibited LPS-induced Akt phosphorylation and inflammatory mediator production in ND7/23 cells. Finally, intrathecal treatment with AKT inhibitor Ⅳ or MK-2206, attenuated CFA-induced mechanical allodynia and thermal hyperalgesia, and simultaneously decreased the mRNA expression of TNF-α, IL-1β, and IL-6 in DRG. Conclusion These data indicate that TPN attenuates CFA-induced pain potentially via inhibiting AKT-mediated pro-inflammatory cytokines production in DRG. TPN may be used for the treatment of chronic inflammatory pain.
Collapse
Affiliation(s)
- Yue-Juan Ling
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Ting-Yu Ding
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Fu-Lu Dong
- School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yong-Jing Gao
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| | - Bao-Chun Jiang
- Institute of Pain Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China.,Institute of Special Environmental Medicine, Nantong University, Nantong, Jiangsu 226019, People's Republic of China
| |
Collapse
|
12
|
Noumi E, Snoussi M, Anouar EH, Alreshidi M, Veettil VN, Elkahoui S, Adnan M, Patel M, Kadri A, Aouadi K, De Feo V, Badraoui R. HR-LCMS-Based Metabolite Profiling, Antioxidant, and Anticancer Properties of Teucrium polium L. Methanolic Extract: Computational and In Vitro Study. Antioxidants (Basel) 2020; 9:E1089. [PMID: 33167507 PMCID: PMC7694502 DOI: 10.3390/antiox9111089] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
In this study, we investigate the phytochemical profile, anticancer, and antioxidant activities of Teucrium polium methanolic extract using both in vitro and in silico approaches. The results showed the identification of 29 phytochemical compounds belonging to 13 classes of compounds and 20 tripeptides using High Resolution-Liquid Chromatography Mass Spectrometry (HR-LCMS). 13R-hydroxy-9E,11Z octadecadienoic acid, dihydrosamidin, valtratum, and cepharantine were the main compounds identified. The tested extract showed promising antioxidant activities (ABTS-IC50 = 0.042 mg/mL; 1,1-diphenyl-2-picrylhydrazyl (DPPH)-IC50 = 0.087 mg/mL, β-carotene-IC50 = 0.101 mg/mL and FRAP-IC50 = 0.292 mg/mL). Using both malignant Walker 256/B and MatLyLu cell lines, T. polium methanolic extract showed a dose/time-dependent antitumor activity. The molecular docking approach revealed that most of the identified molecules were specifically binding with human peroxiredoxin 5, human androgen, and human progesterone receptors with high binding affinity scores. The obtained results confirmed that T. polium is a rich source of bioactive molecules with antioxidant and antitumor potential.
Collapse
Affiliation(s)
- Emira Noumi
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.A.); (V.N.V.); (S.E.); (M.A.); (R.B.)
- Laboratory of Bioressources: Integrative Biology and Recovery, High Institute of Biotechnology-University of Monastir, Monastir 5000, Tunisia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.A.); (V.N.V.); (S.E.); (M.A.); (R.B.)
- Laboratory of Genetics, Biodiversity and Valorisation of Bioressources, High Institute of Biotechnology-University of Monastir, Monastir 5000, Tunisia
| | - El Hassane Anouar
- Department of Chemistry, College of Science and Humanities in Al-Kharj, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Mousa Alreshidi
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.A.); (V.N.V.); (S.E.); (M.A.); (R.B.)
| | - Vajid N. Veettil
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.A.); (V.N.V.); (S.E.); (M.A.); (R.B.)
| | - Salem Elkahoui
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.A.); (V.N.V.); (S.E.); (M.A.); (R.B.)
- Laboratory of Bioactive Substances, Center of Biotechnology of Borj Cedria, BP 901, Hammam lif 2050, Tunisia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.A.); (V.N.V.); (S.E.); (M.A.); (R.B.)
| | - Mitesh Patel
- Bapalal Vaidya Botanical Research Centre, Department of Biosciences, Veer Narmad South Gujarat University, Surat 395007, India;
| | - Adel Kadri
- Department of Chemistry, College of Science and Arts in Baljurashi, Albaha University, Albaha 65527, Saudi Arabia;
- Department of Chemistry, Faculty of Science of Sfax, University of Sfax, BP 1117, Sfax 3000, Tunisia
| | - Kaïss Aouadi
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia;
- Laboratory of Heterocyclic Chemistry, Natural Products and Reactivity, Department of Chemistry, Faculty of Science of Monastir, University of Monastir, Monastir 5019, Tunisia
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, Fisciano, 84084 Salerno, Italy
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha’il 2440, Saudi Arabia; (E.N.); (M.A.); (V.N.V.); (S.E.); (M.A.); (R.B.)
- Section of Histology—Cytology, Medicine College of Tunis, Tunis El Manar University, Road Djebel Lakhdhar, La Rabta-Tunis 1007, Tunisia
- Laboratory of Histo-Embryology and Cytogenetic, Medicine College of Sfax, Sfax University, Sfax 3029, Tunisia
| |
Collapse
|
13
|
Badraoui R, Rebai T, Elkahoui S, Alreshidi M, N. Veettil V, Noumi E, A. Al-Motair K, Aouadi K, Kadri A, De Feo V, Snoussi M. Allium subhirsutum L. as a Potential Source of Antioxidant and Anticancer Bioactive Molecules: HR-LCMS Phytochemical Profiling, In Vitro and In Vivo Pharmacological Study. Antioxidants (Basel) 2020; 9:E1003. [PMID: 33081189 PMCID: PMC7602730 DOI: 10.3390/antiox9101003] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/04/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022] Open
Abstract
This study investigated Allium subhirsutum L. (AS) anticancer and antioxidant effects and inhibition of tumor angiogenesis in a murine model of skeletal metastases due to inoculation of Walker 256/B cells. Phytochemical composition of AS extract (ASE) was studied by High Resolution-Liquid Chromatography Mass Spectroscopy (HR-LCMS). Total phenolic and flavonoid contents (TPC and TFC) were determined. In vitro, the antioxidant properties were evaluated by reducing power and antiradical activity against DPPH. Cancer cells' proliferation, apoptosis, metastatic development and angiogenesis were evaluated using Walker 256/B and MatLyLu cells. The p-coumaric acid was the major phenolic acid (1700 µg/g extract). ASE showed high levels of TPC and TFC and proved potent antioxidant effects. ASE inhibited Walker 256/B and MatLyLu cells' proliferation (Half-maximal inhibitory concentration: IC50 ≃ 150 µg/mL) and induced apoptosis. In silico and in vivo assays confirmed these findings. ASE effectively acts as a chemo-preventive compound, induces apoptosis and attenuates angiogenesis and osteolytic metastases due to Walker 256/B malignant cells.
Collapse
Affiliation(s)
- Riadh Badraoui
- Department of Biology, University of Hail, P.O. Box 2440, Ha’il 81451, Saudi Arabia; (S.E.); (M.A.); (V.N.V.); (E.N.)
- Section of Histology-Cytology, Medicine Faculty of Tunis, University of Tunis El Manar, 1007 La Rabta, Road Djebal Lakhdhar, Tunis 1007, Tunisia
- Department of Histo-Embryology and Cytogenetics, Medicine Faculty of Sfax, University of Sfax, Road of Majida Boulia, Sfax 3029, Tunisia;
| | - Tarek Rebai
- Department of Histo-Embryology and Cytogenetics, Medicine Faculty of Sfax, University of Sfax, Road of Majida Boulia, Sfax 3029, Tunisia;
| | - Salem Elkahoui
- Department of Biology, University of Hail, P.O. Box 2440, Ha’il 81451, Saudi Arabia; (S.E.); (M.A.); (V.N.V.); (E.N.)
- Laboratory of Bioactive Substances, Center of Biotechnology of Borj Cedria (CBBC), BP 901, Hammam Lif 2050, Tunisia
| | - Mousa Alreshidi
- Department of Biology, University of Hail, P.O. Box 2440, Ha’il 81451, Saudi Arabia; (S.E.); (M.A.); (V.N.V.); (E.N.)
| | - Vajid N. Veettil
- Department of Biology, University of Hail, P.O. Box 2440, Ha’il 81451, Saudi Arabia; (S.E.); (M.A.); (V.N.V.); (E.N.)
| | - Emira Noumi
- Department of Biology, University of Hail, P.O. Box 2440, Ha’il 81451, Saudi Arabia; (S.E.); (M.A.); (V.N.V.); (E.N.)
- Laboratory of Bioressources: Integrative Biology & Recovery, High Institute of Biotechnology-University of Monastir, Monastir 5000, Tunisia
| | - Khaled A. Al-Motair
- Molecular Diagnostic and Personalized Therapeutics Unit, University of Ha’il, Ha’il 81451, Saudi Arabia;
| | - Kaïss Aouadi
- Department of Chemistry, College of Science, Qassim University, Buraidah 51452, Saudi Arabia;
- Laboratory of Heterocyclic Chemistry, Natural Products and Reactivity, Department of Chemistry, Faculty of Science of Monastir, University of Monastir, Monastir 5019, Tunisia
| | - Adel Kadri
- Department of Chemistry, College of Science and Arts in Baljurashi, Albaha University, Albaha 65527, Saudi Arabia;
- Department of Chemistry, Faculty of Science of Sfax, University of Sfax, BP 1117, Sfax 3000, Tunisia
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, Fisciano, 84084 Salerno, Italy;
| | - Mejdi Snoussi
- Department of Biology, University of Hail, P.O. Box 2440, Ha’il 81451, Saudi Arabia; (S.E.); (M.A.); (V.N.V.); (E.N.)
- Laboratory of Genetics, Biodiversity and Valorisation of Bioresources, High Institute of Biotechnology-University of Monastir, Monastir 5000, Tunisia
| |
Collapse
|
14
|
Zhang X, Chen C, Zhong Y, Zeng X. lncRNA involved in triptonide-induced cytotoxicity in mouse germ cells. Reprod Toxicol 2020; 98:218-224. [PMID: 33045310 DOI: 10.1016/j.reprotox.2020.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 09/16/2020] [Accepted: 10/04/2020] [Indexed: 01/29/2023]
Abstract
Triptonide reportedly has strong antitumor and anti-inflammatory activities. However, its severe toxicity, including to the reproductive system, has greatly restricted its use in patients who wish to preserve fertility. lncRNAs play crucial roles in male fertility and reportedly regulate triptonide's antitumor activity. However, whether lncRNAs are involved in triptonide-induced reproductive toxicity is unknown. Here, we showed that triptonide induced significant cytotoxicity, as demonstrated by reduced cell viability and induction of apoptosis and autophagy in mouse germ cells (a spermatocyte cell line, GC2). The expression levels of numerous lncRNAs and mRNAs in GC2 cells were altered at the transcriptome level after treatment with triptonide for 24 h, as determined by RNA sequencing. Gene ontology and pathway analyses showed that the functions of the differentially expressed lncRNAs and mRNAs were closely linked with many processes, including gene expression regulation, cell death, cell cycle regulation, cell proliferation and development and others. After validating our RNA-seq data, we selected one lncRNA, Obox4-ps35, dramatically induced by triptonide for further investigation. Obox4-ps35 knock-out aggravated triptonide-induced cytotoxicity by decreasing cell survival and increasing apoptosis and autophagy rates. These data suggest that germ cells exposed to triptonide overexpress Obox4-ps35 to protect against triptonide-induced cytotoxicity. This study provides preliminary evidence and novel directions for exploring roles of lncRNAs in triptonide-induced cytotoxicity, especially in reproductive toxicity.
Collapse
Affiliation(s)
- Xiaoning Zhang
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, China; Institute of Life Science, Nanchang University, Nanchang, China
| | - Chen Chen
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, China
| | - Yuanyuan Zhong
- Institute of Life Science, Nanchang University, Nanchang, China
| | - Xuhui Zeng
- Medical School, Institute of Reproductive Medicine, Nantong University, Nantong, China.
| |
Collapse
|
15
|
Lai M, Ge Y, Chen M, Sun S, Chen J, Cheng R. Saikosaponin D Inhibits Proliferation and Promotes Apoptosis Through Activation of MKK4-JNK Signaling Pathway in Pancreatic Cancer Cells. Onco Targets Ther 2020; 13:9465-9479. [PMID: 33061432 PMCID: PMC7522527 DOI: 10.2147/ott.s263322] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction Pancreatic cancer remains one of the most lethal malignancies and has few treatment options. Saikosaponin D (SSD), a major bioactive triterpene saponin isolated from Bupleurum chinense, has been reported to exert cytotoxicity properties toward many cancer cells. However, the effects of SSD on pancreatic cancer have been little scrutinized. Methods Here, we investigated the effect of SSD on the proliferation and apoptosis of human pancreatic cancer BxPC3 and PANC1 cells and the mouse pancreatic cancer cell line Pan02. Cell viability was determined by MTT assays and cell apoptosis analyzed by DAPI staining and flow cytometry. Expression levels of apoptosis-regulating markers and activity of the MKK4–JNK signaling pathway were determined by Western blotting. The inhibitor SP600125 was applied to confirm the role of the JNK pathway in SSD efficiency. Results SSD significantly inhibited the proliferation of BxPC3, PANC1, and Pan02 cells in a concentration- and time-dependent manner. Flow-cytometry analysis indicated obvious apoptosis induction after SSD exposure. Furthermore, SSD significantly triggered cleavage of caspase 3 and caspase 9 proteins and increased the expression of FoxO3a. In addition, activity of the MKK4–JNK pathway was dramatically increased after treatment with SSD in BxPC3 cells. SSD obviously stimulated phosphorylation of JNK, cJun, and SEK1/MKK4 proteins within 30 minutes. The addition of SP600125 blocked the activation of SSD on the MKK4–JNK regulatory pathway and reversed the effects of SSD on proliferation inhibition and apoptosis induction in BxPC3 cells. Conclusion These results revealed that SSD was capable of suppressing tumor growth and promoting apoptosis of pancreatic cancer cells via targeting the MKK4–JNK signaling pathway, indicating the possibility of further developing SSD as a potential therapeutic candidate for pancreatic cancer.
Collapse
Affiliation(s)
- Mengru Lai
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Yuqing Ge
- First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310006, People's Republic of China
| | - Meng Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Siya Sun
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Jianzhen Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| | - Rubin Cheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, People's Republic of China
| |
Collapse
|
16
|
Gao Y, Chen S, Sun J, Su S, Yang D, Xiang L, Meng X. Traditional Chinese medicine may be further explored as candidate drugs for pancreatic cancer: A review. Phytother Res 2020; 35:603-628. [PMID: 32965773 DOI: 10.1002/ptr.6847] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is a disease with a high mortality rate. Although survival rates for different types of cancers have improved in recent years, the five-year survival rate of pancreatic cancer stands at 8%. Moreover, the current first-line therapy, gemcitabine, results in low remission rates and is associated with drug resistance problems. Alternative treatments for pancreatic cancer such as surgery, chemotherapy and radiation therapy provide marginal remission and survival rates. This calls for the search of more effective drugs or treatments. Traditional Chinese medicine contains numerous bioactive ingredients some of which show activity against pancreatic cancer. In this review, we summarize the mechanisms of five types of traditional Chinese medicine monomers. In so-doing, we provide new potential drug candidates for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yue Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyu Su
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xiang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
17
|
Zhao X, Liu Z, Ren Z, Wang H, Wang Z, Zhai J, Cao D, Lyu S, Li L, Lang R, He Q. Triptolide inhibits pancreatic cancer cell proliferation and migration via down-regulating PLAU based on network pharmacology of Tripterygium wilfordii Hook F. Eur J Pharmacol 2020; 880:173225. [PMID: 32464191 DOI: 10.1016/j.ejphar.2020.173225] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/03/2020] [Accepted: 05/20/2020] [Indexed: 02/07/2023]
Abstract
Tripterygium wilfordii Hook F (TwHF) exhibits anti-tumor efficacy in pancreatic ductal adenocarcinoma (PDAC), however the pharmacological mechanisms are unclear due to complicated formulae and target genes. Using Traditional Chinese Medicine Systems Pharmacology and GeneCards databases, we performed a network pharmacology (NP) of TwHF and screened out 22 ingredients and 25 target genes associated with PDAC. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of the 25 target genes were performed. Using STRING database, protein-protein interaction network of the 25 target genes was constructed, and indicated that triptolide (TL)-plasminogen activator urokinase (PLAU) as a potential target for PDAC treatment. Hence, in vitro experiments were performed and validated that TL inhibited PDAC cell proliferation and migration by suppressing PLAU expression. The results of Western blot suggested that PLAU activated endothelial-mesenchymal transition (EMT) progression. In two Gene Expression Omnibus datasets (GSE16515 and GSE28735), PLAU was up-regulated in tumor tissues, and PLAU overexpression was associated with poor overall survival of PDAC cohort of The Cancer Genome Atlas (P < 0.01). Immunohistochemistry illustrated that overexpression of PLAU protein was related to lymph node metastasis in 20 PDAC patients (P < 0.01). Based on NP of TwHF, we identified and validated that TL-PLAU could serve as a potential target for PDAC treatment.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhe Liu
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhangyong Ren
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Huaguang Wang
- Department of Pharmacology, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zisong Wang
- Department of Traditional Chinese Medicine, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jialei Zhai
- Department of Pathology, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Di Cao
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Shaocheng Lyu
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Lixin Li
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China.
| | - Qiang He
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Yang B, Zhang B, Cao Z, Xu X, Huo Z, Zhang P, Xiang S, Zhao Z, Lv C, Meng M, Zhang G, Dong L, Shi S, Yang L, Zhou Q. The lipogenic LXR-SREBF1 signaling pathway controls cancer cell DNA repair and apoptosis and is a vulnerable point of malignant tumors for cancer therapy. Cell Death Differ 2020; 27:2433-2450. [PMID: 32144382 PMCID: PMC7370224 DOI: 10.1038/s41418-020-0514-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/23/2020] [Accepted: 02/03/2020] [Indexed: 01/13/2023] Open
Abstract
Cancer cells are defective in DNA repair, so they experience increased DNA strand breaks, genome instability, gene mutagenesis, and tumorigenicity; however, multiple classic DNA repair genes and pathways are strongly activated in malignant tumor cells to compensate for the DNA repair deficiency and gain an apoptosis resistance. The mechanisms underlying this phenomenon in cancer are unclear. We speculate that a key DNA repair gene or signaling pathway in cancer has not yet been recognized. Here, we show that the lipogenic liver X receptor (LXR)-sterol response element binding factor-1 (SREBF1) axis controls the transcription of a key DNA repair gene polynucleotide kinase/phosphatase (PNKP), thereby governing cancer cell DNA repair and apoptosis. Notably, the PNKP levels were significantly reduced in 95% of human pancreatic cancer (PC) patients, particularly deep reduction for sixfold in all of the advanced-stage PC cases. PNKP is also deficient in three other types of cancer that we examined. In addition, the expression of LXRs and SREBF1 was significantly reduced in the tumor tissues from human PC patients compared with the adjacent normal tissues. The newly identified LXR-SREBF1-PNKP signaling pathway is deficient in PC, and the defect in the pathway contributes to the DNA repair deficiency in the cancer. Strikingly, further diminution of the vulnerable LXR-SREBF1-PNKP signaling pathway using a small molecule triptonide, a new LXR antagonist identified in this investigation, at a concentration of 8 nM robustly activated tumor-suppressor p53 and readily elevated cancer cell DNA strand breaks over an apoptotic threshold, and selectively induced PC cell apoptosis, resulting in almost complete elimination of tumors in xenograft mice without obvious complications. Our findings provide new insight into DNA repair and apoptosis in cancer, and offer a new platform for developing novel anticancer therapeutics.
Collapse
Affiliation(s)
- Bo Yang
- Department of General Surgery, The Third Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- The First People's Hospital of Changzhou, Changzhou, 213003, P. R. China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
- Suzhou Institute of Systems Medicine, Suzhou, 215123, China
| | - Zhifei Cao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
| | - Xingdong Xu
- Department of General Surgery, The People's Hospital of China, Three Gorges University, Yichang, 443000, P. R. China
- The First People's Hospital of Yichang, Yichang, 443000, P. R. China
| | - Zihe Huo
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Pan Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Shufen Xiang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Zhe Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Chunping Lv
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Gaochuan Zhang
- Department of Bioinformatics, College of Basic Medical Science, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Liang Dong
- Department of Pathology, College of Basic Medical Science, Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Shucheng Shi
- Department of General Surgery, The Third Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- The First People's Hospital of Changzhou, Changzhou, 213003, P. R. China
| | - Lan Yang
- Department of General Surgery, The Third Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, P. R. China
- The First People's Hospital of Changzhou, Changzhou, 213003, P. R. China
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
- State Key Laboratory of Radiation Medicine, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
- Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
- 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, P. R. China.
| |
Collapse
|
19
|
Xiang S, Zhao Z, Zhang T, Zhang B, Meng M, Cao Z, Zhou Q. Triptonide effectively suppresses gastric tumor growth and metastasis through inhibition of the oncogenic Notch1 and NF-κB signaling pathways. Toxicol Appl Pharmacol 2020; 388:114870. [PMID: 31866380 DOI: 10.1016/j.taap.2019.114870] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022]
Abstract
Gastric cancer ranks as the third leading cause of cancer-related death worldwide. The uncontrolled tumor growth and robust metastasis are key factors to cause the cancer patient death. Mechanistically, aberrant activation of Notch and NF-κB signaling pathways plays pivotal roles in the initiation and metastasis of gastric cancer. Despite great efforts have been made in recent decades, the effective drug against the advanced and metastatic gastric cancer is still lacking in the clinical setting. In this study, we found that triptonide, a small molecule (MW358) purified from the traditional Chinese medicinal herb Tripterygium wilfordii Hook F, effectively suppressed tumor growth and metastasis in xenograft mice without obvious toxicity at the doses we tested, resulting in potent anti-gastric cancer effect with low toxicity. Triptonide markedly inhibited human metastatic gastric cancer cell migration, invasion, proliferation, and tumorigenicity. Molecular mechanistic studies revealed that triptonide significantly reduced Notch1 protein levels in metastatic gastric cancer cells through degrading the oncogenic protein Notch1 via the ubiquitin-proteasome pathway. Consequently, the levels of Notch1 downstream proteins RBPJ, IKKα, IKKβ were significantly diminished, and nuclear factor-kappa B (NF-κB) phosphorylation was significantly reduced. Together, triptonide effectively suppresses gastric cancer growth and metastasis via inhibition of the oncogenic Notch1 and NF-κB signaling pathways. Our findings provide a new strategy and drug candidate for treatment of the advanced and metastatic gastric cancer.
Collapse
Affiliation(s)
- Shufen Xiang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Zhe Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Tong Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Zhifei Cao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China.
| |
Collapse
|