1
|
Xing Z, Wei X, Fan Q, Zhao D, He J, Cheng J. Cryptotanshinone promotes ferroptosis in glioblastoma via KEAP1/NRF2/HMOX1 signaling pathway. Biochem Biophys Res Commun 2025; 768:151959. [PMID: 40345007 DOI: 10.1016/j.bbrc.2025.151959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 04/12/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Glioblastoma multiforme (GBM) is a common and highly malignant brain tumor characterized by heterogeneity, invasiveness, and resistance to therapy. Inducing ferroptosis in GBM represents a promising therapeutic strategy that inhibits angiogenesis. Natural ingredients in anti-tumor adjuvants are increasingly reported to promote cell death with fewer side effects. Salvia miltiorrhiza Bunge has been widely proven to have significant anti-tumor activity, but its mechanism remains unclear and not deeply understood. This study aimed to investigate the mechanisms by which the compound cryptotanshinone (CTS) induces cell death in glioblastoma (GBM). Our findings revealed that cryptotanshinone, a lipophilic compound, exhibited the most significant anti-tumor activity against GBM. We observed that cryptotanshinone triggered ferroptosis in GBM cells both in vitro and in vivo. RNA sequencing analysis (RNA-seq) revealed that cryptotanshinone led to the upregulation of heme oxygenase 1 (HMOX1), a key protein that facilitates the release of iron ions, which is essential for the induction of ferroptosis. Knocking down HMOX1 could restore ferrous ion levels and Glutathione peroxidase 4 (GPX4) expression to antagonize GBM ferroptosis induced by cryptotanshinone. An in vivo study also showed that cryptotanshinone inhibited GBM growth and upregulated HMOX1 expression without significant side effects. Mechanistically, we found that cryptotanshinone, acting as a protein-protein interaction (PPI) inhibitor of nuclear factor erythroid 2-related factor 2 (NRF2) and Kelch-like ECH-associated protein 1 (KEAP1), promoted the dissociation of NRF2 from KEAP1, enhancing NRF2 nuclear translocation and the transcription of HMOX1. Together, our results revealed that cryptotanshinone is a novel ferroptosis inducer for GBM treatment.
Collapse
Affiliation(s)
- Zhengcao Xing
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangyun Wei
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuju Fan
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongfeng Zhao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine at Shanghai, Shanghai, China.
| | - Jianli He
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jinke Cheng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Tung MC, Chang GM, Dai WC, Hsu CH, Chang HC, Yang WT, Ho YJ, Lu CH, Chen YH, Chang CC. Cryptotanshinone Suppresses the STAT3/BCL-2 Pathway to Provoke Human Bladder Urothelial Carcinoma Cell Death. ENVIRONMENTAL TOXICOLOGY 2025; 40:624-635. [PMID: 39601353 DOI: 10.1002/tox.24446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Bladder cancer is one of the most common human malignancies worldwide. Aberrant activation of signal transducer and activator of transcription 3 (STAT3) is crucial to driving malignant progression and predicting poor prognosis of multiple human cancers, including bladder cancer, making STAT3 a promising target of cancer therapeutics. Cryptotanshinone (CTS) is an anticancer ingredient of Danshen ( Salvia miltiorrhiza ), a top-graded Chinese medicinal herb. However, whether CTS targets STAT3 to exert its cytotoxic effect on human bladder cancer remains unknown. Herein, we demonstrated that CTS is cytotoxic to multiple human urinary bladder transitional cell carcinoma (TCC) cell lines while sparing normal human urothelial cells. CTS provoked apoptosis-dependent bladder TCC cytotoxicity, as apoptosis blockage by z-VAD-fmk markedly rescued the clonogenicity of CTS-treated cells. Besides, CTS was found to suppress constitutive and interleukin 6-inducible activation of STAT3, evidenced by the downregulation of STAT3 tyrosine 705 phosphorylation and BCL2, a recognized STAT3 transcriptional target. Notably, ectopic expression of a dominant-active STAT3 mutant (STAT3-C) or BCL-2 alleviated CTS-induced apoptosis and clonogenicity inhibition, thus confirming STAT3 blockade as a pivotal mechanism of CTS's cytotoxic action on bladder TCC cells. Lastly, immunoblotting revealed that CTS lowered the levels of active JAK2, an upstream kinase that mediates STAT3 tyrosine 705 phosphorylation. Altogether, we conclude that the blockade of the JAK2/STAT3/BCL-2 antiapoptotic signaling axis is a vital mechanism whereby CTS provokes bladder cancer cytotoxicity. The current evidence implicates CTS's potential to be translated into a bladder cancer therapeutic agent.
Collapse
Affiliation(s)
- Min-Che Tung
- Division of Urology, Department of Surgery, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Ge-Man Chang
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Wen-Chyi Dai
- Doctoral Program in Biotechnology Industrial Innovation and Management, National Chung Hsing University, Taichung, Taiwan
| | - Chen-Hsuan Hsu
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Hsiang-Chun Chang
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Ting Yang
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yann-Jen Ho
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Hsing Lu
- Department of Obstetrics and Gynecology, Taichung Veterans General Hospital, Taichung, Taiwan
- Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Hsin Chen
- Department of Nephrology, Taichung Tzu Chi Hospital, Buddhist Tzu chi Medical Foundation, Taichung, Taiwan
- School of Medicine, Tzu Chi University, Hualein, Taiwan
- Department of Artificial Intelligence and Data Science, National Chung Hsing University, Taichung, Taiwan
| | - Chia-Che Chang
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Biotechnology Industrial Innovation and Management, National Chung Hsing University, Taichung, Taiwan
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Master Program in Precision Health, Rong Hsing Research Center for Translational Medicine, The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
3
|
Lu J, Yu D, Li H, Qin P, Chen H, Chen L. Promising natural products targeting protein tyrosine phosphatase SHP2 for cancer therapy. Phytother Res 2025; 39:1735-1757. [PMID: 38558278 DOI: 10.1002/ptr.8185] [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: 09/01/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
The development of Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) inhibitors is a hot spot in the research and development of antitumor drugs, which may induce immunomodulatory effects in the tumor microenvironment and participate in anti-tumor immune responses. To date, several SHP2 inhibitors have made remarkable progress and entered clinical trials for the treatment of patients with advanced solid tumors. Multiple compounds derived from natural products have been proved to influence tumor cell proliferation, apoptosis, migration and other cellular functions, modulate cell cycle and immune cell activation by regulating the function of SHP2 and its mutants. However, there is a paucity of information about their diversity, biochemistry, and therapeutic potential of targeting SHP2 in tumors. This review will provide the structure, classification, inhibitory activities, experimental models, and antitumor effects of the natural products. Notably, this review summarizes recent advance in the efficacy and pharmacological mechanism of natural products targeting SHP2 in inhibiting the various signaling pathways that regulate different cancers and thus pave the way for further development of anticancer drugs targeting SHP2.
Collapse
Affiliation(s)
- Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danmei Yu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongtao Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Henan University, Kaifeng, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Xie L, Zhong Y, Chen Y, Wang Y, Xian P, Liu S, Xin X, Chen Y, Guan Y, Li K. Cryptotanshinone alleviates immunosuppression in endometriosis by targeting MDSCs through JAK2/STAT3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156227. [PMID: 39580997 DOI: 10.1016/j.phymed.2024.156227] [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: 07/12/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Endometriosis (EMS), a well-recognized chronic inflammatory disorder, characterized by significant immune dysregulation, in which myeloid-derived suppressor cells (MDSCs) are essential for facilitating immunosuppression and driving to disease progression. Cryptotanshinone (CTS) is an active compound capable of modulating MDSC-mediated immunosuppression; however, its therapeutic effects and mechanisms in the treatment of EMS remain unclear. PURPOSE This study aims to investigate the therapeutic potential of CTS in modulating MDSCs through JAK2/STAT3 signaling pathway and to evaluate its effects on immune microenvironment and endometriotic lesion growth in EMS. METHODS Transcriptomic data (GSE141549) and single-cell RNA sequencing data (GSE213216) were analyzed to compare immune cell populations in control endometrium (CE), eutopic endometrium (EuE) and ectopic endometrium (EcE) of patients with EMS. Network pharmacology analysis, surface plasmon resonance (SPR) and cellular thermal shift assay (CETSA) were utilized to explore the molecular mechanism of CTS's effects on MDSCs. A C57BL/6J EMS mice model was established to evaluate CTS's influence on MDSC-mediated immune response in vivo. Flow cytometry and immunofluorescence were used to analyze the immune cell populations, particularly MDSCs and CD8+ T cells. Ex vivo bone marrow (BM)-derived MDSCs were prepared to investigate the modulatory activities of CTS on the frequency and function of MDSCs. The impacts of CTS on JAK2/STAT3 pathway were further examined by western blot. RESULTS Bioinformatic analysis revealed that, among the three progression stages (CE, EuE, and EcE), the EcE stage exhibited a relatively elevated level of MDSCs and a reduced level of CD8+ T cells. Network pharmacological analysis, along with SPR and CETSA identified that CTS potentially modulates MDSCs in EMS by targeting the JAK2/STAT3 pathway. In vivo studies demonstrated that a relatively high dose of CTS treatment (60mg/kg) effectively inhibited lesion growth, reduced the population of MDSCs, and enhanced CD8+ T cell infiltration. Ex vivo experiments showed that CTS decreased the BM-derived MDSC frequency and rescued the suppressive ability of MDSC upon CD8+ T cells in a dose-dependent manner. Further mechanism analysis confirmed that CTS modulates the expression of immunosuppressive genes and proteins associated with MDSCs through JAK2/STAT3 pathway. CONCLUSION This study is the first to demonstrate that CTS is a promising natural compound for EMS treatment by inhibiting MDSC accumulation and modulating MDSC-mediated immune responses. Its therapeutic efficacy is linked to the modulation of the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Linling Xie
- The Third Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Zhong
- The Third Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Chen
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yishu Wang
- The Third Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peiyi Xian
- The Third Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shanjia Liu
- The Third Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyan Xin
- Foshan Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanfen Chen
- Foshan Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongge Guan
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Kunyin Li
- The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
5
|
Wen X, Hu J. Targeting STAT3 signaling pathway in the treatment of Alzheimer's disease with compounds from natural products. Int Immunopharmacol 2024; 141:112936. [PMID: 39163684 DOI: 10.1016/j.intimp.2024.112936] [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: 06/12/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is difficult to cure and of global concern. Neuroinflammation is closely associated with the onset and progression of AD, making its treatment increasingly important. Compounds from natural products, with fewer side effects than synthetic drugs, are of high research interest. STAT3, a multifunctional transcription factor, is involved in various cellular processes including inflammation, cell growth, and apoptosis. Its activation and inhibition can have different effects under various pathological conditions. In AD, the STAT3 protein plays a crucial role in promoting neuroinflammation and contributing to disease progression. This occurs primarily through the JAK2-STAT3 signaling pathway, which impacts microglia, astrocytes, and hippocampal neurons. This paper reviews the STAT3 signaling pathway in AD and 25 compounds targeting STAT3 up to 2024. Notably, Rutin, Paeoniflorin, and Geniposide up-regulate STAT3 in hippocampal and cortex neurons, showing neuroprotective effects in various AD models. Other 23 compounds downregulate AD by suppressing neuroinflammation through inhibition of STAT3 activation in microglia and astrocytes. These findings highlight the potential of compounds from natural products in improving AD by targeting STAT3, offering insights into the prevention and management of AD.
Collapse
Affiliation(s)
- Xiyue Wen
- Department of Clinical Laboratory, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, China
| | - Jinyue Hu
- Medical Research Center, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha 410004, China.
| |
Collapse
|
6
|
Shu C, Chen Y, Wu Z, Zhang W, Zhao J, Wang Y, Zeng Y, Li J, Zhu J, Yan Z, Liu Z. Isotoosendanin exerts anti-tumor effects in NSCLC by enhancing the stability of SHP-2 and inhibiting the JAK/STAT3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155832. [PMID: 38924928 DOI: 10.1016/j.phymed.2024.155832] [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: 01/14/2024] [Revised: 05/23/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Lung cancer has been considered as a serious problem for the public health system. NSCLC is the main type of lung cancer, and finding improved treatments for NSCLC is a pressing concern. In this study, we have explored the efficacy of isotoosendanin (ITSN) for the treatment of NSCLC, and also explored the potential underlying mechanisms. METHODS NSCLC cells were cultured, and colony formation, cell cycle as well as apoptosis assays have been conducted for investigating the biological functions of ITSN on NSCLC cells. Furthermore, target genes of ITSN have been predicted via PharmMapper and SuperPred database, subsequently validated using the drug affinity responsive target stability (DARTS) approach, a cellular thermal shift assay (CETSA) as well as surface plasmon resonance (SPR) analysis. Additionally, ubiquitination experiments have been conducted for the level of ubiquitination of the NSCLC cells. Finally, a nude mouse xenograft model has been established for evaluating the anti-tumor effects of ITSN in vivo. RESULTS ITSN has shown anti-NSCLC activities both in vitro and in vivo. Mechanistically, ITSN interacts with SHP-2 through enhancing its stability and decreases the level of ubiquitination. Notably, ITSN may regulate the behaviors of NSCLC cells via affecting the JAK/STAT3 signaling, and finally, the anti-tumor effects of ITSN was partially reversed by the application of SHP-2 inhibitor or siRNA of SHP-2. CONCLUSIONS ITSN may exert its anti-tumor effects by directly targeting SHP-2, increasing its stability and minimizing its ubiquitination. These results imply that ITSN could be a revolutionary component for treating NSCLC.
Collapse
Affiliation(s)
- Chenying Shu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yuling Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Zhengyan Wu
- Department of Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Weijie Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jian Zhao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Ying Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Yuanyuan Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jianjun Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Jianjie Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China
| | - Zhaowei Yan
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Zeyi Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China; Institute of Respiratory Diseases, Soochow University, Suzhou, 215006, China.
| |
Collapse
|
7
|
Li Z, Zheng Y, Zhang L, Xu E. Cryptotanshinone alleviates liver fibrosis via inhibiting STAT3/CPT1A-dependent fatty acid oxidation in hepatic stellate cells. Chem Biol Interact 2024; 399:111119. [PMID: 38936533 DOI: 10.1016/j.cbi.2024.111119] [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: 04/04/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Hepatic stellate cells (HSCs) are a major source of fibrogenic cells and play a central role in liver fibrogenesis. HSC activation depends on metabolic activation, for which it is well established that fatty acid oxidation (FAO) sustains their rapid proliferative rate. Studies have indicated that tanshinones inhibit HSC activation, however, the anti-fibrosis mechanisms of tanshinones are remain unclear. Herein, we reported that cryptotanshinone (CTS), a lipid-soluble ingredient of Salvia miltiorrhiza Bunge, exhibited the strongest inhibitory effects on HSC-LX2 proliferation and activation. CTS could induce lipocyte phenotype in mouse primary HSC and HSC-LX2. Transcriptomic sequencing and qPCR revealed that CTS regulated fatty acid metabolism and inhibited CPT1A and CPT1B expression. Target prediction suggested CTS regulates lipid metabolism by targeting STAT3. Mechanistically, the level of ATP and acetyl-CoA were reduced by the treatment of CTS, indicating that CTS could inhibit the level of FAO. Furthermore, CTS could inhibit the phosphorylation and nuclear translocation of STAT3. Additionally, CPT1A overexpression reversed the efficacy of CTS. Finally, CTS (40 mg/kg/day) attenuated CCl4-induced liver fibrosis and inhibited collagen production and HSC activation. Moreover, the results of immunofluorescence showed that α-SMA and p-STAT3 were co-located, and CTS could reduce the levels of p-STAT3 and α-SMA. In summary, CTS alleviated liver fibrosis by inhibiting the p-STAT3/CPT1A-dependent FAO both in vitro and in vivo, making it a potential candidate drug for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Zibo Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Yaqiu Zheng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Lin Zhang
- Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Erping Xu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
8
|
Yao C, Zhang H, Wang C. Recent advances in therapeutic engineered extracellular vesicles. NANOSCALE 2024; 16:7825-7840. [PMID: 38533676 DOI: 10.1039/d3nr05470e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Extracellular vesicles (EVs) are natural particles secreted by living cells, which hold significant potential for various therapeutic applications. Native EVs have specific components and structures, allowing them to cross biological barriers, and circulate in vivo for a long time. Native EVs have also been bioengineered to enhance their therapeutic efficacy and targeting affinity. Recently, the therapeutic potential of surface-engineered EVs has been explored in the treatment of tumors, autoimmune diseases, infections and other diseases by ongoing research and clinical trials. In this review, we will introduce the modified methods of engineered EVs, summarize the application of engineered EVs in preclinical and clinical trials, and discuss the opportunities and challenges for the clinical translation of surface-engineered EVs.
Collapse
Affiliation(s)
- Chenlu Yao
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Chao Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
9
|
Khan F, Pandey P, Verma M, Upadhyay TK. Terpenoid-Mediated Targeting of STAT3 Signaling in Cancer: An Overview of Preclinical Studies. Biomolecules 2024; 14:200. [PMID: 38397437 PMCID: PMC10886526 DOI: 10.3390/biom14020200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer has become one of the most multifaceted and widespread illnesses affecting human health, causing substantial mortality at an alarming rate. After cardiovascular problems, the condition has a high occurrence rate and ranks second in terms of mortality. The development of new drugs has been facilitated by increased research and a deeper understanding of the mechanisms behind the emergence and advancement of the disease. Numerous preclinical and clinical studies have repeatedly demonstrated the protective effects of natural terpenoids against a range of malignancies. Numerous potential bioactive terpenoids have been investigated in natural sources for their chemopreventive and chemoprotective properties. In practically all body cells, the signaling molecule referred to as signal transducer and activator of transcription 3 (STAT3) is widely expressed. Numerous studies have demonstrated that STAT3 regulates its downstream target genes, including Bcl-2, Bcl-xL, cyclin D1, c-Myc, and survivin, to promote the growth of cells, differentiation, cell cycle progression, angiogenesis, and immune suppression in addition to chemotherapy resistance. Researchers viewed STAT3 as a primary target for cancer therapy because of its crucial involvement in cancer formation. This therapy primarily focuses on directly and indirectly preventing the expression of STAT3 in tumor cells. By explicitly targeting STAT3 in both in vitro and in vivo settings, it has been possible to explain the protective effect of terpenoids against malignant cells. In this study, we provide a complete overview of STAT3 signal transduction processes, the involvement of STAT3 in carcinogenesis, and mechanisms related to STAT3 persistent activation. The article also thoroughly summarizes the inhibition of STAT3 signaling by certain terpenoid phytochemicals, which have demonstrated strong efficacy in several preclinical cancer models.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India;
| | - Pratibha Pandey
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
- Department of Chemistry, University Institute of Sciences, Chandigarh University, Gharuan, Mohali 140413, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Research and Development Cell, Parul University, Vadodara 391760, India;
| |
Collapse
|
10
|
Wang T, Zhang M, Khan M, Li J, Wu X, Ma T, Li Y. Cryptotanshinone suppresses ovarian cancer via simultaneous inhibition of glycolysis and oxidative phosphorylation. Biomed Pharmacother 2024; 170:115956. [PMID: 38039759 DOI: 10.1016/j.biopha.2023.115956] [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: 07/14/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023] Open
Abstract
Ovarian cancer is one of the most lethal cancers in female reproductive system due to heterogeneity and lack of effective treatment. Targeting aerobic glycolysis, a predominant energy metabolism of cancer cells has been recognized a novel strategy to overcome cancer cell growth. However, the capability of cancer cells to undergo metabolic reprogramming guarantees their survival even when glycolysis is inhibited. Here in this study, we have shown that Cryptotanshinone (CT), a lipid-soluble bioactive anticancer molecule of Salvia miltiorrhiza, inhibits both glycolysis and oxidative phosphorylation (OXPHOS) in ovarian cancer cells leading to growth suppression and apoptosis induction. Our mechanistic study revealed that CT decreased glucose uptake and lactate production, and inhibited the kinase activity of LDHA and HK2. The molecular docking study showed that CT could directly bind with GLUT1, LDHA, HK2, PKM2 and complex-1. The immunoblotting data showed that CT decreased the expression of aberrantly activated glycolytic proteins includingGLUT1, LDHA, HK2, and PKM2. Besides, we found that CT inhibited mitochondrial ComplexⅠ activity, decreased the ratio of NAD+/NADH, and suppressed the generation of ATP and induced activation of AMPK, which controls energy-reducing processes. These in vitro findings were further validated using xenograft model. The findings of in vivo studies were in line with in vitro studies. Taken together, CT effectively suppressed glycolysis and OXPHOS, inhibited growth and induced apoptosis in ovarian cancer cells both in vitro and in vivo study models.
Collapse
Affiliation(s)
- Tong Wang
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengmeng Zhang
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Muhammad Khan
- Cancer Research Lab, Institute of Zoology, University of the Punjab, Quaid-e-Azam Campus, Lahore, Pakistan.
| | - Jingjing Li
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiao Wu
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tonghui Ma
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yongming Li
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
11
|
Wang R, Wang X, Zhao H, Li N, Li J, Zhang H, Di L. Targeted delivery of hybrid nanovesicles for enhanced brain penetration to achieve synergistic therapy of glioma. J Control Release 2024; 365:331-347. [PMID: 38000664 DOI: 10.1016/j.jconrel.2023.11.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 09/05/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Blood-brain barrier (BBB) obstructing brain drug delivery severely hampers the therapeutic efficacy towards glioma. An efficient brain delivery strategy is of paramount importance for the treatment of glioma. Inspired by brain targeting exosome, biomimetic BBB penetrated hybrid (pHybrid) nanovesicles, engineered by membrane fusion between blood exosome and tLyp-1 peptide modified liposome, is explored for brain targeting drug delivery. Transferrin receptor (TfR) on pHybrid nanovesicles facilitates the BBB transcytosis into brain parenchyma, and eventually endocytosed by glioma cells and diffusion to extra-vascular tumor tissues under the guidance of tLyp-1 peptide. pHybrid nanovesicles co-loaded with salvianolic acid B (SAB) and cryptotanshinone (CPT), which is constructed by membrane hybridization of blood exosome loaded with SAB and tLyp-1 modified liposome loaded with CPT, are explored for cytotoxic and anti-angiogenetic therapy towards glioma. Upon accumulation at tumor site, the loaded CPT and SAB shows synergistic effects towards glioma from cytotoxicity on cancer cells and anti-angiogenesis on tumor, respectively. Overall, this study provides a biomimetic nanoplatform for increased BBB transcytosis into brain parenchyma, which serves as a prospective strategy for delivering therapeutic agents against glioma through synergistic mechanisms.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China.
| | - Xue Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Huacong Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Hanwen Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China.
| |
Collapse
|
12
|
Zhu S, Guo J, Yu L, Liu J, Chen J, Xin J, Zhang Y, Luo J, Duan C. Synergistic effect of cryptotanshinone and temozolomide treatment against human glioblastoma cells. Sci Rep 2023; 13:21835. [PMID: 38071213 PMCID: PMC10710453 DOI: 10.1038/s41598-023-48777-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a complex disease to treat owing to its profound chemoresistance. Therefore, we evaluated the combined effect and therapeutic efficacy of temozolomide (TMZ), a potent alkylating agent and the current gold standard therapy for GBM, and cryptotanshinone (CTS), which inhibits glioma cell proliferation in GBM cells. Using LN229 and U87-MG human GBM cells in a short-term stimulation in vitro model, the cytotoxic and anti-proliferative effects of single and combined treatment with 4 μM CTS and 200 μM TMZ were investigated. Furthermore, cell viability, DNA damage, apoptosis rate, and signal transducer and activator of transcription 3 (STAT3) protein were measured using cytotoxic assay, comet assay, flow cytometry, and western blotting analysis, respectively. The two drugs' synergistic interaction was validated using the synergy score. We found that the anti-proliferative effects of combination therapy using the two drugs were greater than that of each agent used alone (CTS or TMZ). Western blot analysis indicated that treatment of GBM cells with CTS combined with TMZ more significantly decreased the expression of MGMT and STAT3, than that with TMZ alone. Combined treatment with CTS and TMZ might be an effective option to overcome the chemoresistance of GBM cells in a long-term treatment strategy.
Collapse
Affiliation(s)
- Songxian Zhu
- Brain Research Institute, Research Center of Neurological Diseases, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Jingjing Guo
- Brain Research Institute, Research Center of Neurological Diseases, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Li Yu
- Brain Research Institute, Research Center of Neurological Diseases, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Jun Liu
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan, 442000, Hubei, China
| | - Jixiang Chen
- Brain Research Institute, Research Center of Neurological Diseases, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Jinxin Xin
- Brain Research Institute, Research Center of Neurological Diseases, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China
| | - Yuqiang Zhang
- Medical Services, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China.
| | - Jie Luo
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan, 442000, Hubei, China.
| | - Chao Duan
- Brain Research Institute, Research Center of Neurological Diseases, Taihe Hospital, Hubei University of Medicine, 32 Renmin South Rd, Shiyan, 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, 442000, Hubei, China.
| |
Collapse
|
13
|
Ahamed A, Hasan M, Samanta A, Alam SSM, Jamil Z, Ali S, Hoque M. Prospective pharmacological potential of cryptotanshinone in cancer therapy. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2023; 9:100308. [DOI: 10.1016/j.prmcm.2023.100308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
14
|
He Y, Nan D, Wang H. Role of Non-Receptor-Type Tyrosine Phosphatases in Brain-Related Diseases. Mol Neurobiol 2023; 60:6530-6541. [PMID: 37458988 DOI: 10.1007/s12035-023-03487-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/05/2023] [Indexed: 09/28/2023]
Abstract
The non-receptor protein tyrosine phosphatase is a class of enzymes that catalyze the dephosphorylation of phosphotyrosines in protein molecules. They are involved in cellular signaling by regulating the phosphorylation status of a variety of receptors and signaling molecules within the cell, thereby influencing cellular physiological and pathological processes. In this article, we detail multiple non-receptor tyrosine phosphatase and non-receptor tyrosine phosphatase genes involved in the pathological process of brain disease. These include PTPN6, PTPN11, and PTPN13, which are involved in glioma signaling; PTPN1, PTPN5, and PTPN13, which are involved in the pathogenesis of Alzheimer's disease Tau protein lesions, PTPN23, which may be involved in the pathogenesis of Epilepsy and PTPN1, which is involved in the pathogenesis of Parkinson's disease. The role of mitochondrial tyrosine phosphatase in brain diseases was also discussed. Non-receptor tyrosine phosphatases have great potential for targeted therapies in brain diseases and are highly promising research areas.
Collapse
Affiliation(s)
- Yatong He
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Ding Nan
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
15
|
Song H, Jiang L, Yang W, Dai Y, Wang Y, Li Z, Liu P, Chen J. Cryptotanshinone alleviates lipopolysaccharide and cigarette smoke-induced chronic obstructive pulmonary disease in mice via the Keap1/Nrf2 axis. Biomed Pharmacother 2023; 165:115105. [PMID: 37399718 DOI: 10.1016/j.biopha.2023.115105] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity worldwide. Cigarette smoking, which leads to abnormalities in the airways or alveoli and persistent obstruction of the airway's flow, is a significant risk factor of COPD. Cryptotanshinone (CTS) is the active ingredient in Salvia miltiorrhiza (Danshen) and has many pharmacological properties including anti-inflammatory, antitumor, and antioxidant properties, but its impact on COPD is uncertain. In the present study, the potential effect of CTS on COPD was investigated in a modified COPD mice model induced with cigarette smoke (CS) and lipopolysaccharide (LPS) exposure. CTS significantly reversed the decline in lung function, emphysema, inflammatory cell infiltration, small airway remodeling, pulmonary pathological damage, and airway epithelial cell proliferation in CS- and LPS-exposed mice. Additionally, CTS decreased inflammatory cytokines such as tumor necrosis factor α (TNF α), interleukins IL-6 and IL-1β, and keratinocyte chemoattractant (KC), increased the activities of superoxide dismutase (SOD), Catalase (CAT) and L-Glutathione (GSH), and repressed the expression of protein hydrolases matrix metalloprotein (MMP)- 9 and - 12 in pulmonary tissue and bronchoalveolar lavage fluid (BALF). The protective effects of CTS were also observed in human bronchial epithelial cell line BEAS-2B simulated with cigarette smoke condensate (CSC) and LPS. Mechanistically, CTS can repress the protein level of Keap1, resulting to activation of erythroid 2-related factor (Nrf2), finally alleviating COPD. In summary, the present findings demonstrated that CTS dramatically ameliorates COPD induced by CS and LPS via activating Keap1/Nrf2 pathway.
Collapse
Affiliation(s)
- Hongjia Song
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| | - Lujing Jiang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| | - Wanchun Yang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| | - Yuxing Dai
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| | - Yao Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China
| | - Zhuoming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China; National and Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Engineering Laboratory of Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China; National and Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Engineering Laboratory of Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Jianwen Chen
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, Guangdong, China; National and Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Engineering Laboratory of Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
16
|
Chen F, Zhu S, Kang R, Tang D, Liu J. ATP6V0D1 promotes alkaliptosis by blocking STAT3-mediated lysosomal pH homeostasis. Cell Rep 2023; 42:111911. [PMID: 36640329 DOI: 10.1016/j.celrep.2022.111911] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/30/2022] Open
Abstract
Alkaliptosis, a type of regulated cell death driven by intracellular alkalization, was first described in pancreatic ductal adenocarcinoma (PDAC) cells after treatment with the opioid analgesic drug JTC801. Here, we used mass-spectrometry-based drug target identification, cellular thermal shift assay, and point mutation technologies to reveal ATP6V0D1 as a direct JTC801 target that drives alkaliptosis in human PDAC cells. Functionally, the protein stability of ATP6V0D1, when mediated by JTC801, increases the interaction between ATP6V0D1 and STAT3, resulting in increased expression and activity of STAT3 for sustaining lysosome homeostasis. Consequently, the pharmacological or genetic inhibition of STAT3 restores the sensitivity of ATP6V0D1-deficient cells to alkaliptosis in vitro or in suitable mouse models. Clinically, a high expression of ATP6V0D1 correlates with prolonged survival of patients with PDAC. Together, these results illustrate a link between ATP6V0D1 and PDAC and advance our understanding of alkaliptosis in targeted therapy.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Shan Zhu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, Guangdong 511436, China.
| |
Collapse
|
17
|
Cryptotanshinone modulates proliferation, apoptosis, and fibrosis through inhibiting AR and EGFR/STAT3 axis to ameliorate benign prostatic hyperplasia progression. Eur J Pharmacol 2022; 938:175434. [PMID: 36462735 DOI: 10.1016/j.ejphar.2022.175434] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/27/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
Benign prostatic hyperplasia (BPH) is a chronic proliferative non-tumorous disease that mainly bothers males older than 50 and significantly disturbs the quality of life. Cryptotanshinone (CTS), a herbal extract, has been proven with therapeutic effects on various diseases. However, the effects and possible mechanisms of CTS in BPH have not yet been elucidated. This study aims to investigate the efficacy of CTS on the BPH-associated pathological processes and the possible mechanisms underlying it. Herein, CTS was intragastrically administrated to estradiol/testosterone (E2/T) (1:100)-induced BPH rats, and finasteride (Fi) was used as the positive control. Human benign prostatic hyperplasia epithelial cells (BPH-1) and normal human prostate stromal cells (WPMY-1) were used for the in vitro experiments. Results indicated that E2/T injection was able to induce BPH manifestation, featured with increased prostate index. Furthermore, it accelerated proliferation, epithelial-mesenchymal transition (EMT), stromal collagen deposition, and inhibited apoptosis of rat prostate. However, the administration of CTS partially reversed the changes mentioned above. The therapeutic effects of CTS on BPH were also confirmed by in vitro experiments. The efficacy of CTS on these processes might be attributed to the suppression of AR and EGFR/STAT3 axis activity. In conclusion, CTS might suppress BPH progression by modulating proliferation, apoptosis, EMT, and stromal collagen deposition via suppressing AR and EGFR/STAT3 axis.
Collapse
|
18
|
Zhao Q, Bai J, Chen Y, Liu X, Zhao S, Ling G, Jia S, Zhai F, Xiang R. An optimized herbal combination for the treatment of liver fibrosis: Hub genes, bioactive ingredients, and molecular mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115567. [PMID: 35870684 DOI: 10.1016/j.jep.2022.115567] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver fibrosis is a chronic liver disease that can lead to cirrhosis, liver failure, and hepatocellular carcinoma, and it is associated with long-term adverse outcomes and mortality. As a primary resource for complementary and alternative medicine, traditional Chinese medicine (TCM) has accumulated a large number of effective formulas for the treatment of liver fibrosis in clinical practice. However, studies on how to systematically optimize TCM formulas are still lacking. AIM OF THE REVIEW To provide a methodological reference for the systematic optimization of TCM formulae against liver fibrosis and explored the underlying molecular mechanisms; To provide an efficient method for searching for lead compounds from natural sources and developing from herbal medicines; To enable clinicians and patients to make more reasonable choices and promote the effective treatment toward those patients with liver fibrosis. MATERIALS AND METHODS TCM formulas related to treating liver fibrosis were collected from the Web of Science, PubMed, the China National Knowledge Infrastructure (CNKI), Wan Fang, and the Chinese Scientific Journals Database (VIP). Furthermore, the TCM compatibility patterns were mined using association analysis. The core TCM combinations were found by designing an optimized formulas algorithm. Finally, the hub target proteins, potential molecular mechanisms, and active compounds were explored through integrative pharmacology and docking-based inverse virtual screening (IVS) approaches. RESULTS We found that the herbs for reinforcing deficiency, activating blood, removing blood stasis, and clearing heat were the basis of TCM formulae patterns. Furthermore, the combination of Salviae Miltiorrhizae (Salvia miltiorrhiza Bunge; Chinese salvia/Danshen), Astragali Radix (Astragalus membranaceus (Fisch.) Bunge; Astragalus/Huangqi), and Radix Bupleuri (Bupleurum chinense DC.; Bupleurum/Chaihu) was identified as core groups. A total of six targets (TNF, STAT3, EGFR, IL2, ICAM1, PTGS2) play a pivotal role in TCM-mediated liver fibrosis inhibition. (-)-Cryptotanshinone, Tanshinaldehyde, Ononin, Thymol, Daidzein, and Formononetin were identified as active compounds in TCM. And mechanistically, TCM could affect the development of liver fibrosis by regulating inflammation, immunity, angiogenesis, antioxidants, and involvement in TNF, MicroRNAs, Jak-STAT, NF-kappa B, and C-type lectin receptors (CLRs) signaling pathways. Molecular docking results showed that key components had good potential to bind to the target genes. CONCLUSION In summary, this study provides a methodological reference for the systematic optimization of TCM formulae and exploration of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Qianqian Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jinwei Bai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yiwei Chen
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xin Liu
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shangfeng Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Guixia Ling
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shubing Jia
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Fei Zhai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Rongwu Xiang
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China; Liaoning Professional Technology Innovation Center on Medical Big Data and Artificial Intelligence, Shenyang, 110016, China.
| |
Collapse
|
19
|
Shakeri A, Hafezian T, Kúsz N, Hohmann J, Boozari M, Mottaghipisheh J, Emami SA, Tayarani-Najaran Z, Asili J. Cytotoxicity, apoptosis inducing activity and Western blot analysis of tanshinone derivatives from Stachys parviflora on prostate and breast cancer cells. Mol Biol Rep 2022; 49:8251-8258. [PMID: 36002657 DOI: 10.1007/s11033-022-07541-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 12/24/2022]
Abstract
Cytotoxic activities of methanolic crude extract of Stachys parviflora (Lamiaceae family) and its sub-fractions were primarily evaluated against human breast adenocarcinoma (MCF-7 and MDA-MB-231) and prostate (PC3) cell lines. The methanolic extract exhibited the highest activity, and was chosen for the isolation procedure. Four diterpenoid quinones, namely miltirone [1], tanshinone IIA [2], 1-hydroxy-tanshinone IIA [3], and cryptotanshinone [4] were isolated. Notably, this is the first report on the isolation and/or characterization of the mentioned diterpenoids from the Stachys genus. In this study, 1-hydroxy-tanshinone IIA [3] displayed the highest cytotoxicity among the isolated compounds. The mechanism of the cytotoxicity of methanolic extract and isolated compounds was further investigated by the utilization of propidium iodide staining (PI) assay. The results showed that the methanolic extract and 1-hydroxy-tanshinone IIA [3] enhanced DNA fragmentation in PC3 and MCF-7 cells. Moreover, the western blotting analysis demonstrated increasing and decreasing protein levels of Bax and Bcl2, respectively, and cleaved poly ADP-ribose polymerase (PARP). Further bioassay-guided phytochemical assessments of S. parviflora can be suggested as a promising approach for discovering potent bioactive secondary metabolites.
Collapse
Affiliation(s)
- Abolfazl Shakeri
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Toktam Hafezian
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Norbert Kúsz
- Department of Pharmacognosy, Interdisciplinary Excellence Centre, University of Szeged, Eötvös u. 6, H-6720, Szeged, Hungary
| | - Judit Hohmann
- Department of Pharmacognosy, Interdisciplinary Excellence Centre, University of Szeged, Eötvös u. 6, H-6720, Szeged, Hungary
| | - Motahare Boozari
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Mottaghipisheh
- Institute of Pharmacy/Pharmacognosy, Center for Molecular Biosciences (CMBI), University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
20
|
Saraf R, Datta A, Sima C, Hua J, Lopes R, Bittner ML, Miller T, Wilson-Robles HM. In Silico Modeling of the Induction of Apoptosis by Cryptotanshinone in Osteosarcoma Cell Lines. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1683-1693. [PMID: 33180729 DOI: 10.1109/tcbb.2020.3037318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor of both children and pet canines. Its characteristic genomic instability and complexity coupled with the dearth of knowledge about its etiology has made improvement in the current treatment difficult. We use the existing literature about the biological pathways active in OS and combine it with the current research involving natural compounds to identify new targets and design more effective drug therapies. The key components of these pathways are modeled as a Boolean network with multiple inputs and multiple outputs. The combinatorial circuit is employed to theoretically predict the efficacies of various drugs in combination with Cryptotanshinone. We show that the action of the herbal drug, Cryptotanshinone on OS cell lines induces apoptosis by increasing sensitivity to TNF-related apoptosis-inducing ligand (TRAIL) through its multi-pronged action on STAT3, DRP1 and DR5. The Boolean framework is used to detect additional drug intervention points in the pathway that could amplify the action of Cryptotanshinone.
Collapse
|
21
|
Osteosarcoma cell proliferation suppression via SHP-2-mediated inactivation of the JAK/STAT3 pathway by tubocapsenolide A. J Adv Res 2022; 34:79-91. [PMID: 35024182 PMCID: PMC8655134 DOI: 10.1016/j.jare.2021.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 05/19/2021] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction Previously, we have reported a withanolide-type steroid, named tubocapsenolide A (TA), which shows potent anti-proliferative activity in several cancer cell lines. However, its inhibitory effect on the Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway and therapeutic potential on osteosarcoma have not been reported. Objectives In the present study, we aimed to investigate the effect and molecular mechanism of TA in osteosarcoma. Methods The biological functions of TA in U2OS cells were investigated using colony formation, 5-ethynyl-20-deoxyuridine (EDU) staining, and cell cycle/apoptosis assays. The interaction between TA and Src homology 2 phosphatase 2 (SHP-2) was detected by enzyme activity and validated by target-identification methods such as drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), and biolayer interferometry (BLI). The in vivo anti-tumor efficacy of TA was analyzed in the xenograft tumor model. Western blotting analysis was performed to detect the protein expression levels. Results TA exhibited antitumor activity against osteosarcoma both in vitro and in vivo by regulating the JAK/STAT3 signaling pathway. Mechanically, TA interacted with SHP-2 directly and activated its phosphatase activity. Importantly, protein tyrosine phosphatase (PTP) inhibitor, SHP-2 inhibitor, and SHP-2 siRNA could reverse the inhibitory effect of TA on the JAK/STAT3 signaling pathway and restored the TA-induced cell death. Conclusion TA activated the phosphatase activity of SHP-2, which resulted in the inhibition of the JAK/STAT3 pathway and contributed to the antitumor efficacy of TA. Collectively, these findings suggested that TA could serve as a novel therapeutic agent for the treatment of osteosarcoma.
Collapse
|
22
|
Meng F, Zhou J, Cheng X, Xu J, Kang L, Li D, Wang D, Bi Y. Design, Synthesis and Cardioprotection of Cryptotanshinone Derivatives. CHINESE J ORG CHEM 2022. [DOI: 10.6023/cjoc202110004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
23
|
Pagano A, Breuzard G, Parat F, Tchoghandjian A, Figarella-Branger D, De Bessa TC, Garrouste F, Douence A, Barbier P, Kovacic H. Tau Regulates Glioblastoma Progression, 3D Cell Organization, Growth and Migration via the PI3K-AKT Axis. Cancers (Basel) 2021; 13:cancers13225818. [PMID: 34830972 PMCID: PMC8616151 DOI: 10.3390/cancers13225818] [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: 09/30/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary The Microtubule-associated protein Tau is expressed in different cancers; however, its role and prognostic value are still debated. In the present work, we evaluated the role of Tau in glioblastoma by down-regulating its expression in glioblastoma cells. We showed that Tau: (1) is required for tumor progression in nude mice; (2) is necessary for glioblastoma 3D cell organization, growth, and migration; and (3) regulates the PI3K/AKT signaling pathway. Abstract The Microtubule-Associated Protein Tau is expressed in several cancers, including low-grade gliomas and glioblastomas. We have previously shown that Tau is crucial for the 2D motility of several glioblastoma cell lines, including U87-MG cells. Using an RNA interference (shRNA), we tested if Tau contributed to glioblastoma in vivo tumorigenicity and analyzed its function in a 3D model of multicellular spheroids (MCS). Tau depletion significantly increased median mouse survival in an orthotopic glioblastoma xenograft model. This was accompanied by the inhibition of MCS growth and cell evasion, as well as decreased MCS compactness, implying N-cadherin mislocalization. Intracellular Signaling Array analysis revealed a defective activation of the PI3K/AKT pathway in Tau-depleted cells. Such a defect in PI3K/AKT signaling was responsible for reduced MCS growth and cell evasion, as demonstrated by the inhibition of the pathway in control MCS using LY294002 or Perifosine, which did not significantly affect Tau-depleted MCS. Finally, analysis of the glioblastoma TCGA dataset showed a positive correlation between the amount of phosphorylated Akt-Ser473 and the expression of MAPT RNA encoding Tau, underlining the relevance of our findings in glioblastoma disease. We suggest a role for Tau in glioblastoma by controlling 3D cell organization and functions via the PI3K/AKT signaling axis.
Collapse
Affiliation(s)
- Alessandra Pagano
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
- Correspondence:
| | - Gilles Breuzard
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Fabrice Parat
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Aurélie Tchoghandjian
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 8, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (A.T.); (D.F.-B.)
| | - Dominique Figarella-Branger
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 8, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (A.T.); (D.F.-B.)
- Service d’Anatomie Pathologique et de Neuropathologie, CHU Timone, APHM, 13005 Marseille, France
| | - Tiphany Coralie De Bessa
- LIM 64: Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-090, SP, Brazil;
| | - Françoise Garrouste
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Alexis Douence
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Pascale Barbier
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| | - Hervé Kovacic
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), Team 9, UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France; (G.B.); (F.P.); (F.G.); (A.D.); (P.B.); (H.K.)
| |
Collapse
|
24
|
Liu Y, Guo G, Lu Y, Chen X, Zhu L, Zhao L, Li C, Zhang Z, Jin X, Dong J, Yang X, Huang Q. Silencing IKBKE inhibits the migration and invasion of glioblastoma by promoting Snail1 degradation. Clin Transl Oncol 2021; 24:816-828. [PMID: 34741724 DOI: 10.1007/s12094-021-02726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is one of the most common malignant brain tumors in adults and has high mortality and relapse rates. Over the past few years, great advances have been made in the diagnosis and treatment of GBM, but unfortunately, the five-year overall survival rate of GBM patients is approximately 5.1%. Inhibitor of nuclear factor kappa-B kinase subunit epsilon (IKBKE) is a major oncogenic protein in tumors and can promote evil development of GBM. Snail1, a key inducer of the epithelial-mesenchymal transition (EMT) transcription factor, is subjected to ubiquitination and degradation, but the mechanism by which Snail1 is stabilized in tumors remains unclear. Our study aimed to investigate the mechanism of IKBKE regulating Snail1 in GBM. METHODS First, we analyzed the correlation between the expression of IKBKE and the tumor grade and prognosis through public databases and laboratory specimen libraries. Second, immunohistochemistry (IHC) and western blot were used to detect the correlation between IKBKE and Snail expression in glioma samples and cell lines. Western blot and immunofluorescence (IF) experiments were used to detect the quality and distribution of IKBKE and Snail1 proteins. Third, In situ animal model of intracranial glioma to detect the regulatory effect of IKBKE on intracranial tumors. RESULTS In this study, Our study reveals a new connection between IKBKE and Snail1, where IKBKE can directly bind to Snail1, translocate Snail1 into the nucleus from the cytoplasm. Downregulation of IKBKE results in Snail1 destabilization and impairs the tumor cell migration and invasion capabilities. CONCLUSION Our studies suggest that the IKBKE-Snail1 axis may serve as a potential therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Y Liu
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China.,Department of Neurosurgery, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - G Guo
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China.,Department of Neurosurgery, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Y Lu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - X Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - L Zhu
- Department of Pathology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - L Zhao
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China.,Department of Neurosurgery, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - C Li
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, Zhengzhou, 450003, Henan, China.,Department of Neurosurgery, Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China
| | - Z Zhang
- Department of Neurosurgery, Ningbo Hospital of Zhejiang University, Ningbo, 315000, Zhejiang, China
| | - X Jin
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300052, China
| | - J Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - X Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.,Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China.,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Q Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China. .,Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China. .,Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China.
| |
Collapse
|
25
|
Sun J, Sun Z, Gareev I, Yan T, Chen X, Ahmad A, Zhang D, Zhao B, Beylerli O, Yang G, Zhao S. Exosomal miR-2276-5p in Plasma Is a Potential Diagnostic and Prognostic Biomarker in Glioma. Front Cell Dev Biol 2021; 9:671202. [PMID: 34141710 PMCID: PMC8204016 DOI: 10.3389/fcell.2021.671202] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/23/2021] [Indexed: 12/18/2022] Open
Abstract
Introduction Exosomal microRNAs (miRNAs) play an essential role in near and distant intercellular communication and are potential diagnostic and prognostic biomarkers for various cancers. This study focused on evaluation of exosomal miR-2276-5p in plasma as a diagnostic and prognostic biomarker for glioma. Methods Plasma exosomes from 124 patients with glioma and 36 non-tumor controls were collected and subjected to quantitative real-time polymerase chain reaction (qRT-PCR) analysis for the exosomal miR-2276-5p expression. Bioinformatic analyses were performed to identify a gene target, and CGGA and TCGA databases were checked for evaluation of prognostic relevance. Results The exosomal miR-2276-5p in glioma patients had a significantly decreased expression, compared with non-glioma patients (p < 0.01). Receiver operating characteristics (ROC) curve analyses were observed to regulate the diagnostic sensitivity and specificity of miR-2276-5p in glioma; the area under the curve (AUC) for miR-2276-5p was 0.8107. The lower expression of exosomal miR-2276-5p in patients with glioma correlated with poorer survival rates. RAB13 was identified as the target of miR-2276-5p which was high in glioma patients, especially those with higher tumor grades and correlated with poor survival. Conclusion The circulating exosomal miR-2276-5p is significantly reduced in the plasma of glioma patients, and thus, it could be a potential biomarker for patients with glioma for diagnostic and/or prognostic purposes.
Collapse
Affiliation(s)
- Jingxian Sun
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Zhenying Sun
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Ilgiz Gareev
- Central Research Laboratory, Bashkir State Medical University, Ufa, Russia
| | - Tao Yan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Aamir Ahmad
- University of Alabama at Birmingham, Birmingham, AL, United States
| | - Daming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Boxian Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Ozal Beylerli
- Central Research Laboratory, Bashkir State Medical University, Ufa, Russia
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| | - Shiguang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Institute of Brain Science, Harbin Medical University, Harbin, China
| |
Collapse
|
26
|
Bittner ML, Lopes R, Hua J, Sima C, Datta A, Wilson-Robles H. Comprehensive live-cell imaging analysis of cryptotanshinone and synergistic drug-screening effects in various human and canine cancer cell lines. PLoS One 2021; 16:e0236074. [PMID: 33544704 PMCID: PMC7864433 DOI: 10.1371/journal.pone.0236074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Background Several studies have highlighted both the extreme anticancer effects of Cryptotanshinone (CT), a Stat3 crippling component from Salvia miltiorrhiza, as well as other STAT3 inhibitors to fight cancer. Methods Data presented in this experiment incorporates 2 years of in vitro studies applying a comprehensive live-cell drug-screening analysis of human and canine cancer cells exposed to CT at 20 μM concentration, as well as to other drug combinations. As previously observed in other studies, dogs are natural cancer models, given to their similarity in cancer genetics, epidemiology and disease progression compared to humans. Results Results obtained from several types of human and canine cancer cells exposed to CT and varied drug combinations, verified CT efficacy at combating cancer by achieving an extremely high percentage of apoptosis within 24 hours of drug exposure. Conclusions CT anticancer efficacy in various human and canine cancer cell lines denotes its ability to interact across different biological processes and cancer regulatory cell networks, driving inhibition of cancer cell survival.
Collapse
Affiliation(s)
- Michael L. Bittner
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Rosana Lopes
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
- * E-mail: (RL); (HWR)
| | - Jianping Hua
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
| | - Chao Sima
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
| | - Aniruddha Datta
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
| | - Heather Wilson-Robles
- College of Veterinary Medicine, Texas A&M University, College Station, TX, United States of America
- * E-mail: (RL); (HWR)
| |
Collapse
|
27
|
Li H, Gao C, Liu C, Liu L, Zhuang J, Yang J, Zhou C, Feng F, Sun C, Wu J. A review of the biological activity and pharmacology of cryptotanshinone, an important active constituent in Danshen. Biomed Pharmacother 2021; 137:111332. [PMID: 33548911 DOI: 10.1016/j.biopha.2021.111332] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cryptotanshinone (IUPAC name: (R)-1,2,6,7,8,9-hexahydro-1,6,6-trimethyl-phenanthro(1,2-b)furan-10,11-dione), a biologically active constituent extracted from the roots and rhizomes of the plant Salvia miltiorrhiza, has been studied in depth as a medicinally active compound and shown to have efficacy in the treatment of numerous diseases and disorders. In this review, we describe in detail the current status of cryptotanshinone research, including findings relating to the structure, pharmacokinetics, pharmacological activity, and derivatives of this compound. Cryptotanshinoneh as a diverse range of pharmacological effects, including anti-cancer, anti-inflammatory, immune regulatory, neuroprotective, and anti-fibrosis activities. Studies on the molecular mechanisms underlying the activities of cryptotanshinone have established that the JAK2/STAT3, PI3K/AKT, NF-κB, AMPK, and cell cycle pathways are involved in the inhibitory and pro-apoptotic effects of cryptotanshinone on different tumor cell lines, these molecular pathways interact in a coordinated manner to inhibit cell proliferation, migration and invasion,and induce transformation, autophagy, necrosis, and cellular immunity. The anti-inflammatory mechanisms of cryptotanshinone have been found to be associated with the TLR4-MyD88/PI3K/Nrf2 and TLR4-MyD88/NF-κB/MAPK pathways, whereasthe Hedgehog, NF-κB, and Nrf-2/HO-1 pathways are regulated by cryptotanshinone to reduce organ fibrosis, and its inhibitory effects on the PI3K/AKT-eNOS pathway have been linked to neuroprotective effects. Given the potential medicinal utility of cryptotanshinone, further research is needed to verify the efficacy and safety of this compound in clinical use, evaluate its pharmacological activity, and identify molecular targets.
Collapse
Affiliation(s)
- Huayao Li
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Lijuan Liu
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Department of Basic Medical Science, Qingdao University, Qingdao, 266071, PR China.
| | - Jing Zhuang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Qingdao Academy of Chinese Medical Sciences, Shandong University of Chinese Medicine, Qingdao, 266112, Shandong, PR China.
| | - Jing Yang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China.
| | - Chao Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China; Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China.
| | - Fubin Feng
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Department of Basic Medical Science, Qingdao University, Qingdao, 266071, PR China.
| | - Changgang Sun
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Chinese Medicine Innovation Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Jibiao Wu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| |
Collapse
|
28
|
Mitra R, Ayyannan SR. Small-Molecule Inhibitors of Shp2 Phosphatase as Potential Chemotherapeutic Agents for Glioblastoma: A Minireview. ChemMedChem 2020; 16:777-787. [PMID: 33210828 DOI: 10.1002/cmdc.202000706] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/13/2020] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is a dreadful cancer characterised by poor prognosis, low survival rate and difficult clinical correlations. Several signalling pathways and molecular mediators are known to precipitate GBM, and small-molecular targets of these mediators have become a favoured thrust area for researchers to develop potent anti-GBM drugs. Shp2, an important phosphatase of the nonreceptor type protein tyrosine phosphatase (PTPN) subfamily is responsible for master regulation of several such signalling pathways in normal and glioma cells. Thus, inhibition of Shp2 is a logical strategy for the design and development of anti-neoplastic drugs against GBM. Though tapping the full potential of Shp2 binding sites has been challenging, nevertheless, many synthetic and natural scaffolds have been documented as possessing potent and selective anti-Shp2 activities in biochemical and cellular assays, through either active-site or allosteric binding. Most of these scaffolds share a few common pharmacophoric features, a thorough study of which is useful in paving the way for the design and development of improved Shp2 inhibitors. This minireview summarizes the current scenario of potent small-molecule Shp2 inhibitors and emphasizes the anti-GBM potential of some important scaffolds that have shown promising GBM-specific activity in in vitro and in vivo models, thus proving their efficacy in GBM therapy. This review could guide researchers to design new and improved anti-Shp2 pharmacophores and develop them as anti-GBM agents by employing GBM-centric drug-discovery protocols.
Collapse
Affiliation(s)
- Rangan Mitra
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| | - Senthil R Ayyannan
- Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, Uttar Pradesh, India
| |
Collapse
|
29
|
He Y, Su W, He X, Chen T, Zeng X, Yan Z, Zhang W, Yang W, Guo J, Wu H. Pharmacokinetics and biotransformation investigation in beagle dog of active compounds from naoxintong capsule. Biomed Pharmacother 2020; 133:110940. [PMID: 33227707 DOI: 10.1016/j.biopha.2020.110940] [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: 07/17/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 10/23/2022] Open
Abstract
Naoxintong Capsule (NXTC), a standardized herbal medicine, has been widely applied in treating cardiovascular and cerebrovascular diseases with remarkable efficacy. However, the efficacy contributing components of NXTC are unclear, and the in vivo absorption and metabolism processes of NXTC remain largely obscured. In this study, using beagle dog as model species, we have identified and tentatively characterized 25 prototype and 15 catabolites of NXTC in beagle dog plasma by ultra-fast liquid chromatography/quadrupole-time-of-flight tandem mass spectrometry (UFLC-Q-TOF-MS/MS). We have proposed the in vivo bio-transformation pathways of these absorbed constituents. In addition, for six crucial components, we have developed a quantitative method and conducted plasma pharmacokinetic study of these six components by rapid resolution liquid chromatography tandem triple quadrupole mass spectrometry (RRLC-QQQ-MS/MS). In conclude, our study provided comprehensive insights into the understanding of the plasma absorbed components profiling of NXTC as well as their in vivo transformation behaviors, which would be of great value for identifying efficacy contributing critical components as well as mechanism related investigations of NXTC in the future.
Collapse
Affiliation(s)
- Yan He
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Su
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiang He
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Taobin Chen
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xuan Zeng
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zenghao Yan
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Weijian Zhang
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei Yang
- Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou, Guangdong, PR China
| | - Jianmin Guo
- Guangdong Lewwin Pharmaceutical Research Institute Co., Ltd., Guangdong Provincial Key Laboratory of Drug Non-Clinical Evaluation and Research, Guangdong Engineering Research Center for Innovative Drug Evaluation and Research, Guangzhou, Guangdong, PR China
| | - Hao Wu
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
30
|
Wu YH, Wu YR, Li B, Yan ZY. Cryptotanshinone: A review of its pharmacology activities and molecular mechanisms. Fitoterapia 2020; 145:104633. [DOI: 10.1016/j.fitote.2020.104633] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
|
31
|
Yi R, Wang H, Deng C, Wang X, Yao L, Niu W, Fei M, Zhaba W. Dihydroartemisinin initiates ferroptosis in glioblastoma through GPX4 inhibition. Biosci Rep 2020; 40:BSR20193314. [PMID: 32452511 PMCID: PMC7313443 DOI: 10.1042/bsr20193314] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/30/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022] Open
Abstract
It has been demonstrated from previous studies about the killing effect of dihydroartemisinin (DHA) on glioblastoma, which involves multiple aspects: cytotoxicity, cell cycle arrest and invasion inhibition. DHA has the advantages of low cytotoxicity to normal cells, selective killing effect and low drug resistance, making it one of the popular anti-tumor research directions. Ferroptosis is a newly discovered form of cell death characterized by iron dependence and lipid reactive oxygen species (ROS) accumulation. In the present study, we found differences in the expression of transferrin receptors in normal human astrocytes (NHA) and glioblastoma cells (U87 and A172), which may be one of the mechanisms of DHA selective killing effect. Through the determination of ferroptosis-related protein expression, we found that the significant decrease of GPX4, accompanied by the constant expression of xCT and ACSL4, suggesting GPX4 was a pivotal target for DHA-activated ferroptosis in glioblastoma. Total and lipid ROS levels were increased and all these results could be reversed by the ferroptosis inhibitor, ferrostatin-1. These findings demonstrated ferroptosis would be a critical component of cell death caused by DHA and GPX4 was the main target. All these results provide a novel treatment direction to glioblastoma. The association between ferroptosis and polyamines is also discussed, which will provide new research directions for ferroptosis caused by DHA in glioblastoma.
Collapse
Affiliation(s)
- Renxin Yi
- Department of Neurosurgery, Jinling Hospital, Southeast University, School of Medicine, Nanjing 210002, P.R. China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Nanjing University, School of Medicine, Nanjing 210002, P.R. China
| | - Chulei Deng
- Department of Neurosurgery, Jinling Hospital, South Medical University, School of Medicine, Nanjing 210002, P.R. China
| | - Xinyue Wang
- State Key Laboratory of Bioelectronics, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210002, P.R. China
| | - Lei Yao
- Department of Neurosurgery, Jinling Hospital, Southeast University, School of Medicine, Nanjing 210002, P.R. China
| | - Wenhao Niu
- Department of Neurosurgery, Jinling Hospital, Southeast University, School of Medicine, Nanjing 210002, P.R. China
| | - Maoxing Fei
- Department of Neurosurgery, Jinling Hospital, Nanjing Medical University, School of Medicine, Nanjing 210002, P.R. China
| | - Wangdui Zhaba
- Department of Neurosurgery, Jinling Hospital, Nanjing University, School of Medicine, Nanjing 210002, P.R. China
| |
Collapse
|
32
|
Sheng J, Meng Q, Yang Z, Guan J, Zhao Y, Zhang J, Wang Y, Zhao L, Wang Y. Identification of cryptotanshinone from Tongmai to inhibit thrombosis in zebrafish via regulating oxidative stress and coagulation cascade. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153263. [PMID: 32563016 DOI: 10.1016/j.phymed.2020.153263] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/22/2020] [Accepted: 06/03/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Thromboembolic events are leading causes of mortality and morbidity all over the world. Tongmai (TM) is a botanical drug with valid clinical efficacy and safety in the management of thrombosis and ischemic cardiovascular diseases, however, its active compounds and underlying mechanism are largely unclear. PURPOSE To investigate the endogenous effects, therapeutic mechanism and active compounds of TM in thrombus formation. STUDY DESIGN Combined with transgenic zebrafish models and high-content imaging system, this study evaluated the endogenous antithrombotic effects of TM and screened for the active compounds. METHODS The PHZ-induced thrombotic model in erythrocytes or platelets labeled transgenic zebrafish were established, to dynamically evaluate the antithrombotic effects of TM. The oxidative damage levels were analyzed by specific fluorescent probes, and the expression levels of key factors in coagulation cascades and platelet activation were examined by QPCR. TM were dissected into fractions by reverse phase chromatography and subsequently screened for their antithrombotic effects in the transgenic fish models. The compounds of the active TM fraction were then analyzed by UPLC-Q-TOF analysis and further verified for their antithrombotic effects and mechanisms. RESULTS In PHZ-induced zebrafish thrombotic model, TM incubation markedly increased cardiac blood flow, decreased peripheral erythrocytes aggregation, and recovered peripheral platelet circulation. Besides, the levels of oxidative stress and lipid peroxidation were increased in the PHZ-induced thrombotic fish, which were greatly decreased by TM treatment. Moreover, TM significantly reduced the expression of coagulation factor II (thrombin) and the downstream fibrinogen. In order to identify the active compounds of TM, four fractions were separated from the extract by reverse phase chromatography, which were subsequently screened for their antithrombotic effects in the fish model. As a result, fraction 4 showed the strongest effect in inhibiting thrombosis. Finally, through UPLC-Q-TOF analysis and endogenous screening, cryptotanshione was identified as the main active compound with antithrombotic effects. CONCLUSION Our study demonstrated the endogenous antithrombotic effects of TM, which is possibly mediated by inhibiting oxidative stress and coagulation cascade. Cryptotanshione was identified as a major compound with antithrombotic activity and is a promising candidate for novel antithrombotic therapy.
Collapse
Affiliation(s)
- Jian Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingfen Meng
- Henan Fusen Pharmaceutical Co., Ltd., Nanyang, 474450, China
| | - Zhenzhong Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianli Guan
- Henan Fusen Pharmaceutical Co., Ltd., Nanyang, 474450, China
| | - Yu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jide Zhang
- Henan Fusen Pharmaceutical Co., Ltd., Nanyang, 474450, China
| | - Yingchao Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
33
|
Quantitative Determination and Validation of Four Ketones in Salvia miltiorrhiza Bunge Using Quantitative Proton Nuclear Magnetic Resonance Spectroscopy. Molecules 2020; 25:molecules25092043. [PMID: 32353926 PMCID: PMC7248917 DOI: 10.3390/molecules25092043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Salvia mltiorrhiza Bunge (SMB) is native to China, whose dried root has been used as medicine. A few chromatographic- or spectrometric-based methods have already been used to analyze the lipid-soluble components in SMB. However, the methodology of qNMR on the extracts of fresh SMB root has not been verified so far. The purpose of this study was to establish a fast and simple method to quantify the tanshinone I, tanshinone IIA, dihydrotanshinone, and cryptotanshinone in fresh Salvia Miltiorrhiza Bunge root without any pre-purification steps using 1H-NMR spectroscopy. The process is as follows: first, 70% methanol aqueous extracts of fresh Salvia Miltiorrhiza Bunge roots were quantitatively analyzed for tanshinone I, tanshinone IIA, dihydrotanshinone, and cryptotanshinone using 1H-NMR spectroscopy. Different internal standards were tested and the validated method was compared with HPLC. 3,4,5-trichloropyridine was chosen as the internal standard. Twelve samples of Salvia Miltiorrhiza Bunge were quantitatively analyzed by qNMR and HPLC respectively. Then, the results were analyzed by chemometric approaches. This NMR method offers a fast, stable, and accurate analysis of four ketones: tanshinone I, tanshinone IIA, dihydrotanshinone, and cryptotanshinone in fresh roots of Salvia Miltiorrhiza Bunge.
Collapse
|
34
|
Fu L, Han B, Zhou Y, Ren J, Cao W, Patel G, Kai G, Zhang J. The Anticancer Properties of Tanshinones and the Pharmacological Effects of Their Active Ingredients. Front Pharmacol 2020; 11:193. [PMID: 32265690 PMCID: PMC7098175 DOI: 10.3389/fphar.2020.00193] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/11/2020] [Indexed: 12/31/2022] Open
Abstract
Cancer is a common malignant disease worldwide with an increasing mortality in recent years. Salvia miltiorrhiza, a well-known traditional Chinese medicine, has been used for the treatment of cardiovascular and cerebrovascular diseases for thousands of years. The liposoluble tanshinones in S. miltiorrhiza are important bioactive components and mainly include tanshinone IIA, dihydrodanshinone, tanshinone I, and cryptotanshinone. Previous studies showed that these four tanshinones exhibited distinct inhibitory effects on tumor cells through different molecular mechanisms in vitro and in vivo. The mechanisms mainly include the inhibition of tumor cell growth, metastasis, invasion, and angiogenesis, apoptosis induction, cell autophagy, and antitumor immunity, and so on. In this review, we describe the latest progress on the antitumor functions and mechanisms of these four tanshinones to provide a deeper understanding of the efficacy. In addition, the important role of tumor immunology is also reviewed.
Collapse
Affiliation(s)
- Li Fu
- School of Life Sciences, Institute of Plant Biotechnology, Shanghai Normal University, Shanghai, China
| | - Bing Han
- Laboratory of Medicinal Plant Biotechnology, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yang Zhou
- School of Life Sciences, Institute of Plant Biotechnology, Shanghai Normal University, Shanghai, China
| | - Jie Ren
- School of Life Sciences, Institute of Plant Biotechnology, Shanghai Normal University, Shanghai, China
| | - Wenzhi Cao
- School of Life Sciences, Institute of Plant Biotechnology, Shanghai Normal University, Shanghai, China
| | - Gopal Patel
- Laboratory of Medicinal Plant Biotechnology, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guoyin Kai
- School of Life Sciences, Institute of Plant Biotechnology, Shanghai Normal University, Shanghai, China.,Laboratory of Medicinal Plant Biotechnology, College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Zhang
- School of Life Sciences, Institute of Plant Biotechnology, Shanghai Normal University, Shanghai, China
| |
Collapse
|
35
|
Li J, He J, Zhang X, Li J, Zhao P, Fei P. TSP1 ameliorates age-related macular degeneration by regulating the STAT3-iNOS signaling pathway. Exp Cell Res 2020; 388:111811. [PMID: 31899207 DOI: 10.1016/j.yexcr.2019.111811] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/25/2019] [Accepted: 12/28/2019] [Indexed: 12/11/2022]
Abstract
Age-related macular degeneration is a progressive ocular disease that is the leading cause of vision loss among elderly. AMD usually is divided into two types: wet and dry AMD, which is linked with inflammation. Choroidal Neovascularization (CNV) formation or wet AMD is also associated with oxidative stress. Previously, TSP1 has been shown to have a significant alleviating effect on CNV in TSP1 knockout (TSP1-/-) mice. However, the mechanism by which TSP1 ameliorates CNV remains unclear. Here we report that TSP1 reduces nitric oxide production to prevent cells from forming tubes formation and reduced the levels of vascular endothelial growth factor (VEGF) and lipid peroxides (LPO) during oxidative stress. We measured RF/6A cell viability by CCK-8 assay and apoptosis by flow cytometry. RF/6A cell were transfected with TSP1 and STAT3 overexpression, and then the mRNA and protein levels of TSP1 and also the signal pathways were detected by qRT-PCR and Western blot analysis. Migration assays were performed using a transwell system. Co-Immunoprecipitation was used to analyze the binding relationship between CD47 and SHP-2. The results show that overexpression of TSP1 alleviated the damage of oxidative stress to RF/6A cells including increased cell activity and migration, decreased apoptosis and reduced migration compared to the control group. SHP-2 was activated by TSP1 through its receptor CD47 and STAT3 phosphorylation was reduced by activation of SHP-2, thereby blocking STAT3-iNOS pathway and reducing NO concentration in RF/6A cells ultimately protecting them from oxidative stress. Finally, the CNV mice model confirmed that TSP1 overexpression could protect the mice against CNV in vivo, modified the antioxidants levels and decreased the expression of TNF-α and IL-6 under laser irradiation. These results indicate a potential mechanism of TSP1 to slow down formation of CNV in wet AMD, which may bring hope for new treatment strategies.
Collapse
Affiliation(s)
- Jing Li
- Department of Ophthalmology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jiaqi He
- Department of General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, 201104, China
| | - Xiang Zhang
- Department of Ophthalmology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jiakai Li
- Department of Ophthalmology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| | - Ping Fei
- Department of Ophthalmology, Xinhua Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
36
|
Wang H, Liu Z, Guan L, Li J, Chen S, Yu W, Lai M. LYW-6, a novel cryptotanshinone derived STAT3 targeting inhibitor, suppresses colorectal cancer growth and metastasis. Pharmacol Res 2020; 153:104661. [PMID: 31982491 DOI: 10.1016/j.phrs.2020.104661] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/15/2019] [Accepted: 01/22/2020] [Indexed: 01/02/2023]
Abstract
The constitutive activation of signal transducer and activator of transcription 3(STAT3) is associated with aggressive development and metastasis in colorectal cancer (CRC), but STAT3-targeting drugs remain elusive in clinic. Here, structure-based strategy was used to remodel the natural compound cryptotanshinone into a more effective STAT3 inhibitor LYW-6. Using the Biolayer Interferometry assay, we observed that LYW-6 exhibited specific interactions with STAT3(KD = 6.6 ± 0.7 μM). Western blot analysis and electrophoretic mobility shift assays (EMSA) showed that LYW-6 inhibited the phosphorylation of STAT3 tyrosine 705 (Tyr-705) and had slight effects on STAT1 and STAT5 phosphorylation. Western blot analysis on the upstream kinases of STAT3 confirmed that the inhibitory mechanism on p-STAT3 was independent of upstream kinases. Further investigation demonstrated that LYW-6 downregulated the expression of downstream oncogenes to inhibit cell viability, cell cycle development, and potently increased cell apoptosis in human CRC cells. The invasion and metastasis linked signaling was also blocked by LYW-6 treatment. LYW-6 was found to reduce the metastasis foci in lung on tail-lung metastasis models. In addition, it was observed that LYW-6 markedly diminished STAT3 phosphorylation in tumor tissue and significantly inhibited tumor growth on xenograft models. Tumor development on chemically-induced colorectal cancer model also significantly inhibited by LYW-6 treatment. These findings provided adequate evidence that STAT3 inhibitor LYW-6 might be a potential candidate agent for CRC treatment.
Collapse
Affiliation(s)
- Huan Wang
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, China
| | - Zhe Liu
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, China
| | - Lingnan Guan
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, China
| | - Jiankang Li
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, China
| | - Siyi Chen
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, China
| | - Wenying Yu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, China.
| | - Maode Lai
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, China; Department of Pathology, School of Medicine, Zhejiang University, China.
| |
Collapse
|
37
|
Li W, Du H, Zhou G, Song D. Euxanthone represses the proliferation, migration, and invasion of glioblastoma cells by modulating STAT3/SHP-1 signaling. Anat Rec (Hoboken) 2020:ar.24363. [PMID: 31922313 DOI: 10.1002/ar.24363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/22/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
Glioblastoma is one of the most prevalent primary malignant brain tumors. Glioblastoma often develops resistance to conventional chemoradiotherapy, and thus, new ways to treat glioblastoma are urgently required. The aim of this study was to investigate the effect of euxanthone on the anticancer activities of glioblastoma and its potential mechanism. The U87 and U251 glioblastoma cell lines were cultured in media containing different concentrations of euxanthone. CCK-8 and colony formation assay were used to evaluate the cell proliferation. Cell migration and invasion were evaluated by wound healing and Transwell assays. Flow cytometry was used to assess the cell cycle and apoptosis rate. TUNEL assay was also employed to evaluate the apoptosis rate. Gene and protein expressions were determined by RT-qPCR and western blotting, respectively. A xenograft model was established to evaluate the efficacy of euxanthone in vivo. Euxanthone significantly repressed cell viability, migration, invasion, and epithelial-to-mesenchymal transition of U87 and U251 cells; and increased the rate of apoptosis. Western blotting results revealed that the levels of p21, p27, cleaved caspase-3, Bax, TIMP-3, and E-cadherin were upregulated while, the levels of CDK4, CDK6, pro-caspase-3, Bcl-2, MMP-2, MMP-9, N-cadherin, and Vimentin were downregulated by euxanthone. In addition, the expression of p-STAT3 was decreased, while the expression of SHP-1 was upregulated by euxanthone. We proposed that euxanthone could repress the malignant behavior of glioblastoma cells through suppression of STAT3 phosphorylation and activation of SHP-1. Further, in vivo data demonstrated that euxanthone repressed tumor growth and promoted apoptosis.
Collapse
Affiliation(s)
- Wen Li
- Jining First People's Hospital, Jining, China
| | - Hongmei Du
- Jining First People's Hospital, Jining, China
| | | | - Daqing Song
- Jining First People's Hospital, Jining, China
| |
Collapse
|
38
|
Wang H, Zhang Y, Zhang Y, Liu W, Wang J. Cryptotanshinone inhibits lung cancer invasion via microRNA-133a/matrix metalloproteinase 14 regulation. Oncol Lett 2019; 18:2554-2559. [PMID: 31452742 DOI: 10.3892/ol.2019.10580] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 04/04/2019] [Indexed: 12/24/2022] Open
Abstract
Cryptotanshinone inhibits the proliferative and colony formation abilities of human non-small cell lung cancer cells (NSCLCs). The present study reported that signal transducer and activator of transcription 3 is not the only target of cryptotanshinone during the inhibition of human NSCLCs. It was identified that cryptotanshinone upregulates the expression levels of microRNA (miR)-30d-5p, miR-126-3p, miR-133a, miR-338-3p and miR-451a, and downregulates miR-21-5p, miR-96-5p, miR-182-5p and miR-205-5p. Among these, miR-133a was the most significantly upregulated. miR-133a targets and downregulates the expression of matrix metalloproteinase (MMP)14; however, MMP15, MMP16 and MMP24 were determined to be unaffected. This process was identified to be independent of tissue inhibitor of metalloproteinases 2. Cryptotanshinone also suppresses the invasion of human NSCLCs, which may be due to the inhibited expression of MMP14. In conclusion, cryptotanshinone may serve as a potential therapeutic agent in the treatment of lung cancer.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Tumor Chemotherapy, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Yanshan Zhang
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Yingguo Zhang
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Wenling Liu
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Jihong Wang
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| |
Collapse
|
39
|
Cryptotanshinone protects against pulmonary fibrosis through inhibiting Smad and STAT3 signaling pathways. Pharmacol Res 2019; 147:104307. [PMID: 31181334 DOI: 10.1016/j.phrs.2019.104307] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/26/2019] [Accepted: 06/06/2019] [Indexed: 01/11/2023]
Abstract
Cryptotanshinone (CTS), a lipophilic compound extracted from root of Salvia miltiorrhiza (Danshen), has demonstrated multiple pharmacological activities, including anti-inflammation, anti-proliferation and anti-infection. However, the effect of CTS on pulmonary fibrosis is unknown. This study aims to investigate the effects of CTS treatment on pulmonary fibrosis and its underlying mechanism. The pulmonary fibrosis model was established by intratracheal instillation of bleomycin (5 mg/kg) in Sprague-Dawley rats (in vivo) and stimulating human fetal lung fibroblasts (HLFs) with transforming growth factor-beta 1 (TGF-β1) (in vitro). CTS (7.5, 15, 30, 60 mg/kg/day) and pirfenidone (150 mg/kg/day, positive control) were administered by oral gavage for 28 days. In this study, we found CTS treatment improved pulmonary function, relieved pathological changes and attenuated the accumulation of extracellular matrix in pulmonary fibrosis rat model induced by bleomycin. Mechanistically, CTS suppressed phosphorylation of Smad2/3 and STAT3 induced by TGF-β1 in HLFs. Stattic, a 1-benzothiophene based small-molecule STAT3 inhibitor, resulted in a significant down-regulation of fibrosis biomarkers including fibronectin, collagen type I and alpha smooth muscle actin (α-SMA). Overexpression of STAT3 promoted expression of fibrosis biomarkers in HLFs cell model induced by TGF-β1 and partially blocked the inhibitory effect of CTS on TGF-β1-induced fibrosis response. Taken together, these results suggested that CTS protects against pulmonary fibrosis via inhibition of Smad and STAT3 signaling pathways. Thus, CTS may represent a promising drug candidate for treating pulmonary fibrosis.
Collapse
|
40
|
Cryptotanshinone ameliorates the pathogenesis of systemic lupus erythematosus by blocking T cell proliferation. Int Immunopharmacol 2019; 74:105677. [PMID: 31177018 DOI: 10.1016/j.intimp.2019.105677] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 11/20/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic, devastating autoimmune disorder associated with severe organ damage. Recently, the role of Signal Transducer and Activator of Transcription 3 (STAT3) in murine lupus has been described, suggesting the involvement of STAT3 signaling in the development of SLE. Cryptotanshinone (CTS) is an effective inhibitor of STAT3; however its potential as a SLE treatment remains to be explored. To determine the function of CTS in SLE, we treated MRL/lpr female mice with CTS. Firstly, we found CTS treatment reversed the elevated STAT3 signaling of spleens in lupus-prone MRL/lpr mice, accompanying with a dramatically decreased number of T cells, especially double-negative (DN) T cells. Further research showed that CTS inhibited T cell proliferation via suppressing of STAT3 activation in vitro and in vivo. Consistently, we also proved that CTS treatment significantly alleviated autoimmune response including notably diminished skin lesions, reduced spleen size and increased life span. In addition, CTS treatment decreased the levels of auto-antibodies and pro-inflammatory cytokines, as well as normalized structure and function of kidneys. All these data suggested that CTS treatment depressed STAT3 phosphorylation, which resulted in blocked DN T cell proliferation and finally attenuated the spontaneous SLE development. Taken together, our data identify CTS as a potential therapeutic drug for SLE patients.
Collapse
|
41
|
Cryptotanshinone Ameliorates Radiation-Induced Lung Injury in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1908416. [PMID: 30915142 PMCID: PMC6402207 DOI: 10.1155/2019/1908416] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/21/2018] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
Abstract
Cryptotanshinone (CTS) was reported to repress a variety of systemic inflammation and alleviate cardiac fibrosis, but it is still unclear whether CTS could prevent radiation-induced lung injury (RILI). Here, we investigated the effects and underlying mechanisms of CTS on a RILI rat model. Our data revealed that CTS could efficiently preserve pulmonary function in RILI rats and reduce early pulmonary inflammation infiltration elicited, along with marked decreased levels of IL-6 and IL-10. Moreover, we found that CTS is superior to prednisone in attenuating collagen deposition and pulmonary fibrosis, in parallel with a marked drop of HYP (a collagen indicator) and α-SMA (a myofibroblast marker). Mechanistically, CTS inhibited profibrotic signals TGF-β1 and NOX-4 expressions, while enhancing the levels of antifibrotic enzyme MMP-1 in lung tissues. It is noteworthy that CTS treatment, in consistent with trichrome staining analysis, exhibited a clear advantage over PND in enhancing MMP-1 levels. However, CTS exhibited little effect on CTGF activation and on COX-2 suppression. Finally, CTS treatment significantly mitigated the radiation-induced activation of CCL3 and its receptor CCR1. In summary, CTS treatment could attenuate RILI, especially pulmonary fibrosis, in rats. The regulation on production and release of inflammatory or fibrotic factors IL-6, IL-10, TGF-β1, NOX-4, and MMP-1, especially MMP-1 and inhibition on CCL3/CCR1 activation, may partly attribute to its attenuating RILI effect.
Collapse
|
42
|
Protein Phosphatases-A Touchy Enemy in the Battle Against Glioblastomas: A Review. Cancers (Basel) 2019; 11:cancers11020241. [PMID: 30791455 PMCID: PMC6406705 DOI: 10.3390/cancers11020241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor arising from brain parenchyma. Although many efforts have been made to develop therapies for GBM, the prognosis still remains poor, mainly because of the difficulty in total resection of the tumor mass from brain tissue and the resistance of the residual tumor against standard chemoradiotherapy. Therefore, novel adjuvant therapies are urgently needed. Recent genome-wide analyses of GBM cases have clarified molecular signaling mechanisms underlying GBM biology. However, results of clinical trials targeting phosphorylation-mediated signaling have been unsatisfactory to date. Protein phosphatases are enzymes that antagonize phosphorylation signaling by dephosphorylating phosphorylated signaling molecules. Recently, the critical roles of phosphatases in the regulation of oncogenic signaling in malignant tumor cells have been reported, and tumorigenic roles of deregulated phosphatases have been demonstrated in GBM. However, a detailed mechanism underlying phosphatase-mediated signaling transduction in the regulation of GBM has not been elucidated, and such information is necessary to apply phosphatases as a therapeutic target for GBM. This review highlights and summarizes the phosphatases that have crucial roles in the regulation of oncogenic signaling in GBM cells.
Collapse
|
43
|
Hu D, Zhang Y. Circular RNA HIPK3 promotes glioma progression by binding to miR-124-3p. Gene 2018; 690:81-89. [PMID: 30576808 DOI: 10.1016/j.gene.2018.11.073] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/13/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023]
Abstract
This study aims to investigate whether circ-HIPK3 could promote the proliferation and invasion of glioma cells by upregulating STAT3 after binding to miR-124-3p, thus participating in the development of glioma. Expression levels of circ-HIPK3, miR-124-3p and STAT3 in glioma cell lines were determined using qRT-PCR. The regulatory effects of circ-HIPK3, miR-124-3p and STAT3 on proliferative and invasive capacities of glioma cells were accessed using EdU assay, CCK-8 assay and invasion assay, respectively. Cell cycle assay and cell apoptosis assay were performed by flow cytometry. Dual-luciferase reporter gene assay was conducted to determine the binding condition among circ-HIPK3, miR-124-3p and STAT3. Rescue experiments were performed in co-transfected glioma cells. QRT-PCR data showed that circ-HIPK3 and STAT3 are highly expressed, whereas miR-124-3p is lowly expressed in glioma cells than those of negative control cell. Knockdown of circ-HIPK3 in U87 and U251 cells inhibited their proliferative and invasive capacities. On the contrary, miR-124-3p knockdown improved proliferative and migratory capacities. Dual-luciferase reporter gene assay exerted that circ-HIPK3 could bind to miR-124-3p and STAT3 is the target gene of miR-124-3p. Western blot results elucidated that circ-HIPK3 stabilizes STAT3 expression, whereas miR-124-3p degrades STAT3 expression. Rescue experiments demonstrated that overexpression of circ-HIPK3 could partially reverse the inhibited proliferative and migratory capacities induced by miR-124-3p in U87 and U251 cells. In summary, we found that overexpression of circ-HIPK3 promotes proliferative and invasive capacities of glioma cells by sponging miR-124-3p to upregulate STAT3 expression.
Collapse
Affiliation(s)
- Daling Hu
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, China
| | - Yin Zhang
- Department of Neurosurgery, Sir Run Run Hospital, Nanjing Medical University, China.
| |
Collapse
|
44
|
Yang Y, Cao Y, Chen L, Liu F, Qi Z, Cheng X, Wang Z. Cryptotanshinone suppresses cell proliferation and glucose metabolism via STAT3/SIRT3 signaling pathway in ovarian cancer cells. Cancer Med 2018; 7:4610-4618. [PMID: 30094960 PMCID: PMC6143944 DOI: 10.1002/cam4.1691] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/20/2018] [Accepted: 06/26/2018] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is the most malignant gynecologic cancer among women worldwide. Cryptotanshinone (CT), isolated from Salvia miltiorrhiza Bunge, has been identified as a potential therapeutic agent in treating several malignant tumors, but the molecular mechanism of CT in ovarian cancer still remains illustrated. Here, we sought to elucidate the regulatory function of CT on cell glucose metabolism in ovarian cancer. The treatment of CT on ovarian cancer cells effectively inhibited glucose uptake and lactate production in ovarian cancer cells. The expression levels of glycolysis-related proteins, such as GLUT1, LDHA, and HK2, were decreased by the treatment of CT detected by qRT-PCR and immunoblotting. Mechanistically, CT exerted its anti-tumor effect by targeting STAT3/SIRT3/HIF-1α signaling pathway in vitro and in vivo, which could be rescued by the introduction of SIRT3 shRNA in ovarian cancer cells. The clinical data showed that the expression level of STAT3 in ovarian cancer patients' sera and tissues was positively correlated with those of GLUT1, LDHA, HK2 and HIF-1α, but negatively with that of SIRT3These findings provide evidence that CT inhibited cellular glycolysis-induced cell growth and proliferation through repression of STAT3/SIRT3/HIF-1α signaling pathway, indicating that CT may be developed as a chemotherapeutic agent to treat ovarian cancer.
Collapse
Affiliation(s)
- Yufei Yang
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Department of Gynecological OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Yue Cao
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
| | - Lihua Chen
- Department of Gynecological OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Fei Liu
- Department of Gynecological OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Zihao Qi
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
| | - Xi Cheng
- Department of Gynecological OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Ziliang Wang
- Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
45
|
Saraf RS, Datta A, Sima C, Hua J, Lopes R, Bittner M. An in-silico study examining the induction of apoptosis by Cryptotanshinone in metastatic melanoma cell lines. BMC Cancer 2018; 18:855. [PMID: 30157799 PMCID: PMC6116360 DOI: 10.1186/s12885-018-4756-0] [Citation(s) in RCA: 12] [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: 04/03/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Metastatic melanoma is an aggressive form of skin cancer that evades various anti-cancer treatments including surgery, radio-,immuno- and chemo-therapy. TRAIL-induced apoptosis is a desirable method to treat melanoma since, unlike other treatments, it does not harm non-cancerous cells. The pro-inflammatory response to melanoma by nF κB and STAT3 pathways makes the cancer cells resist TRAIL-induced apoptosis. We show that due to to its dual action on DR5, a death receptor for TRAIL and on STAT3, Cryptotanshinone can be used to increase sensitivity to TRAIL. METHODS The development of chemoresistance and invasive properties in melanoma cells involves several biological pathways. The key components of these pathways are represented as a Boolean network with multiple inputs and multiple outputs. RESULTS The possible mutations in genes that can lead to cancer are captured by faults in the combinatorial circuit and the model is used to theoretically predict the effectiveness of Cryptotanshinone for inducing apoptosis in melanoma cell lines. This prediction is experimentally validated by showing that Cryptotanshinone can cause enhanced cell death in A375 melanoma cells. CONCLUSION The results presented in this paper facilitate a better understanding of melanoma drug resistance. Furthermore, this framework can be used to detect additional drug intervention points in the pathway that could amplify the action of Cryptotanshinone.
Collapse
Affiliation(s)
- Radhika S. Saraf
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Aniruddha Datta
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Chao Sima
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
| | - Jianping Hua
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
| | - Rosana Lopes
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
| | - Michael Bittner
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
- Translational Genomics Research Institute (TGen), Phoenix, US
| |
Collapse
|
46
|
Ediriweera MK, Tennekoon KH, Samarakoon SR. In vitro assays and techniques utilized in anticancer drug discovery. J Appl Toxicol 2018; 39:38-71. [DOI: 10.1002/jat.3658] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Meran Keshawa Ediriweera
- Institute of Biochemistry, Molecular Biology and Biotechnology; University of Colombo; Colombo 03 Sri Lanka
| | - Kamani Hemamala Tennekoon
- Institute of Biochemistry, Molecular Biology and Biotechnology; University of Colombo; Colombo 03 Sri Lanka
| | | |
Collapse
|
47
|
Ji Q, Qi D, Xu X, Xu Y, Goodman SB, Kang L, Song Q, Fan Z, Maloney WJ, Wang Y. Cryptotanshinone Protects Cartilage against Developing Osteoarthritis through the miR-106a-5p/GLIS3 Axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:170-179. [PMID: 29858052 PMCID: PMC5992348 DOI: 10.1016/j.omtn.2018.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 02/06/2023]
Abstract
Cryptotanshinone (CTS) has emerged as an anti-inflammatory agent in osteoarthritis (OA). However, the molecular mechanism underlying its potent therapeutic effect on OA remains largely unknown. MicroRNAs (miRNAs) act as crucial regulators in maintaining cartilage homeostasis. To investigate whether CTS protects against developing OA through regulation of miRNAs, we examined the potential CTS-mediated miRNA molecules using microarray analysis. We found that CTS significantly promoted miR-106a-5p expression in chondrocytes. Using the OA mouse model created by anterior cruciate ligament transection, we revealed that intra-articular injection of miR-106a-5p agomir attenuated OA. In addition, miR-106a-5p inhibited GLI-similar 3 (GLIS3) production by directly targeting the 3′ untranslated region. CTS promoted miR-106a-5p expression through recruitment of a member of the paired box (PAX) family of transcription factors, PAX5, to the miR-106a-5p promoter. Inhibition of PAX5 mimicked the effect of miR-106a-5p and abolished the CTS ability to regulate miR-106a-5p expression. In OA patients, miR-106-5p is downregulated which is accompanied by downregulation of PAX5 and upregulation of GLIS3. Collectively, these data highlight that the PAX5/miR-106a-5p/GLIS3 axis acts as a novel pleiotropic regulator in CTS-mediated OA cartilage protection, suggesting that miR-106a-5p and PAX5 activation and GLIS3 inhibition might be useful and attractive for therapeutic strategies to treat OA patients.
Collapse
Affiliation(s)
- Quanbo Ji
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, China; Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Dengbin Qi
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yameng Xu
- Department of Traditional Chinese Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Qi Song
- Department of Oncology, General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Zhongyi Fan
- Department of Oncology, General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - William J Maloney
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Yan Wang
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, China.
| |
Collapse
|
48
|
Amlexanox, a selective inhibitor of IKBKE, generates anti-tumoral effects by disrupting the Hippo pathway in human glioblastoma cell lines. Cell Death Dis 2017; 8:e3022. [PMID: 29048430 PMCID: PMC5596579 DOI: 10.1038/cddis.2017.396] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/25/2017] [Accepted: 07/02/2017] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent form of malignant brain tumor. Amlexanox, a novel compound, has been shown to have anti-cancer potential. In this study, the anti-tumoral effects and the underlying mechanisms of amlexanox were investigated. Amlexanox significantly suppressed proliferation and invasion and induced apoptosis in glioblastoma cells. Furthermore, we found that amlexanox altered the protein expression of the Hippo pathway by downregulating IKBKE. Our data indicates that IKBKE directly targets LATS1/2 and induces degradation of LATS1/2, thereby inhibiting the activity of the Hippo pathway. In vivo results further confirmed the tumor inhibitory effect of amlexanox via the downregulation of IKBKE, and amlexanox induced no apparent toxicity. Collectively, our studies suggest that amlexanox is a promising therapeutic agent for the treatment of GBM.
Collapse
|