1
|
Zhang Z, Wang Q, Zhang H, Wang S, Ma X, Wang H. Golm1 facilitates the CaO2-DOPC-DSPE200-PEI -CsPbBr3 QDs -induced apoptotic death of hepatocytes through the stimulation of mitochondrial autophagy and mitochondrial reactive oxygen species production through interactions with P53/Beclin-1/Bcl-2. Chem Biol Interact 2024; 398:111076. [PMID: 38815669 DOI: 10.1016/j.cbi.2024.111076] [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: 01/15/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Mitophagy is a distinct physiological process that can have beneficial or deleterious effects in particular tissues. Prior research suggests that mitophagic activity can be triggered by CaO2-PM-CsPbBr3 QDs, yet the specific role that mitophagy plays in hepatic injury induced by CaO2-PM-CsPbBr3 QDs has yet to be established. Accordingly, in this study a series of mouse model- and cell-based experiments were performed that revealed the ability of CaO2-PM-CsPbBr3 QDs to activate mitophagic activity. Golm1 was upregulated in response to CaO2-PM-CsPbBr3 QDs treatment, and overexpressing Golm1 induced autophagic flux in the murine liver and hepatocytes, whereas knocking down Golm1 had the opposite effect. CaO2-PM-CsPbBr3 QDs were also able to Golm1 expression, in turn promoting the degradation of P53 and decreasing the half-life of this protein. Overexpressing Golm1 was sufficient to suppress the apoptotic death of hepatocytes in vitro and in vivo, whereas the knockdown of Golm1 had the opposite effect. The ability of Golm1 to promote p53-mediated autophagy was found to be associated with the disruption of Beclin-1 binding to Bcl-2, and the Golm1 N-terminal domain was determined to be required for p53 interactions, inducing autophagic activity in a manner independent of helicase activity or RNA binding. Together, these results indicate that inhibiting Golm1 can promote p53-dependent autophagy via disrupting Beclin-1 binding to Bcl-2, highlighting a novel approach to mitigating liver injury induced by CaO2-PM-CsPbBr3 QDs.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China.
| | - Qinglong Wang
- College of Animal Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, Henan Province, China
| | - Haibo Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China
| | - Shengchao Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China
| | - Xia Ma
- College of Animal Medicine, Henan University of Animal Husbandry and Economy, Zhengzhou, 450046, Henan Province, China
| | - Hui Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450045, Henan Province, China.
| |
Collapse
|
2
|
Guan H, Xie L, Ji Z, Song R, Qi J, Nie X. Triptolide inhibits neutrophil extracellular trap formation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1384. [PMID: 34733936 PMCID: PMC8506553 DOI: 10.21037/atm-21-3522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/06/2021] [Indexed: 01/02/2023]
Abstract
Background Triptolide (PG490), as a triterpene dicyclic oxide has been reported to increase the generation of reactive oxygen species (ROS) and nitric oxide (NO) and induce apoptosis of RAW 264.7 cells in a dose-dependent manner. The activity of death NETs plays an important role in anti-bacterial processes in the human body. This study aimed to investigate the effect of triptolide (PG490) on neutrophil extracellular traps (NETs) formation. Methods After isolating peripheral blood neutrophils from healthy volunteers, cells were incubated with PG490 to observe and detect the level of NETs and detect the level of reactive oxygen species (ROS). The cells were cultured, stained and analyzed by fluorescence microscopy. Results Compared with the 12-myristate-13-acetate (PMA) group, the average fluorescence intensity of SYTOX Green in the PG490 + PMA group, as detected by a multifunctional microplate reader, was significantly decreased. Intracellular ROS were labeled by fluorescence, with fluorescence intensity then measured by multifunctional microplate reader and flow cytometry. The results showed that compared with the control group, the fluorescence intensity of the PMA group was significantly increased, while there was no significant difference between PMA group and PG490 + PMA group. Conclusions The production of NETs is inhibited by PG490 in vitro, which is not associated with the level of cellular ROS. This suggests that PG490in Tripterygium wilfordii Hook F can suppress related diseases.
Collapse
Affiliation(s)
- Haiyu Guan
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine and Western Medicine, Southern Medical University, Guangzhou, China
| | - Lifen Xie
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine and Western Medicine, Southern Medical University, Guangzhou, China
| | - Zhenzhen Ji
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine and Western Medicine, Southern Medical University, Guangzhou, China
| | - Rui Song
- Department of Rheumatology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jieying Qi
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine and Western Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoli Nie
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine and Western Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Tang B, Zhu J, Zhang B, Wu F, Wang Y, Weng Q, Fang S, Zheng L, Yang Y, Qiu R, Chen M, Xu M, Zhao Z, Ji J. Therapeutic Potential of Triptolide as an Anti-Inflammatory Agent in Dextran Sulfate Sodium-Induced Murine Experimental Colitis. Front Immunol 2020; 11:592084. [PMID: 33240279 PMCID: PMC7680904 DOI: 10.3389/fimmu.2020.592084] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/12/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn’s disease (CD), is a group of chronic and incurable inflammatory diseases involving the gastrointestinal tract. In this study, we investigated the anti-inflammatory effects of triptolide in a dextran sulfate sodium (DSS)-induced mouse colitis model and LPS-activated macrophages and explored the specific molecular mechanism(s). In mice, triptolide treatment showed significant relief and protection against colitis, and it markedly reduced the inflammatory responses of human monocytes and mouse macrophages. Pharmacological analysis and weighted gene co-expression network analysis (WGCNA) suggested that PDE4B may be an important potential targeting molecule for IBD. Exploration of the specific mechanism of action indicated that triptolide reduced the production of ROS, inhibited macrophage infiltration and M1-type polarization by activating the NRF2/HO-1 signaling pathway, and inhibited the PDE4B/AKT/NF-κB signaling cascade, which may help weaken the intestinal inflammatory response. Our findings laid a theoretical foundation for triptolide as a treatment for IBD and revealed PDE4B as a target molecule, thus providing new ideas for the treatment of IBD.
Collapse
Affiliation(s)
- Bufu Tang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinyu Zhu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Baohui Zhang
- Department of Physiology, School of Life Science, China Medical University, Shenyang, China
| | - Fazong Wu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Yajie Wang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Qiaoyou Weng
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Shiji Fang
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Liyun Zheng
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Yang Yang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Rongfang Qiu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Min Xu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China.,Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| |
Collapse
|
4
|
Tang Y, Liu Q, Feng Y, Zhang Y, Xu Z, Wen C, Zhang Y. Tripterygium Ingredients for Pathogenicity Cells in Rheumatoid Arthritis. Front Pharmacol 2020; 11:583171. [PMID: 33123015 PMCID: PMC7567162 DOI: 10.3389/fphar.2020.583171] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease mainly characterized by chronic polyarthritis. Many types of cells play pivotal roles in the pathogenicity of RA, such as T cells, B cells, macrophages, dendritic cells (DCs), osteoclasts (OCs), and fibroblast-like synoviocytes (FLS). Tripterygium wilfordii Hook f. (TwHf) and its ingredients are able to control disease activity by regulating the functions of cells mentioned above, and the clinical studies have highlighted the importance of TwHf ingredients in RA treatment. They have been demonstrated to improve the RA symptoms of animal models and patients. In this review, we discussed the effect of TwHf ingredients on pathogenicity cells, including disease/cell phenotypes and molecular mechanisms. Here, we constructed a cell-cell interaction network to visualize the effect of TwHf ingredients. We found that TwHf ingredients could inhibit the differentiation and proliferation of the pathogenicity cells. Besides, the components could decrease the levels of pathogenicity cytokines [i.e., interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α)]. Many signaling pathways are involved in the underlying mechanisms, such as PI3K, NF-κB, and MAPK signaling pathways.
Collapse
Affiliation(s)
- Yujun Tang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiuping Liu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuxiang Feng
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Zhang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhenghao Xu
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengping Wen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yun Zhang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
5
|
Tang Y, Zhang Y, Li L, Xie Z, Wen C, Huang L. Kunxian Capsule for Rheumatoid Arthritis: Inhibition of Inflammatory Network and Reducing Adverse Reactions Through Drug Matching. Front Pharmacol 2020; 11:485. [PMID: 32362827 PMCID: PMC7181472 DOI: 10.3389/fphar.2020.00485] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Tripterygium wilfordii Hook.f and Tripterygium hypoglaucum (H.Lév.) Hutch is effective herbs to prevent aggravation of Rheumatoid arthritis (RA). However, both of them show severe side effects in the reproductive system and other systems. Kunxian Capsule (KX), a Traditional Chinese Medicine (TCM) patent prescription, comprised of 4 herbs, including H.Lév. Hutch, is reported to be an available prescription in treating RA with fewer side effects as compares to Tripterygium tablets. To reveal the pharmacological mechanism of KX in RA treatment and side effect alleviation, we collected related information of KX from open-access databases and performed various analyses. 1354 targets were identified in KX. These targets were enriched in the calcium signaling pathway, cAMP signaling pathway, cGMP-PKG signaling pathway and PI3K-AKT signaling pathway, forming biological functions, such as cofactor binding, coenzyme binding, etc. These pathways or functions mostly affect cell cycle, differentiation, and maturation of Th17 cells, macrophage, and synovial fibroblast. These targets also act on the IL-17 signaling pathway, Th17 cell differentiation signaling pathway and TNF signaling pathway, which is related to inflammation response inhibition. Next, a disease network was constructed, which indicated IMPDH2, MTHFD1 are the key genes answering for the side effects of H.Lév. Hutch. The side effect–related genes lead to the negative regulation of nucleic acid, which could be restored by the rest 3 herbs through some positive amino acid metabolism. In conclusion, KX is a relatively safe alternative approach in RA intervention.
Collapse
Affiliation(s)
- Yujun Tang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Zhang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Li
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Xie
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengping Wen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Huang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Yuan K, Li X, Lu Q, Zhu Q, Jiang H, Wang T, Huang G, Xu A. Application and Mechanisms of Triptolide in the Treatment of Inflammatory Diseases-A Review. Front Pharmacol 2019; 10:1469. [PMID: 31866868 PMCID: PMC6908995 DOI: 10.3389/fphar.2019.01469] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/13/2019] [Indexed: 12/19/2022] Open
Abstract
Bioactive compounds from medicinal plants with anti-inflammatory and immunosuppressive effects have been emerging as important sources of drugs for the treatment of inflammatory disorders. Triptolide, a diterpene triepoxide, is a pharmacologically active compound isolated from Tripterygium wilfordii Hook F (TwHF) that is used as a remedy for inflammatory and autoimmune diseases. As the most promising bioactive compound obtained from TwHF, triptolide has attracted considerable interest recently, especially for its potent anti-inflammatory and immunosuppressive activities. Over the past few years, an increasing number of studies have been published emphasizing the value of triptolide in the treatment of diverse inflammatory disorders. Here, we systematically review the mechanism of action and the therapeutic properties of triptolide in various inflammatory diseases according to different systematic organs, including lupus nephritis, inflammatory bowel disease, asthma, and rheumatoid arthritis with pubmed and Embase. Based on this review, potential research strategies might contribute to the clinical application of triptolide in the future.
Collapse
Affiliation(s)
- Kai Yuan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qingyi Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Qingqing Zhu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Haixu Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Anlong Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,State Key Laboratory of Biocontrol, Department of Biochemistry, School of Life Sciences, Sun Yat-Sen (Zhongshan) University, Guangzhou, China
| |
Collapse
|
7
|
Zhang X, Xiao Z, Xu H. A review of the total syntheses of triptolide. Beilstein J Org Chem 2019; 15:1984-1995. [PMID: 31501665 PMCID: PMC6720243 DOI: 10.3762/bjoc.15.194] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 08/03/2019] [Indexed: 12/12/2022] Open
Abstract
Triptolide is a complex triepoxide diterpene natural product that has attracted considerable interest in the organic chemistry and medicinal chemistry societies due to its intriguing structural features and multiple promising biological activities. In this review, progress in the total syntheses of triptolide are systematically summarized. We hope to gain a better understanding of the field and provide constructive suggestions for future studies of triptolide.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zaozao Xiao
- College of Chemical Engineering and Materials Science, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, 201210, China
| |
Collapse
|
8
|
Hou W, Liu B, Xu H. Triptolide: Medicinal chemistry, chemical biology and clinical progress. Eur J Med Chem 2019; 176:378-392. [DOI: 10.1016/j.ejmech.2019.05.032] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/11/2019] [Accepted: 05/11/2019] [Indexed: 12/14/2022]
|
9
|
Wiegand MJ, Kubacki GW, Gilbert JL. Electrochemical potential zone of viability on CoCrMo surfaces is affected by cell type: Macrophages under cathodic bias are more resistant to killing. J Biomed Mater Res A 2018; 107:526-534. [DOI: 10.1002/jbm.a.36567] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/20/2018] [Accepted: 09/28/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Michael J. Wiegand
- Department of Biomedical and Chemical Engineering; Syracuse University; Syracuse New York
- Syracuse Biomaterials Institute, Syracuse University; Syracuse New York
| | - Gregory W. Kubacki
- Department of Biomedical and Chemical Engineering; Syracuse University; Syracuse New York
- Syracuse Biomaterials Institute, Syracuse University; Syracuse New York
| | - Jeremy L. Gilbert
- Department of Biomedical and Chemical Engineering; Syracuse University; Syracuse New York
- Syracuse Biomaterials Institute, Syracuse University; Syracuse New York
- Department of Bioengineering; Clemson University; Clemson South Carolina
- Clemson University-Medical University of South Carolina Program in Bioengineering; Charleston South Carolina
| |
Collapse
|
10
|
Li JL, Yang N, Huang L, Chen D, Zhao Y, Tang MM, Fan H, Bao X. Pyocyanin Inhibits Chlamydia Infection by Disabling Infectivity of the Elementary Body and Disrupting Intracellular Growth. Antimicrob Agents Chemother 2018; 62:e02260-17. [PMID: 29610203 PMCID: PMC5971585 DOI: 10.1128/aac.02260-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/19/2018] [Indexed: 11/20/2022] Open
Abstract
The obligate intracellular bacterium Chlamydia is a widespread human pathogen that causes serious problems, including (but not limited to) infertility and blindness. Our search for novel antichlamydial metabolites from marine-derived microorganisms led to the isolation of pyocyanin, a small compound from Pseudomonas aeruginosa Pyocyanin is an effective antichlamydial for all three Chlamydia spp. tested. It has a 50% inhibitory concentration (IC50) of 0.019 to 0.028 μM, which is comparable to the IC50 of tetracycline. At concentrations as low as 0.0039 μM, pyocyanin disables infectivity of the chlamydial elementary body (EB). At 0.5 μM or higher concentrations, the continuous presence of pyocyanin also inhibits chlamydial growth in the inclusion during later stages of the developmental cycle. Oxidative stress, a major known antimicrobial mechanism of pyocyanin, appears to be responsible only for the inhibition of bacterial growth and not for the disinfection of EBs. Pyocyanin is well-tolerated by probiotic vaginal Lactobacillus spp. Our findings suggest that pyocyanin is of therapeutic value for chlamydial infections and can serve as a valuable chemical probe for studying chlamydial biology.
Collapse
Affiliation(s)
- Jian Lin Li
- School of Pharmacy, Nantong University, Nantong, China
| | - Ningjing Yang
- School of Pharmacy, Nantong University, Nantong, China
| | - Lei Huang
- School of Pharmacy, Nantong University, Nantong, China
- Department of Pharmacy, The First People's Hospital of Yanchen, Yanchen, China
| | - Dandan Chen
- Department of Pharmacy, The Second People's Hospital of Nantong, Nantong, China
| | - Yu Zhao
- School of Pharmacy, Nantong University, Nantong, China
| | - M Matt Tang
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Huizhou Fan
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, China
| |
Collapse
|
11
|
Fan D, Guo Q, Shen J, Zheng K, Lu C, Zhang G, Lu A, He X. The Effect of Triptolide in Rheumatoid Arthritis: From Basic Research towards Clinical Translation. Int J Mol Sci 2018; 19:ijms19020376. [PMID: 29373547 PMCID: PMC5855598 DOI: 10.3390/ijms19020376] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
Triptolide (TP), a major extract of the herb Tripterygium wilfordii Hook F (TWHF), has been shown to exert potent pharmacological effects, especially an immunosuppressive effect in the treatment of rheumatoid arthritis (RA). However, its multiorgan toxicity prevents it from being widely used in clinical practice. Recently, several attempts are being performed to reduce TP toxicity. In this review, recent progress in the use of TP for RA, including its pharmacological effects and toxicity, is summarized. Meanwhile, strategies relying on chemical structural modifications, innovative delivery systems, and drug combinations to alleviate the disadvantages of TP are also reviewed. Furthermore, we also discuss the challenges and perspectives in their clinical translation.
Collapse
Affiliation(s)
- Danping Fan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Qingqing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| | - Jiawen Shen
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| | - Kang Zheng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| |
Collapse
|
12
|
Functional role of PPAR-γ on the proliferation and migration of fibroblast-like synoviocytes in rheumatoid arthritis. Sci Rep 2017; 7:12671. [PMID: 28978936 PMCID: PMC5627284 DOI: 10.1038/s41598-017-12570-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/05/2017] [Indexed: 11/21/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR)-γ is involved in both normal physiological processes and pathology of various diseases. The purpose of this study was to explore the function and underlying mechanisms of PPAR-γ in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLSs) proliferation and migration. In the present study, we found PPAR-γ expression was remarkably reduced in RA synovium patient compare with OA and normal, as well as it was low-expression in Adjuvant-induced arthritis (AA). Moreover, inhibition PPAR-γ expression by T0070907 (12.5 μM) or PPAR-γ siRNA could promote FLSs proliferation and expressions of c-Myc, Cyclin D1, MMP-1, and MMP-9 in AA FLSs, except for TIPM-1. These date indicate that up-regulation of PPAR-γ may play a critical role in RA FLSs. Interestingly, co-incubation FLSs with Pioditazone (25 μM) and over expression vector with pEGFP-N1-PPAR-γ reduced proliferation and expressions of c-Myc, Cyclin D1, MMP-1, and MMP-9 in AA FLSs, besides TIMP-1. Further study indicates that PPAR-γ may induce activation Wnt/β-catenin signaling. In short, these results indicate that PPAR-γ may play a pivotal role during FLSs activation and activation of Wnt/β-catenin signaling pathway.
Collapse
|
13
|
Bei Y, Chen J, Zhou F, Huang Y, Jiang N, Tan R, Shen P. BZ-26, a novel GW9662 derivate, attenuated inflammation by inhibiting the differentiation and activation of inflammatory macrophages. Biomed Pharmacother 2016; 84:730-739. [PMID: 27710897 DOI: 10.1016/j.biopha.2016.08.069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/14/2016] [Accepted: 08/28/2016] [Indexed: 12/14/2022] Open
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARγ) is considered to be an important transcriptional factor in regulation of macrophages differentiation and activation. We have synthesized a series of novel structural molecules based on GW9662's structure (named BZ-24, BZ-25 and BZ-26), and interaction activity was calculated by computational docking. BZ-26 had shown stronger interaction with PPARγ and had higher transcriptional inhibitory activity of PPARγ with lower dosage compared with GW9662. BZ-26 was proved to inhibit inflammatory macrophage differentiation. LPS-induced acute inflammation mouse model was applied to demonstrate its anti-inflammatory activity. And the results showed that BZ-26 administration attenuated plasma tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) secretion, which are vital cytokines in acute inflammation. The anti-inflammatory activity was examined in THP-1 cell line, and TNF-α, IL-6 and MCP-1, were significantly inhibited. The results of Western blot and luciferase reporter assay indicated that BZ-26 not only inhibited NF-κB transcriptional activity, but also abolished LPS-induce nuclear translocation of P65. We also test BZ-26 action in tumor-bearing chronic inflammation mouse model, and BZ-26 was able to alter macrophages phenotype, resulting in antitumor effect. All our data revealed that BZ-26 modulated LPS-induced acute inflammation via inhibiting inflammatory macrophages differentiation and activation, potentially via inhibition of NF-κB signal pathway.
Collapse
Affiliation(s)
- Yuncheng Bei
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210093, China
| | - Jiajia Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210093, China
| | - Feifei Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210093, China
| | - Yahong Huang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210093, China
| | - Nan Jiang
- Institute of Functional Biomolecules, Medical School, Nanjing University, Nanjing, 210093, China
| | - Renxiang Tan
- Institute of Functional Biomolecules, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, 210093, China.
| |
Collapse
|
14
|
Feng X, Weng D, Zhou F, Owen YD, Qin H, Zhao J, WenYu, Huang Y, Chen J, Fu H, Yang N, Chen D, Li J, Tan R, Shen P. Activation of PPARγ by a Natural Flavonoid Modulator, Apigenin Ameliorates Obesity-Related Inflammation Via Regulation of Macrophage Polarization. EBioMedicine 2016; 9:61-76. [PMID: 27374313 PMCID: PMC4972579 DOI: 10.1016/j.ebiom.2016.06.017] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/10/2016] [Accepted: 06/13/2016] [Indexed: 01/04/2023] Open
Abstract
PPARγ has emerged as a master regulator of macrophage polarization and is the molecular target of the thiazolidinedione drugs. Here we show that apigenin binds and activates PPARγ by acting as a modulator. Activation of PPARγ by apigenin blocks p65 translocation into nuclei through inhibition of p65/PPARγ complex translocation into nuclei, thereby decreasing NF-κB activation and favoringM2 macrophage polarization. In HFD and ob/ob mice, apigenin significantly reverses M1 macrophage into M2 and reduces the infiltration of inflammatory cells in liver and adipose tissues, as well as decreases the levels of pro-inflammatory cytokines, thereby alleviating inflammation. Strikingly, apigenin reduces liver and muscular steatosis, decreases the levels of ALT, AST, TC and TG, improving glucose resistance obviously. Unlike rosiglitazone, apigenin does not cause significant weight gain, osteoporosis et al. Our findings identify apigenin as a modulator of PPARγ and a potential lead compound for treatment of metabolic disorders. Apigenin binds and activates PPARγ and significantly reverses the polarization of macrophages from M1 to M2 phenotype. Activation of PPARγ by apigenin blocks p65 translocation through inhibiting p65/PPARγ complex translocation into nucleus. Apigenin significantly attenuates metabolic inflammation and disorders without causing some side effects as TZD drugs do.
PPARγ is the molecular target of the thiazolidinedione drugs to treat type II diabetes. However, TZD drugs have some side effects including cardiovascular failure, liver toxicity, bone fractures and potential carcinogenesis, which have greatly limited their clinical use. Here, we find apigenin, a flavonoid molecule abundant in various fruits and vegetables, can control macrophage fate to inhibit inflammation and metabolic syndrome without causing some side effects as TZD drugs. Further study indicates that apigenin can target PPARγ with a range of beneficial effects and may represent a lead compound for developing new therapies against metabolic disorders.
Collapse
Affiliation(s)
- Xiujing Feng
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Dan Weng
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Feifei Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Young D Owen
- Graduate Medical Education, Virginia Mason Medical Center, Seattle, WA 98101, USA
| | - Haohan Qin
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Jingfa Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - WenYu
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Yahong Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Jiajia Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Haijian Fu
- Key Lab of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210046, China
| | - Nanfei Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Dianhua Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Jianxin Li
- Key Lab of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210046, China
| | - Renxiang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology, School of life Sciences, Nanjing University, Nanjing 210046, China; MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210046, China.
| |
Collapse
|
15
|
Huang C, Yang Y, Li WX, Wu XQ, Li XF, Ma TT, Zhang L, Meng XM, Li J. Hyperin attenuates inflammation by activating PPAR-γ in mice with acute liver injury (ALI) and LPS-induced RAW264.7 cells. Int Immunopharmacol 2015; 29:440-447. [DOI: 10.1016/j.intimp.2015.10.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/14/2015] [Accepted: 10/15/2015] [Indexed: 10/22/2022]
|
16
|
Feng X, Qin H, Shi Q, Zhang Y, Zhou F, Wu H, Ding S, Niu Z, Lu Y, Shen P. Chrysin attenuates inflammation by regulating M1/M2 status via activating PPARγ. Biochem Pharmacol 2014; 89:503-14. [PMID: 24704474 DOI: 10.1016/j.bcp.2014.03.016] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 03/26/2014] [Accepted: 03/26/2014] [Indexed: 12/21/2022]
Abstract
Chrysin (5,7-di-OH-flavone), a widely distributed natural flavonoid, has been well documented for involving in various biological activities, especially in regulation of peroxisome proliferator activated receptor γ (PPARγ) activity as a modest modulator. However, the exact molecular mechanism is still unrevealed. In the current study, for the first time, we discovered that, chrysin not only significantly attenuated inflammation in high-fat feeding mice, but also alleviated high fat diet-induced hepatic, muscular steatosis in obese mice without altering the body weight. Chrysin decreases the infiltration of macrophages into adipose tissue in obese mice. In addition, chrysin was also found to induce an anti-inflammatory M2 phenotype and decreases M1 phenotype, both in peritoneal macrophages of obese mice and cultured macrophages in vitro, and thereby, chrysin changed the M1/M2 status. Our data further showed that chrysin regulated the phenotype of macrophages through enhancing the transcriptional activation of PPARγ and the expression of its target genes. Taken together, we conclude that chrysin may serve as an effective modulator of PPARγ during the pathogenesis of inflammation, thereby our findings shed light on the potential therapeutic feature of chrysin in recovering inflammatory diseases via regulating M1/M2 status.
Collapse
MESH Headings
- Adipose Tissue/drug effects
- Adipose Tissue/immunology
- Adipose Tissue/metabolism
- Adipose Tissue/pathology
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/adverse effects
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Cell Division/drug effects
- Cell Line, Transformed
- Cell Survival/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Fatty Liver/drug therapy
- Fatty Liver/immunology
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Flavonoids/administration & dosage
- Flavonoids/adverse effects
- Flavonoids/pharmacology
- Flavonoids/therapeutic use
- Humans
- Liver/drug effects
- Liver/immunology
- Liver/metabolism
- Liver/pathology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myositis/drug therapy
- Myositis/immunology
- Myositis/metabolism
- Myositis/pathology
- Non-alcoholic Fatty Liver Disease
- Obesity/physiopathology
- PPAR gamma/agonists
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Random Allocation
- Specific Pathogen-Free Organisms
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
- Xiujing Feng
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China
| | - Haohan Qin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China
| | - Qian Shi
- Department of Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), TX 78229, USA
| | - Yang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China
| | - Feifei Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China
| | - Haochen Wu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China
| | - Sen Ding
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China
| | - Zhiyuan Niu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China
| | - Yan Lu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China.
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
17
|
Lu Y, Zhang Y, Li L, Feng X, Ding S, Zheng W, Li J, Shen P. TAB1: A Target of Triptolide in Macrophages. ACTA ACUST UNITED AC 2014; 21:246-56. [DOI: 10.1016/j.chembiol.2013.12.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/21/2013] [Accepted: 12/02/2013] [Indexed: 12/22/2022]
|
18
|
Zhong YY, Chen HP, Tan BZ, Yu HH, Huang XS. Triptolide avoids cisplatin resistance and induces apoptosis via the reactive oxygen species/nuclear factor-κB pathway in SKOV3 PT platinum-resistant human ovarian cancer cells. Oncol Lett 2013; 6:1084-1092. [PMID: 24137468 PMCID: PMC3796418 DOI: 10.3892/ol.2013.1524] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 07/10/2013] [Indexed: 01/15/2023] Open
Abstract
An acquired resistance to platinum-based drugs has emerged as a significant impediment to effective ovarian cancer therapy. The present study explored the anticancer mechanisms of triptolide (TPL) in SKOV3PT platinum-resistant human ovarian cancer cells and observed that TPL activated caspase 3 and induced the dose-dependent apoptosis of the SKOV3PT cells. Furthermore, TPL inhibited complex I of the mitochondrial respiratory chain (MRC) followed by an increase of reactive oxygen species (ROS), which further inhibited nuclear factor (NF)-κB activation and resulted in the downregulation of anti-apoptotic proteins, Bcl-2 and X-linked inhibitor of apoptosis protein (XIAP). Notably, the pre-treatment with N-acetyl-L-cysteine (NAC) abolished the TPL-induced ROS generation, NF-κB inhibition and cell apoptosis, but did not affect the inhibitory effect of TPL on complex I activity. These results suggested that TPL negatively regulated the NF-κB pathway through mitochondria-derived ROS accumulation, promoting the apoptosis of the SKOV3PT cells. Furthermore, TPL synergistically enhanced the cytotoxicity of cisplatin against platinum-resistant ovarian cancer cells. Collectively, these findings suggest that TPL is able to overcome chemoresistance and that it may be an effective treatment for platinum-resistant ovarian cancer, either alone or as an adjuvant therapy.
Collapse
Affiliation(s)
- Yan-Ying Zhong
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang, Jiangxi 330006, P.R. China ; Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | | | | | | | | |
Collapse
|
19
|
TAN BEEJEN, CHIU GIGIN. Role of oxidative stress, endoplasmic reticulum stress and ERK activation in triptolide-induced apoptosis. Int J Oncol 2013; 42:1605-12. [DOI: 10.3892/ijo.2013.1843] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 12/24/2012] [Indexed: 11/06/2022] Open
|
20
|
Yang W, Lu Y, Xu Y, Xu L, Zheng W, Wu Y, Li L, Shen P. Estrogen represses hepatocellular carcinoma (HCC) growth via inhibiting alternative activation of tumor-associated macrophages (TAMs). J Biol Chem 2012; 287:40140-9. [PMID: 22908233 DOI: 10.1074/jbc.m112.348763] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hepatocarcinoma cancer (HCC) occurs more often in men than in women, and little is known about its underlying molecular mechanisms. RESULTS We identify that 17β-estradiol (E2) could suppress tumor growth via regulating the polarization of macrophages. CONCLUSION Estrogen functions as a suppressor for macrophage alternative activation. SIGNIFICANCE These studies introduce a novel mechanism for suppressing male-predominant HCC. Hepatocarcinoma cancer (HCC), one of the most malignant cancers, occurs significantly more often in men than in women; however, little is known about its underlying molecular mechanisms. Here we identified that 17β-estradiol (E2) could suppress tumor growth via regulating the polarization of macrophages. We showed that E2 re-administration reduced tumor growth in orthotopic and ectopic mice HCC models. E2 functioned as a suppressor for macrophage alternative activation and tumor progression by keeping estrogen receptor β (ERβ) away from interacting with ATP5J (also known as ATPase-coupling factor 6), a part of ATPase, thus inhibiting the JAK1-STAT6 signaling pathway. These studies introduce a novel mechanism for suppressing male-predominant HCC.
Collapse
Affiliation(s)
- Weiwei Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center, Nanjing University, Nanjing 210093, China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Xu H, Wei Y, Zhang Y, Xu Y, Li F, Liu J, Zhang W, Han X, Tan R, Shen P. Oestrogen attenuates tumour progression in hepatocellular carcinoma. J Pathol 2012; 228:216-29. [PMID: 22374713 DOI: 10.1002/path.4009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 01/19/2012] [Accepted: 02/15/2012] [Indexed: 01/08/2023]
Abstract
The precise mechanisms underlying gender disparity in hepatocellular carcinoma (HCC) progression and prognosis are not understood. We demonstrate that oestrogen attenuates HCC progression in vitro and in vivo, and this may contribute to the gender differences in HCC behaviour. To investigate the role of oestrogen in HCC progression, we developed an orthotopic homograft tumour model by liver implantation of H22 cells. In combination with male castration, female ovariectomy, and oestrogen treatment, we tested the hypothesis that oestrogen contributes to gender disparity in this model. Pathological analyses were performed to examine the changes in biological behaviour of liver cancer cells, and two cell lines were used to investigate possible molecular mechanisms of the suppressive effect of oestrogen. Our data showed that oestrogen modulates HCC malignancy in vivo by reducing tumour cell invasion, arresting cell cycle progression, and promoting apoptosis, characterized by decreased expression of MMP-2, MMP-9, PCNA, cyclin A, cyclin D1, and Bcl-2, and increased expression in cleaved caspase 3. Through in vitro assays, we further confirmed the changes in expression levels of these related proteins, gained insights into the molecular cascades of oestrogen-induced HCC suppression, and indicated the oestrogen receptor α-mediated inhibition of NF-κB binding activity as a pivotal event in this process. This study represents a novel description of the mechanisms regarding the suppressive effects of oestrogen on HCC, adding a new understanding to the gender disparity in HCC progression. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Hanwen Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Model Animal Research Center, Nanjing University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang Y, Lu JJ, He L, Yu Q. Triptolide (TPL) inhibits global transcription by inducing proteasome-dependent degradation of RNA polymerase II (Pol II). PLoS One 2011; 6:e23993. [PMID: 21931633 PMCID: PMC3172214 DOI: 10.1371/journal.pone.0023993] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 08/03/2011] [Indexed: 11/18/2022] Open
Abstract
Triptolide (TPL), a key biologically active component of the Chinese medicinal herb Tripterygium wilfordii Hook. f., has potent anti-inflammation and anti-cancer activities. Its anti-proliferative and pro-apoptotic effects have been reported to be related to the inhibition of Nuclear Factor κB (NF-κB) and Nuclear Factor of Activated T-cells (NFAT) mediated transcription and suppression of HSP70 expression. The direct targets and precise mechanisms that are responsible for the gene expression inhibition, however, remain unknown. Here, we report that TPL inhibits global gene transcription by inducing proteasome-dependent degradation of the largest subunit of RNA polymerase II (Rpb1) in cancer cells. In the presence of proteosome inhibitor MG132, TPL treatment causes hyperphosphorylation of Rpb1 by activation of upstream protein kinases such as Positive Transcription Elongation Factor b (P-TEFb) in a time and dose dependent manner. Also, we observe that short time incubation of TPL with cancer cells induces DNA damage. In conclusion, we propose a new mechanism of how TPL works in killing cancer. TPL inhibits global transcription in cancer cells by induction of phosphorylation and subsequent proteasome-dependent degradation of Rpb1 resulting in global gene transcription, which may explain the high potency of TPL in killing cancer.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jin-jian Lu
- Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Li He
- Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Qiang Yu
- Department of Pharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
23
|
Triptolide circumvents drug-resistant effect and enhances 5-fluorouracil antitumor effect on KB cells. Anticancer Drugs 2010; 21:502-13. [DOI: 10.1097/cad.0b013e328337337c] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Xu B, Guo X, Mathew S, Armesilla AL, Cassidy J, Darling JL, Wang W. Triptolide simultaneously induces reactive oxygen species, inhibits NF-κB activity and sensitizes 5-fluorouracil in colorectal cancer cell lines. Cancer Lett 2010; 291:200-8. [DOI: 10.1016/j.canlet.2009.10.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 10/14/2009] [Accepted: 10/15/2009] [Indexed: 11/30/2022]
|
25
|
Chen T, Lin X, Xu J, Tan R, Ji J, Shen P. Redox imbalance provokes deactivation of macrophages in sepsis. Proteomics Clin Appl 2009; 3:1000-9. [DOI: 10.1002/prca.200800016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 03/31/2009] [Accepted: 04/08/2009] [Indexed: 11/08/2022]
|
26
|
Shi X, Jin Y, Cheng C, Zhang H, Zou W, Zheng Q, Lu Z, Chen Q, Lai Y, Pan J. Triptolide Inhibits Bcr-Abl Transcription and Induces Apoptosis in STI571-resistant Chronic Myelogenous Leukemia Cells Harboring T315I Mutation. Clin Cancer Res 2009; 15:1686-97. [DOI: 10.1158/1078-0432.ccr-08-2141] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Nicolay JP, Liebig G, Niemoeller OM, Koka S, Ghashghaeinia M, Wieder T, Haendeler J, Busse R, Lang F. Inhibition of suicidal erythrocyte death by nitric oxide. Pflugers Arch 2007; 456:293-305. [PMID: 18058124 DOI: 10.1007/s00424-007-0393-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2007] [Accepted: 11/07/2007] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) is known to counteract apoptosis by S-nitrosylation of protein thiol groups. NO is generated and stored in erythrocytes, which may undergo eryptosis, a suicidal cell death similar to apoptosis of nucleated cells. Eryptosis is triggered by increased cytosolic Ca2+ activity and/or ceramide and characterized by cell shrinkage and phosphatidylserine exposure at the cell surface. The present study explored whether nitric oxide could interfere with the machinery underlying eryptosis. To this end, erythrocyte phosphatidylserine exposure (annexin V-binding) and cell volume (forward scatter) were determined by flow cytometry. The Ca2+ ionophore ionomycin (0.1 microM) increased cytosolic Ca2+ activity, triggered annexin binding, and decreased forward scatter. The annexin binding and decrease of forward scatter but not the increase of cytosolic Ca2+ activity were reversed by the NO-donor nitroprusside (1 microM) and papanonoate (100 microM). Higher concentrations of nitroprusside (0.1 and 1 mM) stimulated eryptosis. Glucose depletion, exposure to C6-ceramide (3 microM), hypertonic (addition of 550 mM sucrose), and isotonic (replacement of Cl- with gluconate) cell shrinkage all triggered annexin V binding, effects all reversed by nitroprusside (1 microM). Dibutyryl-cGMP (1 mM) blunted the ionomycin- but not the ceramide-induced annexin V binding. Ionomycin decreased protein nitrosylation and thioredoxin activity, effects reversed by the NO-donor papanonoate. Clearance of erythrocytes from circulating blood was significantly faster in eNOS knockout mice than in their wild-type littermates. In conclusion, nitric oxide participates in the regulation of erythrocyte survival, an effect partially mimicked by cGMP and paralleled by alterations of protein nitrosylation and thioredoxin activity.
Collapse
Affiliation(s)
- Jan P Nicolay
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|