1
|
Li M, Li J, Tang Q, Zhu Y. Potential antitumor activity of triptolide and its derivatives: Focused on gynecological and breast cancers. Biomed Pharmacother 2024; 180:117581. [PMID: 39427548 DOI: 10.1016/j.biopha.2024.117581] [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: 08/06/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer remains one of the greatest global health concerns. This is especially true for gynecological cancers, which include cervical, ovarian, and endometrial cancers, and breast cancer. Natural products used for cancer treatment offer some unique advantages. Triptolide (TPL) is a biologically active terpenoid extracted from Tripterygium wilfordii, which exhibits anti-inflammatory, immunosuppressive, antitumor, and other pharmacological activities. However, clinical applications of TPL are restricted because of poor water solubility and severe cytotoxicity; to overcome these limitations, various TPL derivatives and drug delivery systems, especially nanocarriers, have been used. Furthermore, various preclinical and clinical studies have demonstrated that TPL and its derivatives exhibit excellent antitumor effects by targeting proteins involved in multiple signaling pathways. Here, we review the progress regarding novel drug delivery systems, antitumor activities, and molecular mechanisms of action of TPL and its derivatives against gynecological and breast cancers. TPL and its derivatives inhibit tumor growth, suppress tumor metastasis, and enhance the drug sensitization of resistant cancers. In addition, TPL and its derivatives exert synergistic antitumor effects against gynecological and breast cancers when combined with existing antitumor drugs, such as carboplatin, cisplatin, and PI3K inhibitors. Moreover, we highlight the clinical potential of TPL analogs against cancer from bench to bedside and their prospects for future applications in gynecologic and breast cancers.
Collapse
Affiliation(s)
- Mengjie Li
- College of Pharmacy, Qinghai University for Nationalities, Xining, China; Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jiamiao Li
- Department of Pharmacy, The Affilliated Chengdu 363 Hospital of Southwest Medical University, Chengdu, China
| | - Qing Tang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yongxia Zhu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
2
|
Koksalar Alkan F, Caglayan AB, Alkan HK, Benson E, Gunduz YE, Sensoy O, Durdagi S, Zarbaliyev E, Dyson G, Assad H, Shull A, Chadli A, Shi H, Ozturk G, Korkaya H. Dual activity of Minnelide chemosensitize basal/triple negative breast cancer stem cells and reprograms immunosuppressive tumor microenvironment. Sci Rep 2024; 14:22487. [PMID: 39341857 PMCID: PMC11439009 DOI: 10.1038/s41598-024-72989-6] [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: 03/18/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
Triple negative breast cancer (TNBC) subtype is characterized with higher EMT/stemness properties and immune suppressive tumor microenvironment (TME). Women with advanced TNBC exhibit aggressive disease and have limited treatment options. Although immune suppressive TME is implicated in driving aggressive properties of basal/TNBC subtype and therapy resistance, effectively targeting it remains a challenge. Minnelide, a prodrug of triptolide currently being tested in clinical trials, has shown anti-tumorigenic activity in multiple malignancies via targeting super enhancers, Myc and anti-apoptotic pathways such as HSP70. Distinct super-enhancer landscape drives cancer stem cells (CSC) in TNBC subtype while inducing immune suppressive TME. We show that Minnelide selectively targets CSCs in human and murine TNBC cell lines compared to cell lines of luminal subtype by targeting Myc and HSP70. Minnelide in combination with cyclophosphamide significantly reduces the tumor growth and eliminates metastasis by reprogramming the tumor microenvironment and enhancing cytotoxic T cell infiltration in 4T1 tumor-bearing mice. Resection of residual tumors following the combination treatment leads to complete eradication of disseminated tumor cells as all mice are free of local and distant recurrences. All control mice showed recurrences within 3 weeks of post-resection while single Minnelide treatment delayed recurrence and one mouse was free of tumor. We provide evidence that Minnelide targets tumor intrinsic pathways and reprograms the immune suppressive microenvironment. Our studies also suggest that Minnelide in combination with cyclophosphamide may lead to durable responses in patients with basal/TNBC subtype warranting its clinical investigation.
Collapse
Affiliation(s)
- Fulya Koksalar Alkan
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, HWCRC 723 4100 John R. Street, Detroit, MI, 48201, USA
| | - Ahmet Burak Caglayan
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, HWCRC 723 4100 John R. Street, Detroit, MI, 48201, USA
| | - Hilmi Kaan Alkan
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, HWCRC 723 4100 John R. Street, Detroit, MI, 48201, USA
| | - Elayne Benson
- Georgia Cancer Center, Department of Biochemistry, Augusta University, Augusta, GA, USA
| | - Yunus Emre Gunduz
- Regenerative and Restorative Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Department of Physiology, International School of Medicine, Medipol University, Istanbul, Turkey
| | - Ozge Sensoy
- Regenerative and Restorative Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Department of Physiology, International School of Medicine, Medipol University, Istanbul, Turkey
| | - Serdar Durdagi
- Department of Pharmaceutical Chemistry, School of Pharmacy, Bahcesehir University, Istanbul, Turkey
| | - Elbrus Zarbaliyev
- Department of Surgery, Gaziosmanpasa Hospital Istanbul, Istanbul Yeni Yuzyil University, Istanbul, Turkey
| | - Greg Dyson
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, HWCRC 723 4100 John R. Street, Detroit, MI, 48201, USA
| | - Hadeel Assad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, HWCRC 723 4100 John R. Street, Detroit, MI, 48201, USA
| | - Austin Shull
- Department of Biology, Presbyterian College, Clinton, SC, USA
| | - Ahmed Chadli
- Georgia Cancer Center, Department of Biochemistry, Augusta University, Augusta, GA, USA
| | - Huidong Shi
- Georgia Cancer Center, Department of Biochemistry, Augusta University, Augusta, GA, USA
| | - Gurkan Ozturk
- Regenerative and Restorative Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Department of Physiology, International School of Medicine, Medipol University, Istanbul, Turkey
| | - Hasan Korkaya
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, HWCRC 723 4100 John R. Street, Detroit, MI, 48201, USA.
| |
Collapse
|
3
|
Xu B, Wang Z, Zhang H, Xu X, Tang M, Wang G, Ding Z, Yu R, Ding M, Zhang T, Shi S. The Cytoprotective Effect of C60 Derivatives in the Self-Microemulsifying Drug Delivery System against Triptolide-Induced Cytotoxicity In Vitro. Molecules 2024; 29:4073. [PMID: 39274920 PMCID: PMC11396586 DOI: 10.3390/molecules29174073] [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: 06/27/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
OBJECTIVE The aim of this study was to optimize the formulation of a C60-modified self-microemulsifying drug delivery system loaded with triptolide (C60-SMEDDS/TP) and evaluate the cytoprotective effect of the C60-SMEDDS/TP on normal human cells. RESULTS The C60-SMEDDS/TP exhibited rapid emulsification, an optimal particle size distribution of 50 ± 0.19 nm (PDI 0.211 ± 0.049), and a near-neutral zeta potential of -1.60 mV. The release kinetics of TP from the C60-SMEDDS/TP exhibited a sustained release profile and followed pseudo-first-order release kinetics. Cellular proliferation and apoptosis analysis indicated that the C60-SMEDDS/TP (with a mass ratio of TP: DSPE-PEG-C60 = 1:10) exhibited lower toxicity towards L02 and GES-1 cells. This was demonstrated by a higher IC50 (40.88 nM on L02 cells and 17.22 nM on GES-1 cells) compared to free TP (21.3 nM and 11.1 nM), and a lower apoptosis rate (20.8% on L02 cells and 26.3% on GES-1 cells, respectively) compared to free TP (50.5% and 47.0%) at a concentration of 50 nM. In comparison to the free TP group, L02 cells and GES-1 cells exposed to the C60-SMEDDS/TP exhibited a significant decrease in intracellular ROS and an increase in mitochondrial membrane potential (ΔψM). On the other hand, the C60-SMEDDS/TP demonstrated a similar inhibitory effect on BEL-7402 cells (IC50 = 28.9 nM) and HepG2 cells (IC50 = 107.6 nM), comparable to that of the free TP (27.2 nM and 90.4 nM). The C60-SMEDDS/TP group also exhibited a similar intracellular level of ROS and mitochondrial membrane potential compared to the SMEDDS/TP and free TP groups. METHOD Fullerenol-Grafted Distearoyl Phosphatidylethanolamine-Polyethylene Glycol (DSPE-PEG-C60) was synthesized and applied in the self-microemulsifying drug delivery system. The C60-SMEDDS/TP was formulated using Cremophor EL, medium-chain triglycerides (MCT), PEG-400, and DSPE-PEG-C60, and loaded with triptolide (TP). The toxicity and bioactivity of the C60-SMEDDS/TP were assessed using normal human liver cell lines (L02 cells), normal human gastric mucosal epithelial cell lines (GES-1 cells), and liver cancer cell lines (BEL-7402 cells and HepG2 cells). The production of reactive oxygen species (ROS) after the C60-SMEDDS/TP treatment was assessed using 2',7'-dichlorofluorescein diacetate (DCFDA) staining. The alterations in mitochondrial membrane potential (ΔψM) were assessed by measuring JC-1 fluorescence. CONCLUSIONS The cytoprotection provided by the C60-SMEDDS/TP favored normal cells (L02 and GES-1) over tumor cells (BEL-7402 and HepG2 cells) in vitro. This suggests a promising approach for the safe and effective treatment of TP.
Collapse
Affiliation(s)
- Beihua Xu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Zhenyu Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Huimin Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Xiao Xu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Mengjie Tang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Gang Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Zhongpeng Ding
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Ruihao Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Meihong Ding
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Ting Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Senlin Shi
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| |
Collapse
|
4
|
Afshari AR, Sanati M, Ahmadi SS, Kesharwani P, Sahebkar A. Harnessing the capacity of phytochemicals to enhance immune checkpoint inhibitor therapy of cancers: A focus on brain malignancies. Cancer Lett 2024; 593:216955. [PMID: 38750720 DOI: 10.1016/j.canlet.2024.216955] [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: 04/05/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/23/2024]
Abstract
Brain cancers, particularly glioblastoma multiforme (GBM), are challenging health issues with frequent unmet aspects. Today, discovering safe and effective therapeutic modalities for brain tumors is among the top research interests. Immunotherapy is an emerging area of investigation in cancer treatment. Since immune checkpoints play fundamental roles in repressing anti-cancer immunity, diverse immune checkpoint inhibitors (ICIs) have been developed, and some monoclonal antibodies have been approved clinically for particular cancers; nevertheless, there are significant concerns regarding their efficacy and safety in brain tumors. Among the various tools to modify the immune checkpoints, phytochemicals show good effectiveness and excellent safety, making them suitable candidates for developing better ICIs. Phytochemicals regulate multiple immunological checkpoint-related signaling pathways in cancer biology; however, their efficacy for clinical cancer immunotherapy remains to be established. Here, we discussed the involvement of immune checkpoints in cancer pathology and summarized recent advancements in applying phytochemicals in modulating immune checkpoints in brain tumors to highlight the state-of-the-art and give constructive prospects for future research.
Collapse
Affiliation(s)
- Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Feineis D, Bringmann G. Structural variety and pharmacological potential of naphthylisoquinoline alkaloids. THE ALKALOIDS. CHEMISTRY AND BIOLOGY 2024; 91:1-410. [PMID: 38811064 DOI: 10.1016/bs.alkal.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Naphthylisoquinoline alkaloids are a fascinating class of natural biaryl compounds. They show characteristic mono- and dimeric scaffolds, with chiral axes and stereogenic centers. Since the appearance of the last comprehensive overview on these secondary plant metabolites in this series in 1995, the number of discovered representatives has tremendously increased to more than 280 examples known today. Many novel-type compounds have meanwhile been discovered, among them naphthylisoquinoline-related follow-up products like e.g., the first seco-type (i.e., ring-opened) and ring-contracted analogues. As highlighted in this review, the knowledge on the broad structural chemodiversity of naphthylisoquinoline alkaloids has been decisively driven forward by extensive phytochemical studies on the metabolite pattern of Ancistrocladus abbreviatus from Coastal West Africa, which is a particularly "creative" plant. These investigations furnished a considerable number of more than 80-mostly new-natural products from this single species, with promising antiplasmodial activities and with pronounced cytotoxic effects against human leukemia, pancreatic, cervical, and breast cancer cells. Another unique feature of naphthylisoquinoline alkaloids is their unprecedented biosynthetic origin from polyketidic precursors and not, as usual for isoquinoline alkaloids, from aromatic amino acids-a striking example of biosynthetic convergence in nature. Furthermore, remarkable botanical results are presented on the natural producers of naphthylisoquinoline alkaloids, the paleotropical Dioncophyllaceae and Ancistrocladaceae lianas, including first investigations on the chemoecological role of these plant metabolites and their storage and accumulation in particular plant organs.
Collapse
Affiliation(s)
- Doris Feineis
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
6
|
Li LB, Yang LX, Liu L, Liu FR, Li AH, Zhu YL, Wen H, Xue X, Tian ZX, Sun H, Li PC, Zhao XG. Targeted inhibition of the HNF1A/SHH axis by triptolide overcomes paclitaxel resistance in non-small cell lung cancer. Acta Pharmacol Sin 2024; 45:1060-1076. [PMID: 38228910 PMCID: PMC11053095 DOI: 10.1038/s41401-023-01219-y] [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: 07/17/2023] [Accepted: 12/17/2023] [Indexed: 01/18/2024] Open
Abstract
Paclitaxel resistance is associated with a poor prognosis in non-small cell lung cancer (NSCLC) patients, and currently, there is no promising drug for paclitaxel resistance. In this study, we investigated the molecular mechanisms underlying the chemoresistance in human NSCLC-derived cell lines. We constructed paclitaxel-resistant NSCLC cell lines (A549/PR and H460/PR) by long-term exposure to paclitaxel. We found that triptolide, a diterpenoid epoxide isolated from the Chinese medicinal herb Tripterygium wilfordii Hook F, effectively enhanced the sensitivity of paclitaxel-resistant cells to paclitaxel by reducing ABCB1 expression in vivo and in vitro. Through high-throughput sequencing, we identified the SHH-initiated Hedgehog signaling pathway playing an important role in this process. We demonstrated that triptolide directly bound to HNF1A, one of the transcription factors of SHH, and inhibited HNF1A/SHH expression, ensuing in attenuation of Hedgehog signaling. In NSCLC tumor tissue microarrays and cancer network databases, we found a positive correlation between HNF1A and SHH expression. Our results illuminate a novel molecular mechanism through which triptolide targets and inhibits HNF1A, thereby impeding the activation of the Hedgehog signaling pathway and reducing the expression of ABCB1. This study suggests the potential clinical application of triptolide and provides promising prospects in targeting the HNF1A/SHH pathway as a therapeutic strategy for NSCLC patients with paclitaxel resistance. Schematic diagram showing that triptolide overcomes paclitaxel resistance by mediating inhibition of the HNF1A/SHH/ABCB1 axis.
Collapse
Affiliation(s)
- Ling-Bing Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Ling-Xiao Yang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Lei Liu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Fan-Rong Liu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Alex H Li
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10010, USA
| | - Yi-Lin Zhu
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Hao Wen
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Xia Xue
- Department of Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Zhong-Xian Tian
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China
| | - Hong Sun
- Division of Environmental Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10010, USA
| | - Pei-Chao Li
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
| | - Xiao-Gang Zhao
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
- Key Laboratory of Chest Cancer, The Second Hospital, Cheeloo College of Medicine, Shandong University, Ji-nan, 250012, China.
| |
Collapse
|
7
|
Chen P, Zhong X, Song Y, Zhong W, Wang S, Wang J, Huang P, Niu Y, Yang W, Ding Z, Luo Q, Yang C, Wang J, Zhang W. Triptolide induces apoptosis and cytoprotective autophagy by ROS accumulation via directly targeting peroxiredoxin 2 in gastric cancer cells. Cancer Lett 2024; 587:216622. [PMID: 38246224 DOI: 10.1016/j.canlet.2024.216622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/08/2023] [Accepted: 12/23/2023] [Indexed: 01/23/2024]
Abstract
Triptolide, a natural bioactive compound derived from herbal medicine Tripterygium wilfordii, has multiple biological activities including anti-cancer effect, which is being tested in clinical trials for treating cancers. However, the exact mechanism by which Triptolide exerts its cytotoxic effects, particularly its specific protein targets, remains unclear. Here, we show that Triptolide effectively induces cytotoxicity in gastric cancer cells by increasing reactive oxygen species (ROS) levels. Further investigations reveal that ROS accumulation contributes to the induction of Endoplasmic Reticulum (ER) stress, and subsequently autophagy induction in response to Triptolide. Meanwhile, this autophagy is cytoprotective. Interestingly, through activity-based protein profiling (ABPP) approach, we identify peroxiredoxins-2 (PRDX2), a component of the key enzyme systems that act in the defense against oxidative stress and protect cells against hydroperoxides, as direct binding target of Triptolide. By covalently binding to PRDX2 to inhibit its antioxidant activity, Triptolide increases ROS levels. Moreover, overexpression of PRDX2 inhibits and knockdown of the expression of PRDX2 increases Triptolide-induced apoptosis. Collectively, these results indicate PRDX2 as a direct target of Triptolides for inducing apoptosis. Our results not only provide novel insight into the underlying mechanisms of Triptolide-induced cytotoxic effects, but also indicate PRDX2 as a promising potential therapeutic target for developing anti-gastric cancer agents.
Collapse
Affiliation(s)
- Pengchen Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China; Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, 523125, Guangdong, China
| | - Xiaoru Zhong
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Yali Song
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, 523125, Guangdong, China
| | - Wenbin Zhong
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, 523125, Guangdong, China
| | - Sisi Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Jinyan Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Pan Huang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Yaping Niu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Wenyue Yang
- Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
| | - Ziyang Ding
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China
| | - Qingming Luo
- Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, 523125, Guangdong, China.
| | - Chuanbin Yang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China.
| | - Jigang Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China; Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan, 523125, Guangdong, China; Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Wei Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital; First Affiliated Hospital of Southern University of Science and Technology; Second Clinical Medical College of Jinan University, Shenzhen, 518020, China.
| |
Collapse
|
8
|
Korkaya H, Koksalar Alkan F, Caglayan A, Alkan H, Benson E, Gunduz Y, Sensoy O, Durdagi S, Zarbaliyev E, Dyson G, Assad H, Shull A, Chadli A, Shi H, Ozturk G. Dual activity of Minnelide chemosensitize basal/triple negative breast cancer stem cells and reprograms immunosuppressive tumor microenvironment. RESEARCH SQUARE 2024:rs.3.rs-3959342. [PMID: 38464167 PMCID: PMC10925405 DOI: 10.21203/rs.3.rs-3959342/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Triple negative breast cancer (TNBC) subtype is characterized with higher EMT/stemness properties and immune suppressive tumor microenvironment (TME). Women with advanced TNBC exhibit aggressive disease and have limited treatment options. Although immune suppressive TME is implicated in driving aggressive properties of basal/TNBC subtype and therapy resistance, effectively targeting it remains a challenge. Minnelide, a prodrug of triptolide currently being tested in clinical trials, has shown anti-tumorigenic activity in multiple malignancies via targeting super enhancers, Myc and anti-apoptotic pathways such as HSP70. Distinct super-enhancer landscape drives cancer stem cells (CSC) in TNBC subtype while inducing immune suppressive TME. We show that Minnelide selectively targets CSCs in human and murine TNBC cell lines compared to cell lines of luminal subtype by targeting Myc and HSP70. Minnelide in combination with cyclophosphamide significantly reduces the tumor growth and eliminates metastasis by reprogramming the tumor microenvironment and enhancing cytotoxic T cell infiltration in 4T1 tumor-bearing mice. Resection of residual tumors following the combination treatment leads to complete eradication of disseminated tumor cells as all mice are free of local and distant recurrences. All control mice showed recurrences within 3 weeks of post-resection while single Minnelide treatment delayed recurrence and one mouse was free of tumor. We provide evidence that Minnelide targets tumor intrinsic pathways and reprograms the immune suppressive microenvironment. Our studies also suggest that Minnelide in combination with cyclophosphamide may lead to durable responses in patients with basal/TNBC subtype warranting its clinical investigation.
Collapse
|
9
|
Jiang S, Feng J, Jiang Y, Lu Z, Kong J, Li X, Lian H, Zhang F, Li Y, Li J. Triptolide attenuates CCL 4-induced liver fibrosis by regulating the differentiation of CD 4+ T cells in mice. Int Immunopharmacol 2023; 125:111206. [PMID: 37956491 DOI: 10.1016/j.intimp.2023.111206] [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: 09/12/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023]
Abstract
Liver fibrosis is a major global health issue, and immune dysregulation is a main contributor. Triptolide is a natural immunosuppressive agent with demonstrated effectiveness in ameliorating liver fibrosis, but whether it exerts anti-liver fibrotic effects via immunoregulation remains obscure. In this study, first, by employing a CCL4-induced liver fibrosis mouse model, we demonstrated that triptolide could alleviate pathological damage to liver tissue and attenuate liver function damaged by CCL4. In addition, triptolide inhibited the expression of liver fibrotic markers such as hydroxyproline, collagen type IV, hyaluronidase, laminin, and procollagen type III, and the protein expression of α-SMA in CCL4-induced liver fibrosis. Second, with the help of network pharmacology, we predicted that triptolide's anti-liver fibrotic effects might occur through the regulation of Th17, Th1, and Th2 cell differentiation, which indicated that triptolide might mitigate liver fibrosis via immunoregulation. Finally, multiplex immunoassays and flow cytometry were adopted to verify this prediction. The results suggested that triptolide could reverse the aberrant expression of inflammatory cytokines caused by CCL4 and regulate the differentiation of Th1, Th2, Th17, and Treg cells. In conclusion, triptolide could attenuate CCL4-induced liver fibrosis by regulating the differentiation of CD4+ T cells. The results obtained in this study extended the application of triptolide and introduced a new mechanism of triptolide's anti-liver fibrotic effects.
Collapse
Affiliation(s)
- Shiyuan Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jing Feng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanling Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihao Lu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingwei Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xueming Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hui Lian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fang Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
10
|
Wang S, Guo Q, Xu R, Lin P, Deng G, Xia X. Combination of ferroptosis and pyroptosis dual induction by triptolide nano-MOFs for immunotherapy of Melanoma. J Nanobiotechnology 2023; 21:383. [PMID: 37858186 PMCID: PMC10585872 DOI: 10.1186/s12951-023-02146-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023] Open
Abstract
Immunotherapy has good potential to eradicate tumors in the long term. However, due to the low immunogenicity of tumor cells, current cancer immunotherapies are not effective. To address this limitation, we constructed a BSA-FA functionalized iron-containing metal-organic framework (TPL@TFBF) that triggers a potent systemic anti-tumor immune response by inducing ferroptosis and pyroptosis in tumor cells and releasing large quantities of damage-associated molecular patterns (DAMPs) to induce immunogenicity, and showing excellent efficacy against melanoma lung metastases in vivo. This nanoplatform forms a metal-organic framework through the coordination between tannic acid (TA) and Fe3+ and is then loaded with triptolide (TPL), which is coated with FA-modified BSA. The nanoparticles target melanoma cells by FA modification, releasing TPL, Fe3+ and TA. Fe3+ is reduced to Fe2+ by TA, triggering the Fenton reaction and resulting in ROS production. Moreover, TPL increases the production of intracellular ROS by inhibiting the expression of nuclear factor erythroid-2 related factor (Nrf2). Such simultaneous amplification of intracellular ROS induces the cells to undergo ferroptosis and pyroptosis, releasing large amounts of DAMPs, which stimulate antigen presentation of dendritic cells (DCs) and the proliferation of cytotoxic T lymphocytes (CD4+/CD8 + T cells) to inhibit tumor and lung metastasis. In addition, combining nanoparticle treatment with immune checkpoint blockade (ICB) further inhibits melanoma growth. This work provides a new strategy for tumor immunotherapy based on various combinations of cell death mechanisms.
Collapse
Affiliation(s)
- Shengmei Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Qiuyan Guo
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Rubing Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Peng Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Guoyan Deng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
11
|
Wang G, Guo H, Ren Y, Chen W, Wang Y, Li J, Liu H, Xing J, Zhang Y, Li N. Triptolide enhances carboplatin-induced apoptosis by inhibiting nucleotide excision repair (NER) activity in melanoma. Front Pharmacol 2023; 14:1157433. [PMID: 37324464 PMCID: PMC10267402 DOI: 10.3389/fphar.2023.1157433] [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: 02/02/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: Carboplatin (CBP) is a DNA damaging drug used to treat various cancers, including advanced melanoma. Yet we still face low response rates and short survival due to resistance. Triptolide (TPL) is considered to have multifunctional antitumor effects and has been confirmed to enhance the cytotoxic effects of chemotherapeutic drugs. Herein, we aimed to investigate the knowledge about the effects and mechanisms for the combined application of TPL and CBP against melanoma. Methods: Melanoma cell lines and xenograft mouse model were used to uncover the antitumor effects and the underlying molecular mechanisms of the alone or combined treatment of TPL and CBP in melanoma. Cell viability, migration, invasion, apoptosis, and DNA damage were detected by conventional methods. The rate-limiting proteins of the NER pathway were quantitated using PCR and Western blot. Fluorescent reporter plasmids were used to test the NER repair capacity. Results: Our results showed that the presence of TPL in CBP treatment could selectively inhibit NER pathway activity, and TPL exerts a synergistic effect with CBP to inhibit viability, migration, invasion, and induce apoptosis of A375 and B16 cells. Moreover, combined treatment with TPL and CBP significantly inhibited tumor progression in nude mice by suppressing cell proliferation and inducing apoptosis. Discussion: This study reveals the NER inhibitor TPL which has great potential in treating melanoma, either alone or in combination with CBP.
Collapse
Affiliation(s)
- Geng Wang
- Health Science Center, Ningbo University, Ningbo, China
| | - Hongmin Guo
- People’s Hospital of Changshou Chongqing, Chongqing, China
| | - Yan Ren
- Health Science Center, Ningbo University, Ningbo, China
| | - Weiyi Chen
- Health Science Center, Ningbo University, Ningbo, China
| | - Yixuan Wang
- Health Science Center, Ningbo University, Ningbo, China
| | - Jianing Li
- Health Science Center, Ningbo University, Ningbo, China
| | - Hua Liu
- Health Science Center, Ningbo University, Ningbo, China
| | - Jingjun Xing
- Health Science Center, Ningbo University, Ningbo, China
| | - Yanru Zhang
- Health Science Center, Ningbo University, Ningbo, China
| | - Na Li
- Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
12
|
Kar RD, Eberhart JK. Predicting Modifiers of Genotype-Phenotype Correlations in Craniofacial Development. Int J Mol Sci 2023; 24:1222. [PMID: 36674738 PMCID: PMC9864425 DOI: 10.3390/ijms24021222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
Most human birth defects are phenotypically variable even when they share a common genetic basis. Our understanding of the mechanisms of this variation is limited, but they are thought to be due to complex gene-environment interactions. Loss of the transcription factor Gata3 associates with the highly variable human birth defects HDR syndrome and microsomia, and can lead to disruption of the neural crest-derived facial skeleton. We have demonstrated that zebrafish gata3 mutants model the variability seen in humans, with genetic background and candidate pathways modifying the resulting phenotype. In this study, we sought to use an unbiased bioinformatic approach to identify environmental modifiers of gata3 mutant craniofacial phenotypes. The LINCs L1000 dataset identifies chemicals that generate differential gene expression that either positively or negatively correlates with an input gene list. These chemicals are predicted to worsen or lessen the mutant phenotype, respectively. We performed RNA-seq on neural crest cells isolated from zebrafish across control, Gata3 loss-of-function, and Gata3 rescue groups. Differential expression analyses revealed 551 potential targets of gata3. We queried the LINCs database with the 100 most upregulated and 100 most downregulated genes. We tested the top eight available chemicals predicted to worsen the mutant phenotype and the top eight predicted to lessen the phenotype. Of these, we found that vinblastine, a microtubule inhibitor, and clofibric acid, a PPAR-alpha agonist, did indeed worsen the gata3 phenotype. The Topoisomerase II and RNA-pol II inhibitors daunorubicin and triptolide, respectively, lessened the phenotype. GO analysis identified Wnt signaling and RNA polymerase function as being enriched in our RNA-seq data, consistent with the mechanism of action of some of the chemicals. Our study illustrates multiple potential pathways for Gata3 function, and demonstrates a systematic, unbiased process to identify modifiers of genotype-phenotype correlations.
Collapse
Affiliation(s)
| | - Johann K. Eberhart
- Department of Molecular Biosciences, College of Natural Sciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
13
|
Zhao L, Lan Z, Peng L, Wan L, Liu D, Tan X, Tang C, Chen G, Liu H. Triptolide promotes autophagy to inhibit mesangial cell proliferation in IgA nephropathy via the CARD9/p38 MAPK pathway. Cell Prolif 2022; 55:e13278. [PMID: 35733381 PMCID: PMC9436901 DOI: 10.1111/cpr.13278] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/04/2022] [Accepted: 05/23/2022] [Indexed: 01/18/2023] Open
Abstract
Background Mesangial cell proliferation is the most basic pathological feature of immunoglobulin A nephropathy (IgAN); however, the specific underlying mechanism and an appropriate therapeutic strategy are yet to be unearthed. This study aimed to investigate the therapeutic effect of triptolide (TP) on IgAN and the mechanism by which TP regulates autophagy and proliferation of mesangial cells through the CARD9/p38 MAPK pathway. Methods We established a TP‐treated IgAN mouse model and produced IgA1‐induced human mesangial cells (HMC) and divided them into control, TP, IgAN, and IgAN+TP groups. The levels of mesangial cell proliferation (PCNA, cyclin D1, cell viability, and cell cycle) and autophagy (P62, LC3 II, and autophagy flux rate) were measured, with the autophagy inhibitor 3‐Methyladenine used to explore the relationship between autophagy and proliferation. We observed CARD9 expression in renal biopsies from patients and analyzed its clinical significance. CARD9 siRNA and overexpression plasmids were constructed to investigate the changes in mesangial cell proliferation and autophagy as well as the expression of CARD9 and p‐p38 MAPK/p38 MAPK following TP treatment. Results Administering TP was safe and effectively alleviated mesangial cell proliferation in IgAN mice. Moreover, TP inhibited IgA1‐induced HMC proliferation by promoting autophagy. The high expression of CARD9 in IgAN patients was positively correlated with the severity of HMC proliferation. CARD9/p38 MAPK was involved in the regulation of HMC autophagy and proliferation, and TP promoted autophagy to inhibit HMC proliferation by downregulating the CARD9/p38 MAPK pathway in IgAN. Conclusion TP promotes autophagy to inhibit mesangial cell proliferation in IgAN via the CARD9/p38 MAPK pathway.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Zhixin Lan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Liang Peng
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Lili Wan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Di Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Xia Tan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
14
|
Li L, He D, Guo Q, Zhang Z, Ru D, Wang L, Gong K, Liu F, Duan Y, Li H. Exosome-liposome hybrid nanoparticle codelivery of TP and miR497 conspicuously overcomes chemoresistant ovarian cancer. J Nanobiotechnology 2022; 20:50. [PMID: 35078498 PMCID: PMC8787930 DOI: 10.1186/s12951-022-01264-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
Background Although cisplatin-based chemotherapy has been used as the first-line treatment for ovarian cancer (OC), tumor cells develop resistance to cisplatin during treatment, causing poor prognosis in OC patients. Studies have demonstrated that overactivation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway is involved in tumor chemoresistance and that overexpression of microRNA-497 (miR497) may overcome OC chemotherapy resistance by inhibiting the mTOR pathway. However, the low transcriptional efficiency and unstable chemical properties of miR497 limit its clinical application. Additionally, triptolide (TP) was confirmed to possess a superior killing effect on cisplatin-resistant cell lines, partially through inhibiting the mTOR pathway. Even so, the clinical applications of TP are restricted by serious systemic toxicity and weak water solubility. Results Herein, whether the combined application of miR497 and TP could further overcome OC chemoresistance by synergically suppressing the mTOR signaling pathway was investigated. Bioinspired hybrid nanoparticles formed by the fusion of CD47-expressing tumor exosomes and cRGD-modified liposomes (miR497/TP-HENPs) were prepared to codeliver miR497 and TP. In vitro results indicated that the nanoparticles were efficiently taken up by tumor cells, thus significantly enhancing tumor cell apoptosis. Similarly, the hybrid nanoparticles were effectively enriched in the tumor areas and exerted significant anticancer activity without any negative effects in vivo. Mechanistically, they promoted dephosphorylation of the overactivated PI3K/AKT/mTOR signaling pathway, boosted reactive oxygen species (ROS) generation and upregulated the polarization of macrophages from M2 to M1 macrophages. Conclusion Overall, our findings may provide a translational strategy to overcome cisplatin-resistant OC and offer a potential solution for the treatment of other cisplatin-resistant tumors. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01264-5.
Collapse
|
15
|
Wu Y, Li J, Zhong X, Shi J, Cheng Y, He C, Li J, Zou L, Fu C, Chen M, Zhang J, Gao H. A pH-sensitive supramolecular nanosystem with chlorin e6 and triptolide co-delivery for chemo-photodynamic combination therapy. Asian J Pharm Sci 2022; 17:206-218. [PMID: 35582637 PMCID: PMC9091603 DOI: 10.1016/j.ajps.2021.12.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/23/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
The combination of Ce6, an acknowledged photosensitizer, and TPL, a natural anticancer agent, has been demonstrated as a useful strategy to reinforce the tumor growth suppression, as well as decrease the systemic side effects compared with their monotherapy. However, in view of the optimal chemo-photodynamic combination efficiency, there is still short of the feasible nanovehicle to steadily co-deliver Ce6 and TPL, and stimuli-responsively burst release drugs in tumor site. Herein, we described the synergistic antitumor performance of a pH-sensitive supramolecular nanosystem, mediated by the host–guest complexing between β-CD and acid pH-responsive amphiphilic co-polymer mPEG-PBAE-mPEG, showing the shell–core structural micelles with the tight β-CD layer coating. Both Ce6 and TPL were facilely co-loaded into the spherical supramolecular NPs (TPL+Ce6/NPs) by one-step nanoprecipitation method, with an ideal particle size (156.0 nm), acid pH-responsive drug release profile, and enhanced cellular internalization capacity. In view of the combination benefit of photodynamic therapy and chemotherapy, as well as co-encapsulation in the fabricated pH-sensitive supramolecular NPs, TPL+Ce6/NPs exhibited significant efficacy to suppress cellular proliferation, boost ROS level, lower MMP, and promote cellular apoptosis in vitro. Particularly, fluorescence imaging revealed that TPL+Ce6/NPs preferentially accumulated in the tumor tissue area, with higher intensity than that of free Ce6. As expected, upon 650-nm laser irradiation, TPL+Ce6/NPs exhibited a cascade of amplified synergistic chemo-photodynamic therapeutic benefits to suppress tumor progression in both hepatoma H22 tumor-bearing mice and B16 tumor-bearing mice. More importantly, lower systemic toxicity was found in the tumor-bearing mice treated with TPL+Ce6/NPs. Overall, the designed supramolecular TPL+Ce6/NPs provided a promising alternative approach for chemo-photodynamic therapy in tumor treatment.
Collapse
Affiliation(s)
- Yihan Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jingjing Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Xuemei Zhong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jinfeng Shi
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanfen Cheng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chenglin He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiaxin Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liang Zou
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Chaomei Fu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Meiwan Chen
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jinming Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Corresponding author.
| | - Huile Gao
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Corresponding author.
| |
Collapse
|
16
|
Sui B, Cheng C, Shi S, Wang M, Xu P. Esterase-activatable and GSH-responsive Triptolide Nano-prodrug for the Eradication of Pancreatic Cancer. ADVANCED NANOBIOMED RESEARCH 2021; 1. [PMID: 34870282 DOI: 10.1002/anbr.202100040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Triptolide (TPL) is a small molecule isolated from a traditional Chinese herb Tripterygium wilfordii Hook F and shows excellent anticancer effect for pancreatic cancer cells. However, the poor water solubility and severe liver toxicity of TPL hindered its clinical application. In this study, TPL was covalently conjugated to a polymer and entrapped inside the core of the TPL nanogel (nTPL) to protect it from premature leakage during blood circulation. With the help of lactobionic acid (LBA), nTPL-LBA could selectively target the tumors in an orthotopic pancreatic cancer mouse model. TPL could be subsequently released intracellularly in its original form due to the presence of elevated intracellular esterase and GSH, and eventually kills cancer cells. nTPL-LBA treatment reduced tumor burden by 99% while not introducing TPL associated liver and kidney toxicities. Most importantly, more than half of the nTPL-LBA treated animals were tumor-free, suggesting that nTPL-LBA is an effective approach in eradicating pancreatic cancer.
Collapse
Affiliation(s)
- Binglin Sui
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Chen Cheng
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Shanshan Shi
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Mingming Wang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| | - Peisheng Xu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter St., Columbia, SC 29208, United States
| |
Collapse
|
17
|
Song H, Xing W, Shi X, Zhang T, Lou H, Fan P. Antitumor and toxicity study of mitochondria-targeted triptolide derivatives using triphenylphosphine (TPP +) as a carrier. Bioorg Med Chem 2021; 50:116466. [PMID: 34700239 DOI: 10.1016/j.bmc.2021.116466] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 01/23/2023]
Abstract
Based on the higher mitochondrial membrane potential (Δψm) of tumor cells than normal cells, a mitochondria-targeting strategy using delocalized lipophilic cations as carriers is a promising way to improve the antitumor effect of small molecules and to reduce toxicity. Triptolide (TP) has a strong antitumor effect but is limited in the clinic due to high systemic toxicity. Mitochondria-targeted TP derivatives were designed and synthesized using triphenylphosphine cations as carriers. The optimal derivative not only maintained the antitumor activity of TP but also showed a tumor cell selectivity trend. Moreover, the optimal derivative increased the release of lactate dehydrogenase and the production of ROS, decreased Δψm, and arrested HepG2 cells in G0/G1 phase. In a zebrafish HepG2 xenograft tumor model, the inhibitory effect of the optimal derivative was comparable to that of TP, while it had no obvious toxic effect on multiple indicators in zebrafish at the test concentrations. This work provided some evidence to support the mitochondria-targeting strategy.
Collapse
Affiliation(s)
- Huina Song
- Department of Natural Product Chemistry, Key Lab of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Wenlan Xing
- Department of Natural Product Chemistry, Key Lab of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Xiaojia Shi
- Department of Natural Product Chemistry, Key Lab of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Tao Zhang
- Shandong Qidu Pharmaceutical Co. Ltd., Neuroprotective Drugs, Zibo 255400, PR China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Lab of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Peihong Fan
- Department of Natural Product Chemistry, Key Lab of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
18
|
Cheng CT, Wang TY, Chen PR, Wu WH, Lai JM, Chang PMH, Hong YR, Huang CYF, Wang FS. Computer-Aided Design for Identifying Anticancer Targets in Genome-Scale Metabolic Models of Colon Cancer. BIOLOGY 2021; 10:biology10111115. [PMID: 34827109 PMCID: PMC8614794 DOI: 10.3390/biology10111115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/21/2023]
Abstract
Simple Summary Discovery of anticancer targets with minimal side effects is a major challenge in drug discovery and development. This study developed a fuzzy optimization framework for identifying anticancer targets. The framework was applied to identify not only gene regulator targets but also metabolite- and reaction-centric targets. The computational results show that the combination of a carbon metabolism target and any one-target gene that participates in the sphingolipid, glycerophospholipid, nucleotide, cholesterol biosynthesis, or pentose phosphate pathways is more effective for treatment than one-target inhibition is, and a two-target combination of 5-FU and folate supplement can improve cell viability, reduce metabolic deviation, and reduce side effects of normal cells. Abstract The efficient discovery of anticancer targets with minimal side effects is a major challenge in drug discovery and development. Early prediction of side effects is key for reducing development costs, increasing drug efficacy, and increasing drug safety. This study developed a fuzzy optimization framework for Identifying AntiCancer Targets (IACT) using constraint-based models. Four objectives were established to evaluate the mortality of treated cancer cells and to minimize side effects causing toxicity-induced tumorigenesis on normal cells and smaller metabolic perturbations. Fuzzy set theory was applied to evaluate potential side effects and investigate the magnitude of metabolic deviations in perturbed cells compared with their normal counterparts. The framework was applied to identify not only gene regulator targets but also metabolite- and reaction-centric targets. A nested hybrid differential evolution algorithm with a hierarchical fitness function was applied to solve multilevel IACT problems. The results show that the combination of a carbon metabolism target and any one-target gene that participates in the sphingolipid, glycerophospholipid, nucleotide, cholesterol biosynthesis, or pentose phosphate pathways is more effective for treatment than one-target inhibition is. A clinical antimetabolite drug 5-fluorouracil (5-FU) has been used to inhibit synthesis of deoxythymidine-5′-triphosphate for treatment of colorectal cancer. The computational results reveal that a two-target combination of 5-FU and a folate supplement can improve cell viability, reduce metabolic deviation, and reduce side effects of normal cells.
Collapse
Affiliation(s)
- Chao-Ting Cheng
- Department of Chemical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan; (C.-T.C.); (T.-Y.W.); (P.-R.C.); (W.-H.W.)
| | - Tsun-Yu Wang
- Department of Chemical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan; (C.-T.C.); (T.-Y.W.); (P.-R.C.); (W.-H.W.)
| | - Pei-Rong Chen
- Department of Chemical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan; (C.-T.C.); (T.-Y.W.); (P.-R.C.); (W.-H.W.)
| | - Wu-Hsiung Wu
- Department of Chemical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan; (C.-T.C.); (T.-Y.W.); (P.-R.C.); (W.-H.W.)
| | - Jin-Mei Lai
- Department of Life Science, Fu-Jen Catholic University, New Taipei City 24205, Taiwan;
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 11211, Taiwan
| | - Yi-Ren Hong
- Department of Biochemistry, Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan;
| | - Chi-Ying F. Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 11211, Taiwan;
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 11211, Taiwan
| | - Feng-Sheng Wang
- Department of Chemical Engineering, National Chung Cheng University, Chiayi 62102, Taiwan; (C.-T.C.); (T.-Y.W.); (P.-R.C.); (W.-H.W.)
- Correspondence: ; Tel.: +886-5-2720411 (ext. 33404)
| |
Collapse
|
19
|
Zheng Y, Kong F, Liu S, Liu X, Pei D, Miao X. Membrane protein-chimeric liposome-mediated delivery of triptolide for targeted hepatocellular carcinoma therapy. Drug Deliv 2021; 28:2033-2043. [PMID: 34569906 PMCID: PMC8477919 DOI: 10.1080/10717544.2021.1983072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Triptolide (TPL) is a diterpenoid triepoxide with broad antitumor efficacy, while lack of mechanism of action, severe systemic toxicity, and poor water solubility of TPL limited its usage. To unveil the mechanism of action and improve the pharmaceutical properties of TPL, here we explored the molecular mechanism of TPL and then fabricated TPL-loaded membrane protein-chimeric liposomes (TPL@MP-LP) and tested its anticancer efficacy against hepatocellular carcinoma (HCC). CCK8 assay, colony formation assay, EdU assay, and flow cytometry were used to examine the activity of TPL. RNA sequence and gain-and-loss of function assays were used to explore the molecular mechanisms. TPL@MP-LP was characterized by size, zeta potential, polydispersity index, and transmission electron microscopy. Cellular uptake and cell viability assay were performed to evaluate the internalization and anticancer efficacy of TPL@MP-LP in vitro. Biodistribution and in vivo antitumor efficacy of TPL@MP-LP were evaluated on orthotopic HCC mice models. TPL robustly inhibited HCC cells by inducing cell proliferation arrest, apoptosis via the mitochondrial pathway, and necroptosis via RIPK1/RIPK3/MLKL signaling. TPL was successfully loaded into MP-LP, with a drug-loading capacity of 5.62 ± 0.80%. MP-LP facilitated TPL internalization and TPL@MP-LP exerted enhanced anticancer efficacy against Huh7 cells. TPL@MP-LP showed targeting ability to the tumor site. More importantly, TPL@MP-LP treatment suppressed tumor growth but showed minimal damage to liver and renal functions. TPL exerted anticancer effects on HCC via inducing cell proliferation arrest, apoptosis, and necroptosis, and the MP-LP might be a promising delivery strategy to improve the antitumor efficacy while mitigating toxicity of TPL for HCC therapy.
Collapse
Affiliation(s)
- Yanwen Zheng
- Department of Liver Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fanhua Kong
- Institute of Hepatobiliary Diseases of Wuhan University, Transplant Centre of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Songyang Liu
- College of Biology, State Key Laboratory of Chemo/Biosensing and Chemometrics, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha, China
| | - Xi Liu
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Dongni Pei
- Department of Liver Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiongying Miao
- Department of Liver Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
20
|
Jiang L, Gu Y, Du Y, Tang X, Wu X, Liu J. Engineering Exosomes Endowed with Targeted Delivery of Triptolide for Malignant Melanoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:42411-42428. [PMID: 34464081 DOI: 10.1021/acsami.1c10325] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Malignant melanoma is considered the most aggressive skin carcinoma with invasive growth patterns. Triptolide (TPL) possesses various biological and pharmacological activities involved in cancer treatment. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce cancer cell apoptosis by binding to DR5 highly expressed on cancer cells. Exosomes are natural nanomaterials with low immunogenicity, nontoxicity, and excellent biocompatibility and have been extensively used as emerging delivery vectors for diverse therapeutic cargos. Herein, a delivery system based on TRAIL-engineered exosomes (TRAIL-Exo) for loading TPL for targeted therapy against malignant melanoma is proposed and systematically investigated. Our results showed that TRAIL-Exo/TPL could improve tumor targetability, enhance cellular uptake, inhibit proliferation, invasion, and migration, and induce apoptosis of A375 cells through activating the extrinsic TRAIL pathway and the intrinsic mitochondrial pathway in vitro. Moreover, intravenous injection of TRAIL-Exo/TPL significantly suppressed tumor progression and reduced the toxicity of TPL in the melanoma nude mouse model. Together, our research presents a novel strategy for high-efficiency exosome-based drug-delivery nanocarriers and provides an alternative dimension for developing a promising approach with synergistic therapeutic efficacy and targeting capacity for melanoma treatment.
Collapse
Affiliation(s)
- Liangdi Jiang
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Yongwei Gu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yue Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Xiaomeng Tang
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xin Wu
- Shanghai Wei Er Biopharmaceutical Technology Co., Ltd., Shanghai 201799, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
21
|
Qiu H, Zhang X, Yu H, Gao R, Shi J, Shen T. Identification of potential targets of triptolide in regulating the tumor microenvironment of stomach adenocarcinoma patients using bioinformatics. Bioengineered 2021; 12:4304-4319. [PMID: 34348580 PMCID: PMC8806726 DOI: 10.1080/21655979.2021.1945522] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
This study aimed to identify potential pharmacological targets of triptolide regulating the tumor microenvironment (TME) of stomach adenocarcinoma (STAD) patients. A total of 343 STAD cases from The Cancer Genome Atlas (TCGA) were assigned into high- or low-score groups applying Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE). Hub genes were identified from differentially expressed genes (DEGs) shared by stromal- and immune-related components in the TME of STAD patients using R software. Cox regression analysis was used to identify genes significantly correlated with STAD patient survival. Triptolide target genes were predicted from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Top 30 genes filtered by Cytohubba from 734 DEGs were screened as hub genes. Forty-two genes were found to be at high risk for STAD prognosis. Thirty-four targets of triptolide were predicted using the TCMSP database. Importantly, C-X-C chemokine receptor type 4 (CXCR4) was identified as a potential target of triptolide associated with the TME in STAD. Analysis of survival highlighted the association between CXCR4 upregulation with STAD progression and poor prognosis. Gene Set Enrichment Analysis (GSEA) confirmed that genes in the CXCR4- upregulated group had significant enrichment in immune-linked pathways. Additionally, triptolide targets were found to be significantly enriched in CXCR4-related chemokine and cancer-related p53 signaling pathways. Molecular docking demonstrated a high affinity between triptolide and CXCR4. In conclusion, CXCR4 may be a therapeutic target of triptolide in the treatment of STAD patients by modulating the TME.
Collapse
Affiliation(s)
- Hairong Qiu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaobo Zhang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Gao
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianglong Shi
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
22
|
Xu Y, Wei H, Gao J. Natural Terpenoids as Neuroinflammatory Inhibitors in LPS-stimulated BV-2 Microglia. Mini Rev Med Chem 2021; 21:520-534. [PMID: 31198113 DOI: 10.2174/1389557519666190611124539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/11/2019] [Accepted: 05/19/2019] [Indexed: 11/22/2022]
Abstract
Neuroinflammation is a typical feature of many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Microglia, the resident immune cells of the brain, readily become activated in response to an infection or an injury. Uncontrolled and overactivated microglia can release pro-inflammatory and cytotoxic factors and are the major culprits in neuroinflammation. Hence, research on novel neuroinflammatory inhibitors is of paramount importance for the treatment of neurodegenerative diseases. Bacterial lipopolysaccharide, widely used in the studies of brain inflammation, initiates several major cellular activities that critically contribute to the pathogenesis of neuroinflammation. This review will highlight the progress on terpenoids, an important and structurally diverse group of natural compounds, as neuroinflammatory inhibitors in lipopolysaccharidestimulated BV-2 microglial cells over the last 20 years.
Collapse
Affiliation(s)
- Yuanzhen Xu
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, China
| | - Hongbo Wei
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, China
| | - Jinming Gao
- Shaanxi Key Laboratory of Natural Products and Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
23
|
Gao J, Zhang Y, Liu X, Wu X, Huang L, Gao W. Triptolide: pharmacological spectrum, biosynthesis, chemical synthesis and derivatives. Theranostics 2021; 11:7199-7221. [PMID: 34158845 PMCID: PMC8210588 DOI: 10.7150/thno.57745] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/29/2021] [Indexed: 12/16/2022] Open
Abstract
Triptolide, an abietane-type diterpenoid isolated from Tripterygium wilfordii Hook. F., has significant pharmacological activity. Research results show that triptolide has obvious inhibitory effects on many solid tumors. Therefore, triptolide has become one of the lead compounds candidates for being the next "blockbuster" drug, and multiple triptolide derivatives have entered clinical research. An increasing number of researchers have developed triptolide synthesis methods to meet the clinical need. To provide new ideas for researchers in different disciplines and connect different disciplines with researchers aiming to solve scientific problems more efficiently, this article reviews the research progress made with analyzes of triptolide pharmacological activity, biosynthetic pathways, and chemical synthesis pathways and reported in toxicological and clinical studies of derivatives over the past 20 years, which have laid the foundation for subsequent researchers to study triptolide in many ways.
Collapse
Affiliation(s)
- Jie Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yifeng Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xihong Liu
- Basic Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xiayi Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Luqi Huang
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
24
|
Yalikong A, Li XQ, Zhou PH, Qi ZP, Li B, Cai SL, Zhong YS. A Triptolide Loaded HER2-Targeted Nano-Drug Delivery System Significantly Suppressed the Proliferation of HER2-Positive and BRAF Mutant Colon Cancer. Int J Nanomedicine 2021; 16:2323-2335. [PMID: 33776436 PMCID: PMC7989962 DOI: 10.2147/ijn.s287732] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/02/2021] [Indexed: 12/15/2022] Open
Abstract
Background Colon cancer (CRC) was a malignant tumor and there were about 25% of patients with tumor metastasis at diagnosis stage. Chemotherapeutic agents for metastatic CRC patients were with great side effects and the clinical treatment results of advanced CRC were still not satisfactory. Human epidermal growth factor receptor 2 (HER2) is overexpressed in some CRC patients and is an effective target for CRC patient treatment. Anti-HER2 therapy had a beneficial role in the treatment of HER2-positive metastatic CRC with fewer side effects. CRC patients with BRAF mutations were resistant to HER2 antibodies treatment. Therefore, there was an urgent need to develop new therapeutic agents. Methods HER2 targeted nanoparticles (TPLNP) drug delivery system loading triptolide (TPL) were prepared and identified. The effects of TPLNP and free TPL on cell viability, targeting and cell cycle progression on HT29 (BRAF mutation) with HER2 overexpression, were evaluated by Cell Counting Kit-8 (CCK8), Fluorescence Activating Cell Sorter (FACS) and immunofluorescence methods, respectively. The anti-tumor efficacies of TPLNP were evaluated in subcutaneous xenograft model of colon cancer and the survival rate, tumor volume, liver and kidney indexes of tumor-bearing mice were measured. Results TPLNP was small in nanosize (73.4±5.2nm) with narrow size distribution (PDI=0.15±0.02) and favorable zeta potential (pH=9.6, zeta potential: −57.3±6.69mV; pH=7.0, zeta potential: −28.7±5.1mV; pH=5.6, zeta potential: −21.1±4.73mV). Comparing with free TPL treatment group, TPLNP developed stranger colon cancer-killing efficiency in a dose- and time-dependent manner detected with CCK8 method; achieved good in vitro colon cancer targeting detected with flow cytometry and immunofluorescence experiments; enhanced more HT29-HER2 apoptosis and induced more cell cycle arrested in G1-S phase detected with FACS in vitro. As for in vivo antitumor response, TPLNP remarkably inhibited the growth of colon cancer in the colon cancer xenograft model, significantly improved the survival rate and did not exhibit significant liver and kidney toxicity in contrast with free TPL in vivo. Conclusion TPLNP was effectively against colon cancer with HER2 overexpression and BRAF mutation in pre-clinical models. In summary, the TPLNP appeared to be a promising treatment option for CRC in clinical application based on improved efficacy and the favorable safety profile.
Collapse
Affiliation(s)
- Ayimukedisi Yalikong
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Xu-Quan Li
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Shanghai Henlius Biopharmaceuticals Co., Ltd., Shanghai, 200033, People's Republic of China
| | - Ping-Hong Zhou
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Zhi-Peng Qi
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Bing Li
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Shi-Lun Cai
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| | - Yun-Shi Zhong
- Endoscopy Center, Zhongshan Hospital of Fudan University, Shanghai, 200032, People's Republic of China.,Endoscopy Research Institute of Fudan University, Shanghai, 200032, People's Republic of China
| |
Collapse
|
25
|
Ren Y, Kinghorn AD. Development of Potential Antitumor Agents from the Scaffolds of Plant-Derived Terpenoid Lactones. J Med Chem 2020; 63:15410-15448. [PMID: 33289552 PMCID: PMC7812702 DOI: 10.1021/acs.jmedchem.0c01449] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Naturally occurring terpenoid lactones and their synthetic derivatives have attracted increasing interest for their promising antitumor activity and potential utilization in the discovery and design of new antitumor agents. In the present perspective article, selected plant-derived five-membered γ-lactones and six-membered δ-lactones that occur with terpenoid scaffolds are reviewed, with their structures, cancer cell line cytotoxicity and in vivo antitumor activity, structure-activity relationships, mechanism of action, and the potential for developing cancer chemotherapeutic agents discussed in each case. The compounds presented include artemisinin (ART, 1), parthenolide (PTL, 2), thapsigargin (TPG, 3), andrographolide (AGL, 4), ginkgolide B (GKL B, 5), jolkinolide B (JKL B, 6), nagilactone E (NGL E, 7), triptolide (TPL, 8), bruceantin (BRC, 9), dichapetalin A (DCT A, 10), and limonin (LMN, 11), and their naturally occurring analogues and synthetic derivatives. It is hoped that this contribution will be supportive of the future development of additional efficacious anticancer agents derived from natural products.
Collapse
Affiliation(s)
- Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A. Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
26
|
Liu Y, Yang S, Wang K, Lu J, Bao X, Wang R, Qiu Y, Wang T, Yu H. Cellular senescence and cancer: Focusing on traditional Chinese medicine and natural products. Cell Prolif 2020; 53:e12894. [PMID: 32881115 PMCID: PMC7574878 DOI: 10.1111/cpr.12894] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is the principal cause of death and a dominant public health problem which seriously threatening human life. Among various ways to treat cancer, traditional Chinese medicine (TCM) and natural products have outstanding anti‐cancer effects with their unique advantages of high efficiency and minimal side effects. Cell senescence is a physiological process of cell growth stagnation triggered by stress, which is an important line of defence against tumour development. In recent years, active ingredients of TCM and natural products, as an interesting research hotspot, can induce cell senescence to suppress the occurrence and development of tumours, by inhibiting telomerase activity, triggering DNA damage, inducing SASP, and activating or inactivating oncogenes. In this paper, the recent research progress on the main compounds derived from TCM and natural products that play anti‐cancer roles by inducing cell senescence is systematically reviewed, aiming to provide a reference for the clinical treatment of pro‐senescent cancer.
Collapse
Affiliation(s)
- Yiman Liu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shenshen Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kailong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Lu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaomei Bao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Tao Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Haiyang Yu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
27
|
Combining triptolide with ABT-199 is effective against acute myeloid leukemia through reciprocal regulation of Bcl-2 family proteins and activation of the intrinsic apoptotic pathway. Cell Death Dis 2020; 11:555. [PMID: 32699295 PMCID: PMC7376040 DOI: 10.1038/s41419-020-02762-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022]
Abstract
Bcl-2 inhibitors display an effective activity in acute myeloid leukemia (AML), but its clinical efficacy as a monotherapy was limited in part owing to failure to target other antiapoptotic Bcl-2 family proteins, such as Mcl-1. In this context, the combination strategy may be a promising approach to overcome this barrier. Here, we report the preclinical efficacy of a novel strategy combining ABT-199 with triptolide (TPL), a natural product extracted from a traditional Chinese medicine, in AML. Combination treatment exhibited markedly increased cytotoxicity in leukemic cells irrespective of p53 status while largely sparing normal cells of the hematopoietic lineage. Moreover, co-administration of ABT-199 with TPL dramatically suppressed leukemia progression as well as prolonged animal survival in a xenograft AML model. The potentiated effect of ABT-199 and TPL against AML was associated with activation of the mitochondrum-related intrinsic apoptotic pathway through a mechanism reciprocally modulating Bcl-2 family proteins. In this case, TPL not only downregulated Mcl-1 but also upregulated proapoptotic BH3-only proteins, thereby overcoming the resistance toward ABT-199. Conversely, ABT-199 abrogated Bcl-2-mediated cytoprotection against TPL. Together, these findings suggest that the regimen combining TPL and ABT-199 might be active against AML by inducing robust apoptosis through reciprocal regulation of anti- and proapoptotic Bcl-2 family proteins, therefore providing a strong rationale for the clinical investigation of this combination regimen for the treatment of AML.
Collapse
|
28
|
Scott AT, Weitz M, Breheny PJ, Ear PH, Darbro B, Brown BJ, Braun TA, Li G, Umesalma S, Kaemmer CA, Maharjan CK, Quelle DE, Bellizzi AM, Chandrasekharan C, Dillon JS, O'Dorisio TM, Howe JR. Gene Expression Signatures Identify Novel Therapeutics for Metastatic Pancreatic Neuroendocrine Tumors. Clin Cancer Res 2020; 26:2011-2021. [PMID: 31937620 PMCID: PMC7165057 DOI: 10.1158/1078-0432.ccr-19-2884] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/19/2019] [Accepted: 01/10/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE Pancreatic neuroendocrine tumors (pNETs) are uncommon malignancies noted for their propensity to metastasize and comparatively favorable prognosis. Although both the treatment options and clinical outcomes have improved in the past decades, most patients will die of metastatic disease. New systemic therapies are needed. EXPERIMENTAL DESIGN Tissues were obtained from 43 patients with well-differentiated pNETs undergoing surgery. Gene expression was compared between primary tumors versus liver and lymph node metastases using RNA-Seq. Genes that were selectively elevated at only one metastatic site were filtered out to reduce tissue-specific effects. Ingenuity pathway analysis (IPA) and the Connectivity Map (CMap) identified drugs likely to antagonize metastasis-specific targets. The biological activity of top identified agents was tested in vitro using two pNET cell lines (BON-1 and QGP-1). RESULTS A total of 902 genes were differentially expressed in pNET metastases compared with primary tumors, 626 of which remained in the common metastatic profile after filtering. Analysis with IPA and CMap revealed altered activity of factors involved in survival and proliferation, and identified drugs targeting those pathways, including inhibitors of mTOR, PI3K, MEK, TOP2A, protein kinase C, NF-kB, cyclin-dependent kinase, and histone deacetylase. Inhibitors of MEK and TOP2A were consistently the most active compounds. CONCLUSIONS We employed a complementary bioinformatics approach to identify novel therapeutics for pNETs by analyzing gene expression in metastatic tumors. The potential utility of these drugs was confirmed by in vitro cytotoxicity assays, suggesting drugs targeting MEK and TOP2A may be highly efficacious against metastatic pNETs. This is a promising strategy for discovering more effective treatments for patients with pNETs.
Collapse
Affiliation(s)
- Aaron T Scott
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Michelle Weitz
- College of Public Health, Department of Biostatistics, University of Iowa, Iowa City, IA
| | - Patrick J Breheny
- College of Public Health, Department of Biostatistics, University of Iowa, Iowa City, IA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
| | - Po Hien Ear
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Benjamin Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Stead Family Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Bart J Brown
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Center for Bioinformatics and Computational Biology, College of Engineering, University of Iowa, Iowa City, IA
| | - Terry A Braun
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Center for Bioinformatics and Computational Biology, College of Engineering, University of Iowa, Iowa City, IA
| | - Guiying Li
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Shaikamjad Umesalma
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Courtney A Kaemmer
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Chandra K Maharjan
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Dawn E Quelle
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA
- Department of Pathology, Carver College of Medicine University of Iowa, Iowa City, IA
| | - Andrew M Bellizzi
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Pathology, Carver College of Medicine University of Iowa, Iowa City, IA
| | - Chandrikha Chandrasekharan
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Joseph S Dillon
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - Thomas M O'Dorisio
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA
| | - James R Howe
- Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
| |
Collapse
|
29
|
Li F, Cui H, Jin X, Gong X, Wang W, Wang J. Triptolide inhibits epithelial‑mesenchymal transition and induces apoptosis in gefitinib‑resistant lung cancer cells. Oncol Rep 2020; 43:1569-1579. [PMID: 32323848 PMCID: PMC7107945 DOI: 10.3892/or.2020.7542] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/29/2020] [Indexed: 01/28/2023] Open
Abstract
The epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI), gefitinib, is used widely to treat non-small cell lung cancer (NSCLC) with EGFR-activating mutations. Unfortunately, the acquired drug resistance promoted by epithelial-mesenchymal transition (EMT) markedly limits the clinical effects and remains a major barrier to a cure. Our previous isobaric tags for relative and absolute quantitation-based proteomics analysis revealed that the E-cadherin protein level was markedly upregulated by triptolide (TP). The present study aimed to determine whether TP reverses the gefitinib resistance of human lung cancer cells by regulating EMT. It was revealed that TP combined with gefitinib synergistically inhibited the migration and invasion of lung adenocarcinoma cell line A549; the combination treatment had a significantly better outcome than that of TP and gefitinib alone. Moreover, TP effectively increased the sensitivity of drug resistant A549 cells to gefitinib by upregulating E-cadherin protein expression and downregulating the MMP9, SNAIL, and vimentin expression levels. The dysregulated E-cadherin expression of gefitinib-sensitive cells induced gefitinib resistance, which could be overcome by TP. Finally, TP combined with gefitinib significantly inhibited the growth of xenograft tumors induced using gefitinib-resistant A549 cells, which was associated with EMT reversal and E-cadherin signaling activation in vivo. The present results indicated that the combination of TP and TKIs may be a promising therapeutic strategy to treat patients with NSCLCs harboring EGFR mutations.
Collapse
Affiliation(s)
- Fangqiong Li
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Huaizhong Cui
- Department of Clinical Laboratory, XiXi Hospital of Hangzhou, Hangzhou, Zhejiang 310023, P.R. China
| | - Xin Jin
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Xiaoting Gong
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Wei Wang
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Juan Wang
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| |
Collapse
|
30
|
Guo P, He Y, Xu T, Pi C, Jiang Q, Wei Y, Zhao L. Co-delivery system of chemotherapy drugs and active ingredients from natural plants: a brief overview of preclinical research for cancer treatment. Expert Opin Drug Deliv 2020; 17:665-675. [PMID: 32149539 DOI: 10.1080/17425247.2020.1739647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Many active ingredients from natural plants (AINPs) have been revealed to possess remarkable anticancer properties. Combination chemotherapy of chemo-drugs and AINPs has also proven to be more advantageous than individual chemo-drug treatment with respect to enhancing efficiency, alleviating toxicity, and controlling the development of multidrug resistance (MDR). Co-delivery is considered a promising method to effectively achieve and manage combination chemotherapy of chemo-drugs and AINPs, and various distinctive and functional co-delivery systems have been designed for these purposes to date.Areas covered: This review focuses on recent preclinical investigations of co-delivery systems for chemo-drugs and AINPs as new cancer treatment modalities. We particularly emphasize the apparent treatment advantages of these approaches, including augmenting efficiency, reducing toxicity, and controlling MDR.Expert opinion: There has already been notable progress in the application of combination chemotherapy with co-delivery systems loaded with chemo-drugs and AINPs based on results with cellular and animal models. The main challenge is to translate these successes into new anticancer compound preparations and promote their clinical application in practice. Nevertheless, continuous efforts with new designs of co-delivery systems remain essential, providing a foundation for future clinical research and development of new anticancer drugs.
Collapse
Affiliation(s)
- Pu Guo
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yingmeng He
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ting Xu
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Pi
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Qingsheng Jiang
- School of International Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Yumeng Wei
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Zhao
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
31
|
Haiaty S, Rashidi MR, Akbarzadeh M, Maroufi NF, Yousefi B, Nouri M. Targeting vasculogenic mimicry by phytochemicals: A potential opportunity for cancer therapy. IUBMB Life 2020; 72:825-841. [PMID: 32026601 DOI: 10.1002/iub.2233] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 01/06/2020] [Indexed: 12/18/2022]
Abstract
Vasculogenic mimicry (VM) is regarded as a process where very aggressive cancer cells generate vascular-like patterns without the presence of endothelial cells. It is considered as the main mark of malignant cancer and has pivotal role in cancer metastasis and progression in various types of cancers. On the other hand, resistance to the antiangiogenesis therapies leads to the cancer recurrence. Therefore, development of novel chemotherapies and their combinations is urgently needed for abolition of VM structures and also for better tumor therapy. Hence, identifying compounds that target VM structures might be superior therapeutic factors for cancers treatment and controlling the recurrence and metastasis. In recent times, naturally occurring compounds, especially phytochemicals have obtained great attention due to their safe properties. Phytochemicals are also capable of targeting VM structure and also their main signaling pathways. Consequently, in this review article, we illustrated key signaling pathways in VM, and the phytochemicals that affect these structures including curcumin, genistein, lycorine, luteolin, columbamine, triptolide, Paris polyphylla, dehydroeffusol, jatrorrhizine hydrochloride, grape seed proanthocyanidins, resveratrol, isoxanthohumol, dehydrocurvularine, galiellalactone, oxacyclododecindione, brucine, honokiol, ginsenoside Rg3, and norcantharidin. The recognition of these phytochemicals and their safety profile may lead to new therapeutic agents' development for VM elimination in different types of tumors.
Collapse
Affiliation(s)
- Sanya Haiaty
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad-Reza Rashidi
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Akbarzadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Nazila F Maroufi
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Li H, Li L, Mei H, Pan G, Wang X, Huang X, Wang T, Jiang Z, Zhang L, Sun L. Antitumor properties of triptolide: phenotype regulation of macrophage differentiation. Cancer Biol Ther 2019; 21:178-188. [PMID: 31663424 DOI: 10.1080/15384047.2019.1679555] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Tumor-associated macrophages (TAMs), which generally exhibit an M2-like phenotype, play a critical role in tumor development. Triptolide exerts a unique bioactive spectrum of anticancer activities. The aim of this study was to determine whether triptolide has any effect on the activation of TAMs and the production of tumor-promoting mediators. ICR-1 mice with azoxymethane/dextran sulfate sodium (AOM/DSS)-induced colon tumors and BALB/c mice co-inoculated with 4T1 cells and M2-polarized RAW264.7 cells were used to examine whether the inhibitory effect of triptolide on tumor progression was mediated by the targeting of TAMs. Real-time PCR, Western blot, immunofluorescence staining, and flow cytometry assays were performed to determine the expression of cell surface markers and cytokine production. The results showed that triptolide inhibited macrophage differentiation toward the M2 phenotype and abolished M2 macrophage-mediated tumor progression. Furthermore, triptolide inhibited the expression of M2 markers, such as CD206, Arginase 1, and CD204, and inhibited the secretion of anti-inflammatory cytokines. Thus our study indicated that triptolide selectively inhibited the functions of M2-polarized macrophages and TAMs, and this inhibitory effect of triptolide on TAM viability, differentiation, and cytokine production might elucidate the major mechanisms underlying its antitumor activity. Our findings provide important information for the potential clinical application of triptolide in cancer therapy.
Collapse
Affiliation(s)
- Han Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Liping Li
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Huifang Mei
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Guofeng Pan
- Department of TCM, Beijing Shijitan Hospital Affiliated with Capital Medical University, Beijing, China
| | - Xinzhi Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Xin Huang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Tao Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China.,Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China.,Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
33
|
Liu R, Li X, Huang N, Fan M, Sun R. Toxicity of traditional Chinese medicine herbal and mineral products. ADVANCES IN PHARMACOLOGY 2019; 87:301-346. [PMID: 32089237 DOI: 10.1016/bs.apha.2019.08.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Traditional Chinese medicine (TCM) has been used to treat numerous kinds of diseases for more than 2000 years in eastern Asian countries. A portion of the TCM herbal and mineral products are believed to be toxic according to modern standards, and are still widely prescribed in the clinic. However, some TCM products considered to be non-toxic or low-toxic have been reported to possess significant toxicological effects on different organs in both animal and human models. In this review, we define the term "toxic" in TCM, and then we summarize the advances in pharmacology and toxicology research of Toxic Traditional Chinese Medicine (TTCM), including Chinese aconite (Fu Zi), Arsenic Trioxide, Tripterygium wilfordii Hook f. (Thunder God Vine), herbal drugs derived from plants in the Aristolochiaceae Juss. family (Ma Dou Ling), and other TCM products. Finally, the compatibility art of TCM and modern pharmaceutical approaches to manage undesired toxicity of TTCM is discussed. Promoting pharmacology and toxicology studies of TTCM and non-toxic TCM is critical for the further development and safety of TCM in clinical practice.
Collapse
Affiliation(s)
- Runping Liu
- Beijing University of Chinese Medicine, Beijing, China
| | | | - Nana Huang
- The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Mengyue Fan
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rong Sun
- The Second Hospital of Shandong University, Shandong University, Jinan, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China; Advanced Medical Research Institute, Shandong University, Jinan, China.
| |
Collapse
|
34
|
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: 2.4] [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
|
35
|
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: 13.0] [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]
|
36
|
Wang D, Wang D, Yan T, Jiang W, Han X, Yan J, Guo Y. Nanostructures assembly and the property of polysaccharide extracted from Tremella Fuciformis fruiting body. Int J Biol Macromol 2019; 137:751-760. [PMID: 31254577 DOI: 10.1016/j.ijbiomac.2019.06.198] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/31/2019] [Accepted: 06/25/2019] [Indexed: 12/19/2022]
Abstract
Polysaccharides from fungi are good free radical scavengers. However, there are no enzymes digesting these polysaccharides in the human body, which limits the use of fungal polysaccharides. Therefore, it is of great significance to study the preparation methods of fungal polysaccharides to improve the utilization rate of fungal polysaccharides. In this paper, the acidic polysaccharide of Tremella fuciformis was extracted by boiling and precipitated by ethanol. The total sugar content obtained by freeze-drying after ion exchange chromatography purification was 93.6%. It is mainly composed of mannose, glucuronic acid, xylose and fucose. According to the peak area, the mass ratio of the substance is about 6.8:1:1.5:0.6, which indicates that TFP is a polysaccharide with mannose as its main chain and glucuronic acid, fucose and xylose as well as a small amount of glucose as the branch chain. Molecular weight is 1.86 × 106 Da. The existence of glucuronic acid endows polysaccharides with negative charge in aqueous solution and can be assembled into nanostructures with chitosan. By measuring the swelling property in aqueous, it shows the TFP separated from Tremella fuciformis fruits is suitable for drug controlled release.
Collapse
Affiliation(s)
- Deqiang Wang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Maanshan 243032, China.
| | - Deguo Wang
- Department of Gerontology, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Tingxuan Yan
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Maanshan 243032, China
| | - Weifeng Jiang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Maanshan 243032, China
| | - Xinya Han
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Maanshan 243032, China
| | - Jvfen Yan
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Maanshan 243032, China
| | - Yanrong Guo
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Maanshan 243032, China
| |
Collapse
|
37
|
Huang Y, Chen Z, Wang Y, Ba X, Huang Y, Shen P, Wang H, Tu S. Triptolide exerts an anti-tumor effect on non‑small cell lung cancer cells by inhibiting activation of the IL‑6/STAT3 axis. Int J Mol Med 2019; 44:291-300. [PMID: 31115521 DOI: 10.3892/ijmm.2019.4197] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/23/2019] [Indexed: 11/05/2022] Open
Abstract
Lung cancer is the leading cause of cancer‑associated mortality and current treatments are not sufficiently effective. Numerous studies have revealed that triptolide (TP), a classical traditional Chinese medicine compound widely used as an anti‑inflammatory and antirheumatic drug, also has an antitumor effect. This effect is hypothesized to be mediated by multiple pathways, with signal transducer and activator of transcription 3 (STAT3) possibly one of them. Evidence indicates that STAT3 participates in the initiation and progression of lung cancer during cell proliferation, apoptosis and migration; however, whether and how TP affects STAT3 and its targets remain unclear. In this study, the potential role of TP in the proliferation, apoptosis, and migration of non‑small cell lung cancer cell lines was investigated and evaluated the impact of TP on the interleukin‑6 (IL‑6)/STAT3 axis. The results showed that TP inhibited cell proliferation and migration and induced apoptosis. TP decreased the phosphorylation of STAT3, inhibited STAT3 translocation into the nucleus, and reduced the expression of STAT3 target genes involved in cell survival, apoptosis and migration, e.g. C‑myc, BCL‑2, myeloid cell leukemia‑1 (MCL‑1), and matrix metallopeptidase 9 (MMP‑9). Additionally, IL‑6‑induced activation of STAT3 target genes (e.g. MCL‑1 and BCL‑2) was attenuated by TP and homoharringtonine. In conclusion, the effect of TP on STAT3 signaling points to a promising strategy for drug development.
Collapse
Affiliation(s)
- Ying Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhe Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yu Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xin Ba
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yao Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Pan Shen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hui Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Shenghao Tu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
38
|
Timmers HTM, Tora L. Transcript Buffering: A Balancing Act between mRNA Synthesis and mRNA Degradation. Mol Cell 2019; 72:10-17. [PMID: 30290147 DOI: 10.1016/j.molcel.2018.08.023] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/20/2018] [Accepted: 08/15/2018] [Indexed: 10/28/2022]
Abstract
Transcript buffering involves reciprocal adjustments between overall rates in mRNA synthesis and degradation to maintain similar cellular concentrations of mRNAs. This phenomenon was first discovered in yeast and encompasses coordination between the nuclear and cytoplasmic compartments. Transcript buffering was revealed by novel methods for pulse labeling of RNA to determine in vivo synthesis and degradation rates. In this Perspective, we discuss the current knowledge of transcript buffering. Emphasis is placed on the future challenges to determine the nature and directionality of the buffering signals, the generality of transcript buffering beyond yeast, and the molecular mechanisms responsible for this balancing.
Collapse
Affiliation(s)
- H Th Marc Timmers
- German Cancer Consortium (DKTK), partner site Freiburg, German Cancer Research Center (DKFZ) Zentrale Klinische Forschung (ZKF), and Medical Faculty-University of Freiburg, Breisacher Str. 66, 79106 Freiburg, Germany.
| | - László Tora
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique, UMR7104, INSERM U1258 and Université de Strasbourg, 67404 Illkirch, France.
| |
Collapse
|
39
|
Jiang W, Chen M, Xiao C, Yang W, Qin Q, Tan Q, Liang Z, Liao X, Mao A, Wei C. Triptolide Suppresses Growth of Breast Cancer by Targeting HMGB1 in Vitro and in Vivo. Biol Pharm Bull 2019; 42:892-899. [PMID: 30956264 DOI: 10.1248/bpb.b18-00818] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Triptolide has been indicated potent anti-cancer effect involving multiple molecular targets and signaling pathways. High-mobility group box 1 (HMGB1) is a highly conserved DNA-binding protein taking part in breast cancer development. The therapeutic effect of triptolide on HMGB1 has not been reported. Thus, our study aims to clarify the role of HMGB1 in triptolide-induced anti-growth effect on breast cancer in vitro and in vivo. We demonstrated that triptolide significantly suppressed growth of breast cancer cells by inhibition of cell viability, clonogenic ability. Further studies evidenced that triptolide treatment not only inhibited HMGB1 mRNA expression, but also decreased supernatant level of HMGB1 in vitro. In line with these observations, exogenous recombinant HMGB1 (rHMGB1) promoted cell proliferation of breast cancer, and triptolide reversed the rHMGB1-promoted proliferative effect. As well, triptolide enhanced the anti-proliferative activity of ethyl pyruvate (EP) (HMGB1 inhibitor). Furthermore, downstream correlation factors (Toll-like receptor 4 (TLR4) and phosphorylated-nuclear factor-kappaB (NF-κB) p65) of HMGB1 were significantly decreased in vitro after triptolide treatment. Consistantly, we confirmed that tumor growth was significantly inhibited after triptolide treatment in vivo. Meanwhile, immunohistochemical analyses showed that triptolide treatment significantly decreased the level of cytoplasmic HMGB1 and TLR4 expression, whereas the expression of NF-κB p65 was relatively higher in cytoplasm, and conversely lower in nucleus as compared to the control group. Collectively, these results demonstrate that triptolide suppresses the growth of breast cancer cells via reduction of HMGB1 expression in vitro and in vivo, which may provide new insights into the treament of breast cancer.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Medical Oncology, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Maojian Chen
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Chanchan Xiao
- Department of Experimental Research, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Weiping Yang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Qinghong Qin
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Qixing Tan
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Zhijie Liang
- Department of Breast and Thyroid Surgery, The Fifth Affliated Hospital of Guangxi Medical University & The First People's Hospital of Nanning
| | - Xiaoli Liao
- Department of Medical Oncology, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Anyun Mao
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| | - Changyuan Wei
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University
| |
Collapse
|
40
|
Li Q, He Z, Liu J, Wu J, Tan G, Jiang J, Su Z, Cao M. Paris polyphylla 26 triggers G2/M phase arrest and induces apoptosis in HepG2 cells via inhibition of the Akt signaling pathway. J Int Med Res 2019; 47:1685-1695. [PMID: 30819018 PMCID: PMC6460622 DOI: 10.1177/0300060519826823] [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] [Indexed: 12/13/2022] Open
Abstract
Objectives Paris polyphylla 26 (PP-26) is a monomer purified from Paris polyphylla, which has traditionally been used as an antimicrobial, hemostatic, and anticancer agent in China. The anti-proliferation effect and underlying molecular mechanism of PP-26 were investigated in vitro. Methods The effects of PP-26 on various tumor cells were detected by MTT assay. PP-26-affected cell cycle and cell cycle-related proteins in HepG2 cells were detected by flow cytometry and western blotting, respectively. Apoptosis in response to PP-26 was assessed by Hoechst 33258 staining and flow cytometry. PP-26-affected apoptosis-related proteins and Akt signaling were detected by western blotting. The inhibitory effect of PP-26 on HepG2 cells, when combined with 5-fluorouracil (5-FU), was also assessed. Results PP-26 inhibited proliferation of HepG2 cells in a dose-dependent manner by triggering G2/M-phase arrest. Moreover, PP-26 induced apoptosis of HepG2 cells. Expression levels of apoptosis proteins caspase 9, caspase 3, PARP, Bcl-2, Bcl-xL, and Mcl-1 were downregulated, while the expression level of apoptosis protein Bax was upregulated. Expression levels of p-Akt, p-GSK-3β, and p-Foxo3 were downregulated. Combination with PP-26 enhanced 5-FU inhibition of HepG2 cell proliferation. Conclusions PP-26 triggers G2/M-phase arrest and induces apoptosis in HepG2 cells via inhibition of the Akt signaling pathway.
Collapse
Affiliation(s)
- Qiang Li
- 1 Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Zifan He
- 2 Department of Biochemistry, Medical College, Jinan University, Guangzhou, China
| | - Jiming Liu
- 3 Department of General Surgery, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jianlong Wu
- 1 Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Guixiang Tan
- 4 School of Nursing, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jianwei Jiang
- 2 Department of Biochemistry, Medical College, Jinan University, Guangzhou, China
| | - Zexuan Su
- 5 Department of Urology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Mingrong Cao
- 1 Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
41
|
Triptolide-targeted delivery methods. Eur J Med Chem 2019; 164:342-351. [DOI: 10.1016/j.ejmech.2018.12.058] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/15/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022]
|
42
|
Lu X, Zhuang H, Yu Q, Zhang X, Wu Z, Zhang L, Xu Y, Wu B, Yang L, Ma A, Gan X, Yu X, Shen J, Xu R. Identification of the UBA2-WTIP fusion gene in acute myeloid leukemia. Exp Cell Res 2018; 371:409-416. [PMID: 30179602 DOI: 10.1016/j.yexcr.2018.08.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/17/2018] [Accepted: 08/31/2018] [Indexed: 10/28/2022]
Abstract
Identifying and targeting oncogenic fusion genes have revolutionized the treatment of leukemia, such as PML-RARα fusion gene in acute promyelocytic leukemia. Here we identified an intrachromosomal fusion gene located on chromosome 19q.13 between UBA2 and WTIP gene in a case of acute myeloid leukemia. The UBA2-WTIP fusion gene contains the N-terminal E1_enzyme_family, VAE_Ubl domains of UBA2, and the C-terminal LIM domains of WTIP. The UBA2-WTIP fusion was detected by reverse transcriptase polymerase chain reaction and Sanger sequencing in 19 of 56 acute myeloid leukemia samples (33.9%). Ectopic expression of the UBA2-WTIP fusion in human acute myeloid leukemia KG-1a cells showed enhanced cell proliferation both in vitro and in vivo. The UBA2-WTIP fusion induced phosphorylation of STAT3, STAT5 and ERK1/2, and abrogates WTIP-mediated mammalian processing body formation. Finally, triptolide displayed selective cytotoxicity against KG-1a cells harboring the UBA2-WTIP fusion. Collectively, our findings suggest that the UBA2-WTIP fusion is an oncogenic fusion gene, as well as a promising therapeutic target for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Xiaoya Lu
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Haifeng Zhuang
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou310009, China
| | - Qingfeng Yu
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Xuzhao Zhang
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Zhaoxing Wu
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Lei Zhang
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Ying Xu
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Bowen Wu
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Linlin Yang
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - An Ma
- Zhejiang Academy of Medical Sciences, Hangzhou 310012, China
| | - Xiaoxian Gan
- Zhejiang Academy of Medical Sciences, Hangzhou 310012, China
| | - Xiaofang Yu
- Cancer Institute, Zhejiang University, Hangzhou 310009, China
| | - Jianping Shen
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou310009, China.
| | - Rongzhen Xu
- Department of Hematology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province), The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; Cancer Institute, Zhejiang University, Hangzhou 310009, China; Institute of Hematology, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
43
|
周 玉, 孙 玉, 李 萍, 秦 国, 程 倩, 刘 宇, 陈 滢, 王 国. [Monoside antagonizes triptolide-induced hepatocyte apoptosis via the anti-oxidative stress pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:949-955. [PMID: 30187882 PMCID: PMC6744036 DOI: 10.3969/j.issn.1673-4254.2018.08.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the protective effect of monoside against triptolide-induced liver injury and explore its molecular mechanism. METHODS BALB/C mice treated with gastric lavage with triptolide and monoside, either alone or in combination, were examined for changes of hepatic biochemical parameters using the serological method. The growth inhibition rate of HepG2 cells treated with triptolide or monoside or both was assessed with MTT assay, and the cell morphological changes were observed using laser confocal microscopy; the expressions of the target proteins in the antioxidative stress pathway were detected using flow cytometry and Western blotting. RESULTS In BALB/C mice, gastric lavage of triptolide induced obvious hepatic damage. In HepG2 cells, treatment with triptolide significantly inhibited the cell growth, resulting in a cell viability as low as 72.83% at 24 h; triptolide also induced obvious cell apoptosis and cell nucleus deformation, causing an apoptosis rate of 43.1% in the cells at 24 h. Triptolide significantly reduced the expressions of Nrf2 and HO-1 proteins related with the oxidative stress pathway. Combined treatment with morroniside obviously reversed these changes, resulting in significantly decreased hepatic biochemical parameters and the liver index in BALB/C mice and in significantly lowered cell apoptosis rate, improved cell morphology, and increased Nrf2 and HO-1 protein expressions in HepG2 cells. CONCLUSIONS Monoside protects against triptolide-induced liver injury possibly by relieving oxidative stress.
Collapse
Affiliation(s)
- 玉燕 周
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
- 安徽省多糖药物药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs/Anhui Provincial Key Laboratory of Active Biological Macro-molecules, Wuhu 241002, China
| | - 玉 孙
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
- 安徽省多糖药物药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs/Anhui Provincial Key Laboratory of Active Biological Macro-molecules, Wuhu 241002, China
| | - 萍 李
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
- 安徽省多糖药物药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs/Anhui Provincial Key Laboratory of Active Biological Macro-molecules, Wuhu 241002, China
| | - 国正 秦
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
- 安徽省多糖药物药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs/Anhui Provincial Key Laboratory of Active Biological Macro-molecules, Wuhu 241002, China
| | - 倩 程
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
| | - 宇 刘
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
| | - 滢俐 陈
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
| | - 国栋 王
- 皖南医学院药物研发中心//药学院,安徽 芜湖 241002Drug Research & Development Center/School of Pharmacy, Wannan Medical College, Wuhu, 241002, China
- 安徽省多糖药物药物工程技术研究中心//活性生物大分子研究安徽省重点实验室,安徽 芜湖 241002Anhui Provincial Engineering Research Center for Polysaccharide Drugs/Anhui Provincial Key Laboratory of Active Biological Macro-molecules, Wuhu 241002, China
| |
Collapse
|