1
|
Feng K, Li X, Bai Y, Zhang D, Tian L. Mechanisms of cancer cell death induction by triptolide: A comprehensive overview. Heliyon 2024; 10:e24335. [PMID: 38293343 PMCID: PMC10826740 DOI: 10.1016/j.heliyon.2024.e24335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
The need for naturally occurring constituents is driven by the rise in the cancer prevalence and the unpleasant side effects associated with chemotherapeutics. Triptolide, the primary active component of "Tripterygium Wilfordii", has exploited for biological mechanisms and therapeutic potential against various tumors. Based on the recent pre-clinical investigations, triptolide is linked to the induction of death of cancerous cells by triggering cellular apoptosis via inhibiting heat shock protein expression (HSP70), and cyclin dependent kinase (CDKs) by up regulating expression of P21. MKP1, histone methyl transferases and RNA polymerases have all recently identified as potential targets of triptolide in cells. Autophagy, AKT signaling pathway and various pathways involving targeted proteins such as A-disintegrin & metalloprotease-10 (ADAM10), Polycystin-2 (PC-2), dCTP pyro-phosphatase 1 (DCTP1), peroxiredoxin-I (Prx-I), TAK1 binding protein (TAB1), kinase subunit (DNA-PKcs) and the xeroderma-pigmentosum B (XPB or ERCC3) have been exploited. Besides that, triptolide is responsible for enhancing the effectiveness of various chemotherapeutics. In addition, several triptolide moieties, including minnelide and LLDT8, have progressed in investigations on humans for the treatment of cancer. Targeted strategies, such as triptolide conjugation with ligands or triptolide loaded nano-carriers, are efficient techniques to confront toxicities associated with triptolide. We expect and anticipate that advances in near future, regarding combination therapies of triptolide, might be beneficial against cancerous cells.
Collapse
Affiliation(s)
- Ke Feng
- Department of General Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Xiaojiang Li
- Department of General Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Yuzhuo Bai
- Department of Breast and Thyroid Surgery Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Dawei Zhang
- Department of General Surgery Baishan Hospital of Traditional Chinese Medicine, Baishan, 134300, China
| | - Lin Tian
- Department of Lung Oncology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| |
Collapse
|
2
|
Yuan C, Liao Y, Liao S, Huang M, Li D, Wu W, Quan Y, Li L, Yu X, Si W. Triptolide inhibits the progression of Glioblastoma U251 cells via targeting PROX1. Front Oncol 2023; 13:1077640. [PMID: 36969058 PMCID: PMC10038275 DOI: 10.3389/fonc.2023.1077640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
BackgroundGlioblastoma multiforme (GBM) is the most lethal brain cancer in adults, characterized by rapid growth, extensive invasiveness, and poor prognosis, and there is still a lack of effective treatments. Here, we aimed to explore the role of triptolide (TPL), purified from Tripterygium wilfordii Hook F, on glioblastoma cell growth, apoptosis, proliferation, migration and invasion, as well as potential underlying mechanisms.MethodsThe publicly available clinical data of Brain Lower Grade Glioma (LGG) from The Cancer Genome Atlas (TCGA) had been screened to observe PROX1 expression. The Kaplan-Meier analysis was used to analyze the relationship between PROX1 expression and GBM prognosis. CCK8, cell cycle, EDU, apoptosis, wound healing, and transwell assays were performed to detect the effects of TPL on glioblastoma U251 cell viability, cell cycle, proliferation, apoptosis, migration and invasion, respectively. Further, a soft agar colony assay was used to calculate the growth of glioblastoma cells. The qRT-PCR and western blot were conducted to quantify PROX1 mRNA and protein levels. The transcriptional regulation of TPL was detected by Dual luciferase reporter assay.ResultsWe found that TPL inhibited glioblastoma cell viability, proliferation, cell cycle, migration and invasion, but enhanced apoptosis in a dose-dependent manner. The expression of cell cycle inhibitor, P21, and pro-apoptosis factor, Bax was increased, while invasion-related factors MMP2 and MMP9 were silenced after TPL treatments. Mechanistically, TPL showed transcriptional inhibition of PROX1 appearance. Moreover, ectopic expression of PROX1 partially rescued the effects of TPL on glioblastoma cell viability, proliferation, apoptosis, migration and invasion, and on the expression of cell function-related genes.ConclusionThis study verified that TPL inhibited the progression of glioblastoma cells by transcriptionally depressing the expression of PROX1.
Collapse
Affiliation(s)
- Chao Yuan
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
- Department of Oncology, Zhaoqing First People’s Hospital Affiliated to Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Yanli Liao
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Shengjie Liao
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Mi Huang
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Duanzhuo Li
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Weibin Wu
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Yi Quan
- Department of Oncology, Zhaoqing First People’s Hospital Affiliated to Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Liqiang Li
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
- Department of Oncology, Zhaoqing First People’s Hospital Affiliated to Zhaoqing Medical College, Zhaoqing, Guangdong, China
| | - Xin Yu
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
- *Correspondence: Wenxia Si, ; ; Xin Yu, ;
| | - Wenxia Si
- Department of Scientific Research and Experiment Center, Zhaoqing Medical College, Zhaoqing, Guangdong, China
- Department of Oncology, Zhaoqing First People’s Hospital Affiliated to Zhaoqing Medical College, Zhaoqing, Guangdong, China
- *Correspondence: Wenxia Si, ; ; Xin Yu, ;
| |
Collapse
|
3
|
Chen LJ, Hsu TC, Yeh PJ, Yow JL, Chang CL, Lin CH, Tzang BS. Differential Effects of Wedelia chinensis on Human Glioblastoma Multiforme Cells. Integr Cancer Ther 2021; 20:15347354211000119. [PMID: 33729002 PMCID: PMC7983241 DOI: 10.1177/15347354211000119] [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: 12/22/2022] Open
Abstract
Introduction: Glioblastoma multiforme (GBM) is the most aggressive glioma, and its diffuse
nature makes resection of it difficult. Moreover, even with the
administration of postoperative radiotherapy and chemotherapy, prolonged
remission is often not achieved. Hence, innovative or alternative treatments
for GBM are urgently required. Traditional Chinese herbs and their
functional components have long been used in the treatment of various
cancers, including GBM. The current study investigated the antitumor
activity of Wedelia chinensis and its major functional
components, luteolin and apigenin, on GBM. Materials and Methods: MTT assay, Transwell migration assay, and flow cytometry analysis were
adopted to assess the cell viability, invasive capability, and cell cycle.
Immunofluorescence staining and Western blotting were used to detect the
expressions of apoptotic and autophagy-related signaling molecules. Results: The W. chinensis extract (WCE) significantly inhibited the
proliferation and invasive ability of both GBM8401 and U-87MG cells in a
dose-dependent manner. Moreover, differential effects of WCE on GBM8401 and
U-87MG cells were observed: WCE induced apoptosis in GBM8401 cells and
autophagy in U-87MG cells. Notably, WCE had significant effects in reducing
the cell survival and invasive capability of both GBM8401 and U-87MG cells
than the combination of luteolin and apigenin. Conclusions: Taken together, these findings indicate the potential of using WCE and the
combination of luteolin and apigenin for GBM treatment. However, further
investigations are warranted before considering recommending the clinical
use of WCE or the combination of luteolin and apigenin as the standard for
GBM treatment.
Collapse
Affiliation(s)
- Li-Jeng Chen
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan, R.O.C
| | - Tsai-Ching Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan, R.O.C.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, Taiwan, R.O.C.,Immunology Research Center, Chung Shan Medical University, Taichung city, Taiwan, R.O.C
| | - Pei-Jung Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan, R.O.C
| | - Jia Le Yow
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan, R.O.C
| | - Chia-Ling Chang
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan, R.O.C
| | - Cheng-Hui Lin
- Division of Rheumatology Immunology Clinic, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung City, Taiwan, R.O.C
| | - Bor-Show Tzang
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan, R.O.C.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung City, Taiwan, R.O.C.,Immunology Research Center, Chung Shan Medical University, Taichung city, Taiwan, R.O.C.,Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung City, Taiwan, R.O.C
| |
Collapse
|
4
|
Liaw K, Sharma R, Sharma A, Salazar S, Appiani La Rosa S, Kannan RM. Systemic dendrimer delivery of triptolide to tumor-associated macrophages improves anti-tumor efficacy and reduces systemic toxicity in glioblastoma. J Control Release 2021; 329:434-444. [PMID: 33290796 PMCID: PMC7904646 DOI: 10.1016/j.jconrel.2020.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Novel delivery strategies are necessary to effectively address glioblastoma without systemic toxicities. Triptolide is a therapy derived from the thunder god vine that has shown potent anti-proliferative and immunosuppressive properties but exhibits significant adverse systemic effects. Dendrimer-based nanomedicines have shown great potential for clinical translation of systemic therapies targeting neuroinflammation and brain tumors. Here we present a novel dendrimer-triptolide conjugate that specifically targets tumor-associated macrophages (TAMs) in glioblastoma from systemic administration and exhibits triggered release under intracellular and intratumor conditions. This targeted delivery improves phenotype switching of TAMs from pro- towards anti-tumor expression in vitro. In an orthotopic model of glioblastoma, dendrimer-triptolide achieved significantly improved amelioration of tumor burden compared to free triptolide. Notably, the triggered release mechanism of dendrimer-mediated triptolide delivery significantly reduced triptolide-associated hepatic and cardiac toxicities. These results demonstrate that dendrimers are a promising targeted delivery platform to achieve effective glioblastoma treatment by improving efficacy while reducing systemic toxicities.
Collapse
Affiliation(s)
- Kevin Liaw
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Rishi Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Anjali Sharma
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Sebastian Salazar
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Santiago Appiani La Rosa
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD, 21205, USA.
| |
Collapse
|
5
|
Wang J, Qi F, Wang Z, Zhang Z, Pan N, Huai L, Qu S, Zhao L. A review of traditional Chinese medicine for treatment of glioblastoma. Biosci Trends 2019; 13:476-487. [PMID: 31866614 DOI: 10.5582/bst.2019.01323] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) is the most common primary malignant intracranial tumor. Due to its high morbidity, high mortality, high recurrence rate, and low cure rate, it has brought great difficulty for treatment. Although the current treatment is multimodal, including surgical resection, radiotherapy, and chemotherapy, it does not significantly improve survival time. The dismal prognosis and inevitable recurrence as well as resistance to chemoradiotherapy may be related to its highly cellular heterogeneity and multiple subclonal populations. Traditional Chinese medicine has its own unique advantages in the prevention and treatment of it. A comprehensive literature search of anti-glioblastoma active ingredients and derivatives from traditional Chinese medicine was carried out in literature published in PubMed, Scopus, Web of Science Cochrane library, CNKI, Wanfang, and VIP database. Hence, this article systematically reviews experimental research progress of some traditional Chinese medicine in treatment of glioblastoma from two aspects: strengthening vital qi and eliminating pathogenic qi. Among, strengthening vital qi medicine includes panax ginseng, licorice, lycium barbarum, angelica sinensis; eliminating pathogenic medicine includes salvia miltiorrhiza bunge, scutellaria baicalensis, coptis rhizoma, thunder god vine, and sophora flavescens. We found that the same active ingredient can act on different signaling pathways, such as ginsenoside Rg3 inhibited proliferation and induced apoptosis via the AKT, MEK signal pathway. Hence, this multi-target, multi-level pathway may bring on a new dawn for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Jinjing Wang
- Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Fanghua Qi
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affili-ated to Shandong University, Ji'nan, China
| | - Zhixue Wang
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affili-ated to Shandong University, Ji'nan, China
| | - Zhikun Zhang
- Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Ni Pan
- Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Lei Huai
- Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Shuyu Qu
- Shandong University of Traditional Chinese Medicine, Ji'nan, China
| | - Lin Zhao
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital affili-ated to Shandong University, Ji'nan, China
| |
Collapse
|
6
|
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
|
7
|
Wang F, Zheng Z, Guan J, Qi D, Zhou S, Shen X, Wang F, Wenkert D, Kirmani B, Solouki T, Fonkem E, Wong ET, Huang JH, Wu E. Identification of a panel of genes as a prognostic biomarker for glioblastoma. EBioMedicine 2018; 37:68-77. [PMID: 30341039 PMCID: PMC6284420 DOI: 10.1016/j.ebiom.2018.10.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/31/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is a fatal disease without effective therapy. Identification of new biomarkers for prognosis would enable more rational selections of strategies to cure patients with GBM and prevent disease relapse. Methods Seven datasets derived from GBM patients using microarray or next generation sequencing in R2 online database (http://r2.amc.nl) were extracted and then analyzed using JMP software. The survival distribution was calculated according to the Kaplan-Meier method and the significance was determined using log-rank statistics. The sensitivity of a panel of GBM cell lines in response to temozolomide (TMZ), salinomycin, celastrol, and triptolide treatments was evaluated using MTS and tumor-sphere formation assay. Findings We identified that CD44, ATP binding cassette subfamily C member 3 (ABCC3), and tumor necrosis factor receptor subfamily member 1A (TNFRSF1A) as highly expressed genes in GBMs are associated with patients' poor outcomes and therapy resistance. Furthermore, these three markers combined with MGMT, a conventional GBM marker, can classify GBM patients into five new subtypes with different overall survival time in response to treatment. The four-gene signature and the therapy response of GBMs to a panel of therapeutic compounds were confirmed in a panel of GBM cell lines. Interpretation The data indicate that the four-gene panel can be used as a therapy response index for GBM patients and potential therapeutic targets. These results provide important new insights into the early diagnosis and the prognosis for GBM patients and introduce potential targets for GBM therapeutics. Fund Baylor Scott & White Health Startup Fund (E.W.); Collaborative Faculty Research Investment Program (CFRIP) of Baylor University, Baylor Scott & White Health, and Baylor College of Medicine (E.W., T.S., J.H.H.); NIH R01 NS067435 (J.H.H.); Scott & White Plummer Foundation Grant (J.H.H.); National Natural Science Foundation of China 816280007 (J.H.H. and Fu.W.).
Collapse
Affiliation(s)
- Fengfei Wang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Neurology, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA.
| | - Zheng Zheng
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Psychology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jitian Guan
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Shuang Zhou
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Xin Shen
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Anesthesiology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Fushun Wang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Psychology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14643, USA
| | - David Wenkert
- Department of Medicine, Division of Endocrinology, Baylor Scott & White Health, Temple, TX 76508, USA; Department of Medicine, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA
| | - Batool Kirmani
- Department of Neurology, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Neurology, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA
| | - Touradj Solouki
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76706, USA
| | - Ekokobe Fonkem
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Neurology, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA; LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Eric T Wong
- Brain Tumor Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA.
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA; LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutical Sciences, Texas A & M Health Science Center, College of Pharmacy, College Station, TX 77843, USA.
| |
Collapse
|
8
|
Li C, Zhang B, Lv W, Lai C, Chen Z, Wang R, Long X, Feng X. Triptolide inhibits cell growth and GRP78 protein expression but induces cell apoptosis in original and radioresistant NPC cells. Oncotarget 2018; 7:49588-49596. [PMID: 27391061 PMCID: PMC5226531 DOI: 10.18632/oncotarget.10412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 06/16/2016] [Indexed: 01/23/2023] Open
Abstract
The radioresistance is the key factor to hamper curative effect and survival of nasopharyngeal carcinoma (NPC) patients. Nature triptolide (TPL) has been found to circumvent drug-resistant effect of cancer, but its effect on NPC radioresistance has been rarely studied. In the present study, the 10 Gy-resistant CNE2 subclones (CNE2-SR) were used as a NPC radioresistant model. The IC50 of TPL in CNE2 and CNE2-SR cells was measured by MTT assay, cell cycle was analyzed by flow cytometry, and protein expression was examined by western blot. Our data showed that TPL treatment decreased the percentage of viable cells, and IC50 value in CNE2 and CNE2-SR cells was 23.6 ± 1.41 nmol/L and 31.2 ± 1.16 nmol/L, respectively. Six Gy was a moderate dosage of X-ray for CNE2, and 25 nM TPL was close to IC50 value of CNE2 and CNE2-SR. Six Gy X-ray and/or 25 nM TPL significantly inhibited tumor growth in nude mice. Furthermore, 6 Gy X-ray and/or 25 nM TPL significantly inhibited cell growth and induced cell apoptosis and M/G2 phase arrest in CNE2 and CNE2-SR cells. Moreover, TPL treatment significantly inhibited the expression of GRP78 protein in CNE2 and CNE2-SR cells. These results suggest that TPL may serve as a potential radiosensitizer agent for NPC treatment.
Collapse
Affiliation(s)
- Chengmin Li
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Bin Zhang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410008, China
| | - Wuwu Lv
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Chen Lai
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Zhikang Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Ran Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Xueying Long
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| | - Xueping Feng
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, China
| |
Collapse
|
9
|
Cheung So E, Lo YC, Chen LT, Kao CA, Wu SN. High effectiveness of triptolide, an active diterpenoid triepoxide, in suppressing Kir-channel currents from human glioma cells. Eur J Pharmacol 2014; 738:332-41. [DOI: 10.1016/j.ejphar.2014.05.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/23/2014] [Accepted: 05/23/2014] [Indexed: 11/17/2022]
|
10
|
Sai K, Li WY, Chen YS, Wang J, Guan S, Yang QY, Guo CC, Mou YG, Li WP, Chen ZP. Triptolide Synergistically Enhances Temozolomide-Induced Apoptosis and Potentiates Inhibition of NF-κB Signaling in Glioma Initiating Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:485-503. [DOI: 10.1142/s0192415x14500323] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Glioblastoma multiforme (GBM) is a lethal solid cancer in adults. Temozolomide (TMZ) is a first-line chemotherapeutic agent but the efficacy is limited by intrinsic and acquired resistance in GBM. Triptolide (TPL), a derivative from traditional Chinese medicine, demonstrated anti-tumor activity. In this study, we explored the interaction of TPL and TMZ in glioma-initiating cells (GICs) and the potential mechanism. A GIC line (GIC-1) was successfully established. Cell viability of GIC-1 after treatment was measured using a CCK-8 assay. The interaction between TPL and TMZ was calculated from Chou–Talalay equations and isobologram. Self-renewal was evaluated with tumor sphere formation assay. Apoptosis was assessed with flow cytometry and western blot. Luciferase assay was employed to measure NF-κB transcriptional activity. The expression of NF-κB downstream genes, NF-κB nuclear translocalization and phoshorylation of IκBα and p65 were evaluated using western blot. We found that GIC-1 cells were resistant to TMZ, with the expected IC50 of 705.7 μmol/L. Co-treatment with TPL yielded a more than three-fold dose reduction of TMZ. TPL significantly increased the percentage of apoptotic cells and suppressed the tumor sphere formation when combined with TMZ. Phosphorylation of IκBα and p65 coupled with NF-κB nuclear translocalization were notably inhibited after a combined treatment. Co-incubation synergistically repressed NF-κB transcriptional activity and downstream gene expression. TPL sensitizes GICs to TMZ by synergistically enhancing apoptosis, which is likely resulting from the augmented repression of NF-κB signaling. TPL is therefore a potential chemosensitizer in the treatment of GBM.
Collapse
Affiliation(s)
- Ke Sai
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Wen-Yu Li
- Guangzhou Medical University, Guangzhou 510182, China
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen 518029, China
| | - Yin-Sheng Chen
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Jian Wang
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Su Guan
- School of Bioscience and Bioengineering, South China University of Technology, Higher Education Mega Center, Guangzhou 510006, China
| | - Qun-Ying Yang
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Cheng-Cheng Guo
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Yong-Gao Mou
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Wei-Ping Li
- Guangzhou Medical University, Guangzhou 510182, China
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen 518029, China
| | - Zhong-Ping Chen
- Department of Neurosurgery/Neuro-Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- State Key Laboratory of Oncology in South China, Guangzhou, China
| |
Collapse
|
11
|
Nakazato T, Sagawa M, Kizaki M. Triptolide induces apoptotic cell death of multiple myeloma cells via transcriptional repression of Mcl-1. Int J Oncol 2014; 44:1131-8. [PMID: 24481531 DOI: 10.3892/ijo.2014.2280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 12/27/2013] [Indexed: 11/06/2022] Open
Abstract
Triptolide, a diterpenoid trioxide purified from the Chinese herb Tripterygium wilfordii Hook F, has been used as a natural medicine in China for hundreds of years. Several reports have demonstrated that triptolide inhibits the proliferation of cancer cells in vitro and reduces the growth of several types of tumors in vivo. To address the potential of triptolide as a novel therapeutic agent for patients with multiple myeloma, we investigated the effects of triptolide on the induction of apoptosis in human multiple myeloma cells in vitro. Triptolide rapidly induces apoptotic cell death in various myeloma cell lines. Triptolide-induced apoptosis in myeloma cells is associated with the loss of mitochondrial transmembrane potential (∆ψm), the release of cytochrome c and Smac/DIABLO from mitochondria into the cytosol, and the activation of caspase-3 and caspase-9. Furthermore, triptolide induces a rapid decline in the levels of Mcl-1 protein that correlates with caspase activation and induction of apoptosis. Inhibition of Mcl-1 synthesis by triptolide occurs at the level of mRNA transcription and is associated with an inhibition of phosphorylation of RNA polymerase II CTD. These results indicate that Mcl-1 is an important target for triptolide-induced apoptosis in myeloma cells that occurs via inhibition of Mcl-1 mRNA transcription coupled with rapid protein degradation through the ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Tomonori Nakazato
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Kanagawa 240-8550, Japan
| | - Morihiko Sagawa
- Division of Hematology, Keio University School of Medicine, Tokyo 160-0001, Japan
| | - Masahiro Kizaki
- Department of Hematology, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
| |
Collapse
|
12
|
Triptolide induces growth inhibition and apoptosis of human laryngocarcinoma cells by enhancing p53 activities and suppressing E6-mediated p53 degradation. PLoS One 2013; 8:e80784. [PMID: 24244715 PMCID: PMC3828261 DOI: 10.1371/journal.pone.0080784] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 10/07/2013] [Indexed: 12/20/2022] Open
Abstract
Triptolide, an active compound extracted from Chinese herb Leigongteng (Tripterygium wilfordii Hook F.), shows a broad-spectrum of anticancer activity through its cytotoxicity. However, the efficacy of triptolide on laryngocarcinoma rarely been evaluated, and the mechanism by which triptolide-induced cellular apoptosis is still not well understood. In this study, we found that triptolide significantly inhibited the laryngocarcinoma HEp-2 cells proliferation, migration and survivability. Triptolide induces HEp-2 cell cycle arrest at the G1 phase and apoptosis through intrinsic and extrinsic pathways since both caspase-8 and -9 are activated. Moreover, triptolide enhances p53 expression by increasing its stability via down-regulation of E6 and E6AP. Increased p53 transactivates down-stream target genes to initiate apoptosis. In addition, we found that short time treatment with triptolide induced DNA damage, which was consistent with the increase in p53. Furthermore, the cytotoxicity of triptolide is decreased by p53 knockdown or use of caspases inhibitor. In conclusion, our results demonstrated that triptolide inhibits cell proliferation and induces apoptosis in laryngocarcinoma cells by enhancing p53 expression and activating p53 functions through induction of DNA damage and suppression of E6 mediated p53 degradation. These studies indicate that triptolide is a potential anti-laryngocarcinoma drug.
Collapse
|
13
|
Triptolide-Mediated Apoptosis by Suppression of Focal Adhesion Kinase through Extrinsic and Intrinsic Pathways in Human Melanoma Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:172548. [PMID: 25945102 PMCID: PMC4402567 DOI: 10.1155/2013/172548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 03/29/2013] [Accepted: 04/07/2013] [Indexed: 02/03/2023]
Abstract
Triptolide (TPL) has been shown to inhibit cell proliferation and induce apoptosis in various human cancer cells; however, the precise mechanism of apoptosis induced by TPL in human melanoma cells has not yet been elucidated. In this study, we investigated the precise mechanism underlying cytocidal effects of TPL on human melanoma cells. Treatment of human melanoma cells with TPL significantly inhibited cell growth and induced apoptosis, as evidenced by flow cytometry and annexin V-fluorescein isothiocyanate analyses. TPL increased the levels of Fas and Fas-associated death domain (FADD) and induced cleavage of Bid by activation of caspase-8 and cytochrome c release from mitochondria to the cytosol, which resulted in activation of caspase-9 and caspase-3. Moreover, TPL-induced apoptosis in SK-MEL-2 cells was mediated through dephosphorylation of focal adhesion kinase (FAK) and its cleavage by caspase-8-mediated caspase-3 activation via upregulation of Fas expression. We also found that TPL mediated the dissociation of receptor-interacting protein (RIP) from FAK and enhanced the formation of RIP/Fas complex formation initiating cell death. In conclusion, our data firstly demonstrated that TPL induces apoptosis by both extrinsic and intrinsic apoptosis pathways in human melanoma cells and identified that RIP shuttles between Fas and FAK to mediate apoptosis.
Collapse
|
14
|
Zheng Y, Zhang WJ, Wang XM. Triptolide with potential medicinal value for diseases of the central nervous system. CNS Neurosci Ther 2012; 19:76-82. [PMID: 23253124 DOI: 10.1111/cns.12039] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 11/01/2012] [Accepted: 11/02/2012] [Indexed: 11/29/2022] Open
Abstract
Tripterygium wilfordii Hook.f. (TWHF) has a long history as a Traditional Chinese Medicine (TCM) herb that aids in treating inflammatory and autoimmune diseases. The major bioactive component of TWHF is triptolide, which has been recognized to possess a broad spectrum of biological profiles including antiinflammatory, immunosuppressive, antifertility, and antitumor activities, as well as neurotrophic and neuroprotective effects. Limitation of triptolide, such as poor water solubility and severe systemic toxicity, has postponed clinical development and trials; however, the wide range of medicinal value of triptolide has been drawing intensive worldwide attention. In particular, triptolide has been shown to have significant effects on central nervous system (CNS) diseases, such as Parkinson's disease, Alzheimer's disease, spinal cord and brain injury, and multiple sclerosis. This review focuses on the potential therapeutic role of triptolide on CNS diseases, and discusses the structural features, potential modifications, and the other pharmacological activities of triptolide.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Physiology, Capital Medical University, Beijing
| | | | | |
Collapse
|
15
|
Zhang H, Zhu W, Su X, Wu S, Lin Y, Li J, Wang Y, Chen J, Zhou Y, Qiu P, Yan G, Zhao S, Hu J, Zhang J. Triptolide inhibits proliferation and invasion of malignant glioma cells. J Neurooncol 2012; 109:53-62. [PMID: 22562416 DOI: 10.1007/s11060-012-0885-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 04/16/2012] [Indexed: 12/29/2022]
Abstract
Malignant glioma is the most devastating and aggressive tumor in brain, characterized by rapid proliferation and diffuse invasion. Chemotherapy and radiotherapy are the pivotal strategies after surgery; however, high drug resistance of malignant glioma and the blood-brain barrier usually render chemotherapy drugs ineffective. Here, we find that triptolide, a small molecule with high lipid solubility, is capable of inhibiting proliferation and invasion of malignant glioma cells effectively. In both investigated malignant glioma cell lines, triptolide repressed cell proliferation via inducing cell cycle arrest in G0/G1 phase, associated with downregulation of G0/G1 cell cycle regulators cyclin D1, CDK4, and CDK6 followed by reduced phosphorylation of retinoblastoma protein (Rb). In addition, triptolide induced morphological change of C6 cells through downregulation of protein expression of MAP-2 and inhibition of activities of GTPases Cdc42 and Rac1/2/3, thus significantly suppressing migratory and invasive capacity. Moreover, in an in vivo tumor model, triptolide delayed growth of malignant glioma xenografts. These findings suggest an important inhibitory action of triptolide on proliferation and invasion of malignant glioma, and encourage triptolide as a candidate for glioma therapy.
Collapse
Affiliation(s)
- Haipeng Zhang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, 74 Zhongshan Road II, Guangzhou, 510080, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Liu Y, Chen HL, Yang G. Extract of Tripterygium wilfordii Hook F Protect Dopaminergic Neurons Against Lipopolysaccharide-Induced Inflammatory Damage. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 38:801-14. [DOI: 10.1142/s0192415x10008251] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Activation of microglia is a critical pathological marker of Parkinson's disease. Activated microglia produces proinflammatory and neurotoxic factors, which cause neurons to induce neurodegeneration. Although it is believed that Chinese herbs, such as Tripterygium wilfordii Hook F, can ease inflammatory diseases, little is known about its benefit to neurodegenerative disease, like Parkinson's disease. In this study, we report the extract of Tripterygium wilfordii Hook F with a novel extraction method significantly protected dopaminergic neurons from LPS-induced degeneration in rat mesencephalic neuron-glia cultures. Cells pretreated with the extract have shown dose-dependent inhibition of LPS-induced TNFα and excessive NO production. More importantly, the total number of activated microglia was greatly reduced in these pretreated cells. Our results suggest that the extract of Tripterygium wilfordii Hook F has a strong bioactive function to diminish the pro-inflammatory factors, such as TNFα and NO. These data might also shed light for future neurodegenerative disease therapy.
Collapse
Affiliation(s)
- Yuxin Liu
- Laboratory of Cell Pharmacology, College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei Province, 071002, P. R. China
| | - Hui-Ling Chen
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, D.C. 20010, USA
| | - Gengliang Yang
- Drug Quality Control Key Laboratory of Hebei Province, Baoding, Hebei Province, 071002, P. R. China
| |
Collapse
|
17
|
Triptolide induces cell-cycle arrest and apoptosis of human multiple myeloma cells in vitro via altering expression of histone demethylase LSD1 and JMJD2B. Acta Pharmacol Sin 2012; 33:109-19. [PMID: 22120968 DOI: 10.1038/aps.2011.145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM To elucidate the relationship between triptolide-induced changes in histone methylation and its antitumor effect on human multiple myeloma (MM) cells in vitro. METHODS Human multiple myeloma cell line RPMI8226 was used. Apoptosis was evaluated using Annexin-V-FITC/PI-labeled flow cytometry, Hoechst 33258 staining, and transmission electron microscopy. Flow cytometry was used to detect the cell cycle distribution of the apoptotic cells. The presence of the LSD1, JMJD2B, H3K4me2, H3K9me2, and H3K36me2 proteins was verified by Western blot analysis. Semi-quantitative real-time PCR was performed to examine the expression of LSD1 and JMJD2B. RESULTS Triptolide (10-160 nmol/L) suppressed the proliferation of MM cells in a dose- and time-dependent manner with an IC(50) value of 99.2 ± 9.0 nmol/L at 24 h. Triptolide (50 nmol/L) induced G(0)/G(1) cell cycle arrest in MM cells. The agent (50-150 nmol/L) induced apoptosis of MM cells in a dose-dependent manner. The same concentrations of triptolide suppressed the expression of dimethylated H3K4, dimethylated H3K9 and dimethylated H3K36 by altering the expression of histone demethylase LSD1 and JMJD2B without affecting the expression of histone demethylase LSD1. CONCLUSION Triptolide potently inhibits the growth of MM cells via regulating the expression of histone demethylase LSD1 and JMJD2B, which lead to abnormal histone methylation.
Collapse
|
18
|
Liu Q. Triptolide and its expanding multiple pharmacological functions. Int Immunopharmacol 2011; 11:377-83. [PMID: 21255694 DOI: 10.1016/j.intimp.2011.01.012] [Citation(s) in RCA: 256] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 01/11/2011] [Indexed: 01/14/2023]
Abstract
Triptolide, a diterpene triepoxide, is a major active component of extracts derived from the medicinal plant Tripterygium wilfordii Hook F (TWHF). Triptolide has multiple pharmacological activities including anti-inflammatory, immune modulation, antiproliferative and proapoptotic activity. So, triptolide has been widely used to treat inflammatory diseases, autoimmune diseases, organ transplantation and even tumors. Triptolide cannot only induce tumor cell apoptosis directly, but can also enhance apoptosis induced by cytotoxic agents such as TNF-α, TRAIL and chemotherapeutic agents regardless of p53 phenotype by inhibiting NFκB activation. Recently, the cellular targets of triptolide, such as MKP-1, HSP, 5-Lox, RNA polymerase and histone methyl-transferases had been demonstrated. However, the clinical use of triptolide is often limited by its severe toxicity and water-insolubility. New water-soluble triptolide derivatives have been designed and synthesized, such as PG490-88 or F60008, which have been shown to be safe and potent antitumor agent. Importantly, PG490-88 has been approved entry into Phase I clinical trial for treatment of prostate cancer in USA. This review will focus on these breakthrough findings of triptolide and its implications.
Collapse
Affiliation(s)
- Qiuyan Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China.
| |
Collapse
|
19
|
Zhao F, Zeng LL, Chen Y, Li R, Liu Y, Wen L, Cheng YQ, Zhang C. Effects of triptolide on histone acetylation and HDAC8 expression in multiple myeloma in vitro. Chin J Cancer Res 2010. [DOI: 10.1007/s11670-010-0148-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
20
|
Zhao F, Chen Y, Zeng LL, Li R, Zeng R, Wen L, Liu Y, Zhang C. Effects of triptolide on RIZ1 expression, proliferation, and apoptosis in multiple myeloma U266 cells. Acta Pharmacol Sin 2010; 31:733-40. [PMID: 20523343 DOI: 10.1038/aps.2010.49] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM To investigate the effects of triptolide on proliferation and apoptosis as well as on the expression of RIZ1 in the human multiple myeloma cell line U266 in vitro. METHODS The effect of triptolide on the growth of U266 cells was studied by MTT assay. Apoptosis was detected by Hoechst 33258 staining and Annexin V/PI double-labeled flow cytometry, and caspase-3 mRNA was measured by RT-PCR. Western blotting, flow cytometry and RT-PCR were used to assess the expression of RIZ1, and the location and expression of H3K9me1 were detected by confocal microscopy and Western blotting. RESULTS Triptolide significantly inhibited the proliferation of U266 cells in a time- and concentration-dependent manner (the IC(50) value for a 24-h exposure was 157.19+/-0.38 nmol/L). Triptolide induced typical apoptotic morphological changes. Triptolide 40, 80, and 160 nmol/L treatment induced significant caspase-3-dependent apoptosis compared with control group (10.5%+/-1.23%, 37.9%+/-2.45%, and 40.5%+/-2.30% vs 3.8%+/-1.98%, P<0.05). Compared with peripheral blood monocular cells (PBMC) from healthy donors, the protein expression of RIZ1 in U266 cells was relatively low, but the mRNA and protein expression of RIZ1 were strikingly increased by triptolide in a concentration-dependent manner. Triptolide increased the protein expression of RIZ1 and RIZ1 methylates histone H3 lysine 9 in U266 cells. CONCLUSION Triptolide increased the protein expression of RIZ1, inhibited the proliferation, and induced caspase-dependent apoptosis in U266 cells.
Collapse
|
21
|
Zhao F, Chen Y, Li R, Liu Y, Wen L, Zhang C. Triptolide alters histone H3K9 and H3K27 methylation state and induces G0/G1 arrest and caspase-dependent apoptosis in multiple myeloma in vitro. Toxicology 2010; 267:70-9. [DOI: 10.1016/j.tox.2009.10.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2009] [Revised: 10/17/2009] [Accepted: 10/19/2009] [Indexed: 12/15/2022]
|
22
|
Chen YW, Lin GJ, Chia WT, Lin CK, Chuang YP, Sytwu HK. Triptolide exerts anti-tumor effect on oral cancer and KB cells in vitro and in vivo. Oral Oncol 2009; 45:562-8. [PMID: 19359213 DOI: 10.1016/j.oraloncology.2008.10.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 10/07/2008] [Accepted: 10/08/2008] [Indexed: 01/11/2023]
Abstract
Triptolide (TPL), a diterpenoid triepoxide purified from the Chinese herb Tripterygium wilfordii Hook F, has been reported to potentiate the anti-tumor effect in various cancer cells. However, the effect of TPL on oral cancers is not yet evaluated. Herein we first demonstrate that TPL induces prominent growth inhibition and apoptosis in two oral cancer cell lines, SCC25 and OEC-M1 and in KB cells. Our results indicate that TPL induces a dose-dependent apoptosis of these cells at nanomolar concentration. Apoptosis signalings are both activated through time upon TPL treatment detected by elevated caspase-3, 8, 9 activities. In xenograft tumor mouse model, TPL injection successfully inhibits the tumor growth via apoptosis induction which was demonstrated by TUNEL assay. These results demonstrate that TPL exerts anti-tumor effect on oral cancer and KB cells and suggest further the potential of TPL combining with other chemotherapeutic agents or radiotherapy for advanced oral cancer.
Collapse
Affiliation(s)
- Yuan-Wu Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, No. 161, Section 6, Min-Chuan East Road, Neihu 114, Taipei 114, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
The recent article by Zhou et al was highly interesting and thought provoking. The authors have clearly shown that triptolide administration is associated with up-regulation of the Bax gene, resulting in an attenuating effect on cell growth in gastrointestinal malignancies such as pancreatic carcinomas. The article by Zhou et al is all the more important because it highlights the rapidly increasing role of triplodide in the management of systemic malignancies. For instance, triptolide acts on the PI3K/Akt/NF-κB pathway, thereby enhancing apoptosis secondary to the administration of bortezomib in multiple myeloma cells. Similar synergisms are seen when triptolide is administered along with 5-fluoruracil for the management of colonic carcinomas. Similarly, triptolide causes down-regulation of the Bcl-2 gene, resulting in control of cell growth in tumors, such as glioblastoma multiformes.
Collapse
|
24
|
Zhou GX, Ding XL, Huang JF, Zhang H, Wu SB, Cheng JP, Wei Q. Apoptosis of human pancreatic cancer cells induced by Triptolide. World J Gastroenterol 2008; 14:1504-9. [PMID: 18330938 PMCID: PMC2693742 DOI: 10.3748/wjg.14.1504] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM: To investigate apoptosis in human pancreatic cancer cells induced by Triptolide (TL), and the relationship between this apoptosis and expression of caspase-3’ bcl-2 and bax.
METHODS: Human pancreatic cancer cell line SW1990 was cultured in DMEM media for this study. MTT assay was used to determine the cell growth inhibitory rate in vitro. Flow cytometry and TUNEL assay were used to detect the apoptosis of human pancreatic cancer cells before and after TL treatment. RT-PCR was used to detect the expression of apoptosis-associated gene caspase-3’ bcl-2 and bax.
RESULTS: TL inhibited the growth of human pancreatic cancer cells in a dose-and time-dependent manner. TL induced human pancreatic cancer cells to undergo apoptosis with typically apoptotic characteristics. TUNEL assay showed that after the treatment of human pancreatic cancer cells with 40 ng/mL TL for 12 h and 24 h, the apoptotic rates of human pancreatic cancer cells increased significantly. RT-PCR demonstrated that caspase-3 and bax were significantly up-regulated in SW1990 cells treated with TL while bcl-2 mRNA was not.
CONCLUSION: TL is able to induce the apoptosis in human pancreatic cancer cells. This apoptosis may be mediated by up-regulating the expression of apoptosis-associated caspase-3 and bax gene.
Collapse
|