1
|
Gorrie D, Bravo M, Fan L. The Yin and Yang of the Natural Product Triptolide and Its Interactions with XPB, an Essential Protein for Gene Expression and DNA Repair. Genes (Basel) 2024; 15:1287. [PMID: 39457411 PMCID: PMC11507457 DOI: 10.3390/genes15101287] [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: 08/31/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
Triptolide, a bioactive diterpene tri-epoxide extracted from Tripterygium wilfordii Hook F (TWHF), exhibits notable pharmacological activities, including anti-inflammatory, immunosuppressive, antifertility, and anticancer effects. Despite its promising therapeutic potential, clinical applications of triptolide are significantly limited by its poor water solubility and substantial toxicity, particularly hepatotoxicity, nephrotoxicity, and cardiotoxicity. These toxic effects are difficult to separate from many of its desired therapeutic effects, the Yin and Yang of triptolide applications. Triptolide's therapeutic and toxic effects are linked to its inhibitory interactions with XPB, a DNA helicase essential for transcription by RNA polymerase II (RNAPII) and nucleotide excision repair (NER). By irreversibly binding to XPB, triptolide inhibits its ATPase activity, leading to global repression of transcription and impaired NER, which underlies its cytotoxic and antitumor properties. Recent developments, including triptolide prodrugs such as Minnelide and derivatives like glutriptolides, aim to enhance its pharmacokinetic properties and reduce toxicity. This review critically examines triptolide's chemical structure, therapeutic applications, toxicological profile, and molecular interactions with XPB and other protein targets to inform future strategies that maximize therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
| | | | - Li Fan
- Department of Biochemistry, University of California, 900 University Ave, Riverside, CA 92521, USA; (D.G.); (M.B.)
| |
Collapse
|
2
|
Yuan L, Jiang X, Jia G, Li Z, Wang M, Hu S, Yang J, Liang F, Zhang F, Gao L, Gao N. Minnelide exhibits antileukemic activity by targeting the Ars2/miR-190a-3p axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155724. [PMID: 38759317 DOI: 10.1016/j.phymed.2024.155724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/25/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND The identification of a novel and effective strategy for the clinical treatment of acute leukemia (AL) is a long-term goal. Minnelide, a water-soluble prodrug of triptolide, has recently been evaluated in phase I and II clinical trials in patients with multiple cancers and has shown promise as an antileukemic agent. However, the molecular mechanism underlying minnelide's antileukemic activity remains unclear. PURPOSE To explore the molecular mechanisms by which minnelide exhibits antileukemic activity. METHODS AL cells, primary human leukemia cells, and a xenograft mouse model were treated with triptolide and minnelide. The molecular mechanism was elucidated using western blotting, immunoprecipitation, flow cytometry, GSEA and liquid chromatography-mass spectrometry analysis. RESULTS Minnelide was highly effective in inhibiting leukemogenesis and improving survival in two complementary AL mouse models. Triptolide, an active form of minnelide, causes cell cycle arrest in G1 phase and induces apoptosis in both human AL cell lines and primary AL cells. Mechanistically, we identified Ars2 as a new chemotherapeutic target of minnelide for AL treatment. We found that triptolide directly targeted Ars2, resulting in the downregulation of miR-190a-3p, which led to the disturbance of PTEN/Akt signaling and culminated in G1 cell cycle arrest and apoptosis. CONCLUSIONS Our findings demonstrate that targeting Ars2/miR-190a-3p signaling using minnelide could represent a novel chemotherapeutic strategy for AL treatment and support the evaluation of minnelide for the treatment of AL in clinical trials.
Collapse
Affiliation(s)
- Liang Yuan
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, PR China
| | - Xiuxing Jiang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, PR China
| | - Guanfei Jia
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, PR China
| | - Zhiqiang Li
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing 400038, PR China
| | - Mei Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, PR China
| | - Siyi Hu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, PR China
| | - Jiawang Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, PR China
| | - Feng Liang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, PR China
| | - Fenglin Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, PR China
| | - Lu Gao
- Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, PR China.
| | - Ning Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563006, PR China.
| |
Collapse
|
3
|
Pan W, Yang B, He D, Chen L, Fu C. Functions and targets of miRNAs in pharmacological and toxicological effects of major components of Tripterygium wilfordii Hook F. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1997-2019. [PMID: 37831113 DOI: 10.1007/s00210-023-02764-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
Tripterygium wilfordii Hook F (TwHF) has a long history of use as a traditional Chinese medicine and has been widely administered to treat various inflammatory and autoimmune diseases. MicroRNAs (miRNAs) are endogenous, short, non-coding RNAs that regulate gene expression post-transcriptionally. They participate in the efficacies and even toxicities of the components of TwHF, rendering miRNAs an appealing therapeutic strategy. This review summarizes the recent literature related to the roles and mechanisms of miRNAs in the pharmacological and toxicological effects of main components of TwHF, focusing on two active compounds, triptolide (TP) and celastrol (CEL). Additionally, the prospects for the "You Gu Wu Yun" theory regarding TwHF nephrotoxicity are presented.
Collapse
Affiliation(s)
- Wei Pan
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Bo Yang
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Dongxiu He
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China
| | - Chengxiao Fu
- Institute of Pharmacy and Pharmacology, College of Basic Medical Science, Hengyang Medical School, University of South China, Hengyang, 421200, Hunan, People's Republic of China.
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
4
|
AbdulHussein AH, Al-Taee MM, Radih ZA, Aljuboory DS, Mohammed ZQ, Hashesh TS, Riadi Y, Hadrawi SK, Najafi M. Mechanisms of cancer cell death induction by triptolide. Biofactors 2023; 49:718-735. [PMID: 36876465 DOI: 10.1002/biof.1944] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/07/2023]
Abstract
Drug resistance is a hot topic issue in cancer research and therapy. Although cancer therapy including radiotherapy and anti-cancer drugs can kill malignant cells within the tumor, cancer cells can develop a wide range of mechanisms to resist the toxic effects of anti-cancer agents. Cancer cells may provide some mechanisms to resist oxidative stress and escape from apoptosis and attack by the immune system. Furthermore, cancer cells may resist senescence, pyroptosis, ferroptosis, necroptosis, and autophagic cell death by modulating several critical genes. The development of these mechanisms leads to resistance to anti-cancer drugs and also radiotherapy. Resistance to therapy can increase mortality and reduce survival following cancer therapy. Thus, overcoming mechanisms of resistance to cell death in malignant cells can facilitate tumor elimination and increase the efficiency of anti-cancer therapy. Natural-derived molecules are intriguing agents that may be suggested to be used as an adjuvant in combination with other anticancer drugs or radiotherapy to sensitize cancer cells to therapy with at least side effects. This paper aims to review the potential of triptolide for inducing various types of cell death in cancer cells. We review the induction or resistance to different cell death mechanisms such as apoptosis, autophagic cell death, senescence, pyroptosis, ferroptosis, and necrosis following the administration of triptolide. We also review the safety and future perspectives for triptolide and its derivatives in experimental and human studies. The anticancer potential of triptolide and its derivatives may make them effective adjuvants for enhancing tumor suppression in combination with anticancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Salema K Hadrawi
- Refrigeration and Air-Conditioning Technical Engineering Department, College of Technical Engineering, The Islamic University, Najaf, Iraq
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
5
|
Wu J, Deng Y, Zhang X, Ma J, Zheng X, Chen Y. Suchilactone inhibits the growth of acute myeloid leukaemia by inactivating SHP2. PHARMACEUTICAL BIOLOGY 2022; 60:144-153. [PMID: 34962431 PMCID: PMC8725822 DOI: 10.1080/13880209.2021.2017467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 12/07/2021] [Indexed: 05/12/2023]
Abstract
CONTEXT Suchilactone, a lignan compound extracted from Monsonia angustifolia E.Mey. ex A.Rich. (Geraniaceae), has little research on pharmacological activity; whether suchilactone has inhibitory effect on acute myeloid leukaemia (AML) is unclear. OBJECTIVE To investigate the antitumor effect of suchilactone and its mechanism in AML. MATERIALS AND METHODS The effects of suchilactone on cell growth were detected by CCK-8 and flow cytometry. Network pharmacology was conducted to explore target of suchilactone. Gene expression was detected by western blot and RT-PCR. SHI-1 cells (1 × 106 cell per mouse) were subcutaneously inoculated into the female SCID mice. Suchilactone (15 and 30 mg/kg) was dissolved in PBS with 0.5% carboxymethylcellulose sodium and administered (i.g.) to mice once a day for 19 days, while the control group received PBS with 0.5% carboxymethylcellulose sodium. Tumour tissues were stained with Ki-67 and TUNEL. RESULTS Suchilactone exerted an effective inhibition on the growth of SHI-1 cells with IC50 of 17.01 μM. Then, we found that suchilactone binds to the SHP2 protein and inhibits its activation, and suchilactone interacted with SHP2 to inhibit cell proliferation and promote cell apoptosis via blocking the activation of SHP2. Moreover, Suchilaction inhibited tumour growth of AML xenografts in mice, as the tumour weight decreased from 0.618 g (control) to 0.35 g (15 mg/kg) and 0.258 g (30 mg/kg). Suchilactone inhibited Ki-67 expression and increased TUNEL expression in tumour tissue. DISCUSSION AND CONCLUSIONS Our study is the first to demonstrate suchilactone inhibits AML growth, suggesting that suchilactone is a candidate drug for the treatment of AML.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Mice
- Antineoplastic Agents, Phytogenic/administration & dosage
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Geraniaceae/chemistry
- Leukemia, Myeloid, Acute/drug therapy
- Mice, Inbred BALB C
- Mice, SCID
- Network Pharmacology
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai’an, China
| | - Yuan Deng
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai’an, China
| | - Xin Zhang
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai’an, China
| | - Jingjing Ma
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai’an, China
| | - Xinqi Zheng
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai’an, China
| | - Yue Chen
- Department of Hematology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai’an, China
| |
Collapse
|
6
|
Du Y, Geng G, Zhao C, Gao T, Wei B. LncRNA MEG3 promotes cisplatin sensitivity of cervical cancer cells by regulating the miR-21/PTEN axis. BMC Cancer 2022; 22:1145. [DOI: 10.1186/s12885-022-10188-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022] Open
Abstract
Abstract
Background
Cervical cancer (CC) is a common gynecological malignancy worldwide. Some patients perform serious resistance after chemotherapy, and long-stranded non-coding RNA MEG3 is reported to be involved in the regulation of chemoresistance in many solid tumors. However, its involvement in cervical adenocarcinoma has not been reported.
Methods
Hela cell lines, cisplatin-resistant cell lines (Hela-CR) and nude mice were used in this study. After MEG3 was overexpressed or knocked down in cells by the lentivirus vector, cell growth was detected by the CCK-8 assay, and cell migration was evaluated using Transwell assay. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to examine the expression of MEG3, miR-21 and PTEN mRNA. Apoptosis was detected by flow cytometry. The targeting relationship between mRNAs was predicted and verified using dual-luciferase reporter gene experiments. Western blot was executed to examine Bax, cleaved-caspase 3, Bcl-2, PTEN and GAPDH expression. Cells were injected into the mice to form xenograft tumors to compare tumorigenesis capacity.
Results
We demonstrated that MEG3 was down-regulated in cervical cancer by analyzing the TCGA database. Moreover, knockdown of MEG3 promoted CC cell proliferation, migration and inhibited the apoptosis. These changes of CC cells were more pronounced under cisplatin treatment. Further studies showed that the MEG3/miR-21/PTEN axis affected cisplatin sensitivity in cervical cancer cells, and these results of recue assay were used to confirm this conclusion.
Conclusions
MEG3 performing as ceRNA promotes cisplatin sensitivity in CC cells through the miR-21/PTEN axis.
Collapse
|
7
|
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: 2.5] [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
|
8
|
Ganesan M, Kanimozhi G, Pradhapsingh B, Khan HA, Alhomida AS, Ekhzaimy A, Brindha GR, Prasad NR. Phytochemicals reverse P-glycoprotein mediated multidrug resistance via signal transduction pathways. Biomed Pharmacother 2021; 139:111632. [PMID: 34243600 DOI: 10.1016/j.biopha.2021.111632] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
P-glycoprotein, encoded by ATP-binding cassette transporters B1 gene (ABCB1), renders multidrug resistance (MDR) during cancer chemotherapy. Several synthetic small molecule inhibitors affect P-glycoprotein (P-gp) transport function in MDR tumor cells. However, inhibition of P-gp transport function adversely accumulates chemotherapeutic drugs in non-target normal tissues. Moreover, most small-molecule P-gp inhibitors failed in the clinical trials due to the low therapeutic window at the maximum tolerated dose. Therefore, downregulation of ABCB1-gene expression (P-gp) in tumor tissues seems to be a novel approach rather than inhibiting its transport function for the reversal of multidrug resistance (MDR). Several plant-derived phytochemicals modulate various signal transduction pathways and inhibit translocation of transcription factors, thereby reverses P-gp mediated MDR in tumor cells. Therefore, phytochemicals may be considered an alternative to synthetic small molecule P-gp inhibitors for the reversal of MDR in cancer cells. This review discussed the role of natural phytochemicals that modulate ABCB1 expression through various signal transduction pathways in MDR cancer cells. Therefore, modulating the cell signaling pathways by phytochemicals might play crucial roles in modulating ABCB1 gene expression and the reversal of MDR.
Collapse
Affiliation(s)
- M Ganesan
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - G Kanimozhi
- Department of Biochemistry, Dharmapuram Gnanambigai Government Arts College for Women, Mayiladuthurai, Tamil Nadu, India
| | - B Pradhapsingh
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S Alhomida
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Aishah Ekhzaimy
- Division of Endocrinology, Department of Medicine, King Khalid University Hospital, Riyadh 12372, Saudi Arabia
| | - G R Brindha
- School of Computing, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| | - N Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamil Nadu, India.
| |
Collapse
|
9
|
Zhou K, Chang Y, Han B, Li R, Wei Y. MicroRNAs as crucial mediators in the pharmacological activities of triptolide (Review). Exp Ther Med 2021; 21:499. [PMID: 33791008 PMCID: PMC8005665 DOI: 10.3892/etm.2021.9930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
Triptolide is the main bioactive constituent isolated from the Chinese herb Tripterygium wilfordii Hook F., which possesses a variety of pharmacological properties. MicroRNAs (miRNAs/miRs) are short non-coding RNAs that regulate gene expression post-transcriptionally. miRNAs are implicated in several intracellular processes, whereby their dysregulation contributes to pathogenesis of various diseases. Thus, miRNAs have great potential as biomarkers and therapeutic targets for diseases, and are implicated in drug treatment. Previous studies have reported that specific miRNAs are targeted, and their expression levels can be altered following exposure to triptolide. Thus, miRNAs are emerging as crucial mediators in the pharmacological activities of triptolide. The present review summarizes current literature on miRNAs as target molecules in the pharmacological activities of triptolide, including antitumor, anti-inflammatory, immunosuppressive, renal protective, cardioprotective, antiangiogenesis activities and multiorgan toxicity effects. In addition, the diverse signaling pathways involved are discussed to provide a comprehensive understanding of the underlying molecular mechanisms of triptolide in the regulation of target miRNAs.
Collapse
Affiliation(s)
- Kun Zhou
- Shanxi Institute of Energy, Taiyuan, Shanxi 030600, P.R. China
| | - Yinxia Chang
- College of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, P.R. China
| | - Bo Han
- College of Basic Medicine, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, P.R. China
| | - Rui Li
- College of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, P.R. China
| | - Yanming Wei
- College of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, P.R. China
| |
Collapse
|
10
|
Peixoto da Silva S, Caires HR, Bergantim R, Guimarães JE, Vasconcelos MH. miRNAs mediated drug resistance in hematological malignancies. Semin Cancer Biol 2021; 83:283-302. [PMID: 33757848 DOI: 10.1016/j.semcancer.2021.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/11/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022]
Abstract
Despite improvements in the therapeutic approaches for hematological malignancies in the last decades, refractory disease still occurs, and cancer drug resistance still remains a major hurdle in the clinical management of these cancer patients. The investigation of this problem has been extensive and different mechanism and molecules have been associated with drug resistance. MicroRNAs (miRNAs) have been described as having an important action in the emergence of cancer, including hematological tumors, and as being major players in their progression, aggressiveness and response to treatments. Moreover, miRNAs have been strongly associated with cancer drug resistance and with the modulation of the sensitivity of cancer cells to a wide array of anticancer drugs. Furthermore, this role has also been reported for miRNAs packaged into extracellular vesicles (EVs-miRNAs), which in turn have been described as essential for the horizontal transfer of drug resistance to sensitive cells. Several studies have been suggesting the use of miRNAs as biomarkers for drug response and clinical outcome prediction, as well as promising therapeutic tools in hematological diseases. Indeed, the combination of miRNA-based therapeutic tools with conventional drugs contributes to overcome drug resistance. This review addresses the role of miRNAs in the pathogenesis of hematological malignances, namely multiple myeloma, leukemias and lymphomas, highlighting their important action (either in their cell-free circulating form or within circulating EVs) in drug resistance and their potential clinical applications.
Collapse
Affiliation(s)
- Sara Peixoto da Silva
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal
| | - Hugo R Caires
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal
| | - Rui Bergantim
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal; Clinical Hematology, Hospital São João, 4200-319, Porto, Portugal; Clinical Hematology, FMUP - Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | - José E Guimarães
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal; Clinical Hematology, FMUP - Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal; Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário, IUCSCESPU, 4585-116, Gandra, Paredes, Portugal
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
11
|
Peng F, Xie X, Peng C. Chinese Herbal Medicine-Based Cancer Therapy: Novel Anticancer Agents Targeting MicroRNAs to Regulate Tumor Growth and Metastasis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1711-1735. [PMID: 31801358 DOI: 10.1142/s0192415x19500873] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
MicroRNAs, small non-coding RNA molecules, have gained a reputation of the most substantial regulators in gene network with the ability to down-regulate their targets. Accumulating evidence shifted insight toward microRNAs regulation as the key element of cancer initiation, development, and aggression. Recent studies have attached the importance of traditional Chinese medicine (TCM) to the treatment of various cancers, and the functional natural compounds have been considered as novel anticancer agents to directly inhibit tumor progression. In more recent decades, a wide range of biologically active components of TCM has gained increasing attention to their applications in the modulation of microRNAs. This review is on the purpose of demonstrating the significance of TCM bioactive ingredients in microRNAs regulation for cancer treatment according to the reports mainly in the recent six years, providing the evidence of efficient Chinese herbal medicine-based therapy and effective pro-diagnosis focusing on microRNAs expression of cancer patients.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, P. R. China
| | - Xiaofang Xie
- Chengdu University of Traditional Chinese Medicine, Chengdu, P. R. China.,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Sichuan Province and Ministry of Science and Technology, Chengdu, P. R. China
| | - Cheng Peng
- Chengdu University of Traditional Chinese Medicine, Chengdu, P. R. China.,State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Sichuan Province and Ministry of Science and Technology, Chengdu, P. R. China
| |
Collapse
|
12
|
Yao C, Li H, Zhang W. Triptolide inhibits benign prostatic epithelium viability and migration and induces apoptosis via upregulation of microRNA-218. Int J Immunopathol Pharmacol 2019; 32:2058738418812349. [PMID: 30453799 PMCID: PMC6247479 DOI: 10.1177/2058738418812349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Benign prostatic hypertrophy (BPH) has become a troublesome disease for elder
men. Triptolide (TPL) has been reported to be a potential anticancer agent.
However, the potential effects of TPL on BPH have not been shown out. BPH-1
cells were treated with different concentrations of TPL and/or transfected with
microRNA-218 (miR-218) inhibitor, pc-survivin, sh-survivin, or their
corresponding controls (NC). Thereafter, cell viability was determined by CCK-8
assay. Cell migration was accessed by modified two-chamber migration assay. Cell
apoptosis was checked by propidium iodide (PI) and fluorescein isothiocyanate
(FITC)-conjugated Annexin V staining. In addition, messenger RNA (mRNA) and
protein levels were detected using quantitative real-time polymerase chain
reaction (qRT-PCR) and western blot analysis, respectively. BPH-1 cell viability
and migration were significantly decreased, while cell apoptosis and expression
of miR-218 were statistically enhanced by TPL (P < 0.05 or
P < 0.01). However, downregulation of miR-218 increased
cell viability and migration, while decreased cell apoptosis compared with the
negative control group (P < 0.05 or
P < 0.01). Furthermore, the expression of cell cycle–related
proteins and cell apoptosis–related proteins were also led to the opposite
results with NC. In addition, we found that miR-218 negatively regulated the
expression of survivin (P < 0.01) and suppression of
survivin significantly enhanced cell apoptosis (P < 0.01).
Moreover, the results demonstrated that TPL could inactivate mammalian target of
rapamycin (mTOR) pathway, while inhibition of miR-218 alleviated the effects.
TPL inhibits viability and migration of BPH-1 cells and induces cell apoptosis
and also inactivates mTOR signal pathway via upregulation of miR-218. This study
provides evidence for the further studies representing triptolide as a potential
agent in the treatment of human BPH.
Collapse
Affiliation(s)
- Changlei Yao
- 1 Department of Urinary Surgery, People's Hospital of Rizhao, Rizhao, China
| | - Hongfa Li
- 1 Department of Urinary Surgery, People's Hospital of Rizhao, Rizhao, China
| | - Weitao Zhang
- 2 Department of Urinary Surgery, Affiliated Hospital of Taishan Medical University, Taian, China
| |
Collapse
|
13
|
Zhu J, Wang H, Chen F, Lv H, Xu Z, Fu J, Hou Y, Xu Y, Pi J. Triptolide enhances chemotherapeutic efficacy of antitumor drugs in non-small-cell lung cancer cells by inhibiting Nrf2-ARE activity. Toxicol Appl Pharmacol 2018; 358:1-9. [PMID: 30196066 DOI: 10.1016/j.taap.2018.09.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 12/20/2022]
Abstract
Non-small cell lung cancer (NSCLC) has a high mortality rate worldwide. Various treatments strategies have been used against NSCLC including individualized chemotherapies, but innate or acquired cancer cell drug resistance remains a major obstacle. Recent studies revealed that the Kelch-like ECH associated protein 1/Nuclear factor erythroid 2-related factor 2 (Keap1/Nrf2) pathway is intimately involved in cancer progression and chemoresistance. Thus, antagonizing Nrf2 would seem to be a viable strategy in cancer therapy. In the present study a traditional Chinese medicine, triptolide, was identified that markedly inhibited expression and transcriptional activity of Nrf2 in various cancer cells, including NSCLC and liver cancer cells. Consequently, triptolide made cancer cells more chemosensitivity toward antitumor drugs both in vitro and in a xenograft tumor model system using lung carcinoma cells. These results suggest that triptolide blocks chemoresistance in cancer cells by targeting the Nrf2 pathway. Triptolide should be further investigated in clinical cancer trials.
Collapse
Affiliation(s)
- Jiayu Zhu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China
| | - Huihui Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China.
| | - Feng Chen
- Department of Interventional Radiology, The First Affiliated Hospital of China Medical University, No. 155 Nanjing North Road, Heping Area, Shenyang, 110001, China; Interventional Department, Qianfoshan Hospital, Shandong University, No.16766 Jingshi Road, Jinan 250014, China
| | - Hang Lv
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China
| | - Zijin Xu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China
| | - Yongyong Hou
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China
| | - Yuanyuan Xu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New area, Shenyang 110122, China.
| |
Collapse
|
14
|
Broad targeting of triptolide to resistance and sensitization for cancer therapy. Biomed Pharmacother 2018; 104:771-780. [DOI: 10.1016/j.biopha.2018.05.088] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/06/2018] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
|
15
|
Ding B, Wahid MA, Wang Z, Xie C, Thakkar A, Prabhu S, Wang J. Triptolide and celastrol loaded silk fibroin nanoparticles show synergistic effect against human pancreatic cancer cells. NANOSCALE 2017; 9:11739-11753. [PMID: 28782773 PMCID: PMC5648537 DOI: 10.1039/c7nr03016a] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Pancreatic cancer is a lethal disease with a dreadful 5-year survival rate of only 5%. In spite of several treatment options, the prognosis still remains extremely poor. Therefore, novel therapy strategies with combinations of drugs are urgently required to combat this fatal disease. Triptolide (TPL) and celastrol (CL), two main compounds in traditional Chinese medicine isolated from Thunder God Vine, have a broad range of bioactivities including anticancer activity. Silk fibroin (SF), a naturally occurring protein with several unique properties, is an ideal carrier material. In this study, we prepared TPL and CL loaded silk fibroin nanoparticles (TPL-SFNPs and CL-SFNPs) by a modified desolvation method and evaluated their synergistic effects against human pancreatic cancer cells. Both SFNPs were characterized for particle size and zeta potential. The entrapment efficiency, drug loading, and drug release profiles were evaluated by HPLC. The cytotoxicity and synergistic effect of SFNPs were investigated in MIA PaCa-2 and PANC-1 human pancreatic cells. The results showed that the particle sizes of TPL-SFNPs and CL-SFNPs were 166.4 ± 4.6 nm and 170.4 ± 2.3 nm, with a mean zeta potential -27.2 ± 2.0 mV and -25.5 ± 2.57 mV, respectively. TPL-SFNPs and CL-SFNPs have a drug loading of 57.0 ± 4.7 μg mg-1 and 63.5 ± 3.8 μg mg-1 along with an encapsulation efficiency of 81.8 ± 2.8% and 87.0 ± 5.1%, respectively. Drug release studies revealed that a rapid release of the drugs from SFNPs was observed at pH 4.5 (lysosomal pH) and a delayed release was observed at pH 7.4 (plasma pH). TPL-SFNPs (IC50 3.80 and 4.75 nM) and CL-SFNPs (IC50 0.38 and 0.64 μM) were 2-3 fold more potent against MIA PaCa-2 and PANC-1 cells than free TPL (IC50 11.25 and 11.58 nM) and CL (IC50 0.84 and 1.23 μM). Furthermore, co-treatment with TPL-SFNPs and CL-SFNPs increased the growth inhibition of the same cells significantly in comparison with TPL-SFNPs or CL-SFNPs alone. Almost all combination index (CI) values, calculated using the CompuSyn software, were <1, suggesting that the growth inhibition effect of TPL-SFNPs in combination with CL-SFNPs was synergistic rather than additive, further suggesting that this novel combination may offer a potential treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Baoyue Ding
- Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, PR China
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| | - Md Arif Wahid
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| | - Zhijun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| | - Chen Xie
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| | - Arvind Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| | - Sunil Prabhu
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| | - Jeffrey Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
16
|
张 瑞, 许 建, 孙 珏, 梁 芳, 张 勇, 余 倩, 范 忠. 肠胃清方对人结肠癌HCT116/L-OHP裸鼠皮下移植瘤miR-30a/Beclin1通路的影响. Shijie Huaren Xiaohua Zazhi 2017; 25:1061-1070. [DOI: 10.11569/wcjd.v25.i12.1061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
目的 本课题旨在分子生物学技术和中医基础理论指导下, 从miRNAs与自噬相关联的角度, 讨论miR-30a与自噬基因Beclin1在结肠癌化疗耐药中的相关性, 并探讨肠胃清对miR-30a介导自噬的调控作用及逆转结肠癌耐药的分子机制.
方法 建立人结肠癌耐奥沙利铂(L-OHP)细胞株HCT116/L-OHP裸鼠皮下移植瘤模型, 随机分为空白组、L-OHP组、肠胃清方组、肠胃清方低剂量+L-OHP组、肠胃清方高剂量+L-OHP组. 治疗结束后采用RT-PCR、免疫组织化学、Tunnel等研究肠胃清对瘤体组织中miR-30a、Beclin1、LC3的表达及细胞凋亡的影响.
结果 奥沙利铂组可见Beclin1、LC3的上调, miR-30a的下调, 细胞凋亡的减少. 而肠胃清联用奥沙利铂组可见Beclin1、LC3的下调, miR-30a的上调, 细胞凋亡的增加(P<0.05或P<0.01).
结论 奥沙利铂诱导保护性自噬导致细胞凋亡减少可能是其耐药的机制. 肠胃清可通过调控miR-30a/Beclin1通路而抑制自噬逆转结肠癌耐药.
Collapse
|
17
|
Jia Y, Liu J, Xu J. Influence of grapefruit juice on pharmacokinetics of triptolide in rats grapefruit juice on the effects of triptolide. Xenobiotica 2017; 48:407-411. [PMID: 28359180 DOI: 10.1080/00498254.2017.1313470] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Yuzhen Jia
- Department of Pediatrics, Yidu Central Hospital of Weifang, China,
| | - Jie Liu
- Yidu Central Hospital of Weifang, China, and
| | - Jisen Xu
- Department of Neurology, Yidu Central Hospital of Weifang, China
| |
Collapse
|
18
|
Jiang QW, Cheng KJ, Mei XL, Qiu JG, Zhang WJ, Xue YQ, Qin WM, Yang Y, Zheng DW, Chen Y, Wei MN, Zhang X, Lv M, Chen MW, Wei X, Shi Z. Synergistic anticancer effects of triptolide and celastrol, two main compounds from thunder god vine. Oncotarget 2016; 6:32790-804. [PMID: 26447544 PMCID: PMC4741730 DOI: 10.18632/oncotarget.5411] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/25/2015] [Indexed: 02/07/2023] Open
Abstract
Triptolide and celastrol are two main active compounds isolated from Thunder God Vine with the potent anticancer activity. However, the anticancer effect of triptolide in combination with celastrol is still unknown. In the present study, we demonstrated that the combination of triptolide with celastrol synergistically induced cell growth inhibition, cell cycle arrest at G2/M phase and apoptosis with the increased intracellular ROS accumulation in cancer cells. Pretreatment with ROS scavenger N-acetyl-L-cysteine dramatically blocked the apoptosis induced by co-treatment with triptolide and celastrol. Treatment with celastrol alone led to the decreased expressions of HSP90 client proteins including survivin, AKT, EGFR, which was enhanced by the addition of triptolide. Additionally, the celastrol-induced expression of HSP70 and HSP27 was abrogated by triptolide. In the nude mice with xenograft tumors, the lower-dose combination of triptolide with celastrol significantly inhibited the growth of tumors without obvious toxicity. Overall, triptolide in combination with celastrol showed outstanding synergistic anticancer effect in vitro and in vivo, suggesting that this beneficial combination may offer a promising treatment option for cancer patients.
Collapse
Affiliation(s)
- Qi-Wei Jiang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Ke-Jun Cheng
- Chemical Biology Center, Lishui Institute of Agricultural Sciences, Lishui, Zhejiang, China.,National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiao-Long Mei
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Jian-Ge Qiu
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Wen-Ji Zhang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - You-Qiu Xue
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Wu-Ming Qin
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Yang Yang
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Di-Wei Zheng
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Yao Chen
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Meng-Ning Wei
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xu Zhang
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Min Lv
- Institute of Materia Medica, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mei-Wan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xing Wei
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Li X, Zang A, Jia Y, Zhang J, Fan W, Feng J, Duan M, Zhang L, Huo R, Jiao J, Zhu X. Triptolide reduces proliferation and enhances apoptosis of human non-small cell lung cancer cells through PTEN by targeting miR-21. Mol Med Rep 2016; 13:2763-8. [PMID: 26847601 DOI: 10.3892/mmr.2016.4844] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 12/01/2015] [Indexed: 11/06/2022] Open
Abstract
Triptolide is used in traditional Chinese medicine. It has the advantages of a unique mechanism of action, a wide antitumor spectrum, multiple targets, multi-channel effects and low toxicity. The current study was conducted to evaluate whether the potential anticancer effects of triptolide reduces proliferation and enhances apoptosis of human non‑small cell lung cancer (NSCLC) cells, and to assess the underlying anticancer mechanisms. In PC‑9 cells, treatment with triptolide reduced cell proliferation and increased cell apoptosis and caspase‑3 and 9 activity. Triptolide treatment reduced miR‑21 expression and enhanced phosphatase and tensin homolog (PTEN) protein expression levels in the PC‑9 cells. Furthermore, the upregulation of miR‑21 expression levels suppressed the effect of triptolide on cell viability and PTEN protein expression levels in PC‑9 cells. To the best of our knowledge, the present study is the first to demonstrate that triptolide reduced the proliferation and enhanced the apoptosis of human NSCLC cells through PTEN by targeting miR-21.
Collapse
Affiliation(s)
- Xiaofang Li
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Aimin Zang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Youchao Jia
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Jinchao Zhang
- College of Chemistry and Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding, Hebei 071002, P.R. China
| | - Wufang Fan
- College of Life Sciences, Hebei University, Baoding, Hebei 071002, P.R. China
| | - Jia Feng
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Mindan Duan
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Lei Zhang
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Ran Huo
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Jin Jiao
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Xiaowei Zhu
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
20
|
Jiang N, Dong XP, Zhang SL, You QY, Jiang XT, Zhao XG. Triptolide reverses the Taxol resistance of lung adenocarcinoma by inhibiting the NF-κB signaling pathway and the expression of NF-κB-regulated drug-resistant genes. Mol Med Rep 2015; 13:153-9. [PMID: 26531258 PMCID: PMC4686042 DOI: 10.3892/mmr.2015.4493] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 09/24/2015] [Indexed: 12/26/2022] Open
Abstract
Paclitaxel (or Taxol®) is a first-line chemotherapeutic drug for the treatment of non-small cell lung cancer; however, resistance to the drug is an important factor, which influences the outcome of chemotherapy. The present study aimed to investigate the role of triptolide (TPL) in reversing Taxol‑resistant human lung adenocarcinoma and to elucidate the underlying molecular mechanism of resistance reversal mediated by TPL. It was hypothesized that this experimental approach would assist in solving the problem of chemotherapeutic resistance in non‑small cell lung cancer, thereby improving the clinical outcomes. The human Taxol‑resistant lung adenocarcinoma cell line, A549/Taxol, was established. The resistance index of the cell line was calculated, according to the half maximal inhibitory concentration (IC50) of A549/Taxol IC50 of A549, to be 51.87. The levels of apoptosis and the cell cycle in the A549/Taxol cell line were assessed to confirm the effects of TPL at three different concentrations (0.03, 0.3 and 3 µmol/l) and treatment durations (2, 4, 6 and 12 h) by flow cytometric analysis, and the inhibition of the NF‑κB signaling pathway and the expression of NF‑κB‑regulated drug‑resistant proteins were determined by immunofluorescence and western blotting, respectively. The administration of TPL promoted cell apoptosis in the A549/Taxol lung adenocarcinoma Taxol‑resistant cell line and also promoted cell cycle regulation. The drug was also able to elicit a reversal of the drug resistance. TPL inhibited the nuclear factor‑κB (NF‑κB) signaling pathway and the expression of NF‑κB‑regulated drug‑resistant genes, including those for FLICE‑like inhibitory protein, X‑linked inhibitor of apoptosis protein, Bcl‑2, Bcl‑xL and cyclo‑oxygenase‑2. TPL exerted a marked drug‑resistance‑reversal effect on human lung adenocarcinoma Taxol resistance, and the effect was revealed to be dose‑ and time‑dependent. In conclusion, TPL exerted its role in the process of resistance reversal by inhibiting the NF‑κB signaling pathway, and the transcription and expression of NF-κB-regulated drug-resistant genes.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiao-Peng Dong
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Suo-Lin Zhang
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Qing-Yong You
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xing-Tao Jiang
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiao-Gang Zhao
- Department of Thoracic Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
21
|
Gui Z, Li S, Liu X, Xu B, Xu J. Oridonin alters the expression profiles of microRNAs in BxPC-3 human pancreatic cancer cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:117. [PMID: 25880988 PMCID: PMC4399397 DOI: 10.1186/s12906-015-0640-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/30/2015] [Indexed: 12/17/2022]
Abstract
Background Oridonin, an ingredient used in traditional Chinese medicine, has been demonstrated to play an important role in antitumour effects, but the mechanism underlying its antitumour properties is still not clear. Methods To verify the anti-cancer effects of oridonin via a miRNA-dependent mechanism, comprehensive miRNA expression profiling of oridonin-treated BxPC-3 human pancreatic cancer cells was performed using a miRNA microarray assay based on Sanger miR-Base Release 20, followed by a validation using real-time PCR. MicroRNA target prediction and Gene Ontology and KEGG pathway analysis were performed to investigate possible pathways involved. Results The results showed that 105 miRNAs were significantly differentially expressed (signal reading >500, p ≤ 0.01, |Log2-value| ≥1) in oridonin-treated BxPC-3 human pancreatic cancer cells. Conclusions Our data indicates that oridonin inhibits BxPC-3 cells probably through regulating the expression of miRNAs. Interruption of miRNA profiling may provide new therapeutic methods for the clinical treatment of pancreatic cancer.
Collapse
|
22
|
Drug resistance-related microRNAs in hematological malignancies: Translating basic evidence into therapeutic strategies. Blood Rev 2015; 29:33-44. [DOI: 10.1016/j.blre.2014.09.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/25/2014] [Accepted: 09/09/2014] [Indexed: 12/12/2022]
|
23
|
Chen RH, Tian YJ. Enhanced anti-tumor efficacy of aspirin combined with triptolide in cervical cancer cells. Asian Pac J Cancer Prev 2014; 14:3041-4. [PMID: 23803076 DOI: 10.7314/apjcp.2013.14.5.3041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The non-steroidal anti-inflammatory drug (NSAID) aspirin (acetylsalicylic acid) is an inhibitor of cyclooxygenase enzymes. Recent studies have shown that aspirin could be used as an anti-tumor drug. Triptolide, the major compound extracted from the Chinese herb Tripteryglum wilfordii Hook.f, has now been shown that it can inhibit tumor growth. The aim of this study was to analyze the anti-tumor efficiency of aspirin and triptolide in cervical cancer cells. METHODS Viability of cervical cancer cell lines was assessed by the MTT method at various concentrations of aspirin and triptolide. Siha and HeLa cell apoptotic analysis was performed by flow cytometry. Real time-PCR and Western Blotting were used to analyze the expression of Bcl-2/Bax, Cyclin D1 and p16. RESULTS Viability in the combination group was significantly decreased as compared with either drug used alone. Expression change of Bcl-2/Bax, CyclinD1 and p16 appeared to play an important role in the synergistic killing effect on cervical cancer cell apoptosis. CONCLUSION Aspirin and triptolide combination treatment may have synergistic anti-tumor effects on cervical cancer cells.
Collapse
Affiliation(s)
- Rong-Hui Chen
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, China
| | | |
Collapse
|
24
|
Liu Y, Cui YF. Synergism of cytotoxicity effects of triptolide and artesunate combination treatment in pancreatic cancer cell lines. Asian Pac J Cancer Prev 2014; 14:5243-8. [PMID: 24175808 DOI: 10.7314/apjcp.2013.14.9.5243] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Triptolide, extracted from the herb Tripteryglum wilfordii Hook.f that has long been used as a natural medicine in China, has attracted much interest for its anti-cancer effects against some kinds of tumours in recent years. Artesunate, extracted from the Chinese herb Artemisia annua, has proven to be effective and safe as an anti-malarial drug that possesses anticancer potential. The present study attempted to clarify if triptolide enhances artesunate-induced cytotoxicity in pancreatic cancer cell lines in vitro and in vivo. METHODS In vitro, to test synergic actions, cell viability and apoptosis were analyzed after treatment of pancreatic cancer cell lines with the two agents singly or in combination. The molecular mechanisms of apoptotic effects were also explored using qRT-PCR and Western blotting. In vivo, a tumor xenograft model was established in nude mice, for assessment of inhibitory effects of triptolide and artesunate. RESULTS We could show that the combination of triptolide and artesunate could inhibit pancreatic cancer cell line growth, and induce apoptosis, accompanied by expression of HSP 20 and HSP 27, indicating important roles in the synergic effects. Moreover, tumor growth was decreased with triptolide and artesunate synergy. CONCLUSION Our result indicated that triptolide and artesunate in combination at low concentrations can exert synergistic anti-tumor effects in pancreatic cancer cells with potential clinical applications.
Collapse
Affiliation(s)
- Yao Liu
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China E-mail :
| | | |
Collapse
|
25
|
Hsp90 inhibitor BIIB021 enhances triptolide-induced apoptosis of human T-cell acute lymphoblastic leukemia cells in vitro mainly by disrupting p53-MDM2 balance. Acta Pharmacol Sin 2013; 34:1545-53. [PMID: 24241349 DOI: 10.1038/aps.2013.124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/02/2013] [Indexed: 01/02/2023] Open
Abstract
AIM To investigate the effects of BIIB021, an inhibitor of heat shock protein 90 (Hsp90) alone or in combination with triptolide (TPL) on T-cell acute lymphoblastic leukemia (T-ALL) and the mechanisms of action. METHODS Human T-ALL cells line Molt-4 was examined. The cell viability was measured using MTT assay. Apoptotic cells were studied with Hoechst 33258 staining. Cell apoptosis and cell cycle were analyzed using flow cytometry with Annexin V/PI staining and PI staining, respectively. The levels of multiple proteins, including Akt, p65, CDK4/6, p18, Bcl-2 family proteins, MDM2, and p53, were examined with Western blotting. The level of MDM2 mRNA was determined using RT-PCR. RESULTS Treatment of Molt-4 cells with BIIB021 (50-800 nmol/L) inhibited the cell growth in a dose-dependent manner (the IC50 value was 384.6 and 301.8 nmol/L, respectively, at 48 and 72 h). BIIB021 dose-dependently induced G0/G1 phase arrest, followed by apoptosis of Molt-4 cells. Furthermore, BIIB021 increased the expression of p18, decreased the expression of CDK4/6, and activated the caspase pathway in Molt-4 cells. Moreover, BIIB021 (50-400 nmol/L) dose-dependently decreased the phospho-MDM2 and total MDM2 protein levels, but slightly increased the phospho-p53 and total p53 protein levels, whereas TPL (5-40 nmol/L) dose-dependently enhanced p53 activation without affecting MDM2 levels. Co-treatment with BIIB021 and TPL showed synergic inhibition on Molt-4 cell growth. The co-treatment disrupted p53-MDM2 balance, thus markedly enhanced p53 activation. In addition, the co-treatment increased the expression of Bak and Bim, followed by increased activation of caspase-9. CONCLUSION The combination of BIIB021 and TPL may provide a novel strategy for treating T-ALL by overcoming multiple mechanisms of apoptosis resistance.
Collapse
|
26
|
Chung WM, Chang WC, Chen L, Chang YY, Shyr CR, Hung YC, Ma WL. MicroRNA-21 promotes the ovarian teratocarcinoma PA1 cell line by sustaining cancer stem/progenitor populations in vitro. Stem Cell Res Ther 2013; 4:88. [PMID: 23890123 PMCID: PMC3854706 DOI: 10.1186/scrt247] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 07/24/2013] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Resistance of cancer stem/progenitor cells (CSPCs) to chemotherapy can lead to cancer relapse. Ovarian teratocarcinoma (OVTC) arises from germ cells and comprises pluripotent cells that can be used to study cancer cell stemness. In this study, we evaluated whether microRNA-21 (miR-21) promotes ovarian teratocarcinoma by maintaining cancer stem/progenitor populations. METHODS The lentiviral delivery system was used to upregulate or to suppress the expression of miR-21 in the human ovarian teratocarcinoma cell line PA1 and cell growth assays were used to monitor the expression of miR-21 at different time points. Antibodies directed toward CD133, a stem cell marker, were used to identify CSPCs in the PA1 cell population, and the level of miR-21 expression was determined in enriched CSPCs. Stem cell functional assays (sphere assay and assays for CD133 expression) were used to assess the effects of miR-21 on progression of the CD133+ population. RESULTS Knockdown of miR-21 in PA1 cells attenuated growth of PA1 cells whereas overexpression of miR-21 promoted cell growth. Moreover, knockdown of miR-21 resulted in a marked reduction in the CD133+ population and sphere formation of CSPCs. In contrast, overexpression of miR-21 resulted in a marked increase in the population of CD133+ cells as well as sphere formation of CSPCs. CONCLUSIONS MicroRNA-21 plays a significant role in cancer growth by regulating stemness in cancer cells.
Collapse
|
27
|
Hong L, Han Y, Zhang Y, Zhang H, Zhao Q, Wu K, Fan D. MicroRNA-21: a therapeutic target for reversing drug resistance in cancer. Expert Opin Ther Targets 2013; 17:1073-80. [PMID: 23865553 DOI: 10.1517/14728222.2013.819853] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Drug resistance is a major clinical obstacle to the successful treatment of human cancer. The microRNAs-21 (miR-21), an oncomiR, may play an important role in the progress of drug resistance. AREAS COVERED This review covers all related literature on miR-21 in drug resistance of human cancers and analyzes the expression, biological functions and targets of it. This study also envisages future developments toward its clinical and therapeutic applications in cancer treatment. EXPERT OPINION The miR-21 may promote the drug resistance of various cancers. Inhibitors of miR-21 may function as effective approaches for reversing drug resistance in cancer cells. There is a tough way from discovering the function of miR-21 to clinical use. Further understanding of miR-21-mediated signaling pathways will help to promote the therapeutic-clinical use of miR-21 in cancer.
Collapse
Affiliation(s)
- Liu Hong
- Fourth Military Medical University, State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Xi'an, 710032, Shaanxi Province, China.
| | | | | | | | | | | | | |
Collapse
|
28
|
Xie Z, Cao L, Zhang J. miR-21 modulates paclitaxel sensitivity and hypoxia-inducible factor-1α expression in human ovarian cancer cells. Oncol Lett 2013; 6:795-800. [PMID: 24137413 PMCID: PMC3789026 DOI: 10.3892/ol.2013.1432] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 06/07/2013] [Indexed: 01/11/2023] Open
Abstract
Drug resistance is a major problem encountered in the treatment of ovarian cancer. Previous studies have demonstrated that in several types of cancer the overexpression of the multidrug resistance 1 (MDR1) gene is mainly associated with drug resistance. The present study aimed to investigate the role of miR-21 in the development of drug resistance in ovarian cancer cells. The expression levels of miR-21 in the ovarian cancer A2780 and A2780/taxol cell lines were detected by stem-loop real-time PCR. A2780 and A2780/taxol cells were transfected with mimics or inhibitors of miR-21 or negative control RNA. The expression levels of P-glycoprotein (P-gp) and hypoxia-inducible factor-1α (HIF-1α) proteins were assessed by western blot analysis. Drug sensitivity was analyzed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The expression levels of miR-21 and P-gp were upregulated to a greater extent in the paclitaxel-resistant ovarian cancer A2780/taxol cell line compared with the parental A2780 cell line. Transfection of A2780/taxol cells with inhibitors of miR-21 decreased the expression levels of the P-gp and HIF-1α proteins, and increased the sensitivity of the A2780/taxol cells to paclitaxel. The expression levels of P-gp were additionally increased; however, the sensitivity of the miR-21 mimic-treated A2780 cells to paclitaxel was decreased. miR-21 may be involved in the development of drug resistance and the regulation of MDR1/P-gp expression, at least in part, by targeting HIF-1α in ovarian cancer cells.
Collapse
Affiliation(s)
- Zhongbin Xie
- Department of Clinical Laboratory Medicine, Yulin Number Two Hospital, Yulin 719000, P.R. China
| | | | | |
Collapse
|
29
|
MacKenzie TN, Mujumdar N, Banerjee S, Sangwan V, Sarver A, Vickers S, Subramanian S, Saluja AK. Triptolide induces the expression of miR-142-3p: a negative regulator of heat shock protein 70 and pancreatic cancer cell proliferation. Mol Cancer Ther 2013; 12:1266-75. [PMID: 23635652 DOI: 10.1158/1535-7163.mct-12-1231] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest malignancies, is resistant to current chemotherapies. We previously showed that triptolide inhibits PDAC cell growth in vitro and blocks metastatic spread in vivo. Triptolide downregulates HSP70, a molecular chaperone upregulated in several tumor types. This study investigates the mechanism by which triptolide inhibits HSP70. Because microRNAs (miRNA) are becoming increasingly recognized as negative regulators of gene expression, we tested whether triptolide regulates HSP70 via miRNAs. Here, we show that triptolide as well as quercetin, but not gemcitabine, upregulated miR-142-3p in PDAC cells (MIA PaCa-2, Capan-1, and S2-013). Ectopic expression of miR-142-3p inhibited cell proliferation, measured by electric cell-substrate impedance sensing, and decreased HSP70 expression, measured by real-time PCR and immunoblotting, compared with controls. We showed that miR-142-3p directly binds to the 3'UTR of HSP70, and that this interaction is important as HSP70 overexpression rescued miR-142-3p-induced cell death. We found that miR-142-3p regulates HSP70 independently of heat shock factor 1. Furthermore, Minnelide, a water-soluble prodrug of triptolide, induced the expression of miR-142-3p in vivo. This is the first description of an miRNA-mediated mechanism of HSP70 regulation in cancer, making miR-142-3p an attractive target for PDAC therapeutic intervention.
Collapse
Affiliation(s)
- Tiffany N MacKenzie
- Department of Pharmacology, University of Minnesota, MMC 195, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|