1
|
Qu X, Wang Y, Xu Y, Xu L, Ye X, Cai H, Bu L, Zeng Z, Zhou H. Aromatic nitroolefin with inhibition efficacy in triple-negative breast cancer cells by dual targeting RXRα and tubulins. Eur J Med Chem 2025; 289:117486. [PMID: 40090298 DOI: 10.1016/j.ejmech.2025.117486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
We previously identified that 1-(2-Nitrovinyl)naphthalene (Z-10) is a ligand of retinoid x receptor α (RXRα) with a potent anti-breast cancer efficacy and revealed that nitro group is an essential pharmacophore in Z-10. In this study, we defined that the double bond of the nitrovinyl group is also vital for Z-10 to bind and activate RXRα. Mechanistically, the double bond has a chemical ability to mediate Z-10's covalent binding of RXRα via the Michael addition reaction with Cys432. By retaining the nitrovinyl group, a series of Z-10 analogues with different aromatic groups and different aromatic ring-positions of nitrovinyl group and alkoxy groups were designed and synthesized. We found that some analogues including compound 30 show stronger ability than Z-10 in inhibiting TNFα survival signal in MDA-MB-231 breast cancer cells. Interestingly, these RXRα ligands also bind to tubulins likely through the similar covalent interaction and induce the degradation of tubulins and cell cycle arrest in MDA-MB-231 cells, of which 30 displays the strongest efficacy. Importantly, these analogues and TNFα exhibit synergistic effects in inducing breast cancer cell apoptosis, of which 30 shows greater efficacy than Z-10. Together, our study provides a theoretical basis for the RXRα and tubulin dual-targeting drug design for breast cancer treatment.
Collapse
Affiliation(s)
- Xiaofang Qu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yanxia Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunqing Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiaohong Ye
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hongchen Cai
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Liang Bu
- Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
2
|
Zhu B, Chen ZC, Du W, Chen YC. Facile construction of benzofulvene frameworks via a palladium-catalysed three-component reaction. Org Biomol Chem 2024; 22:8397-8400. [PMID: 39329403 DOI: 10.1039/d4ob01414f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Here we report a three-component reaction of 2-formylarylboronic acids, N-sulfonyl amines and 1,3-enynes, proceeding through a cascade imine formation/Pd0-catalysed vinylogous addition/intramolecular Suzuki coupling/isomerization process. This protocol exhibited broad substrate scope and good functionality tolerance, and a spectrum of multifunctionalised benzofulvene derivatives were furnished in moderate to good yields and E/Z-selectivity.
Collapse
Affiliation(s)
- Bo Zhu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Zhi-Chao Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Wei Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Ying-Chun Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
- College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
3
|
Zhang W, Hu H, Zhu Y, He Y, Yu M, Du W, Huang J. In silico study of androgen receptor N-terminal domain and exploration of its modulators. J Biomol Struct Dyn 2024:1-13. [PMID: 38661004 DOI: 10.1080/07391102.2024.2333454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/15/2024] [Indexed: 04/26/2024]
Abstract
The androgen receptor (AR, Uniprot: P10275) signaling plays a key role in the progression of prostate cancer, various AR-related ligands have been reported to treat prostate cancer. However, some resistance mechanisms limited the treating effect of these ligands. Since DBD binding or the allosteric binding sites in LBD of AR may allow the circumvention of some drug resistance mechanisms, anti-resistance is expected especially through the NTD (N-terminal domain) targeting. What's more, studies have shown that compounds including EPI-001 and its derivatives which bind to the Tau-5 region on NTD could be promising molecules for AR-based therapeutics. Herein, we employed aMD (accelerated molecular dynamics) simulation to fold Tau-5 unit proteins into native structure correctly. Subsequently, based on the predicted structural features of Tau-5, the virtual screening was conducted to discover new compounds targeting AR-NTD. We picked up 8 compounds (according to their docking scores and partly similar structural consists as known AR ligands) and analyzed their interaction with Tau-5, compared with the positive control EPI-001, four of the pick-up compounds showed better glide scores. Interestingly, although compound 8 had a lower docking score, it consisted of a similar component as the ligand EIQPN and the amide derivatives, this predicts that compound 8 has also the potential to be modified into an excellent AR-NTD binding molecule. These 8 compounds were all commercially available and could be tested to check whether there was a hit compound to bind the AR-NTD and to regulate its bio-activities. Together, this study described an in silico VLS approach to discover AR-NTD ligands and provided more choices for developing AR-targeted therapies.
Collapse
Affiliation(s)
- Weidong Zhang
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Hongyu Hu
- Xingzhi College, Zhejiang Normal University, Lanxi, China
| | - Yalan Zhu
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yiling He
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Mingyue Yu
- Xingzhi College, Zhejiang Normal University, Lanxi, China
| | - Wenjun Du
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jiangang Huang
- Xingzhi College, Zhejiang Normal University, Lanxi, China
| |
Collapse
|
4
|
Gu W, Bai J, Chen J. Application of thermally treated sludge residues on an e-waste contaminated soil: effects on PTE bioavailability, soil physicochemical and biological properties, and L. perenne growth. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:21962-21972. [PMID: 38400963 DOI: 10.1007/s11356-024-32179-3] [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: 06/08/2023] [Accepted: 01/21/2024] [Indexed: 02/26/2024]
Abstract
In the context of sustainable development, potentially toxic element (PTE) contamination of soil and large-scale disposal of sludge are two major environmental issues that need to be addressed urgently. It is of great significance to develop efficient and green technologies to solve these problems simultaneously. This study investigated the effects of a 5% addition of thermally treated sludge residues (fermentation and pyrolysis residues) in synergy with L. perenne on soil organic matter, mineral nutrients, PTE speciation, and PTE uptake and transport by L. perenne in an e-waste-contaminated soil through pot experiments. The results showed that the thermally treated sludge residues significantly increased soil electrical conductivity, cation exchange capacity, organic matter, available phosphorus, and exchangeable potassium contents. New PTE-containing crystalline phases were detected, and dissolved humic substances were found. Sludge fermentation residue significantly increased dissolved organic matter content, whereas sludge pyrolysis residue showed no significant effect. The combination of thermally treated sludge residues and L. perenne increased the residual fractions of Cu, Zn, Pb, and Cd. The thermally treated sludge residues promoted L. perenne growth, increasing fresh weight, plant height, and phosphorus and potassium uptake. The uptake of Cu, Zn, Pb, and Cd by L. perenne was significantly reduced. This approach has the potential for applications in the ecological restoration of e-waste-contaminated soils.
Collapse
Affiliation(s)
- Weihua Gu
- College of Environment, Zhejiang University of Technology, Hangzhou, 310014, China
- School of Resources and Environmental Engineering, Shanghai Polytechnic University, Shanghai, 201209, China
| | - Jianfeng Bai
- School of Resources and Environmental Engineering, Shanghai Polytechnic University, Shanghai, 201209, China
| | - Jianmeng Chen
- College of Environment, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
5
|
Li XM, Yang Y, Jiang FQ, Hu G, Wan S, Yan WY, He XS, Xiao F, Yang XM, Guo X, Lu JH, Yang XQ, Chen JJ, Ye WL, Liu Y, He K, Duan HX, Zhou YJ, Gan WJ, Liu F, Wu H. Histone lactylation inhibits RARγ expression in macrophages to promote colorectal tumorigenesis through activation of TRAF6-IL-6-STAT3 signaling. Cell Rep 2024; 43:113688. [PMID: 38245869 DOI: 10.1016/j.celrep.2024.113688] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Macrophages are phenotypically and functionally diverse in the tumor microenvironment (TME). However, how to remodel macrophages with a protumor phenotype and how to manipulate them for therapeutic purposes remain to be explored. Here, we show that in the TME, RARγ is downregulated in macrophages, and its expression correlates with poor prognosis in patients with colorectal cancer (CRC). In macrophages, RARγ interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6), which prevents TRAF6 oligomerization and autoubiquitination, leading to inhibition of nuclear factor κB signaling. However, tumor-derived lactate fuels H3K18 lactylation to prohibit RARγ gene transcription in macrophages, consequently enhancing interleukin-6 (IL-6) levels in the TME and endowing macrophages with tumor-promoting functions via activation of signal transducer and activator of transcription 3 (STAT3) signaling in CRC cells. We identified that nordihydroguaiaretic acid (NDGA) exerts effective antitumor action by directly binding to RARγ to inhibit TRAF6-IL-6-STAT3 signaling. This study unravels lactate-driven macrophage function remodeling by inhibition of RARγ expression and highlights NDGA as a candidate compound for treating CRC.
Collapse
Affiliation(s)
- Xiu-Ming Li
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Yun Yang
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Fu-Quan Jiang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Guang Hu
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Shan Wan
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Ying Yan
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Xiao-Shun He
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Fei Xiao
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Xue-Mei Yang
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Xin Guo
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Jun-Hou Lu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Jun-Jie Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Wen-Long Ye
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Yue Liu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Kuang He
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Han-Xiao Duan
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Yu-Jia Zhou
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Juan Gan
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China.
| | - Feng Liu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China.
| | - Hua Wu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China.
| |
Collapse
|
6
|
Chen J, Zhao T, He F, Zhong Y, Wang S, Tang Z, Qiu Y, Wu Z, Fang M. Discovery of bipyridine amide derivatives targeting pRXRα-PLK1 interaction for anticancer therapy. Eur J Med Chem 2023; 254:115341. [PMID: 37058970 DOI: 10.1016/j.ejmech.2023.115341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/23/2023] [Accepted: 04/02/2023] [Indexed: 04/16/2023]
Abstract
Retinoid X receptor alpha (RXRα) is an important therapeutic target of cancer. Recently, small molecules (e.g.,XS-060 and its derivatives), which can significantly induce RXRα-dependent mitotic arrest by inhibiting pRXRα-PLK1 interaction, have been demonstrated as excellent anticancer agents. To further obtain novel RXR-targeted antimitotic agents with excellent bioactivity and drug-like properties, we herein synthesized two new series of bipyridine amide derivatives with XS-060 as the lead compound. In the reporter gene assay, most synthesized compounds showed antagonistic activity against RXRα. The most active compound, bipyridine amide B9 (BPA-B9), showed better activity than XS-060, with excellent RXRα-binding affinity (KD = 39.29 ± 1.12 nM) and anti-proliferative activity against MDA-MB-231 (IC50 = 16 nM, SI > 3). Besides, a docking study revealed a proper fitting of BPA-B9 into the coactivator binding site of RXRα, rationalizing its potent antagonistic effect on RXRα transactivation. Further, the mechanism studies revealed that the anticancer activity of BPA-B9 was dependent on its cellular RXRα-targeted mechanism, such as inhibiting pRXRα-PLK1 interaction and inducing RXRα-dependent mitotic arrest. Besides, BPA-B9 displayed better pharmacokinetics than the lead XS-060. Further, animal assays indicated BPA-B9 had significant anticancer efficacy in vivo with no considerable side effects. Together, our study reveals a novel RXRα ligand BPA-B9 targeting the pRXRα-PLK1 interaction, with great potential as a promising anticancer drug candidate for further development.
Collapse
Affiliation(s)
- Jun Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Taige Zhao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Fengming He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Yijing Zhong
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Susu Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Ziqing Tang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Yingkun Qiu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Zhen Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China.
| | - Meijuan Fang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
7
|
Xie CL, Zhang D, Guo KQ, Yan QX, Zou ZB, He ZH, Wu Z, Zhang XK, Chen HF, Yang XW. Meroterpenthiazole A, a unique meroterpenoid from the deep-sea-derived Penicillium allii-sativi, significantly inhibited retinoid X receptor (RXR)-α transcriptional effect. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.09.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Zhu X, Li J, Ning H, Yuan Z, Zhong Y, Wu S, Zeng JZ. α-Mangostin Induces Apoptosis and Inhibits Metastasis of Breast Cancer Cells via Regulating RXRα-AKT Signaling Pathway. Front Pharmacol 2021; 12:739658. [PMID: 34539418 PMCID: PMC8444262 DOI: 10.3389/fphar.2021.739658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/18/2021] [Indexed: 11/28/2022] Open
Abstract
Mangostin, which has the function of anti-inflammatory, antioxidant, and anticancer, etc, is one of the main active ingredients of the hull of the mangosteen. The main objective of the study was to elucidate its anti-cancer function and possible mechanism. α-Mangostin was separated and structurally confirmed. MTT method was used to check the effect of mangostin on breast cancer cell proliferation. Then the effect of α-Mangostin on the transcriptional activity of RXRα was tested by dual-luciferase reporter gene assay. And Western blot (WB) was used to detect the expression of apoptosis-related proteins or cell cycle-associated proteins after treatment. Also, this study was to observe the effects of α-Mangostin on the invasion of breast cancer cell line MDA-MB-231. α-Mangostin regulates the downstream effectors of the PI3K/AKT signaling pathway by degrading RXRα/tRXRα. α-Mangostin can trigger PARP cleavage and induce apoptosis, which may be related to the induction of upregulated BAX expression and downregulation of BAD and cleaved caspase-3 expression in MDA-MB-231 cells through blockade of AKT signaling. The experiments verify that α-Mangostin have evident inhibition effects of invasion and metastasis of MDA-MB-231 cells. Cyclin D1 was involved in the anticancer effects of α-Mangostin on the cell cycle in MDA-MB-231 cells. α-Mangostin induces apoptosis, suppresses the migration and invasion of breast cancer cells through the PI3K/AKT signaling pathway by targeting RXRα, and cyclin D1 has involved in this process.
Collapse
Affiliation(s)
- Xiuzhi Zhu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jialin Li
- School of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Huiting Ning
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Zhidong Yuan
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yue Zhong
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Suzhen Wu
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jin-Zhang Zeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
9
|
Watanabe M, Fujihara M, Motoyama T, Kawasaki M, Yamada S, Takamura Y, Ito S, Makishima M, Nakano S, Kakuta H. Discovery of a "Gatekeeper" Antagonist that Blocks Entry Pathway to Retinoid X Receptors (RXRs) without Allosteric Ligand Inhibition in Permissive RXR Heterodimers. J Med Chem 2020; 64:430-439. [PMID: 33356247 DOI: 10.1021/acs.jmedchem.0c01354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Retinoid X receptor (RXR) heterodimers such as PPAR/RXR, LXR/RXR, and FXR/RXR can be activated by RXR agonists alone and are therefore designated as permissive. Similarly, existing RXR antagonists show allosteric antagonism toward partner receptor agonists in these permissive RXR heterodimers. Here, we show 1-(3-(2-ethoxyethoxy)-5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-2-(trifluoromethyl)-1H-benzo[d]imidazole-5-carboxylic acid (14, CBTF-EE) as the first RXR antagonist that does not show allosteric inhibition in permissive RXR heterodimers. This compound was designed based on the hypothesis that RXR antagonists that do not induce conformational changes of RXR would not exhibit such allosteric inhibition. CD spectra and X-ray co-crystallography of the complex of 14 and the RXR ligand binding domain (LBD) confirmed that 14 does not change the conformation of hRXR-LBD. The X-ray structure analysis revealed that 14 binds at the entrance of the ligand binding pocket (LBP), blocking access to the LBP and thus serving as a "gatekeeper".
Collapse
Affiliation(s)
- Masaki Watanabe
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Michiko Fujihara
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.,AIBIOS Company. Ltd., Tri-Seven Roppongi 8F 7-7-7 Roppongi, Minato-ku, Tokyo 106-0032, Japan
| | - Tomoharu Motoyama
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Mayu Kawasaki
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Shoya Yamada
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.,Research Fellowship Division, Japan Society for the Promotion of Science, Sumitomo-Ichibancho FS Bldg., 8 Ichibancho, Chiyoda-ku, Tokyo 102-8472, Japan
| | - Yuta Takamura
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Sohei Ito
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Shogo Nakano
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
10
|
Qiu DR, Zhou M, Liu XZ, Chen JJ, Wang GH, Lin T, Yu FR, Ding R, Sun CL, Tian WJ, Chen HF. Cytotoxic polyprenylated phloroglucinol derivatives from Hypericum elodeoides Choisy modulating the transactivation of RXRα. Bioorg Chem 2020; 107:104578. [PMID: 33383323 DOI: 10.1016/j.bioorg.2020.104578] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/03/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022]
Abstract
Hyperelodione D (1), an undescribed polyprenylated phloroglucinol derivative possessing 6/6/5/5 fused tetracyclic core, together with hyperelodiones E-F (2-3), two unreported analogues bearing 6/5/5 fused tricyclic structure, were isolated from Hypericum elodeoides Choisy. Their planar structures were elucidated by spectroscopic analysis (HRESIMS, 1D and 2D NMR) and their absolute configurations were determined by comparison of experimental and calculated ECD data. The cytotoxicity and retinoid X receptor-α (RXRα) related activities of the isolates were evaluated and the plausible biogenetic pathways of 1-3 were proposed.
Collapse
Affiliation(s)
- Da-Ren Qiu
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China; Sichuan Province College Key Laboratory of Structure-Specific Small Molecule Drugs, School of Pharmacy, Chengdu Medical College, Chengdu 610500, People's Republic of China
| | - Mi Zhou
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Xiang-Zhong Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Jun-Jie Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Guang-Hui Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Ting Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Fu-Rong Yu
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Rong Ding
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Cui-Ling Sun
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China
| | - Wen-Jing Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China.
| | - Hai-Feng Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, People's Republic of China.
| |
Collapse
|
11
|
Xie G, Zhou Y, Tu X, Ye X, Xu L, Xiao Z, Wang Q, Wang X, Du M, Chen Z, Chi X, Zhang X, Xia J, Zhang X, Zhou Y, Li Z, Xie C, Sheng L, Zeng Z, Zhou H, Yin Z, Su Y, Xu Y, Zhang XK. Centrosomal Localization of RXRα Promotes PLK1 Activation and Mitotic Progression and Constitutes a Tumor Vulnerability. Dev Cell 2020; 55:707-722.e9. [PMID: 33321102 DOI: 10.1016/j.devcel.2020.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/15/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
Retinoid X receptor alpha (RXRα), a nuclear receptor of transcription factor, controls various physiological and pathological pathways including cellular growth, proliferation, differentiation, and apoptosis. Here, we report that RXRα is phosphorylated at its N-terminal A/B domain by cyclin-dependent kinase 1 (Cdk1) at the onset of mitosis, triggering its translocation to the centrosome, where phosphorylated-RXRα (p-RXRα) interacts with polo-like kinase 1 (PLK1) through its N-terminal A/B domain by a unique mechanism. The interaction promotes PLK1 activation, centrosome maturation, and mitotic progression. Levels of p-RXRα are abnormally elevated in cancer cell lines, during carcinogenesis in animals, and in clinical tumor tissues. An RXRα ligand XS060, which specifically inhibits p-RXRα/PLK1 interaction but not RXRα heterodimerization, promotes mitotic arrest and catastrophe in a tumor-specific manner. These findings unravel a transcription-independent action of RXRα at the centrosome during mitosis and identify p-RXRα as a tumor-specific vulnerability for developing mitotic drugs with improved therapeutic index.
Collapse
Affiliation(s)
- Guobin Xie
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Yuqi Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China; NucMito Pharmaceuticals Co. Ltd., Xiamen 361101, Fujian, China
| | - Xuhuang Tu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Xiaohong Ye
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Zhijian Xiao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Qiqiang Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Mingxuan Du
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Ziwen Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China; NucMito Pharmaceuticals Co. Ltd., Xiamen 361101, Fujian, China
| | - Xiaoqin Chi
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital Xiamen University, Xiamen 361004, Fujian, China
| | - Xiaoli Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Ji Xia
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Xiaowei Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Yunxia Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Zongxi Li
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Chengrong Xie
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital Xiamen University, Xiamen 361004, Fujian, China
| | - Luoyan Sheng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Zhenyu Yin
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital Xiamen University, Xiamen 361004, Fujian, China
| | - Ying Su
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China; NucMito Pharmaceuticals Co. Ltd., Xiamen 361101, Fujian, China
| | - Yang Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
12
|
Wu Y, Liao H, Liu LY, Sun F, Chen HF, Jiao WH, Zhu HR, Yang F, Huang G, Zeng DQ, Zhou M, Wang SP, Lin HW. Phakefustatins A–C: Kynurenine-Bearing Cycloheptapeptides as RXRα Modulators from the Marine Sponge Phakellia fusca. Org Lett 2020; 22:6703-6708. [PMID: 32701300 DOI: 10.1021/acs.orglett.0c01586] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ying Wu
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hongze Liao
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Li-Yun Liu
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fan Sun
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hai-Feng Chen
- School of Pharmaceutical Sciences, Xiamen University, South Xiangan Road, Xiamen, Fujian 361102, China
| | - Wei-Hua Jiao
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hong-Rui Zhu
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fan Yang
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - De-Quan Zeng
- School of Pharmaceutical Sciences, Xiamen University, South Xiangan Road, Xiamen, Fujian 361102, China
| | - Mi Zhou
- School of Pharmaceutical Sciences, Xiamen University, South Xiangan Road, Xiamen, Fujian 361102, China
| | - Shu-Ping Wang
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hou-Wen Lin
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
13
|
Leijten-van de Gevel IA, Brunsveld L. Delineation of the molecular determinants of the unique allosteric binding site of the orphan nuclear receptor RORγt. J Biol Chem 2020; 295:9183-9191. [PMID: 32439807 PMCID: PMC7335795 DOI: 10.1074/jbc.ra120.013581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/17/2020] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptors (NRs) are high-interest targets in drug discovery because of their involvement in numerous biological processes and diseases. Classically, NRs are targeted via their hydrophobic, orthosteric pocket. Although successful, this approach comes with challenges, including off-target effects due to lack of selectivity. Allosteric modulation of NR activity constitutes a promising pharmacological strategy. The retinoic acid receptor-related orphan receptor-γt (RORγt) is a constitutively active NR that positively regulates the expression of interleukin-17 in T helper 17 cells. Inhibiting this process is an emerging strategy for managing autoimmune diseases. Recently, an allosteric binding pocket in the C-terminal region of the ligand-binding domain (LBD) of RORγt was discovered that is amenable to small-molecule drug discovery. Compounds binding this pocket induce a reorientation of helix 12, thereby preventing coactivator recruitment. Therefore, inverse agonists binding this site with high affinity are actively being pursued. To elucidate the pocket formation mechanism, verify the uniqueness of this pocket, and substantiate the relevance of targeting this site, here we identified the key characteristics of the RORγt allosteric region. We evaluated the effects of substitutions in the LBD on coactivator, orthosteric, and allosteric ligand binding. We found that two molecular elements unique to RORγt, the length of helix 11' and a Gln-487 residue, are crucial for the formation of the allosteric pocket. The unique combination of elements present in RORγt suggests a high potential for subtype-selective targeting of this NR to more effectively treat patients with autoimmune diseases.
Collapse
Affiliation(s)
- Iris A Leijten-van de Gevel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Eindhoven, the Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Eindhoven, the Netherlands.
| |
Collapse
|
14
|
Yamada S, Kawasaki M, Fujihara M, Watanabe M, Takamura Y, Takioku M, Nishioka H, Takeuchi Y, Makishima M, Motoyama T, Ito S, Tokiwa H, Nakano S, Kakuta H. Competitive Binding Assay with an Umbelliferone-Based Fluorescent Rexinoid for Retinoid X Receptor Ligand Screening. J Med Chem 2019; 62:8809-8818. [PMID: 31483660 DOI: 10.1021/acs.jmedchem.9b00995] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ligands for retinoid X receptors (RXRs), "rexinoids", are attracting interest as candidates for therapy of type 2 diabetes and Alzheimer's and Parkinson's diseases. However, current screening methods for rexinoids are slow and require special apparatus or facilities. Here, we created 7-hydroxy-2-oxo-6-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-2H-chromene-3-carboxylic acid (10, CU-6PMN) as a new fluorescent RXR agonist and developed a screening system of rexinoids using 10. Compound 10 was designed based on the fact that umbelliferone emits strong fluorescence in a hydrophilic environment, but the fluorescence intensity decreases in hydrophobic environments such as the interior of proteins. The developed assay using 10 enabled screening of rexinoids to be performed easily within a few hours by monitoring changes of fluorescence intensity with widely available fluorescence microplate readers, without the need for processes such as filtration.
Collapse
Affiliation(s)
- Shoya Yamada
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan.,Research Fellowship Division , Japan Society for the Promotion of Science , Sumitomo-Ichibancho FS Bldg., 8 Ichibancho , Chiyoda-ku, Tokyo 102-8472 , Japan
| | - Mayu Kawasaki
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences , University of Shizuoka , 52-1 Yada , Suruga-ku, Shizuoka 422-8526 , Japan
| | - Michiko Fujihara
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan.,AIBIOS Co. Ltd. , Tri-Seven Roppongi 8F 7-7-7 Roppongi , Minato-ku, Tokyo 106-0032 Japan
| | - Masaki Watanabe
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan
| | - Yuta Takamura
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan
| | - Maho Takioku
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan
| | - Hiromi Nishioka
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan
| | - Yasuo Takeuchi
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences , Nihon University School of Medicine , 30-1 Oyaguchi-kamicho , Itabashi-ku, Tokyo 173-8610 , Japan
| | - Tomoharu Motoyama
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences , University of Shizuoka , 52-1 Yada , Suruga-ku, Shizuoka 422-8526 , Japan
| | - Sohei Ito
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences , University of Shizuoka , 52-1 Yada , Suruga-ku, Shizuoka 422-8526 , Japan
| | | | - Shogo Nakano
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences , University of Shizuoka , 52-1 Yada , Suruga-ku, Shizuoka 422-8526 , Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , 1-1-1, Tsushima-naka , Kita-ku, Okayama 700-8530 , Japan
| |
Collapse
|
15
|
Shen L, Sun Z, Nie P, Yuan R, Cai Z, Wu C, Hu L, Jin S, Zhou H, Zhang X, He B. Sulindac-derived retinoid X receptor-α modulator attenuates atherosclerotic plaque progression and destabilization in ApoE -/- mice. Br J Pharmacol 2019; 176:2559-2572. [PMID: 30943581 PMCID: PMC6592870 DOI: 10.1111/bph.14682] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis is a chronic inflammatory disease, and retinoid X receptor-α (RXRα) is an intriguing anti-atherosclerosis target. This study investigated whether and how an RXRα modulator, K-80003, derived from a non-steroidal anti-inflammatory drug attenuates atherosclerotic plaque progression and destabilization. EXPERIMENTAL APPROACH Our previously established ApoE-/- mouse model of carotid vulnerable plaque progression was treated with K-80003 or vehicle for 4 or 8 weeks. Samples of carotid arteries and serum were collected to determine atherosclerotic lesion size, histological features, expression of related proteins, and lipid profiles. In vitro studies were carried out in 7-ketocholesterol (7-KC)-stimulated macrophages treated with or without K-80003. KEY RESULTS K-80003 significantly reduced lesion size, plaque rupture, macrophage infiltration, and inflammatory cytokine levels. Plaque macrophages positive for nuclear p65 (RelA) NF-κB subunit were markedly reduced after K-80003 treatment. Also, K-80003 treatment inhibited 7-KC-induced p65 nuclear translocation, IκBα degradation, and transcription of NF-κB target genes. In addition, K-80003 inhibited NF-κB pathway mainly through the reduction of p62/sequestosome 1 (SQSTM1), probably due to promotion of autophagic flux by K-80003. Mechanistically, cytoplasmic localization of RXRα was associated with decreased autophagic flux. Increasing cytoplasmic RXRα expression by overexpression of RXRα/385 mutant decreased autophagic flux in RAW264.7 cells. Finally, K-80003 strongly inhibited 7-KC-induced RXRα cytoplasmic translocation. CONCLUSIONS AND IMPLICATIONS K-80003 suppressed atherosclerotic plaque progression and destabilization by promoting macrophage autophagic flux and consequently inhibited the p62/SQSTM1-mediated NF-κB proinflammatory pathway. Thus, targeting RXRα-mediated autophagy-inflammation axis by its noncanonical modulator may represent a promising strategy to treat atherosclerosis.
Collapse
Affiliation(s)
- Linghong Shen
- Department of CardiologyShanghai Chest Hospital, Shanghai Jiaotong UniversityShanghaiChina
| | - Zhe Sun
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
| | - Peng Nie
- Department of CardiologyRenji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Ruosen Yuan
- Department of CardiologyRenji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Zhaohua Cai
- Department of CardiologyRenji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Caizhe Wu
- Department of CardiologyRenji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Liuhua Hu
- Department of CardiologyRenji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Shuxuan Jin
- Department of CardiologyRenji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Hu Zhou
- School of Pharmaceutical SciencesXiamen UniversityXiamenChina
| | - Xiaokun Zhang
- School of Pharmaceutical SciencesXiamen UniversityXiamenChina
- Cancer CenterSandford Burnham Prebys Medical Discovery InstituteLa JollaCA
| | - Ben He
- Department of CardiologyShanghai Chest Hospital, Shanghai Jiaotong UniversityShanghaiChina
| |
Collapse
|
16
|
Krężel W, Rühl R, de Lera AR. Alternative retinoid X receptor (RXR) ligands. Mol Cell Endocrinol 2019; 491:110436. [PMID: 31026478 DOI: 10.1016/j.mce.2019.04.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/06/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022]
Abstract
Retinoid X receptors (RXRs) control a wide variety of functions by virtue of their dimerization with other nuclear hormone receptors (NRs), contributing thereby to activities of different signaling pathways. We review known RXR ligands as transcriptional modulators of specific RXR-dimers and the associated biological processes. We also discuss the physiological relevance of such ligands, which remains frequently a matter of debate and which at present is best met by member(s) of a novel family of retinoids, postulated as Vitamin A5. Through comparison with other natural, but also with synthetic ligands, we discuss high diversity in the modes of ligand binding to RXRs resulting in agonistic or antagonistic profiles and selectivity towards specific subtypes of permissive heterodimers. Despite such diversity, direct ligand binding to the ligand binding pocket resulting in agonistic activity was preferentially preserved in the course of animal evolution pointing to its functional relevance, and potential for existence of other, species-specific endogenous RXR ligands sharing the same mode of function.
Collapse
Affiliation(s)
- Wojciech Krężel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U 1258, Illkirch, France; Université de Strasbourg, Illkirch, France.
| | - Ralph Rühl
- Paprika Bioanalytics BT, Debrecen, Hungary
| | - Angel R de Lera
- Departamento de Química Orgánica, Facultade de Química, Lagoas-Marcosende, 36310, Vigo, Spain
| |
Collapse
|
17
|
Meijer FA, Leijten-van de Gevel IA, de Vries RMJM, Brunsveld L. Allosteric small molecule modulators of nuclear receptors. Mol Cell Endocrinol 2019; 485:20-34. [PMID: 30703487 DOI: 10.1016/j.mce.2019.01.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 02/08/2023]
Abstract
Nuclear Receptors (NRs) are multi-domain proteins, whose natural regulation occurs via ligands for a classical, orthosteric, binding pocket and via intra- and inter-domain allosteric mechanisms. Allosteric modulation of NRs via synthetic small molecules has recently emerged as an interesting entry to address the need for small molecules targeting NRs in pathology, via novel modes of action and with beneficial profiles. In this review the general concept of allosteric modulation in drug discovery is first discussed, serving as a background and inspiration for NRs. Subsequently, the review focuses on examples of small molecules that allosterically modulate NRs, with a strong focus on structural information and the ligand binding domain. Recently discovered nanomolar potent allosteric site NR modulators are catapulting allosteric targeting of NRs to the center of attention. The obtained insights serve as a basis for recommendations for the next steps to take in allosteric small molecular targeting of NRs.
Collapse
Affiliation(s)
- Femke A Meijer
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Den Dolech 2, 5612AZ, Eindhoven, the Netherlands
| | - Iris A Leijten-van de Gevel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Den Dolech 2, 5612AZ, Eindhoven, the Netherlands
| | - Rens M J M de Vries
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Den Dolech 2, 5612AZ, Eindhoven, the Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Technische Universiteit Eindhoven, Den Dolech 2, 5612AZ, Eindhoven, the Netherlands.
| |
Collapse
|
18
|
Oncogenic potential of truncated RXRα during colitis-associated colorectal tumorigenesis by promoting IL-6-STAT3 signaling. Nat Commun 2019; 10:1463. [PMID: 30931933 PMCID: PMC6443775 DOI: 10.1038/s41467-019-09375-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/18/2019] [Indexed: 12/22/2022] Open
Abstract
Retinoid X receptor-alpha (RXRα) is a potent regulator of inflammatory responses; however, its therapeutic potential for inflammatory cancer remains to be explored. We previously discovered that RXRα is abnormally cleaved in tumor cells and tissues, producing a truncated RXRα (tRXRα). Here, we show that transgenic expression of tRXRα in mice accelerates the development of colitis-associated colon cancer (CAC). The tumorigenic effect of tRXRα is primarily dependent on its expression in myeloid cells, which results in interleukin-6 (IL-6) induction and STAT3 activation. Mechanistic studies reveal an extensive interaction between tRXRα and TRAF6 in the cytoplasm of macrophages, leading to TRAF6 ubiquitination and subsequent activation of the NF-κB inflammatory pathway. K-80003, a tRXRα modulator derived from nonsteroidal anti-inflammatory drug (NSAID) sulindac, suppresses the growth of tRXRα-mediated colorectal tumor by inhibiting the NF-κB-IL-6-STAT3 signaling cascade. These results provide new insight into tRXRα action and identify a promising tRXRα ligand for treating CAC.
Collapse
|
19
|
Chen X, Cao X, Tu X, Alitongbieke G, Xia Z, Li X, Chen Z, Yin M, Xu D, Guo S, Li Z, Chen L, Zhang X, Xu D, Gao M, Liu J, Zeng Z, Zhou H, Su Y, Zhang XK. BI1071, a Novel Nur77 Modulator, Induces Apoptosis of Cancer Cells by Activating the Nur77-Bcl-2 Apoptotic Pathway. Mol Cancer Ther 2019; 18:886-899. [PMID: 30926635 DOI: 10.1158/1535-7163.mct-18-0918] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/28/2018] [Accepted: 03/14/2019] [Indexed: 11/16/2022]
Abstract
Nur77 (also called TR3 or NGFI-B), an orphan member of the nuclear receptor superfamily, induces apoptosis by translocating to mitochondria where it interacts with Bcl-2 to convert Bcl-2 from an antiapoptotic to a pro-apoptotic molecule. Nur77 posttranslational modification such as phosphorylation has been shown to induce Nur77 translocation from the nucleus to mitochondria. However, small molecules that can bind directly to Nur77 to trigger its mitochondrial localization and Bcl-2 interaction remain to be explored. Here, we report our identification and characterization of DIM-C-pPhCF3 +MeSO3 - (BI1071), an oxidized product derived from indole-3-carbinol metabolite, as a modulator of the Nur77-Bcl-2 apoptotic pathway. BI1071 binds Nur77 with high affinity, promotes Nur77 mitochondrial targeting and interaction with Bcl-2, and effectively induces apoptosis of cancer cells in a Nur77- and Bcl-2-dependent manner. Studies with animal model showed that BI1071 potently inhibited the growth of tumor cells in animals through its induction of apoptosis. Our results identify BI1071 as a novel Nur77-binding modulator of the Nur77-Bcl-2 apoptotic pathway, which may serve as a promising lead for treating cancers with overexpression of Bcl-2.
Collapse
Affiliation(s)
- Xiaohui Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Xihua Cao
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Xuhuang Tu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Gulimiran Alitongbieke
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Zebin Xia
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Xiaotong Li
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Ziwen Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | | | - Dan Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Shangjie Guo
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Zongxi Li
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Liqun Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Xindao Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Dingyu Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Meichun Gao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Jie Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China
| | - Ying Su
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, China. .,Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| |
Collapse
|
20
|
Abstract
Tetrazole derivatives are a prime class of heterocycles, very important to medicinal chemistry and drug design due to not only their bioisosterism to carboxylic acid and amide moieties but also to their metabolic stability and other beneficial physicochemical properties. Although more than 20 FDA-approved drugs contain 1 H- or 2 H-tetrazole substituents, their exact binding mode, structural biology, 3D conformations, and in general their chemical behavior is not fully understood. Importantly, multicomponent reaction (MCR) chemistry offers convergent access to multiple tetrazole scaffolds providing the three important elements of novelty, diversity, and complexity, yet MCR pathways to tetrazoles are far from completely explored. Here, we review the use of multicomponent reactions for the preparation of substituted tetrazole derivatives. We highlight specific applications and general trends holding therein and discuss synthetic approaches and their value by analyzing scope and limitations, and also enlighten their receptor binding mode. Finally, we estimated the prospects of further research in this field.
Collapse
Affiliation(s)
- Constantinos G. Neochoritis
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9700 AD Groningen, The Netherlands
| | - Ting Zhao
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9700 AD Groningen, The Netherlands
| | - Alexander Dömling
- Drug Design Group, Department of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9700 AD Groningen, The Netherlands
| |
Collapse
|
21
|
Eberhardt J, McEwen AG, Bourguet W, Moras D, Dejaegere A. A revisited version of the apo structure of the ligand-binding domain of the human nuclear receptor retinoic X receptor α. Acta Crystallogr F Struct Biol Commun 2019; 75:98-104. [PMID: 30713160 PMCID: PMC6360438 DOI: 10.1107/s2053230x18018022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/20/2018] [Indexed: 11/10/2022] Open
Abstract
The retinoic X receptor (RXR) plays a crucial role in the superfamily of nuclear receptors (NRs) by acting as an obligatory partner of several nuclear receptors; its role as a transcription factor is thus critical in many signalling pathways, such as metabolism, cell development, differentiation and cellular death. The first published structure of the apo ligand-binding domain (LBD) of RXRα, which is still used as a reference today, contained inaccuracies. In the present work, these inaccuracies were corrected using modern crystallographic tools. The most important correction concerns the presence of a π-bulge in helix H7, which was originally built as a regular α-helix. The presence of several CHAPS molecules, which are visible for the first time in the electron-density map and which stabilize the H1-H3 loop, which contains helix H2, are also revealed. The apo RXR structure has played an essential role in deciphering the molecular mode of action of NR ligands and is still used in numerous biophysical studies. This refined structure should be used preferentially in the future in interpreting experiments as well as for modelling and structural dynamics studies of the apo RXRα LBD.
Collapse
Affiliation(s)
- Jérôme Eberhardt
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Alastair G. McEwen
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - William Bourguet
- Centre de Biochimie Structurale (CBS), CNRS, Inserm, Université de Montpellier, Montpellier, France
| | - Dino Moras
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Annick Dejaegere
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
- Université de Strasbourg, Illkirch, France
| |
Collapse
|
22
|
Yan Z, Chong S, Lin H, Yang Q, Wang X, Zhang W, Zhang X, Zeng Z, Su Y. Design, synthesis and biological evaluation of tetrazole-containing RXRα ligands as anticancer agents. Eur J Med Chem 2019; 164:562-575. [DOI: 10.1016/j.ejmech.2018.12.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022]
|
23
|
Wang X, Chong S, Lin H, Yan Z, Huang F, Zeng Z, Zhang X, Su Y. Discovery of atorvastatin as a tetramer stabilizer of nuclear receptor RXRα through structure-based virtual screening. Bioorg Chem 2019; 85:413-419. [PMID: 30665035 DOI: 10.1016/j.bioorg.2019.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/30/2018] [Accepted: 01/06/2019] [Indexed: 01/10/2023]
Abstract
Retinoid X receptor alpha (RXRα), a central member of the nuclear receptor superfamily and a key regulator of many signal transduction pathways, has been an attractive drug target. We previously discovered that an N-terminally truncated form of RXRα can be induced by specific ligands to form homotetramers, which, as a result of conformational selection, forms the basis for inhibiting the nongenomic activation of RXRα. Here, we report the identification and characterization of atorvastatin as a new RXRα tetramer stabilizer by using structure-based virtual screening and demonstrate that virtual library screening can be used to aid in identifying RXRα ligands that can induce its tetramerization. In this study, docking was applied to screen the FDA-approved small molecule drugs in the DrugBank 4.0 collection. Two compounds were selected and purchased for testing. We showed that the selected atorvastatin could bind to RXRα to promote RXRα-LBD tetramerization. We also showed that atorvastatin possessed RXRα-dependent apoptotic effects. In addition, we used a chemical approach to aid in the studies of the binding mode of atorvastatin.
Collapse
Affiliation(s)
- Xin Wang
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Fujian 361002, China
| | - Shuyi Chong
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Fujian 361002, China
| | - Huiyun Lin
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Fujian 361002, China
| | - Zhiqiang Yan
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Fujian 361002, China
| | - Fengyu Huang
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Fujian 361002, China
| | - Zhiping Zeng
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Fujian 361002, China
| | - Xiaokun Zhang
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Fujian 361002, China; Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Ying Su
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
24
|
Abstract
Retinoid X receptors (RXRs) are promiscuous partners of heterodimeric associations with other members of the Nuclear Receptor (NR) superfamily. RXR ligands ("rexinoids") either transcriptionally activate the "permissive" subclass of heterodimers or synergize with partner ligands in the "nonpermissive" subclass of heterodimers. The rationale for rexinoid design with a wide structural diversity going from the structures of existing complexes with RXR determined by X-Ray, to natural products and other ligands discovered by high-throughput screening (HTS), mere serendipity, and rationally designed based on Molecular Modeling, will be described. Included is the new generation of ligands that modulate the structure of specific receptor surfaces that serve to communicate with other regulators. The panel of the known RXR agonists, partial (ant)agonists, and/or heterodimer-selective rexinoids require the exploration of their therapeutic potential in order to overcome some of the current limitations of rexinoids in therapy.
Collapse
Affiliation(s)
- Claudio Martínez
- Departamento de Química Orgánica, Facultade de Química, CINBIO and IBIV, Universidade de Vigo, Vigo, Spain
| | - José A Souto
- Departamento de Química Orgánica, Facultade de Química, CINBIO and IBIV, Universidade de Vigo, Vigo, Spain
| | - Angel R de Lera
- Departamento de Química Orgánica, Facultade de Química, CINBIO and IBIV, Universidade de Vigo, Vigo, Spain.
| |
Collapse
|
25
|
Ren G, Bao W, Zeng Z, Zhang W, Shang C, Wang M, Su Y, Zhang XK, Zhou H. Retinoid X Receptor Alpha Nitro-ligand Z-10 and Its Optimized Derivative Z-36 Reduce β-Amyloid Plaques in Alzheimer's Disease Mouse Model. Mol Pharm 2018; 16:480-488. [PMID: 29995422 DOI: 10.1021/acs.molpharmaceut.8b00096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bexarotene, an agonist of retinoid X receptor alpha (RXRα), has been shown to increase the expression of apoE, ABCA1, and ABCG1 by activating RXR/LXR and RXR/PPAR heterodimers, resulting in amyloid β (Aβ)-protein clearance in the brain of an Alzheimer's disease (AD) mouse model and reversal of mouse cognitive deficits. Nitrostyrene derivative Z-10 is the first identified nitro-ligand of RXRα. We hypothesized that Z-10 and its derivatives have the similar effect as bexarotene. A series of Z-10 derivatives were synthesized by introducing methoxyl, hydroxyl, and methoxy groups in 2- or 4-position of naphthalene ring, respectively. Our reporter gene assays showed that the derivatives with substituted groups of methyl and methoxyl in position 2 were more potent to activate Gal4-DBD/RXRα-LBD and RXRα homodimer as well as RXRα heterodimers than the corresponding 4-substituted derivatives. The derivatives with hydroxyl substitution in either 2- or 4-position failed to activate RXRα. Consistently, the derivatives with stronger potency of RXRα activation had higher RXRα binding affinity. Z-10 and its 2-ethyoxyl substituted derivative Z-36 reduced Aβ plaques in both hippocampus and cortex of AD mouse model significantly, of which Z-36 had stronger efficacy. This may due to the stronger ability of Z-36 than Z-10 in activating RXR/LXR and RXR/PPAR heterodimers and inducing ABCA1 and ABCG1 expressions. Thus, the 2- rather than 4-position was the better site for Z-10 modification as to RXRα transactivation, and Z-36 is an optimized derivative of Z-10 as to reducing Aβ plaques in AD mouse model.
Collapse
Affiliation(s)
- Gaoang Ren
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China
| | - Wei Bao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China
| | - Weidong Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China
| | - Ce Shang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China
| | - Maosi Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China
| | - Ying Su
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China.,Cancer Center , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , California 92037 , United States
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China.,Cancer Center , Sanford Burnham Prebys Medical Discovery Institute , La Jolla , California 92037 , United States
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research , Xiamen University , Xiamen , Fujian 361102 , China
| |
Collapse
|
26
|
Miyashita Y, Numoto N, Arulmozhiraja S, Nakano S, Matsuo N, Shimizu K, Shibahara O, Fujihara M, Kakuta H, Ito S, Ikura T, Ito N, Tokiwa H. Dual conformation of the ligand induces the partial agonistic activity of retinoid X receptor α (RXRα). FEBS Lett 2018; 593:242-250. [DOI: 10.1002/1873-3468.13301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/01/2018] [Accepted: 11/14/2018] [Indexed: 01/29/2023]
Affiliation(s)
- Yurina Miyashita
- Department of Chemistry; Rikkyo University; Tokyo Japan
- AMED-CREST; Japan Agency for Medical Research and Development (AMED); Tokyo Japan
- Department of Structural Biology; Medical Research Institute; Tokyo Medical and Dental University (TMDU); Japan
| | - Nobutaka Numoto
- Department of Structural Biology; Medical Research Institute; Tokyo Medical and Dental University (TMDU); Japan
| | - Sundaram Arulmozhiraja
- Department of Chemistry; Rikkyo University; Tokyo Japan
- AMED; Japan Agency for Medical Research and Development (AMED); Tokyo Japan
| | - Shogo Nakano
- School of Food and Nutritional Sciences; University of Shizuoka; Japan
| | - Naoya Matsuo
- Department of Chemistry; Rikkyo University; Tokyo Japan
| | | | - Osamu Shibahara
- Division of Pharmaceutical Sciences; Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences; Japan
| | - Michiko Fujihara
- Division of Pharmaceutical Sciences; Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences; Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences; Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences; Japan
| | - Sohei Ito
- School of Food and Nutritional Sciences; University of Shizuoka; Japan
| | - Teikichi Ikura
- Department of Structural Biology; Medical Research Institute; Tokyo Medical and Dental University (TMDU); Japan
| | - Nobutoshi Ito
- Department of Structural Biology; Medical Research Institute; Tokyo Medical and Dental University (TMDU); Japan
| | - Hiroaki Tokiwa
- Department of Chemistry; Rikkyo University; Tokyo Japan
- AMED-CREST; Japan Agency for Medical Research and Development (AMED); Tokyo Japan
- AMED; Japan Agency for Medical Research and Development (AMED); Tokyo Japan
- Research Center for Smart Molecules; Rikkyo University; Tokyo Japan
| |
Collapse
|
27
|
LncRNA DANCR upregulates PI3K/AKT signaling through activating serine phosphorylation of RXRA. Cell Death Dis 2018; 9:1167. [PMID: 30518934 PMCID: PMC6281578 DOI: 10.1038/s41419-018-1220-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/01/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022]
Abstract
Conventional therapies and novel molecular targeted therapies against breast cancer have gained great advances over the past two decades. However, poor prognosis and low survival rate are far from expectation for improvement, particularly in patients with triple negative breast cancer (TNBC). Here, we found that lncRNA DANCR was significantly overregulated in TNBC tissues and cell lines compared with normal breast tissues or other type of breast cancer. Knockdown of DANCR suppressed TNBC proliferation both in vitro and in vivo. Further study of underlying mechanisms demonstrated that DANCR bound with RXRA and increased its serine 49/78 phosphorylation via GSK3β, resulting in activating PIK3CA transcription, and subsequently enhanced PI3K/AKT signaling and TNBC tumorigenesis. Taken together, Our findings identified DANCR as an pro-oncogene and uncoverd a new working pattern of lncRNA to mediate TNBC tumorigenesis, which may be a potential therapeutic target for improving treatment of TNBC.
Collapse
|
28
|
Watanabe M, Kakuta H. Retinoid X Receptor Antagonists. Int J Mol Sci 2018; 19:ijms19082354. [PMID: 30103423 PMCID: PMC6121510 DOI: 10.3390/ijms19082354] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 12/18/2022] Open
Abstract
Retinoid X receptor (RXR) antagonists are not only useful as chemical tools for biological research, but are also candidate drugs for the treatment of various diseases, including diabetes and allergies, although no RXR antagonist has yet been approved for clinical use. In this review, we present a brief overview of RXR structure, function, and target genes, and describe currently available RXR antagonists, their structural classification, and their evaluation, focusing on the latest research.
Collapse
Affiliation(s)
- Masaki Watanabe
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
29
|
Chen L, Wu L, Zhu L, Zhao Y. Overview of the structure-based non-genomic effects of the nuclear receptor RXRα. Cell Mol Biol Lett 2018; 23:36. [PMID: 30093910 PMCID: PMC6080560 DOI: 10.1186/s11658-018-0103-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Abstract
The nuclear receptor RXRα (retinoid X receptor-α) is a transcription factor that regulates the expression of multiple genes. Its non-genomic function is largely related to its structure, polymeric forms and modification. Previous research revealed that some non-genomic activity of RXRα occurs via formation of heterodimers with Nur77. RXRα-Nur77 heterodimers translocate from the nucleus to the mitochondria in response to certain apoptotic stimuli and this activity correlates with cell apoptosis. More recent studies revealed a significant role for truncated RXRα (tRXRα), which interacts with the p85α subunit of the PI3K/AKT signaling pathway, leading to enhanced activation of AKT and promoting cell growth in vitro and in animals. We recently reported on a series of NSAID sulindac analogs that can bind to tRXRα through a unique binding mechanism. We also identified one analog, K-80003, which can inhibit cancer cell growth by inducing tRXRα to form a tetramer, thus disrupting p85α-tRXRα interaction. This review analyzes the non-genomic effects of RXRα in normal and tumor cells, and discusses the functional differences based on RXRα protein structure (structure source: the RCSB Protein Data Bank).
Collapse
Affiliation(s)
- Liqun Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108 China
| | - Lingjuan Wu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108 China
| | - Linyan Zhu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108 China
| | - Yiyi Zhao
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108 China
| |
Collapse
|
30
|
Celastrol-Induced Nur77 Interaction with TRAF2 Alleviates Inflammation by Promoting Mitochondrial Ubiquitination and Autophagy. Mol Cell 2017; 66:141-153.e6. [PMID: 28388439 DOI: 10.1016/j.molcel.2017.03.008] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 12/19/2016] [Accepted: 03/10/2017] [Indexed: 01/09/2023]
Abstract
Mitochondria play an integral role in cell death, autophagy, immunity, and inflammation. We previously showed that Nur77, an orphan nuclear receptor, induces apoptosis by targeting mitochondria. Here, we report that celastrol, a potent anti-inflammatory pentacyclic triterpene, binds Nur77 to inhibit inflammation and induce autophagy in a Nur77-dependent manner. Celastrol promotes Nur77 translocation from the nucleus to mitochondria, where it interacts with tumor necrosis factor receptor-associated factor 2 (TRAF2), a scaffold protein and E3 ubiquitin ligase important for inflammatory signaling. The interaction is mediated by an LxxLL motif in TRAF2 and results not only in the inhibition of TRAF2 ubiquitination but also in Lys63-linked Nur77 ubiquitination. Under inflammatory conditions, ubiquitinated Nur77 resides at mitochondria, rendering them sensitive to autophagy, an event involving Nur77 interaction with p62/SQSTM1. Together, our results identify Nur77 as a critical intracellular target for celastrol and unravel a mechanism of Nur77-dependent clearance of inflamed mitochondria to alleviate inflammation.
Collapse
|
31
|
Berberine binds RXRα to suppress β-catenin signaling in colon cancer cells. Oncogene 2017; 36:6906-6918. [PMID: 28846104 PMCID: PMC5735301 DOI: 10.1038/onc.2017.296] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/22/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Berberine, an isoquinoline alkaloid, is a traditional oriental medicine used to treat diarrhea and gastroenteritis. Recently, we reported that it could inhibit the growth of intestinal polyp in animals and in patients with the familial adenomatous polyposis by downregulating β-catenin signaling. However, the intracellular target mediating the effects of berberine remains elusive. Here, we provide evidence that berberine inhibits β-catenin function via directly binding to a unique region comprising residues Gln275, Arg316 and Arg371 in nuclear receptor retinoid X receptor alpha (RXRα), where berberine concomitantly binding to and synergistically activating RXRα with 9-cis-retinoic acid (9-cis-RA), a natural ligand binding to the classical ligand-binding pocket of RXRα. Berberine binding promotes RXRα interaction with nuclear β-catenin, leading to c-Cbl mediated degradation of β-catenin, and consequently inhibits the proliferation of colon cancer cells. Furthermore, berberine suppresses the growth of human colon carcinoma xenograft in nude mice in an RXRα-dependent manner. Together, our study not only identifies RXRα as a direct protein target for berberine but also dissects their binding mode and validates that berberine indeed suppresses β-catenin signaling and cell growth in colon cancer via binding RXRα, which provide new strategies for the design of new RXRα-based antitumor agents and drug combinations.
Collapse
|
32
|
Chen L, Aleshin AE, Alitongbieke G, Zhou Y, Zhang X, Ye X, Hu M, Ren G, Chen Z, Ma Y, Zhang D, Liu S, Gao W, Cai L, Wu L, Zeng Z, Jiang F, Liu J, Zhou H, Cadwell G, Liddington RC, Su Y, Zhang XK. Modulation of nongenomic activation of PI3K signalling by tetramerization of N-terminally-cleaved RXRα. Nat Commun 2017; 8:16066. [PMID: 28714476 PMCID: PMC5520057 DOI: 10.1038/ncomms16066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 05/24/2017] [Indexed: 12/14/2022] Open
Abstract
Retinoid X receptor-alpha (RXRα) binds to DNA either as homodimers or heterodimers, but it also forms homotetramers whose function is poorly defined. We previously discovered that an N-terminally-cleaved form of RXRα (tRXRα), produced in tumour cells, activates phosphoinositide 3-kinase (PI3K) signalling by binding to the p85α subunit of PI3K and that K-80003, an anti-cancer agent, inhibits this process. Here, we report through crystallographic and biochemical studies that K-80003 binds to and stabilizes tRXRα tetramers via a ‘three-pronged’ combination of canonical and non-canonical mechanisms. K-80003 binding has no effect on tetramerization of RXRα, owing to the head–tail interaction that is absent in tRXRα. We also identify an LxxLL motif in p85α, which binds to the coactivator-binding groove on tRXRα and dissociates from tRXRα upon tRXRα tetramerization. These results identify conformational selection as the mechanism for inhibiting the nongenomic action of tRXRα and provide molecular insights into the development of RXRα cancer therapeutics. The transcription factor retinoid X receptor-alpha (RXRα) can also form homotetramers. Here the authors show that the anti-cancer agent K-80003 selectively inhibits the nongenomic action of N-terminally-cleaved RXRα in tumour cells by stabilizing its tetramerization but not that of full-length RXRα.
Collapse
Affiliation(s)
- Liqun Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China.,College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China.,Sanford Burnham Prebys Medical Discovery Institute, 10901, North Torrey Pines Road, La Jolla, California 92037, USA
| | - Alexander E Aleshin
- Sanford Burnham Prebys Medical Discovery Institute, 10901, North Torrey Pines Road, La Jolla, California 92037, USA
| | - Gulimiran Alitongbieke
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Yuqi Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Xindao Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Xiaohong Ye
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Mengjie Hu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Gaoang Ren
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Ziwen Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Yue Ma
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Duo Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Shuai Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Weiwei Gao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Lijun Cai
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Lingjuan Wu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Fuquan Jiang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Jie Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Gregory Cadwell
- Sanford Burnham Prebys Medical Discovery Institute, 10901, North Torrey Pines Road, La Jolla, California 92037, USA
| | - Robert C Liddington
- Sanford Burnham Prebys Medical Discovery Institute, 10901, North Torrey Pines Road, La Jolla, California 92037, USA
| | - Ying Su
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China.,Sanford Burnham Prebys Medical Discovery Institute, 10901, North Torrey Pines Road, La Jolla, California 92037, USA
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China.,Sanford Burnham Prebys Medical Discovery Institute, 10901, North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
33
|
Xu D, Cai L, Guo S, Xie L, Yin M, Chen Z, Zhou H, Su Y, Zeng Z, Zhang X. Virtual screening and experimental validation identify novel modulators of nuclear receptor RXRα from Drugbank database. Bioorg Med Chem Lett 2017; 27:1055-1061. [DOI: 10.1016/j.bmcl.2016.12.058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 11/16/2022]
|
34
|
Tian WJ, Qiu YQ, Chen JJ, Yao XJ, Wang GH, Dai Y, Chen HF, Yao XS. Norsampsone E, an unprecedented decarbonyl polycyclic polyprenylated acylphloroglucinol with a homoadamantyl core from Hypericum sampsonii. RSC Adv 2017. [DOI: 10.1039/c7ra05947g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Norsampsone E, an unprecedented polycyclic polyprenylated acylphloroglucinol from Hypericum sampsonii, featured a homoadamantyl skeleton with the loss of the C-4 carbonyl.
Collapse
Affiliation(s)
- Wen-Jing Tian
- Fujian Provincial Key Laboratory of Innovative Drug Target
- School of Pharmaceutical Sciences
- Xiamen University
- Xiamen 361005
- P. R. China
| | - Yu-Qin Qiu
- Institute of Traditional Chinese Medicine & Natural Products
- Jinan University
- Guangzhou 510632
- P. R. China
| | - Jun-Jie Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target
- School of Pharmaceutical Sciences
- Xiamen University
- Xiamen 361005
- P. R. China
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine
- Macau Institute for Applied Research in Medicine and Health
- Macau University of Science and Technology
- Taipa
- P. R. China
| | - Guang-Hui Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target
- School of Pharmaceutical Sciences
- Xiamen University
- Xiamen 361005
- P. R. China
| | - Yi Dai
- Institute of Traditional Chinese Medicine & Natural Products
- Jinan University
- Guangzhou 510632
- P. R. China
| | - Hai-Feng Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target
- School of Pharmaceutical Sciences
- Xiamen University
- Xiamen 361005
- P. R. China
| | - Xin-Sheng Yao
- Institute of Traditional Chinese Medicine & Natural Products
- Jinan University
- Guangzhou 510632
- P. R. China
| |
Collapse
|
35
|
Xu D, Guo S, Chen Z, Bao Y, Huang F, Xu D, Zhang X, Zeng Z, Zhou H, Zhang X, Su Y. Binding characterization, synthesis and biological evaluation of RXRα antagonists targeting the coactivator binding site. Bioorg Med Chem Lett 2016; 26:3846-9. [DOI: 10.1016/j.bmcl.2016.07.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 07/10/2016] [Accepted: 07/11/2016] [Indexed: 11/16/2022]
|
36
|
Tice CM, Zheng YJ. Non-canonical modulators of nuclear receptors. Bioorg Med Chem Lett 2016; 26:4157-64. [PMID: 27503683 DOI: 10.1016/j.bmcl.2016.07.067] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/19/2016] [Accepted: 07/27/2016] [Indexed: 12/13/2022]
Abstract
Like G protein-coupled receptors (GPCRs) and protein kinases, nuclear receptors (NRs) are a rich source of pharmaceutical targets. Over 80 NR-targeting drugs have been approved for 18 NRs. The focus of drug discovery in NRs has hitherto been on identifying ligands that bind to the canonical ligand binding pockets of the C-terminal ligand binding domains (LBDs). Due to the development of drug resistance and selectivity concerns, there has been considerable interest in exploring other, non-canonical ligand binding sites. Unfortunately, the potencies of compounds binding at other sites have generally not been sufficient for clinical development. However, the situation has changed dramatically over the last 3years, as compounds with sufficient potency have been reported for several NR targets. Here we review recent developments in this area from a medicinal chemistry point of view in the hope of stimulating further interest in this area of research.
Collapse
Affiliation(s)
- Colin M Tice
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, PA 19034, United States
| | - Ya-Jun Zheng
- Vitae Pharmaceuticals, Inc., 502 West Office Center Drive, Fort Washington, PA 19034, United States
| |
Collapse
|
37
|
Zhang X, Zhou H, Su Y. Targeting truncated RXRα for cancer therapy. Acta Biochim Biophys Sin (Shanghai) 2016; 48:49-59. [PMID: 26494413 DOI: 10.1093/abbs/gmv104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/24/2015] [Indexed: 01/08/2023] Open
Abstract
Retinoid X receptor-alpha (RXRα), a unique member of the nuclear receptor superfamily, is a well-established drug target, representing one of the most important targets for pharmacologic interventions and therapeutic applications for cancer. However, how RXRα regulates cancer cell growth and how RXRα modulators suppress tumorigenesis are poorly understood. Altered expression and aberrant function of RXRα are implicated in the development of cancer. Previously, several studies had demonstrated the presence of N-terminally truncated RXRα (tRXRα) proteins resulted from limited proteolysis of RXRα in tumor cells. Recently, we discovered that overexpression of tRXRα can promote tumor growth by interacting with tumor necrosis factor-alpha-induced phosphoinositide 3-kinase and NF-κB signal transduction pathways. We also identified nonsteroidal anti-inflammatory drug Sulindac and analogs as effective inhibitors of tRXRα activities via a unique binding mechanism. This review discusses the emerging roles of tRXRα and modulators in the regulation of cancer cell survival and death as well as inflammation and our recent understanding of tRXRα regulation by targeting the alternate binding sites on its surface.
Collapse
Affiliation(s)
- Xiaokun Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| | - Hu Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ying Su
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| |
Collapse
|
38
|
Zeng Z, Sun Z, Huang M, Zhang W, Liu J, Chen L, Chen F, Zhou Y, Lin J, Huang F, Xu L, Zhuang Z, Guo S, Alitongbieke G, Xie G, Xu Y, Lin B, Cao X, Su Y, Zhang XK, Zhou H. Nitrostyrene Derivatives Act as RXRα Ligands to Inhibit TNFα Activation of NF-κB. Cancer Res 2015; 75:2049-60. [PMID: 25795708 DOI: 10.1158/0008-5472.can-14-2435] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 01/21/2015] [Indexed: 11/16/2022]
Abstract
Retinoid X receptor alpha (RXRα) and its N-terminally truncated version, tRXRα, are widely implicated in cancer development and represent intriguing targets for cancer prevention and treatment. Successful manipulation of RXRα and tRXRα requires the identification of their modulators that could produce therapeutic effects. Here, we report that a class of nitrostyrene derivatives bind to RXRα by a unique mechanism, of which the nitro group of nitrostyrene derivatives and Cys432 of RXRα are required for binding. The binding results in the potent activation of Gal4-DBD-RXRα-LBD transactivation. However, the binding inhibits the transactivation of RXRα homodimer, which might be due to the distinct conformation of RXRα homodimer induced by these nitrostyrene derivatives. Two RXRα point mutants with Cys432 substituted with Tyr and Trp, respectively, could mimic the bindings of two nitrostyrene derivatives and have the ability of autotransactivation. In studying the functional consequences of the binding, we show that these nitrostyrene derivatives could potently inhibit the TNFα/NFκB signaling pathway in a tRXRα-dependent manner. tRXRα promotes TNFα-induced NF-κB activation through its interaction with TRAF2 and enhances TNFα-induced ubiquitination of RIP1, which is strongly inhibited by nitrostyrene derivatives. The inhibition of TNFα-induced NF-κB activation results in the synergistic effect of the combination of nitrostyrene derivatives and TNFα on the induction of cancer cell apoptosis. Together, our results show a new class of RXRα modulators that induce apoptosis of cancer cells through their unique binding mode and new mechanism of action.
Collapse
Affiliation(s)
- Zhiping Zeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe Sun
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Mingfeng Huang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Weidong Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jie Liu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Liqun Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Fan Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yuqi Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jiacheng Lin
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Fengyu Huang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zixing Zhuang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Shangjie Guo
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | | | - Guobin Xie
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yang Xu
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Bingzhen Lin
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Xihua Cao
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Ying Su
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China. Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Xiao-Kun Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China. Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California.
| | - Hu Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
39
|
Zhang XK, Su Y, Chen L, Chen F, Liu J, Zhou H. Regulation of the nongenomic actions of retinoid X receptor-α by targeting the coregulator-binding sites. Acta Pharmacol Sin 2015; 36:102-12. [PMID: 25434990 DOI: 10.1038/aps.2014.109] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 09/28/2014] [Indexed: 12/31/2022]
Abstract
Retinoid X receptor-α (RXRα), a unique member of the nuclear receptor superfamily, represents an intriguing and unusual target for pharmacologic interventions and therapeutic applications in cancer, metabolic disorders and neurodegenerative diseases. Despite the fact that the RXR-based drug Targretin (bexarotene) is currently used for treating human cutaneous T-cell lymphoma and the fact that RXRα ligands (rexinoids) show beneficial effects in the treatment of cancer and diseases, the therapeutic potential of RXRα remains unexplored. In addition to its conventional transcription regulation activity in the nucleus, RXRα can act in the cytoplasm to modulate important biological processes, such as mitochondria-dependent apoptosis, inflammation, and phosphatidylinositol 3-kinase (PI3K)/AKT-mediated cell survival. Recently, new small-molecule-binding sites on the surface of RXRα have been identified, which mediate the regulation of the nongenomic actions of RXRα by a class of small molecules derived from the nonsteroidal anti-inflammatory drug (NSAID) Sulindac. This review discusses the emerging roles of the nongenomic actions of RXRα in the RXRα signaling network, and their possible implications in cancer, metabolic and neurodegenerative disorders, as well as our current understanding of RXRα regulation by targeting alternate binding sites on its surface.
Collapse
|