1
|
Li SC, Wang B, Zhang M, Yin Q, Yang ZY, Li XT, Liang G. Induction of cytochrome P450 via upregulation of CAR and PXR: a potential mechanism for altered florfenicol metabolism by macranthoidin B in vivo. Front Pharmacol 2024; 15:1460948. [PMID: 39444610 PMCID: PMC11496122 DOI: 10.3389/fphar.2024.1460948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Macranthoidin B (MB) is a primary active component of Flos Lonicerae. In Chinese veterinary clinics, Flos Lonicerae is frequently used in combination with florfenicol to prevent and treat infections in livestock and poultry. However, potential interactions between Flos Lonicerae and florfenicol remain unclear. To systematically study these interactions, it is crucial to investigate the individual phytochemicals within Flos Lonicerae. Therefore, MB was selected for this study to assess its effect on the pharmacokinetics of florfenicol in vivo and to explore the underlying mechanisms involved. Methods Male Sprague-Dawley rats were administered MB (60 mg/kg BW) or sterile water orally for 7 consecutive days. On the 8th day, a single oral dose of florfenicol (25 mg/kg BW) was given. Florfenicol pharmacokinetics were analyzed using ultra-high performance liquid chromatography. The hepatic expression levels of cytochrome P450 (CYP1A2, CYP2C11, CYP3A1), UDP-glucuronosyltransferase (UGT1A1), P-glycoprotein (P-gp), and nuclear receptors, including constitutive androstane receptor (CAR), pregnane X receptor (PXR), and retinoid X receptor alpha (RXRα), were quantified via reverse transcription-quantitative polymerase chain reaction and Western blotting (WB). Hepatic CYP1A2 and CYP2C11 activities were measured using a cocktail method. Additionally, the subcellular expression and localization of CAR, PXR, and RXRαin hepatocytes was assessed using WB and immunofluorescence staining. Results MB significantly reduces the AUC(0-∞) and MRT(0-∞) of florfenicol. MB also markedly upregulates the mRNA and protein expression of hepatic CYP1A2 and CYP2C11, along with their catalytic activities. Substantial upregulation of CAR and PXR proteins occurs in the hepatocyte nucleus, along with significant nuclear colocalization of the transcriptionally active CAR/RXRα and PXR/RXRαheterodimers, indicating MB-induced nuclear translocation of both CAR and PXR. Discussion These findings suggest that MB-induced alterations in florfenicol pharmacokinetics, particularly its accelerated elimination, may be due to increased expression and activities of CYP1A2 and CYP2C11, with CAR and PXR potentially involved in these regulatory effects. Further investigation is yet needed to fully elucidate the clinical implications of these interactions concerning the efficacy of florfenicol in veterinary medicine.
Collapse
Affiliation(s)
- Si-cong Li
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Veterinary Natural Medicine Research and Good Clinical Practice Experimental Animal Centre, Lezhi, China
| | - Bin Wang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Veterinary Natural Medicine Research and Good Clinical Practice Experimental Animal Centre, Lezhi, China
| | - Min Zhang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Veterinary Natural Medicine Research and Good Clinical Practice Experimental Animal Centre, Lezhi, China
| | - Qin Yin
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
| | - Zi-yi Yang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Veterinary Natural Medicine Research and Good Clinical Practice Experimental Animal Centre, Lezhi, China
| | - Xu-ting Li
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Veterinary Natural Medicine Research and Good Clinical Practice Experimental Animal Centre, Lezhi, China
| | - Ge Liang
- Animal Breeding and Genetics key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
- Veterinary Natural Medicine Research and Good Clinical Practice Experimental Animal Centre, Lezhi, China
| |
Collapse
|
2
|
Al-Bari MAA, Peake N, Eid N. Tuberculosis-diabetes comorbidities: Mechanistic insights for clinical considerations and treatment challenges. World J Diabetes 2024; 15:853-866. [PMID: 38766427 PMCID: PMC11099355 DOI: 10.4239/wjd.v15.i5.853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/08/2024] [Accepted: 03/21/2024] [Indexed: 05/10/2024] Open
Abstract
Tuberculosis (TB) remains a leading cause of death among infectious diseases, particularly in poor countries. Viral infections, multidrug-resistant and ex-tensively drug-resistant TB strains, as well as the coexistence of chronic illnesses such as diabetes mellitus (DM) greatly aggravate TB morbidity and mortality. DM [particularly type 2 DM (T2DM)] and TB have converged making their control even more challenging. Two contemporary global epidemics, TB-DM behaves like a syndemic, a synergistic confluence of two highly prevalent diseases. T2DM is a risk factor for developing more severe forms of multi-drug resistant-TB and TB recurrence after preventive treatment. Since a bidirectional relationship exists between TB and DM, it is necessary to concurrently treat both, and promote recommendations for the joint management of both diseases. There are also some drug-drug interactions resulting in adverse treatment outcomes in TB-DM patients including treatment failure, and reinfection. In addition, autophagy may play a role in these comorbidities. Therefore, the TB-DM comorbidities present several health challenges, requiring a focus on multidisciplinary collaboration and integrated strategies, to effectively deal with this double burden. To effectively manage the comorbidity, further screening in affected countries, more suitable drugs, and better treatment strategies are required.
Collapse
Affiliation(s)
| | - Nicholas Peake
- Biosciences and Chemistry and Biomolecular Research Centre, Sheffield Hallam University, Sheffield S1 1WB, United Kingdom
| | - Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
3
|
Structural Biology Inspired Development of a Series of Human Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) Ligands: From Agonist to Antagonist. Int J Mol Sci 2023; 24:ijms24043940. [PMID: 36835351 PMCID: PMC9960108 DOI: 10.3390/ijms24043940] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Recent progress in the structural and molecular pharmacological understanding of the nuclear receptor, peroxisome proliferator-activated receptor gamma (hPPARγ)-a transcription factor with pleiotropic effects on biological responses-has enabled the investigation of various graded hPPARγ ligands (full agonist, partial agonist, and antagonist). Such ligands are useful tools to investigate the functions of hPPARγ in detail and are also candidate drugs for the treatment of hPPARγ-mediated diseases, such as metabolic syndrome and cancer. This review summarizes our medicinal chemistry research on the design, synthesis, and pharmacological evaluation of a covalent-binding and non-covalent-binding hPPARγ antagonist, both of which have been created based on our working hypothesis of the helix 12 (H12) holding induction/inhibition concept. X-ray crystallographic analyses of our representative antagonists complexed with an hPPARγ ligand binding domain (LBD) indicated the unique binding modes of hPPARγ LBD, which are quite different from the binding modes observed for hPPARγ agonists and partial agonists.
Collapse
|
4
|
Iijima T, Katoh M, Takedomi K, Yamamoto Y, Akatsuka H, Shirata N, Nishi A, Takakuwa M, Watanabe Y, Munakata H, Koyama N, Ikeda T, Iguchi T, Kato H, Kikkawa K, Kawaguchi T. Discovery of Apararenone (MT-3995) as a Highly Selective, Potent, and Novel Nonsteroidal Mineralocorticoid Receptor Antagonist. J Med Chem 2022; 65:8127-8143. [PMID: 35652647 DOI: 10.1021/acs.jmedchem.2c00402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Overactivation of the mineralocorticoid receptor (MR) is involved in many diseases, such as hypertension, kidney disease, and heart failure. Thus, MR antagonists (MRAs) are expected to be beneficial to patients with these diseases. In order to identify novel nonsteroidal MRAs that overcome the issues of already marketed steroidal MRAs, we searched for new compounds guided by our hypothesis that T-shaped compounds with a hydrophobic core structure, two polar functional groups at both extremities able to interact with MR, and a bulky substituent that can interfere with the folding of the C-terminal helix 12 may exhibit antagonist activity toward MR. We discovered that the novel 1,4-benzoxazin-3-one derivative 19 (apararenone: MT-3995) acted as a highly selective and potent nonsteroidal MRA. Apararenone exhibited a more potent antihypertensive and organ-protective activity than steroidal MRA eplerenone in a primary aldosteronism rat model obtained by infusing aldosterone in uninephrectomized rats.
Collapse
Affiliation(s)
- Toru Iijima
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan.,Lead Exploration Unit, Drug Discovery Initiative, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Makoto Katoh
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Kei Takedomi
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Yasuo Yamamoto
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Hidenori Akatsuka
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Naritoshi Shirata
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Akito Nishi
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Misae Takakuwa
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Yoshinori Watanabe
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Hitomi Munakata
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Naomi Koyama
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Tomoko Ikeda
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Taku Iguchi
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Harutoshi Kato
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Kohei Kikkawa
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| | - Takayuki Kawaguchi
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama 227-0033, Japan
| |
Collapse
|
5
|
Miyachi H. Structural Biology-Based Exploration of Subtype-Selective Agonists for Peroxisome Proliferator-Activated Receptors. Int J Mol Sci 2021; 22:ijms22179223. [PMID: 34502131 PMCID: PMC8430769 DOI: 10.3390/ijms22179223] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Progress in understanding peroxisome proliferator-activated receptor (PPAR) subtypes as nuclear receptors that have pleiotropic effects on biological responses has enabled the exploration of new subtype-selective PPAR ligands. Such ligands are useful chemical biology/pharmacological tools to investigate the functions of PPARs and are also candidate drugs for the treatment of PPAR-mediated diseases, such as metabolic syndrome, inflammation and cancer. This review summarizes our medicinal chemistry research of more than 20 years on the design, synthesis, and pharmacological evaluation of subtype-selective PPAR agonists, which has been based on two working hypotheses, the ligand superfamily concept and the helix 12 (H12) holding induction concept. X-ray crystallographic analyses of our agonists complexed with each PPAR subtype validate our working hypotheses.
Collapse
Affiliation(s)
- Hiroyuki Miyachi
- Lead Exploration Unit, Drug Discovery Initiative, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
6
|
Yoshikawa C, Ishida H, Ohashi N, Itoh T. Synthesis of a Coumarin-Based PPARγ Fluorescence Probe for Competitive Binding Assay. Int J Mol Sci 2021; 22:4034. [PMID: 33919837 PMCID: PMC8070791 DOI: 10.3390/ijms22084034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/10/2021] [Accepted: 04/11/2021] [Indexed: 12/28/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a molecular target of metabolic syndrome and inflammatory disease. PPARγ is an important nuclear receptor and numerous PPARγ ligands were developed to date; thus, efficient assay methods are important. Here, we investigated the incorporation of 7-diethylamino coumarin into the PPARγ agonist rosiglitazone and used the compound in a binding assay for PPARγ. PPARγ-ligand-incorporated 7-methoxycoumarin, 1, showed weak fluorescence intensity in a previous report. We synthesized PPARγ-ligand-incorporating coumarin, 2, in this report, and it enhanced the fluorescence intensity. The PPARγ ligand 2 maintained the rosiglitazone activity. The obtained partial agonist 6 appeared to act through a novel mechanism. The fluorescence intensity of 2 and 6 increased by binding to the ligand binding domain (LBD) of PPARγ and the affinity of reported PPARγ ligands were evaluated using the probe.
Collapse
Affiliation(s)
| | | | | | - Toshimasa Itoh
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan; (C.Y.); (H.I.); (N.O.)
| |
Collapse
|
7
|
Mays SG, Stec J, Liu X, D'Agostino EH, Whitby RJ, Ortlund EA. Enantiomer-specific activities of an LRH-1 and SF-1 dual agonist. Sci Rep 2020; 10:22279. [PMID: 33335203 PMCID: PMC7747700 DOI: 10.1038/s41598-020-79251-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/09/2020] [Indexed: 12/28/2022] Open
Abstract
Chirality is an important consideration in drug development: it can influence recognition of the intended target, pharmacokinetics, and off-target effects. Here, we investigate how chirality affects the activity and mechanism of action of RJW100, a racemic agonist of the nuclear receptors liver receptor homolog-1 (LRH-1) and steroidogenic factor-1 (SF-1). LRH-1 and SF-1 modulators are highly sought as treatments for metabolic and neoplastic diseases, and RJW100 has one of the few scaffolds shown to activate them. However, enantiomer-specific effects on receptor activation are poorly understood. We show that the enantiomers have similar binding affinities, but RR-RJW100 stabilizes both receptors and is 46% more active than SS-RJW100 in LRH-1 luciferase reporter assays. We present an LRH-1 crystal structure that illuminates striking mechanistic differences: SS-RJW100 adopts multiple configurations in the pocket and fails to make an interaction critical for activation by RR-RJW100. In molecular dynamics simulations, SS-RJW100 attenuates intramolecular signalling important for coregulator recruitment, consistent with previous observations that it weakly recruits coregulators in vitro. These studies provide a rationale for pursuing enantiomerically pure RJW100 derivatives: they establish RR-RJW100 as the stronger LRH-1 agonist and identify a potential for optimizing the SS-RJW100 scaffold for antagonist design.
Collapse
Affiliation(s)
- Suzanne G Mays
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
- Centre for Genomic Regulation, Carrer Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Józef Stec
- School of Chemistry, University of Southampton, Southampton, Hants, SO17, United Kingdom
- Department of Pharmaceutical Sciences, College of Pharmacy, Marshall B. Ketchum University, 2575 Yorba Linda Blvd, Fullerton, CA, 82831, USA
| | - Xu Liu
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Emma H D'Agostino
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Richard J Whitby
- School of Chemistry, University of Southampton, Southampton, Hants, SO17, United Kingdom
| | - Eric A Ortlund
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Kaupang Å, Hansen TV. The PPAR Ω Pocket: Renewed Opportunities for Drug Development. PPAR Res 2020; 2020:9657380. [PMID: 32695150 PMCID: PMC7351019 DOI: 10.1155/2020/9657380] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
The past decade of PPARγ research has dramatically improved our understanding of the structural and mechanistic bases for the diverging physiological effects of different classes of PPARγ ligands. The discoveries that lie at the heart of these developments have enabled the design of a new class of PPARγ ligands, capable of isolating central therapeutic effects of PPARγ modulation, while displaying markedly lower toxicities than previous generations of PPARγ ligands. This review examines the emerging framework around the design of these ligands and seeks to unite its principles with the development of new classes of ligands for PPARα and PPARβ/δ. The focus is on the relationships between the binding modes of ligands, their influence on PPAR posttranslational modifications, and gene expression patterns. Specifically, we encourage the design and study of ligands that primarily bind to the Ω pockets of PPARα and PPARβ/δ. In support of this development, we highlight already reported ligands that if studied in the context of this new framework may further our understanding of the gene programs regulated by PPARα and PPARβ/δ. Moreover, recently developed pharmacological tools that can be utilized in the search for ligands with new binding modes are also presented.
Collapse
Affiliation(s)
- Åsmund Kaupang
- Section for Pharmaceutical Chemistry, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway
| | - Trond Vidar Hansen
- Section for Pharmaceutical Chemistry, Department of Pharmacy, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
9
|
Ammazzalorso A, Bruno I, Florio R, De Lellis L, Laghezza A, Cerchia C, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Tortorella P, Veschi S, Loiodice F, Lavecchia A, Cama A, Amoroso R. Sulfonimide and Amide Derivatives as Novel PPARα Antagonists: Synthesis, Antiproliferative Activity, and Docking Studies. ACS Med Chem Lett 2020; 11:624-632. [PMID: 32435362 PMCID: PMC7236056 DOI: 10.1021/acsmedchemlett.9b00666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/03/2020] [Indexed: 12/30/2022] Open
Abstract
An agonist-antagonist switching strategy was performed to discover novel PPARα antagonists. Phenyldiazenyl derivatives of fibrates were developed, bearing sulfonimide or amide functional groups. A second series of compounds was synthesized, replacing the phenyldiazenyl moiety with amide or urea portions. Final compounds were screened by transactivation assay, showing good PPARα antagonism and selectivity at submicromolar concentrations. When tested in cancer cell models expressing PPARα, selected derivatives induced marked effects on cell viability. Notably, 3c, 3d, and 10e displayed remarkable antiproliferative effects in two paraganglioma cell lines, with CC50 lower than commercial PPARα antagonist GW6471 and a negligible toxicity on normal fibroblast cells. Docking studies were also performed to elucidate the binding mode of these compounds and to help interpretation of SAR data.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Isabella Bruno
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Rosalba Florio
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Laura De Lellis
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Antonio Laghezza
- Department of Pharmacy-Drug Science, University of Bari “Aldo Moro”, Via E. Orabona 4, 70126 Bari, Italy
| | - Carmen Cerchia
- Department of Pharmacy, “Drug Discovery” Laboratory, University of Napoli “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy
| | - Barbara De Filippis
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Marialuigia Fantacuzzi
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Letizia Giampietro
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Cristina Maccallini
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Paolo Tortorella
- Department of Pharmacy-Drug Science, University of Bari “Aldo Moro”, Via E. Orabona 4, 70126 Bari, Italy
| | - Serena Veschi
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| | - Fulvio Loiodice
- Department of Pharmacy-Drug Science, University of Bari “Aldo Moro”, Via E. Orabona 4, 70126 Bari, Italy
| | - Antonio Lavecchia
- Department of Pharmacy, “Drug Discovery” Laboratory, University of Napoli “Federico II”, Via D. Montesano 49, 80131 Napoli, Italy
| | - Alessandro Cama
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
- Center for Advanced Studies and Technology CAST, Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via Dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
10
|
Dart DA, Ashelford K, Jiang WG. AR mRNA stability is increased with AR-antagonist resistance via 3'UTR variants. Endocr Connect 2020; 9:9-19. [PMID: 31778359 PMCID: PMC6933836 DOI: 10.1530/ec-19-0340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022]
Abstract
Advanced prostate cancer is often treated with AR antagonists which target the androgen receptor (AR) on which the growth of the tumour depends. Prostate cancer often develops AR-antagonist resistance via a plethora of mechanisms, many of which are as yet unknown, but it is thought that AR upregulation or AR ligand-binding site mutations, may be responsible. Here we describe the production of cell lines based on LNCaP and VCaP, with acquired resistance to the clinically relevant AR antagonists, bicalutamide and enzalutamide. In these resistant cells, we observed, via RNA-seq, that new variants in the 3'UTR of the AR mRNA were detectable and that the levels were increased both with AR-antagonist treatment and with hormonal starvation. Around 20% of AR transcripts showed a 3 kb deletion within the 6.7 kb 3'UTR sequence. Actinomycin D and luciferase fusion studies indicated that this shorter mRNA variant was inherently more stable in anti-androgen-resistant cell lines. Of additional interest was that the AR UTR variant could be detected in the sera of prostate cancer patients in a cohort of serum samples collected from patients of Gleason grades 6-10, with an increasing level correlated to increasing grade. We hypothesise that the shorter AR UTR variant is a survival adaptation to low hormone levels and/or AR-antagonist treatment in these cells, where a more stable mRNA may allow higher levels of AR expression under these conditions.
Collapse
Affiliation(s)
- D A Dart
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, Wales, UK
- Imperial College London, London, UK
| | - K Ashelford
- Division of Cancer and Genetics, Wales Gene Park, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - W G Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, Wales, UK
| |
Collapse
|
11
|
Liu YY, Feng XY, Jia WQ, Jing Z, Xu WR, Cheng XC. Virtual identification of novel PPARα/γ dual agonists by 3D-QSAR, molecule docking and molecular dynamics studies. J Biomol Struct Dyn 2019; 38:2672-2685. [DOI: 10.1080/07391102.2019.1656110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Ya-Ya Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiao-Yan Feng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wen-Qing Jia
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Zhi Jing
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wei-Ren Xu
- Tianjin Key Laboratory of Molecular Design and Drug Discovery, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Xian-Chao Cheng
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
12
|
Wang F, Wang H, Wu Y, Wang L, Zhang L, Ye X, Peng D, Chen W. Activation of Pregnane X Receptor-Cytochrome P450s Axis: A Possible Reason for the Enhanced Accelerated Blood Clearance Phenomenon of PEGylated Liposomes In Vivo. Drug Metab Dispos 2019; 47:785-793. [PMID: 31118196 DOI: 10.1124/dmd.119.086769] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/16/2019] [Indexed: 12/30/2022] Open
Abstract
Recently, we reported that repeated injection of PEGylated liposomes (PEG-L) at certain intervals to the same rat lead to the disappearance of their long-circulation properties, referred to as the "accelerated blood clearance (ABC) phenomenon". Evidence from our recent studies suggested that cytochrome P450s (P450s) contribute to induction of the ABC phenomenon, a possibility that had been previously ignored. However, few details are known about the mechanism for induction of P450s. The present study was undertaken to investigate the roles in the ABC phenomenon of pregnane X receptor (PXR) and constitutive androstane receptor (CAR), the major upstream transcriptional regulators of the P450 genes, including CYP3A1, CYP2C6, and CYP1A2. The results demonstrated that expression of rat PXR and CAR was significantly increased in the ABC phenomenon and was accompanied by elevated CYP3A1, CYP2C6, and CYP1A2 levels. Further findings revealed that PXR but not CAR protein was substantially upregulated in the hepatocyte nucleus, together with marked nuclear colocalization of the PXR-retinoid X receptor alpha (RXRα) transcriptionally active heterodimer, indicating that nuclear translocation of PXR was induced in the ABC phenomenon, whereas nuclear translocation of CAR was not observed. Notably, pretreatment with the specific PXR inducer dexamethasone significantly induced accelerated systemic clearance of the subsequent injection of PEG-L, associating with increased nuclear colocalization of PXR-RXRα These results revealed that the induction of P450s in the ABC phenomenon may be attributable largely to the activation of PXR induced by sequential injections of PEG-L, thus confirming the crucial involvement of the PXR-P450s axis in promoting the ABC phenomenon. SIGNIFICANCE STATEMENT: The results of this study revealed that the induction of P450s in the ABC phenomenon may be largely attributable to the activation of PXR induced by sequential injections of PEG-L, thus confirming the crucial involvement of the PXR-P450s axis in promoting the ABC phenomenon. The data may help to extend our insights into 1) the role of P450s, which are regulated by the liver-enriched nuclear receptor PXR, in the ABC phenomenon, and 2) the therapeutic potential of targeting the PXR-P450 axis for reducing the magnitude of the ABC phenomenon in clinical practice.
Collapse
Affiliation(s)
- Fengling Wang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Huihui Wang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Yifan Wu
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Lei Wang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Ling Zhang
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Xi Ye
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Daiyin Peng
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| | - Weidong Chen
- Institute of Drug Metabolism (F.W., H.W., Y.W., L.W., L.Z., D.P., W.C.) and Institute of Pharmaceutics (W.C.), School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China; Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China (L.W., L.Z., D.P., W.C.); and Synergetic Innovation Center of Anhui Authentic Chinese Medicine Quality Improvement, Hefei, China (D.P., W.C.)
| |
Collapse
|
13
|
Dart DA, Kandil S, Tommasini-Ghelfi S, Serrano de Almeida G, Bevan CL, Jiang W, Westwell AD. Novel Trifluoromethylated Enobosarm Analogues with Potent Antiandrogenic Activity In Vitro and Tissue Selectivity In Vivo. Mol Cancer Ther 2018; 17:1846-1858. [PMID: 29895558 DOI: 10.1158/1535-7163.mct-18-0037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/10/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022]
Abstract
Prostate cancer often develops antiandrogen resistance, possibly via androgen receptor (AR) mutations, which change antagonists to agonists. Novel therapies with increased anticancer activity, while overcoming current drug resistance are urgently needed. Enobosarm has anabolic effects on muscle and bone while having no effect on the prostate. Here, we describe the activity of novel chemically modified enobosarm analogues. The rational addition of bis-trifluoromethyl groups into ring B of enobosarm, profoundly modified their activity, pharmacokinetic and tissue distribution profiles. These chemical structural modifications resulted in an improved AR binding affinity-by increasing the molecular occupational volume near helix 12 of AR. In vitro, the analogues SK33 and SK51 showed very potent antiandrogenic activity, monitored using LNCaP/AR-Luciferase cells where growth, PSA and luciferase activity were used as AR activity measurements. These compounds were 10-fold more potent than bicalutamide and 100-fold more potent than enobosarm within the LNCaP model. These compounds were also active in LNCaP/BicR cells with acquired bicalutamide resistance. In vivo, using the AR-Luc reporter mice, these drugs showed potent AR inhibitory activity in the prostate and other AR-expressing tissues, e.g., testes, seminal vesicles, and brain. These compounds do not inhibit AR activity in the skeletal muscle, and spleen, thus indicating a selective tissue inhibitory profile. These compounds were also active in vivo in the Pb-Pten deletion model. SK33 and SK51 have significantly different and enhanced activity profiles compared with enobosarm and are ideal candidates for further development for prostate cancer therapy with potentially fewer side effects. Mol Cancer Ther; 17(9); 1846-58. ©2018 AACR.
Collapse
Affiliation(s)
- D Alwyn Dart
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, Wales, United Kingdom. .,Androgen Signaling Laboratory, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Sahar Kandil
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Serena Tommasini-Ghelfi
- Androgen Signaling Laboratory, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Gilberto Serrano de Almeida
- Androgen Signaling Laboratory, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Charlotte L Bevan
- Androgen Signaling Laboratory, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Wenguo Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, Wales, United Kingdom
| | - Andrew D Westwell
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| |
Collapse
|
14
|
Wang Y, Cai W, Tang T, Liu Q, Yang T, Yang L, Ma Y, Zhang G, Huang Y, Song X, Orband-Miller LA, Wu Q, Zhou L, Xiang Z, Xiang JN, Leung S, Shao L, Lin X, Lobera M, Ren F. From RORγt Agonist to Two Types of RORγt Inverse Agonists. ACS Med Chem Lett 2018; 9:120-124. [PMID: 29456799 DOI: 10.1021/acsmedchemlett.7b00476] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/22/2018] [Indexed: 11/28/2022] Open
Abstract
Biaryl amides as new RORγt modulators were discovered. The crystal structure of biaryl amide agonist 6 in complex with RORγt ligand binding domain (LBD) was resolved, and both "short" and "long" inverse agonists were obtained by removing from 6 or adding to 6 a proper structural moiety. While "short" inverse agonist (8) recruits a corepressor peptide and dispels a coactivator peptide, "long" inverse agonist (9) dispels both. The two types of inverse agonists can be utilized as potential tools to study mechanisms of Th17 transcriptional network inhibition and related disease biology.
Collapse
Affiliation(s)
- Yonghui Wang
- School
of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong, Shanghai 201203, China
| | - Wei Cai
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Ting Tang
- School
of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong, Shanghai 201203, China
| | - Qian Liu
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Ting Yang
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Liuqing Yang
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Yingli Ma
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Guifeng Zhang
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Yafei Huang
- School
of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong, Shanghai 201203, China
| | - Xiaoxia Song
- School
of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong, Shanghai 201203, China
| | - Lisa A. Orband-Miller
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Qianqian Wu
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Ling Zhou
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Zhijun Xiang
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Jia-Ning Xiang
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Stewart Leung
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Liming Shao
- School
of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong, Shanghai 201203, China
| | - Xichen Lin
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| | - Mercedes Lobera
- Research
and Development, GlaxoSmithKline, 200 Technology Square, Suite 602, Cambridge, Massachusetts 02139, United States
| | - Feng Ren
- Research and Development, GlaxoSmithKline, No. 3 Building, 898 Halei Road, Pudong, Shanghai 201203, China
| |
Collapse
|
15
|
De Lellis L, Cimini A, Veschi S, Benedetti E, Amoroso R, Cama A, Ammazzalorso A. The Anticancer Potential of Peroxisome Proliferator-Activated Receptor Antagonists. ChemMedChem 2018; 13:209-219. [PMID: 29276815 DOI: 10.1002/cmdc.201700703] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/17/2017] [Indexed: 12/13/2022]
Abstract
The effects on cancer-cell proliferation and differentiation mediated by peroxisome proliferator-activated receptors (PPARs) have been widely studied, and pleiotropic outcomes in different cancer models and under different experimental conditions have been obtained. Interestingly, few studies report and little preclinical evidence supports the potential antitumor activity of PPAR antagonists. This review focuses on recent findings on the antitumor in vitro and in vivo effects observed for compounds able to inhibit the three PPAR subtypes in different tumor models, providing a rationale for the use of PPAR antagonists in the treatment of tumors expressing the corresponding receptors.
Collapse
Affiliation(s)
- Laura De Lellis
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi (Aq), Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, 1900 N. 12th Street, Philadelphia, PA, 19122, USA
| | - Serena Veschi
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rosa Amoroso
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | | |
Collapse
|
16
|
Identification of a novel selective PPARγ ligand with a unique binding mode and improved therapeutic profile in vitro. Sci Rep 2017; 7:41487. [PMID: 28128331 PMCID: PMC5270246 DOI: 10.1038/srep41487] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 12/21/2016] [Indexed: 01/11/2023] Open
Abstract
Thiazolidinediones (TZD) function as potent anti-diabetic drugs through their direct action on the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ), but their therapeutic benefits are compromised by severe side effects. To address this concern, here we developed a potent “hit” compound, VSP-51, which is a novel selective PPARγ-modulating ligand with improved therapeutic profiles in vitro compared to the multi-billion dollar TZD drug rosiglitazone (Rosi). Unlike Rosi, VSP-51 is a partial agonist of PPARγ with improved insulin sensitivity due to its ability to bind PPARγ with high affinity without stimulating adipocyte differentiation and the expression of adipogenesis-related genes. We have determined the crystal structure of the PPARγ ligand-binding domain (LBD) in complex with VSP-51, which revealed a unique mode of binding for VSP-51 and provides the molecular basis for the discrimination between VSP-51 from TZDs and other ligands such as telmisartan, SR1663 and SR1664. Taken together, our findings demonstrate that: a) VSP-51 can serve as a promising candidate for anti-diabetic drug discovery; and b) provide a rational basis for the development of future pharmacological agents targeting PPARγ with advantages over current TZD drugs.
Collapse
|
17
|
Zhu J, Janesick A, Wu L, Hu L, Tang W, Blumberg B, Shi H. The unexpected teratogenicity of RXR antagonist UVI3003 via activation of PPARγ in Xenopus tropicalis. Toxicol Appl Pharmacol 2017; 314:91-97. [PMID: 27894914 PMCID: PMC5183475 DOI: 10.1016/j.taap.2016.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/31/2016] [Accepted: 11/23/2016] [Indexed: 01/28/2023]
Abstract
The RXR agonist (triphenyltin, TPT) and the RXR antagonist (UVI3003) both show teratogenicity and, unexpectedly, induce similar malformations in Xenopus tropicalis embryos. In the present study, we exposed X. tropicalis embryos to UVI3003 in seven specific developmental windows and identified changes in gene expression. We further measured the ability of UVI3003 to activate Xenopus RXRα (xRXRα) and PPARγ (xPPARγ) in vitro and in vivo. We found that UVI3003 activated xPPARγ either in Cos7 cells (in vitro) or Xenopus embryos (in vivo). UVI3003 did not significantly activate human or mouse PPARγ in vitro; therefore, the activation of Xenopus PPARγ by UVI3003 is novel. The ability of UVI3003 to activate xPPARγ explains why UVI3003 and TPT yield similar phenotypes in Xenopus embryos. Our results indicate that activating PPARγ leads to teratogenic effects in Xenopus embryos. More generally, we infer that chemicals known to specifically modulate mammalian nuclear hormone receptors cannot be assumed to have the same activity in non-mammalian species, such as Xenopus. Rather they must be tested for activity and specificity on receptors of the species in question to avoid making inappropriate conclusions.
Collapse
Affiliation(s)
- Jingmin Zhu
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Amanda Janesick
- Department of Developmental and Cell Biology, University of California, Irvine, California 92697-2300, USA
| | - Lijiao Wu
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Lingling Hu
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China
| | - Weiyi Tang
- Department of Developmental and Cell Biology, University of California, Irvine, California 92697-2300, USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, University of California, Irvine, California 92697-2300, USA
| | - Huahong Shi
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
18
|
Ammazzalorso A, Carrieri A, Verginelli F, Bruno I, Carbonara G, D'Angelo A, De Filippis B, Fantacuzzi M, Florio R, Fracchiolla G, Giampietro L, Giancristofaro A, Maccallini C, Cama A, Amoroso R. Synthesis, in vitro evaluation, and molecular modeling investigation of benzenesulfonimide peroxisome proliferator-activated receptors α antagonists. Eur J Med Chem 2016; 114:191-200. [PMID: 26974385 DOI: 10.1016/j.ejmech.2016.02.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 02/02/2023]
Abstract
Recent evidences suggest a moderate activation of Peroxisome Proliferator-Activated Receptors (PPARs) could be favorable in metabolic diseases, reducing side effects given from full agonists. PPAR partial agonists and antagonists represent, to date, interesting tools to better elucidate biological processes modulated by these receptors. In this work are reported new benzenesulfonimide compounds able to block PPARα, synthesized and tested by transactivation assays and gene expression analysis. Some of these compounds showed a dose-dependent antagonistic behavior on PPARα, submicromolar potency, different profiles of selectivity versus PPARγ, and a repressive effect on CPT1A expression. Dockings and molecular dynamics on properly selected benzenesulfonimide derivatives furnished fresh insights into the molecular determinant most likely responsible for PPARα antagonism.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy.
| | - Antonio Carrieri
- Dipartimento di Farmacia-Scienze del Farmaco, Università di Bari, Italy
| | - Fabio Verginelli
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy; Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Isabella Bruno
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | | | - Alessandra D'Angelo
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Barbara De Filippis
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Marialuigia Fantacuzzi
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Rosalba Florio
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy; Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | | | - Letizia Giampietro
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Antonella Giancristofaro
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Cristina Maccallini
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy
| | - Alessandro Cama
- Unità di Patologia Generale, Dipartimento di Farmacia, Università "G. d'Annunzio", Chieti, Italy; Aging Research Center (Ce.S.I.), Università "G. d'Annunzio", Chieti, Italy
| | - Rosa Amoroso
- Unità di Chimica Farmaceutica, Dipartimento di Farmacia, Università"G. d'Annunzio", Chieti, Italy.
| |
Collapse
|
19
|
Ammazzalorso A, Tricca ML, Bruno I, De Filippis B, Di Matteo M, Fantacuzzi M, Giampietro L, Maccallini C, Mollica A, Amoroso R. Titanium-Promoted Acylation of Sulfonamides toN-Acylsulfonamide PPARαAntagonists. SYNTHETIC COMMUN 2015. [DOI: 10.1080/00397911.2015.1092552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
20
|
Kaupang Å, Paulsen SM, Steindal CC, Ravna AW, Sylte I, Halvorsen TG, Thoresen GH, Hansen TV. Synthesis, biological evaluation and molecular modeling studies of the PPARβ/δ antagonist CC618. Eur J Med Chem 2015; 94:229-36. [PMID: 25768705 DOI: 10.1016/j.ejmech.2015.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 02/06/2023]
Abstract
Herein, we describe the synthesis, biological evaluation and molecular docking of the selective PPARβ/δ antagonist (4-methyl-2-(4-(trifluoromethyl)phenyl)-N-(2-(5-(trifluoromethyl)-pyridin-2-ylsulfonyl)ethyl)thiazole-5-carboxamide)), CC618. Results from in vitro luciferase reporter gene assays against the three known human PPAR subtypes revealed that CC618 selectively antagonizes agonist-induced PPARβ/δ activity with an IC50 = 10.0 μM. As observed by LC-MS/MS analysis of tryptic digests, the treatment of PPARβ/δ with CC618 leads to a covalent modification of Cys249, located centrally in the PPARβ/δ ligand binding pocket, corresponding to the conversion of its thiol moiety to a 5-trifluoromethyl-2-pyridylthioether. Finally, molecular docking is employed to shed light on the mode of action of the antagonist and its structural consequences for the PPARβ/δ ligand binding pocket.
Collapse
Affiliation(s)
- Åsmund Kaupang
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO BOX 1068, Blindern, N-0316 Oslo, Norway
| | - Steinar Martin Paulsen
- MabCent-SFI, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Calin C Steindal
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO BOX 1068, Blindern, N-0316 Oslo, Norway
| | - Aina W Ravna
- Medical Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Ingebrigt Sylte
- Medical Pharmacology and Toxicology, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, N-9037 Tromsø, Norway
| | - Trine G Halvorsen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO BOX 1068, Blindern, N-0316 Oslo, Norway
| | - G Hege Thoresen
- Department of Pharmaceutical Biosciences, School of Pharmacy, Faculty of Medicine, University of Oslo, PO BOX 1068, Blindern, N-0316 Oslo, Norway; Department of Pharmacology, Institute of Clinical Medicine, Faculty of Medicine, Oslo University Hospital, PO Box 1057, Blindern, N-0316 Oslo, Norway
| | - Trond Vidar Hansen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, PO BOX 1068, Blindern, N-0316 Oslo, Norway.
| |
Collapse
|
21
|
Ohta K, Chiba Y, Kaise A, Endo Y. Structure-activity relationship study of diphenylamine-based estrogen receptor (ER) antagonists. Bioorg Med Chem 2015; 23:861-7. [PMID: 25614118 DOI: 10.1016/j.bmc.2014.12.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 12/31/2022]
Abstract
We have reported the design and synthesis of novel estrogen receptor (ER) agonists with a diphenylamine skeleton, which has several advantages over the formerly used diphenylmethane skeleton for drug development. Here, we confirmed the versatility of the diphenylamine skeleton by designing and synthesizing ER antagonist candidates bearing a basic alkylamino side chain on one of the two phenol groups of the diphenylamine agonist core structure. Among the tested compounds, cyclic alkylamine-containing derivatives showed more potent ER-antagonistic activity than the corresponding acyclic derivatives in cell proliferation assay using the MCF-7 cell line. Compound 5e showed the most potent antiestrogenic activity (IC50: 1.3×10(-7)M), being 10times more potent than tamoxifen.
Collapse
Affiliation(s)
- Kiminori Ohta
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Yuki Chiba
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Asako Kaise
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| | - Yasuyuki Endo
- Faculty of Pharmaceutical Sciences, Tohoku Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.
| |
Collapse
|
22
|
Ohashi M, Gamo K, Tanaka Y, Waki M, Beniyama Y, Matsuno K, Wada J, Tenta M, Eguchi J, Makishima M, Matsuura N, Oyama T, Miyachi H. Structural design and synthesis of arylalkynyl amide-type peroxisome proliferator-activated receptor γ (PPARγ)-selective antagonists based on the helix12-folding inhibition hypothesis. Eur J Med Chem 2015; 90:53-67. [DOI: 10.1016/j.ejmech.2014.11.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/05/2014] [Accepted: 11/06/2014] [Indexed: 01/13/2023]
|
23
|
Ammazzalorso A, De Filippis B, Giampietro L, Amoroso R. Blocking the peroxisome proliferator-activated receptor (PPAR): an overview. ChemMedChem 2013; 8:1609-16. [PMID: 23939910 DOI: 10.1002/cmdc.201300250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Indexed: 12/19/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) have been studied extensively over the last few decades and have been assessed as molecular targets for the development of drugs against metabolic disorders. A rapid increase in understanding of the physiology and pharmacology of these receptors has occurred, together with the identification of novel chemical structures that are able to activate the various PPAR subtypes. More recent evidence suggests that moderate activation of these receptors could be favorable in pathological situations due to a decrease in the side effects brought about by PPAR agonists. PPAR partial agonists and antagonists are interesting tools that are currently used to better elucidate the biological processes modulated by this family of nuclear receptors. Herein we present an overview of the various molecular structures that are able to block each of the PPAR subtypes, with a focus on promising therapeutic applications.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Dipartimento di Farmacia, Università "G. d'Annunzio" via dei Vestini 31, 66100 Chieti (Italy)
| | | | | | | |
Collapse
|
24
|
Nakamura M, Makishima M, Hashimoto Y. Development of silicon-containing bis-phenol derivatives as androgen receptor antagonists: selectivity switching by C/Si exchange. Bioorg Med Chem 2013; 21:1643-51. [PMID: 23462715 DOI: 10.1016/j.bmc.2013.01.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 10/27/2022]
Abstract
We previously reported that bis-phenol derivatives, including LG190178 (3a), possess not only vitamin D receptor (VDR) agonistic activity, but also androgen receptor (AR) antagonistic activity. Here, we describe the design, synthesis and evaluation of silicon-containing bis-phenol derivatives, with the objective of obtaining increased selectivity toward VDR or AR. We found that replacement of the quaternary carbon in the bis-phenol skeleton with silicon increased AR-antagonistic activity and reduced VDR-agonistic activity, that is, the AR selectivity of the silicon-containing compounds was higher than that of corresponding carbon compounds. To our knowledge, this is the first report of nuclear receptor (NR) selectivity switching by sila-substitution (C/Si exchange). Among the compounds synthesized, AR-selective ligand (S,R)-3b exhibited more potent anti-androgenic activity (IC50=0.072 μM) than hydroxyflutamide, a well-known androgen antagonist (IC50=1.4 μM), in SC-3 cell proliferation assay. These results suggest that sila-substitution is a useful approach for structural development of selective AR ligands.
Collapse
Affiliation(s)
- Masaharu Nakamura
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| | | | | |
Collapse
|
25
|
Mita Y, Dodo K, Noguchi-Yachide T, Hashimoto Y, Ishikawa M. Structure–activity relationship of benzodiazepine derivatives as LXXLL peptide mimetics that inhibit the interaction of vitamin D receptor with coactivators. Bioorg Med Chem 2013; 21:993-1005. [DOI: 10.1016/j.bmc.2012.11.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 11/14/2012] [Accepted: 11/20/2012] [Indexed: 11/15/2022]
|
26
|
Yamamoto S, Kobayashi H, Kaku T, Aikawa K, Hara T, Yamaoka M, Kanzaki N, Hasuoka A, Baba A, Ito M. Design, synthesis, and biological evaluation of 3-aryl-3-hydroxy-1-phenylpyrrolidine derivatives as novel androgen receptor antagonists. Bioorg Med Chem 2013. [DOI: 10.1016/j.bmc.2012.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
27
|
Design, synthesis and in vitro antimicrobial activity of novel phenylbenzamido-aminothiazole-based azasterol mimics. Med Chem Res 2012. [DOI: 10.1007/s00044-012-0314-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
28
|
Ohashi M, Nakagome I, Kasuga JI, Nobusada H, Matsuno K, Makishima M, Hirono S, Hashimoto Y, Miyachi H. Design, synthesis and in vitro evaluation of a series of α-substituted phenylpropanoic acid PPARγ agonists to further investigate the stereochemistry–activity relationship. Bioorg Med Chem 2012; 20:6375-83. [DOI: 10.1016/j.bmc.2012.08.061] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 08/28/2012] [Accepted: 08/29/2012] [Indexed: 01/15/2023]
|
29
|
Estrogenic activity of B-fluorinated o-carborane-1,2-bisphenol synthesized via SNAr reaction. Bioorg Med Chem Lett 2012; 22:4728-30. [DOI: 10.1016/j.bmcl.2012.05.068] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 05/17/2012] [Accepted: 05/18/2012] [Indexed: 11/21/2022]
|
30
|
Rey J, O'Riordan TJC, Hu H, Snyder JP, White AJP, Barrett AGM. Design and Diastereoselective Synthesis of C-2,C-20-Diaryl Steroidal Derivatives. European J Org Chem 2012. [DOI: 10.1002/ejoc.201200190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
31
|
Yamamoto S, Tomita N, Suzuki Y, Suzaki T, Kaku T, Hara T, Yamaoka M, Kanzaki N, Hasuoka A, Baba A, Ito M. Design, synthesis, and biological evaluation of 4-arylmethyl-1-phenylpyrazole and 4-aryloxy-1-phenylpyrazole derivatives as novel androgen receptor antagonists. Bioorg Med Chem 2012; 20:2338-52. [DOI: 10.1016/j.bmc.2012.02.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 02/02/2012] [Accepted: 02/02/2012] [Indexed: 11/28/2022]
|
32
|
Ammazzalorso A, Giancristofaro A, D'Angelo A, Filippis BD, Fantacuzzi M, Giampietro L, Maccallini C, Amoroso R. Benzothiazole-based N-(phenylsulfonyl)amides as a novel family of PPARα antagonists. Bioorg Med Chem Lett 2011; 21:4869-72. [PMID: 21742490 DOI: 10.1016/j.bmcl.2011.06.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/04/2011] [Accepted: 06/08/2011] [Indexed: 11/18/2022]
Abstract
The discovery of PPAR antagonists is emerging as an useful tool for elucidating the biological role of the receptor. Here we report the identification of N-(phenylsulfonyl)amides containing the benzothiazole scaffold, a novel class of potent PPARα antagonists obtained from chemical modification of carboxylic acid agonists. In this work, a group of phenylsulfonamides were synthesized and in vitro evaluated against the agonistic effect of GW7647; they showed an inhibitory effect on PPARα activation, with best compounds revealing a dose-dependent antagonistic profile. Some of these antagonists showed also an inhibitory effect on CPT1A pattern expression.
Collapse
|
33
|
Dawson MI, Xia Z. The retinoid X receptors and their ligands. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:21-56. [PMID: 22020178 DOI: 10.1016/j.bbalip.2011.09.014] [Citation(s) in RCA: 264] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 08/23/2011] [Accepted: 09/23/2011] [Indexed: 12/12/2022]
Abstract
This chapter presents an overview of the current status of studies on the structural and molecular biology of the retinoid X receptor subtypes α, β, and γ (RXRs, NR2B1-3), their nuclear and cytoplasmic functions, post-transcriptional processing, and recently reported ligands. Points of interest are the different changes in the ligand-binding pocket induced by variously shaped agonists, the communication of the ligand-bound pocket with the coactivator binding surface and the heterodimerization interface, and recently identified ligands that are natural products, those that function as environmental toxins or drugs that had been originally designed to interact with other targets, as well as those that were deliberately designed as RXR-selective transcriptional agonists, synergists, or antagonists. Of these synthetic ligands, the general trend in design appears to be away from fully aromatic rigid structures to those containing partial elements of the flexible tetraene side chain of 9-cis-retinoic acid. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).
Collapse
Affiliation(s)
- Marcia I Dawson
- Cancer Center, Sanford-Burn Medical Research Institute, 10901 North Torrey Pines Rd., La Jolla, CA 93207, USA.
| | | |
Collapse
|
34
|
Structural Development Studies of Subtype-Selective Ligands for Peroxisome Proliferator-Activated Receptors (PPARs) Based on the 3,4-Disubstituted Phenylpropanoic Acid Scaffold as a Versatile Template. PPAR Res 2011; 2008:689859. [PMID: 18566690 PMCID: PMC2430034 DOI: 10.1155/2008/689859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2007] [Revised: 11/09/2007] [Accepted: 12/26/2007] [Indexed: 12/22/2022] Open
Abstract
Improvements in our understanding of the functions of the nuclear receptor peroxisome proliferator-activated receptor (PPAR) subtypes as pleiotropic regulators of biological responses, including lipid, lipoprotein, glucose homeostasis, inflammation, differentiation and proliferation of various cancer cells, and memory, have provided an opportunity to develop novel PPAR ligands with characteristic subtype selectivity. Such ligands are not only chemical tools to investigate the functions of PPARs, but also candidates for the treatment of PPAR-mediated diseases, including metabolic syndrome, inflammation, dementia, and cancer. This minireview summarizes our work on the design, synthesis, and pharmacological evaluation of subtype-selective PPAR agonists based on the use of 3,4-disubstituted phenylpropanoic acid as a versatile template.
Collapse
|
35
|
Crystal structure, docking study and structure–activity relationship of carborane-containing androgen receptor antagonist 3-(12-hydroxymethyl-1,12-dicarba-closo-dodecaboran-1-yl)benzonitrile. Bioorg Med Chem 2011; 19:3540-8. [DOI: 10.1016/j.bmc.2011.04.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 04/06/2011] [Accepted: 04/08/2011] [Indexed: 01/03/2023]
|
36
|
Ohashi M, Oyama T, Nakagome I, Satoh M, Nishio Y, Nobusada H, Hirono S, Morikawa K, Hashimoto Y, Miyachi H. Design, Synthesis, and Structural Analysis of Phenylpropanoic Acid-Type PPARγ-Selective Agonists: Discovery of Reversed Stereochemistry−Activity Relationship. J Med Chem 2010; 54:331-41. [DOI: 10.1021/jm101233f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Masao Ohashi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Takuji Oyama
- Institute for Protein Research, Osaka University, 6-2-3, Furuedai, Suita, Osaka 565-0874, Japan
| | - Izumi Nakagome
- School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Mayumi Satoh
- Tumor Therapy Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo Metropolitan Organization for Medical Research, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Yoshino Nishio
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Hiromi Nobusada
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Shuichi Hirono
- School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kosuke Morikawa
- Institute for Protein Research, Osaka University, 6-2-3, Furuedai, Suita, Osaka 565-0874, Japan
| | - Yuichi Hashimoto
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Hiroyuki Miyachi
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
37
|
Design and synthesis of an androgen receptor pure antagonist (CH5137291) for the treatment of castration-resistant prostate cancer. Bioorg Med Chem 2010; 18:8150-7. [DOI: 10.1016/j.bmc.2010.10.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 10/08/2010] [Accepted: 10/09/2010] [Indexed: 11/24/2022]
|
38
|
Sato Y, Ramalanjaona N, Huet T, Potier N, Osz J, Antony P, Peluso-Iltis C, Poussin-Courmontagne P, Ennifar E, Mély Y, Dejaegere A, Moras D, Rochel N. The "Phantom Effect" of the Rexinoid LG100754: structural and functional insights. PLoS One 2010; 5:e15119. [PMID: 21152046 PMCID: PMC2994906 DOI: 10.1371/journal.pone.0015119] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 10/22/2010] [Indexed: 12/29/2022] Open
Abstract
Retinoic acid receptors (RARs) and Retinoid X nuclear receptors (RXRs) are ligand-dependent transcriptional modulators that execute their biological action through the generation of functional heterodimers. RXR acts as an obligate dimer partner in many signalling pathways, gene regulation by rexinoids depending on the liganded state of the specific heterodimeric partner. To address the question of the effect of rexinoid antagonists on RAR/RXR function, we solved the crystal structure of the heterodimer formed by the ligand binding domain (LBD) of the RARα bound to its natural agonist ligand (all-trans retinoic acid, atRA) and RXRα bound to a rexinoid antagonist (LG100754). We observed that RARα exhibits the canonical agonist conformation and RXRα an antagonist one with the C-terminal H12 flipping out to the solvent. Examination of the protein-LG100754 interactions reveals that its propoxy group sterically prevents the H12 associating with the LBD, without affecting the dimerization or the active conformation of RAR. Although LG100754 has been reported to act as a ‘phantom ligand’ activating RAR in a cellular context, our structural data and biochemical assays demonstrate that LG100754 mediates its effect as a full RXR antagonist. Finally we show that the ‘phantom ligand effect’ of the LG100754 is due to a direct binding of the ligand to RAR that stabilizes coactivator interactions thus accounting for the observed transcriptional activation of RAR/RXR.
Collapse
Affiliation(s)
- Yoshiteru Sato
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Nick Ramalanjaona
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Tiphaine Huet
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Noelle Potier
- Institut de Chimie LC3 - CNRS- UMR 7177, ISIS, Strasbourg, France
| | - Judit Osz
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Pierre Antony
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Carole Peluso-Iltis
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Pierre Poussin-Courmontagne
- Plate-forme technologique de Biologie et Génomique structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Eric Ennifar
- Architecture et réactivité de l'ARN, Université de Strasbourg, CNRS, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Yves Mély
- Laboratoire de Biophotonique et Pharmacologie, Faculté de Pharmacie, UMR 7213 du CNRS, Université de Strasbourg, Illkirch, France
| | - Annick Dejaegere
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Dino Moras
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
| | - Natacha Rochel
- Département de Biologie et de Génomique Structurales, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de Santé et de Recherche Médicale (INSERM) U964/Centre National de Recherche Scientifique (CNRS) UMR 1704/Université de Strasbourg, Illkirch, France
- * E-mail:
| |
Collapse
|
39
|
Lamblin M, Spingarn R, Wang TT, Burger MC, Dabbas B, Moitessier N, White JH, Gleason JL. An o-aminoanilide analogue of 1α,25-dihydroxyvitamin D(3) functions as a strong vitamin D receptor antagonist. J Med Chem 2010; 53:7461-5. [PMID: 20883026 DOI: 10.1021/jm1007159] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vitamin D receptor (VDR) antagonists have therapeutic potential in treatment of allergic conditions and hypercalcemia driven by granulomatous diseases. We have identified an o-aminoanilide analogue of the hormonal form of vitamin D with a dienyl side chain that functions as a strong VDR antagonist. Modeling studies indicate that antagonism arises from side chain rigidity, when compared to a more flexible saturated analogue, which interferes with H12 folding/alignment.
Collapse
Affiliation(s)
- Marc Lamblin
- Departments of Chemistry, McGill University, Montreal, Quebec, H3A 2K6, Canada
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Maruyama K, Noguchi-Yachide T, Sugita K, Hashimoto Y, Ishikawa M. Novel selective anti-androgens with a diphenylpentane skeleton. Bioorg Med Chem Lett 2010; 20:6661-6. [DOI: 10.1016/j.bmcl.2010.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 08/30/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
|
41
|
Mukherjee S, Mani S. Orphan nuclear receptors as targets for drug development. Pharm Res 2010; 27:1439-68. [PMID: 20372994 PMCID: PMC3518931 DOI: 10.1007/s11095-010-0117-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Accepted: 03/04/2010] [Indexed: 12/31/2022]
Abstract
Orphan nuclear receptors regulate diverse biological processes. These important molecules are ligand-activated transcription factors that act as natural sensors for a wide range of steroid hormones and xenobiotic ligands. Because of their importance in regulating various novel signaling pathways, recent research has focused on identifying xenobiotics targeting these receptors for the treatment of multiple human diseases. In this review, we will highlight these receptors in several physiologic and pathophysiologic actions and demonstrate how their functions can be exploited for the successful development of newer drugs.
Collapse
Affiliation(s)
- Subhajit Mukherjee
- Departments of Medicine, Genetics and Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Chanin 302-D1, Bronx, New York 10461, USA
| | - Sridhar Mani
- Departments of Medicine, Genetics and Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Chanin 302-D1, Bronx, New York 10461, USA
| |
Collapse
|
42
|
Takizawa D, Kakizaki S, Horiguchi N, Tojima H, Yamazaki Y, Ichikawa T, Sato K, Mori M. Histone deacetylase inhibitors induce cytochrome P450 2B by activating nuclear receptor constitutive androstane receptor. Drug Metab Dispos 2010; 38:1493-8. [PMID: 20516253 DOI: 10.1124/dmd.110.032854] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Valproic acid, a histone deacetylase (HDAC) inhibitor, induces the cytochrome P450 2B subfamily. However, the effects of HDAC inhibitors on CYP2B induction are still not fully understood. Nuclear receptor constitutive androstane receptor (CAR) is a key regulator of CYP2B induction. In this study, we investigated the effect of HDAC inhibitors on CAR-mediated CYP2B induction. The expression of CYP2B6 mRNA was induced in HepG2 cells stably expressing mouse CAR (Ym17) by HDAC inhibitors including valproic acid, phenylbutyrate, and trichostatin A. HDAC inhibitors activated the phenobarbital-responsive enhancer module of the CYP2B6 promoter in transient transfection reporter assays with Ym17 cells. Furthermore, HDAC inhibitors synergistically augmented the effect of the CAR ligand, 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene, in the transactivation of CYP2B6 mRNA and the promoter assay in Ym17 cells. Intraperitoneal injection of HDAC inhibitors induced Cyp2b10 mRNA in wild-type mice. However, such induction was not observed in CAR(-/-) mice. Immunoprecipitation demonstrated that CAR formed a complex with HDACs. HDAC inhibitors diminished the binding between CAR and HDAC1 and augmented the binding of steroid receptor coactivator-1 (SRC-1) to CAR. Furthermore, small interfering RNA knockdown of HDAC1 increased CYP2B6 mRNA expression. These results provide novel insight into the mechanism by which HDAC inhibitors affect gene expression of CYP2B6. HDAC inhibitors have the potential to up-regulate CYP2B6 through the dissociation of HDAC1 and recruitment of SRC-1 to receptor CAR.
Collapse
Affiliation(s)
- Daichi Takizawa
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Structure–activity relationships of bioisosteric replacement of the carboxylic acid in novel androgen receptor pure antagonists. Bioorg Med Chem 2010; 18:3159-68. [DOI: 10.1016/j.bmc.2010.03.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 03/13/2010] [Accepted: 03/16/2010] [Indexed: 11/20/2022]
|
44
|
Abstract
Nuclear receptors (NRs) are ligand-inducible transcription factors that regulate a plethora of cell biological phenomena, thus orchestrating complex events like development, organ homeostasis, immune function, and reproduction. Due to their regulatory potential, NRs are major drug targets for a variety of diseases, including cancer and metabolic diseases, and had a major societal impact following the development of contraceptives and abortifacients. Not surprisingly in view of this medical and societal importance, a large amount of diverse NR ligands have been generated and the corresponding structural and functional analyses have provided a deep insight into the molecular basis of ligand action. What we have learnt is that ligands regulate, via allosteric conformational changes, the ability of NRs to interact with different sets of coregulators which in turn recruit enzymatically active complexes, the workhorses of the ligand-induced epigenetic and transcription-regulatory events. Thus, ligands essentially direct the communication of a given NR with its intracellular environment at the chromatin and extragenomic level to modulate gene programs directly at the chromatin level or via less well-understood extranuclear actions. Here we will review our current structural and mechanistic insight into the functionalities of subsets of retinoid and rexinoid ligands that act generically as antagonists but follow different mechanistic principles, resulting in "classical" or neutral antagonism, or inverse agonism. In addition, we describe the chemical features and guidelines for the synthesis of retinoids/rexinoids that exert specific functions and we provide protocols for a number of experimental approaches that are useful for studies of the agonistic and antagonistic features of NR ligands.
Collapse
Affiliation(s)
- William Bourguet
- INSERM U554 and CNRS UMR5048, Centre de Biochimie Structurale, Universités Montpellier 1 & 2, Montpellier, France
| | | | | |
Collapse
|
45
|
Casey FP, Pihan E, Shields DC. Discovery of Small Molecule Inhibitors of Protein−Protein Interactions Using Combined Ligand and Target Score Normalization. J Chem Inf Model 2009; 49:2708-17. [DOI: 10.1021/ci900294x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Fergal P. Casey
- UCD Complex and Adaptive Systems Laboratory, UCD Conway Institute, and School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Emilie Pihan
- UCD Complex and Adaptive Systems Laboratory, UCD Conway Institute, and School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| | - Denis C. Shields
- UCD Complex and Adaptive Systems Laboratory, UCD Conway Institute, and School of Medicine and Medical Sciences, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
46
|
Kasuga JI, Ishida S, Yamasaki D, Makishima M, Doi T, Hashimoto Y, Miyachi H. Novel biphenylcarboxylic acid peroxisome proliferator-activated receptor (PPAR) delta selective antagonists. Bioorg Med Chem Lett 2009; 19:6595-9. [PMID: 19853439 DOI: 10.1016/j.bmcl.2009.10.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 10/04/2009] [Accepted: 10/06/2009] [Indexed: 11/17/2022]
Abstract
We designed and synthesized novel PPARdelta antagonists based on the crystal structure of the PPARdelta full agonist TIPP-204 bound to the PPARdelta ligand-binding domain, in combination with our nuclear receptor helix 12 folding modification hypothesis. Representative compound 3a exhibits PPARdelta-preferential antagonistic activity.
Collapse
Affiliation(s)
- Jun-ichi Kasuga
- Institute of Molecular and Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Fujimoto H, Noguchi T, Kobayashi H, Miyachi H, Hashimoto Y. Effects of immunomodulatory derivatives of thalidomide (IMiDs) and their analogs on cell-differentiation, cyclooxygenase activity and angiogenesis. Chem Pharm Bull (Tokyo) 2009; 54:855-60. [PMID: 16755058 DOI: 10.1248/cpb.54.855] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Various analogs of known immunomodulatory derivatives of thalidomide (1) (IMiDs: 3, 5) were synthesized, focusing on cell-differentiation-inducing, cyclooxygenase-inhibitory and anti-angiogenesis activities. Among the prepared compounds, NIDO-33 (14) showed cell differentiation-inducing activity on HL-60 cells and anti-angiogenic activity on human umbilical vein endothelial cells (HUVEC). AIDO-00 (7) also showed anti-angiogenic activity. NIDO-11 (8) showed an enhancing effect on all-trans retinoic acid (ATRA)-induced HL-60 cell differentiation, and AIDO-30 (13) exhibited cyclooxygenase (COX)-inhibitory activity.
Collapse
Affiliation(s)
- Haruka Fujimoto
- Institute of Molecular & Cellular Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | | | | | | | | |
Collapse
|
48
|
Alvarez S, Alvarez R, Khanwalkar H, Germain P, Lemaire G, Rodríguez-Barrios F, Gronemeyer H, de Lera AR. Retinoid receptor subtype-selective modulators through synthetic modifications of RARgamma agonists. Bioorg Med Chem 2009; 17:4345-59. [PMID: 19482478 DOI: 10.1016/j.bmc.2009.05.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 05/06/2009] [Accepted: 05/11/2009] [Indexed: 12/31/2022]
Abstract
A series of retinoids designed to interfere with the repositioning of H12 have been synthesized to identify novel RARgamma antagonists based on the structure of known RARgamma agonists. The transcriptional activities of the novel ligands were revealed by cell-based reporting assays, using engineered cells containg RAR subtype-selective fusions of the RAR ligand-binding domains with the yeast GAL4 activator DNA-binding domain and the cognate luciferase reporter gene. Whereas none of the ligands exhibited features of a selective RARgamma antagonist, some of them are endowed with interesting activities. In particular 24a acts as a pan-RAR agonist that induces at high concentration a higher transactivation potential on RARalpha than TTNPB and synergizes at low concentration with TTNPB-bound RARalpha but not RARbeta or RARgamma. Similarly, 24c synergizes with TTNPB-bound RARgamma and exhibits RARalpha,beta antagonist activity. Compounds 24b and 25b are strong RARalpha,beta-selective antagonists without agonist or antagonist activities for RARgamma. Compounds 24b and 24c display weak RXR antagonist activity. In addition several pan-antagonists and partial agonist/antagonists have been defined.
Collapse
Affiliation(s)
- Susana Alvarez
- Departamento de Química Orgánica, Universidade de Vigo, Vigo, Spain
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Jiang X, Lee GT, Prasad K, Repič O. A Practical Synthesis of a Diazepinylbenzoic Acid, a Retinoid X Receptor Antagonist. Org Process Res Dev 2008. [DOI: 10.1021/op800142b] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xinglong Jiang
- Process Research & Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, U.S.A
| | - George T. Lee
- Process Research & Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, U.S.A
| | - Kapa Prasad
- Process Research & Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, U.S.A
| | - Oljan Repič
- Process Research & Development, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, New Jersey 07936, U.S.A
| |
Collapse
|
50
|
SAR-oriented discovery of peroxisome proliferator-activated receptor pan agonist with a 4-adamantylphenyl group as a hydrophobic tail. Bioorg Med Chem Lett 2008; 18:1110-5. [DOI: 10.1016/j.bmcl.2007.12.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 11/15/2007] [Accepted: 12/01/2007] [Indexed: 01/08/2023]
|