1
|
Zhang Y, Xiao B, Liu Y, Wu S, Xiang Q, Xiao Y, Zhao J, Yuan R, Xie K, Li L. Roles of PPAR activation in cancer therapeutic resistance: Implications for combination therapy and drug development. Eur J Pharmacol 2024; 964:176304. [PMID: 38142851 DOI: 10.1016/j.ejphar.2023.176304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 12/26/2023]
Abstract
Therapeutic resistance is a major obstacle to successful treatment or effective containment of cancer. Peroxisome proliferator-activated receptors (PPARs) play an essential role in regulating energy homeostasis and determining cell fate. Despite of the pleiotropic roles of PPARs in cancer, numerous studies have suggested their intricate relationship with therapeutic resistance in cancer. In this review, we provided an overview of the roles of excessively activated PPARs in promoting resistance to modern anti-cancer treatments, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy. The mechanisms through which activated PPARs contribute to therapeutic resistance in most cases include metabolic reprogramming, anti-oxidant defense, anti-apoptosis signaling, proliferation-promoting pathways, and induction of an immunosuppressive tumor microenvironment. In addition, we discussed the mechanisms through which activated PPARs lead to multidrug resistance in cancer, including drug efflux, epithelial-to-mesenchymal transition, and acquisition and maintenance of the cancer stem cell phenotype. Preliminary studies investigating the effect of combination therapies with PPAR antagonists have suggested the potential of these antagonists in reversing resistance and facilitating sustained cancer management. These findings will provide a valuable reference for further research on and clinical translation of PPAR-targeting treatment strategies.
Collapse
Affiliation(s)
- Yanxia Zhang
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China; Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yunduo Liu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Shunhong Wu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Qin Xiang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yuhan Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Junxiu Zhao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Ruanfei Yuan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Keping Xie
- School of Medicine, The South China University of Technology, Guangzhou, 510006, China.
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
2
|
Chen ZH, Xu RM, Zheng GH, Jin YZ, Li Y, Chen XY, Tian YS. Development of Combretastatin A-4 Analogues as Potential Anticancer Agents with Improved Aqueous Solubility. Molecules 2023; 28:molecules28041717. [PMID: 36838705 PMCID: PMC9963121 DOI: 10.3390/molecules28041717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Combretastatin A-4 (CA-4) is a potent tubulin polymerisation inhibitor. However, the clinical application of CA-4 is limited owing to its low aqueous solubility and the easy conversion of the olefin double bond from the more active cis- to the less active trans-configuration. Several structural modifications were investigated to improve the solubility of CA-4 derivatives. Among the compounds we synthesized, the kinetic solubility assay revealed that the solubility of compounds containing a piperazine ring increased the most, and the solubility of compounds 12a1, 12a2, 15 and 18 was increased 230-2494 times compared with that of the control compound (Z)-3-(4-aminophenyl)-2-(3,4,5-trimethoxyphenyl)acrylonitrile (9a). In addition, these synthesised stilbene nitriles had high anticancer cell (AGS, BEL-7402, MCF-7, and HCT-116) selectivity over L-02 and MCF-10A normal cells while maintaining micromolar activity against cancer cells. The most cytotoxic compound is 9a, and the IC50 value is 20 nM against HCT-116 cancer cells. Preliminary studies indicated that compound 12a1 had excellent plasma stability and moderate binding to rat plasma proteins, suggesting it is a promising lead compound for the development of an anticancer agent.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yu-Shun Tian
- Correspondence: ; Tel.: +864332436028; Fax: +864332435026
| |
Collapse
|
3
|
The Role of PPARs in Breast Cancer. Cells 2022; 12:cells12010130. [PMID: 36611922 PMCID: PMC9818187 DOI: 10.3390/cells12010130] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/07/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is a malignant tumor with high morbidity and lethality. Its pathogenesis is related to the abnormal expression of many genes. The peroxisome proliferator-activated receptors (PPARs) are a class of ligand-dependent transcription factors in the nuclear receptor superfamily. They can regulate the transcription of a large number of target genes, which are involved in life activities such as cell proliferation, differentiation, metabolism, and apoptosis, and regulate physiological processes such as glucose metabolism, lipid metabolism, inflammation, and wound healing. Further, the changes in its expression are associated with various diseases, including breast cancer. The experimental reports related to "PPAR" and "breast cancer" were retrieved from PubMed since the discovery of PPARs and summarized in this paper. This review (1) analyzed the roles and potential molecular mechanisms of non-coordinated and ligand-activated subtypes of PPARs in breast cancer progression; (2) discussed the correlations between PPARs and estrogen receptors (ERs) as the nuclear receptor superfamily; and (3) investigated the interaction between PPARs and key regulators in several signaling pathways. As a result, this paper identifies PPARs as targets for breast cancer prevention and treatment in order to provide more evidence for the synthesis of new drugs targeting PPARs or the search for new drug combination treatments.
Collapse
|
4
|
Kadu R, Savani C, Roy H, Soni P, Singh AK, Vennapu DR, Singh VK. Impact of N-substituents on crystal packing of N-alkyl-N′-tosylpiperazines and development of new polymorph of tosylbis(2-(tosyloxy)ethyl)amine: Synthesis, DFT, photophysical, cytotoxic property. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
5
|
Arslan B, Gülcemal S. α-Alkylation of arylacetonitriles with primary alcohols catalyzed by backbone modified N-heterocyclic carbene iridium(i) complexes. Dalton Trans 2021; 50:1788-1796. [DOI: 10.1039/d0dt04082g] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
α-Alkylation of arylacetonitriles with primary alcohols was achieved by using backbone-modified NHC–IrI complexes as catalysts with turnover numbers of up to 960.
Collapse
Affiliation(s)
- Burcu Arslan
- Department of Chemistry
- Ege University
- 35100 Bornova
- Turkey
| | | |
Collapse
|
6
|
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
|
7
|
Allam HA, Aly EE, Farouk AK, El Kerdawy AM, Rashwan E, Abbass SE. Design and Synthesis of some new 2,4,6-trisubstituted quinazoline EGFR inhibitors as targeted anticancer agents. Bioorg Chem 2020; 98:103726. [DOI: 10.1016/j.bioorg.2020.103726] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/20/2020] [Accepted: 03/04/2020] [Indexed: 12/30/2022]
|
8
|
Sirim MM, Krishna VS, Sriram D, Unsal Tan O. Novel benzimidazole-acrylonitrile hybrids and their derivatives: Design, synthesis and antimycobacterial activity. Eur J Med Chem 2020; 188:112010. [DOI: 10.1016/j.ejmech.2019.112010] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/04/2019] [Accepted: 12/23/2019] [Indexed: 01/06/2023]
|
9
|
Legrand N, Bretscher CL, Zielke S, Wilke B, Daude M, Fritz B, Diederich WE, Adhikary T. PPARβ/δ recruits NCOR and regulates transcription reinitiation of ANGPTL4. Nucleic Acids Res 2019; 47:9573-9591. [PMID: 31428774 PMCID: PMC6765110 DOI: 10.1093/nar/gkz685] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/20/2019] [Accepted: 07/28/2019] [Indexed: 12/24/2022] Open
Abstract
In the absence of ligands, the nuclear receptor PPARβ/δ recruits the NCOR and SMRT corepressors, which form complexes with HDAC3, to canonical target genes. Agonistic ligands cause dissociation of corepressors and enable enhanced transcription. Vice versa, synthetic inverse agonists augment corepressor recruitment and repression. Both basal repression of the target gene ANGPTL4 and reinforced repression elicited by inverse agonists are partially insensitive to HDAC inhibition. This raises the question how PPARβ/δ represses transcription mechanistically. We show that the PPARβ/δ inverse agonist PT-S264 impairs transcription initiation by decreasing recruitment of activating Mediator subunits, RNA polymerase II, and TFIIB, but not of TFIIA, to the ANGPTL4 promoter. Mass spectrometry identifies NCOR as the main PT-S264-dependent interactor of PPARβ/δ. Reconstitution of knockout cells with PPARβ/δ mutants deficient in basal repression results in diminished recruitment of NCOR, SMRT, and HDAC3 to PPAR target genes, while occupancy by RNA polymerase II is increased. PT-S264 restores binding of NCOR, SMRT, and HDAC3 to the mutants, resulting in reduced polymerase II occupancy. Our findings corroborate deacetylase-dependent and -independent repressive functions of HDAC3-containing complexes, which act in parallel to downregulate transcription.
Collapse
Affiliation(s)
- Nathalie Legrand
- Department of Medicine, Institute for Molecular Biology and Tumour Research, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Clemens L Bretscher
- Department of Medicine, Institute for Molecular Biology and Tumour Research, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Svenja Zielke
- Department of Medicine, Institute for Molecular Biology and Tumour Research, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Bernhard Wilke
- Department of Medicine, Institute for Molecular Biology and Tumour Research, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany.,Department of Medicine, Institute for Medical Bioinformatics and Biostatistics, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Michael Daude
- Core Facility Medicinal Chemistry, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Barbara Fritz
- Centre for Human Genetics, Universitätsklinikum Giessen und Marburg GmbH, Baldingerstrasse, 35043 Marburg, Germany
| | - Wibke E Diederich
- Core Facility Medicinal Chemistry, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany.,Department of Pharmacy, Institute for Pharmaceutical Chemistry, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Till Adhikary
- Department of Medicine, Institute for Molecular Biology and Tumour Research, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany.,Department of Medicine, Institute for Medical Bioinformatics and Biostatistics, Centre for Tumour Biology and Immunology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| |
Collapse
|
10
|
Gong X, Li G, Gan Z, Yan Q, Dou X, Yang D. Sulfonylacetonitriles as Building Blocks in Copper‐Catalyzed Domino Reactions: An Efficient Apporach to Sulfonated Isoquinolin‐1(2
H
)‐ones. ASIAN J ORG CHEM 2019. [DOI: 10.1002/ajoc.201900382] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Xueqin Gong
- School of Chemistry and Chemical EngineeringQufu Normal University Jingxuan Road Qufu 273165 Shandong P. R. China
| | - Guoqing Li
- School of Chemistry and Chemical EngineeringQufu Normal University Jingxuan Road Qufu 273165 Shandong P. R. China
| | - Ziyu Gan
- School of Chemistry and Chemical EngineeringQufu Normal University Jingxuan Road Qufu 273165 Shandong P. R. China
| | - Qiuli Yan
- State Key Laboratory Base of Eco-Chemical Engineering College of Chemistry and Molecular EngineeringQingdao University of Science and Technology Qingdao 266042 P. R. China
- School of Chemistry and Chemical EngineeringQufu Normal University Jingxuan Road Qufu 273165 Shandong P. R. China
| | - Xiaomeng Dou
- School of Chemistry and Chemical EngineeringHunan University of Science and Technology Xiangtan 411201 P. R. China
| | - Daoshan Yang
- State Key Laboratory Base of Eco-Chemical Engineering College of Chemistry and Molecular EngineeringQingdao University of Science and Technology Qingdao 266042 P. R. China
- School of Chemistry and Chemical EngineeringQufu Normal University Jingxuan Road Qufu 273165 Shandong P. R. China
| |
Collapse
|
11
|
Fantacuzzi M, De Filippis B, Amoroso R, Giampietro L. PPAR Ligands Containing Stilbene Scaffold. Mini Rev Med Chem 2019; 19:1599-1610. [PMID: 31161987 DOI: 10.2174/1389557519666190603085026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/18/2019] [Accepted: 05/19/2019] [Indexed: 01/26/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are transcriptional factors which belong to the ligand-activated nuclear receptor superfamily. They are ubiquitously expressed throughout the body. So far, three major subtypes have been identified, PPARα, PPARβ/δ and PPARγ. They are crucial for lipid and glucose metabolism and are also involved in the regulation of several types of tumors, inflammation, cardiovascular diseases and infertility. The importance of these transcription factors in physiology and pathophysiology has been largely investigated. Synthetic PPAR ligands are widely used in the treatment of dyslipidemia (e.g. fibrates - PPARα activators) or in diabetes mellitus (e.g. thiazolidinediones - PPARγ agonists) while a new generation of dual agonists reveals hypolipemic, hypotensive, antiatherogenic, anti-inflammatory and anticoagulant action. Many natural ligands, including polyphenolic compounds, influence the expression of these receptors. They have several health-promoting properties, including antioxidant, anti-inflammatory, and antineoplastic activities. Resveratrol, a stilbene polyphenol, is a biological active modulator of several signaling proteins, including PPARs. Given the enormous pharmacological potential of resveratrol, stilbene-based medicinal chemistry had a rapid increase covering various areas of research. The present review discusses ligands of PPARs that contain stilbene scaffold and summarises the different types of compounds on the basis of chemical structure.
Collapse
Affiliation(s)
| | - Barbara De Filippis
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Letizia Giampietro
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
12
|
Xin YB, Li JJ, Zhang HJ, Ma J, Liu X, Gong GH, Tian YS. Synthesis and characterisation of (Z)-styrylbenzene derivatives as potential selective anticancer agents. J Enzyme Inhib Med Chem 2018; 33:1554-1564. [PMID: 30244610 PMCID: PMC6161602 DOI: 10.1080/14756366.2018.1513925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/31/2018] [Accepted: 08/16/2018] [Indexed: 01/06/2023] Open
Abstract
To identify anticancer agents with high potency and low toxicity, a series of (Z)-styrylbenzene derivatives were synthesised and evaluated for anticancer activities using a panel of nine cancer cell lines and two noncancerous cell lines. Most derivatives exhibited significant anti-proliferative activities against five cancer cell lines, including MGC-803 and BEL-7402. (Z)-3-(p-Tolyl)-2-(3,4,5-trimethoxyphenyl)acrylonitrile (6h) showed a strong inhibitory effect on MGC-803 cells (IC50 < 0.01 µM) and exhibited stronger anti-proliferative activity than taxol (IC50 < 0.06 ± 0.01 µM). The IC50 value of 6h in L-02 cells was 10,000-fold higher than in MGC-803 cells. Compound 6h inhibited proliferation of BEL-7402 cells by arresting at the G2/M phase through up-regulation of cyclin B1 expression, down-regulation of cyclin A and D1 expression, and induction of apoptosis. In addition, 6h inhibited the migration of BEL-7402 cells and the formation of cell colonies.
Collapse
Affiliation(s)
- Ya-Bing Xin
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Jia-Jun Li
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Hong-Jian Zhang
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Jun Ma
- Jiangsu Hansoh Pharmaceutical Group Co., Ltd., Lianyungang, P.R. China
| | - Xin Liu
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| | - Guo-Hua Gong
- First Clinical Medical College of Inner Mongolia University for Nationalities, Tongliao, P.R. China
- Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Inner Mongolia University for Nationalities, Tongliao, P.R. China
| | - Yu-Shun Tian
- Key Laboratory of Natural Resources and Functional Molecules of the Changbai Mountain, Affiliated Ministry of Education, College of Pharmacy, Yanbian University, Yanji, P.R. China
| |
Collapse
|
13
|
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
|
14
|
Venkatesan P, Cerón M, Thamotharan S, Robles F, Percino MJ. Quantitative analysis of weak non-covalent interactions in (Z)-3-(4-halophenyl)-2-(pyridin-2/3/4-yl)acrylonitriles. CrystEngComm 2018. [DOI: 10.1039/c7ce02096a] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A detailed experimental and theoretical investigation on the intermolecular interactions in (Z)-3-(4-halophenyl)-2-(pyridin-2/3/4-yl)acrylonitriles is reported and different π staking motifs observed in these structures.
Collapse
Affiliation(s)
- Perumal Venkatesan
- Laboratorio de Polímeros
- Centro de Química
- Instituto de Ciencias
- Benemérita Universidad Autónoma de Puebla
- Complejo de Ciencias
| | - Margarita Cerón
- Laboratorio de Polímeros
- Centro de Química
- Instituto de Ciencias
- Benemérita Universidad Autónoma de Puebla
- Complejo de Ciencias
| | - Subbiah Thamotharan
- Biomolecular Crystallography Laboratory
- Department of Bioinformatics
- School of Chemical and Biotechnology
- SASTRA Deemed University
- Thanjavur 613 401
| | - Fernando Robles
- Laboratorio Nacional del Sureste de México
- BUAP
- C.P. 72500 Puebla
- Mexico
| | - M. Judith Percino
- Laboratorio de Polímeros
- Centro de Química
- Instituto de Ciencias
- Benemérita Universidad Autónoma de Puebla
- Complejo de Ciencias
| |
Collapse
|
15
|
PPAR-delta modulates membrane cholesterol and cytokine signaling in malignant B cells. Leukemia 2017; 32:184-193. [PMID: 28555083 DOI: 10.1038/leu.2017.162] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/12/2022]
Abstract
A deeper understanding of the mechanisms that underlie aberrant signal transduction in B-cell cancers such as chronic lymphocytic leukemia (CLL) may reveal new treatment strategies. The lipid-activated nuclear receptor peroxisome proliferator-activated receptor delta (PPARδ) accounts for a number of properties of aggressive cancers and was found to enhance Janus kinase (JAK)-mediated phosphorylation of signal transducer and activator of transcription (STAT) proteins in B lymphoma cell lines and primary CLL cells. Autocrine production of cytokines such as IL10 and interferon-beta was not increased by PPARδ but signaling responses to these cytokines were amplified and associated with increased cholesterol biosynthesis and plasma membrane levels. Plasmalemmal cholesterol and STAT phosphorylation from type 1 interferons (IFNs) were increased by PPARδ agonists, transgenes and exogenous cholesterol, and decreased by cyclodextrin, PPARD deletion and chemical PPARδ inhibitors. Functional consequences of PPARδ-mediated perturbation of IFN signaling included impaired upregulation of co-stimulatory molecules. These observations suggest PPARδ modulates signaling processes in malignant B cells in part by altering cholesterol metabolism and changes the outcomes of signaling from cytokines such as IFNs. PPARδ antagonists may have therapeutic activity as anti-leukemic signal transduction modulators.
Collapse
|
16
|
Yao PL, Chen L, Dobrzański TP, Zhu B, Kang BH, Müller R, Gonzalez FJ, Peters JM. Peroxisome proliferator-activated receptor-β/δ inhibits human neuroblastoma cell tumorigenesis by inducing p53- and SOX2-mediated cell differentiation. Mol Carcinog 2017; 56:1472-1483. [PMID: 27996177 DOI: 10.1002/mc.22607] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/22/2016] [Accepted: 12/15/2016] [Indexed: 12/12/2022]
Abstract
Neuroblastoma is a common childhood cancer typically treated by inducing differentiation with retinoic acid (RA). Peroxisome proliferator-activated receptor-β/δ, (PPARβ/δ) is known to promote terminal differentiation of many cell types. In the present study, PPARβ/δ was over-expressed in three human neuroblastoma cell lines, NGP, SK-N-BE(2), and IMR-32, that exhibit high, medium, and low sensitivity, respectively, to retinoic acid-induced differentiation to determine if PPARβ/δ and retinoic acid receptors (RARs) could be jointly targeted to increase the efficacy of treatment. All-trans-RA (atRA) decreased expression of SRY (sex determining region Y)-box 2 (SOX2), a stem cell regulator and marker of de-differentiation, in NGP and SK-N-BE(2) cells with inactive or mutant tumor suppressor p53, respectively. However, atRA did not suppress SOX2 expression in IMR-32 cells carrying wild-type p53. Over-expression and/or ligand activation of PPARβ/δ reduced the average volume and weight of ectopic tumor xenografts from NGP, SK-N-BE(2), or IMR-32 cells compared to controls. Compared with that found with atRA, PPARβ/δ suppressed SOX2 expression in NGP and SK-N-BE(2) cells and ectopic xenografts, and was also effective in suppressing SOX2 expression in IMR-32 cells that exhibit higher p53 expression compared to the former cell lines. Combined, these observations demonstrate that activating or over-expressing PPARβ/δ induces cell differentiation through p53- and SOX2-dependent signaling pathways in neuroblastoma cells and tumors. This suggests that combinatorial activation of both RARα and PPARβ/δ may be suitable as an alternative therapeutic approach for RA-resistant neuroblastoma patients.
Collapse
Affiliation(s)
- Pei-Li Yao
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania
| | - Liping Chen
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania
| | - Tomasz P Dobrzański
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania
| | - Bokai Zhu
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania
| | - Boo-Hyon Kang
- Non-clinical Research Institute, Chemon, Jeil-Ri, Yangji-Myeon, Cheoin-Gu, Yongin-Si, Gyeonggi-Do, Korea
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University, Marburg, Germany
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
17
|
PPAR-delta promotes survival of chronic lymphocytic leukemia cells in energetically unfavorable conditions. Leukemia 2017; 31:1905-1914. [PMID: 28050012 DOI: 10.1038/leu.2016.395] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 12/07/2016] [Accepted: 12/09/2016] [Indexed: 12/15/2022]
Abstract
Targeting the mechanisms that allow chronic lymphocytic leukemia (CLL) cells to survive in harsh cancer microenvironments should improve patient outcomes. The nuclear receptor peroxisome proliferator activated receptor delta (PPARδ) sustains other cancers, and in silico analysis showed higher PPARD expression in CLL cells than normal lymphocytes and other hematologic cancers. A direct association was found between PPARδ protein levels in CLL cells and clinical score. Transgenic expression of PPARδ increased the growth and survival of CD5+ Daudi cells and primary CLL cells in stressful conditions including exhausted tissue culture media, low extracellular glucose, hypoxia and exposure to cytotoxic drugs. Glucocorticoids and synthetic PPARδ agonists up-regulated PPARD expression and also protected Daudi and primary CLL cells from metabolic stressors. Survival in low glucose was related to increased antioxidant expression, substrate utilization and mitochondrial performance, and was reversed by genetic deletion and synthetic PPARδ antagonists. These findings suggest PPARδ conditions CLL cells to survive in harsh microenvironmental conditions by reducing oxidative stress and increasing metabolic efficiency. Targeting PPARδ may be beneficial in the treatment of CLL.
Collapse
|
18
|
Müller R. PPARβ/δ in human cancer. Biochimie 2016; 136:90-99. [PMID: 27916645 DOI: 10.1016/j.biochi.2016.10.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 10/06/2016] [Accepted: 10/19/2016] [Indexed: 12/31/2022]
Abstract
The nuclear receptor factor peroxisome proliferator-activated receptor (PPARβ/δ) can regulate its target genes by transcriptional activation or repression through both ligand-dependent and independent mechanism as well as by interactions with other transcription factors. PPARβ/δ exerts essential regulatory functions in intermediary metabolism that have been elucidated in detail, but clearly also plays a role in inflammation, differentiation, apoptosis and other cancer-associated processes, which is, however, mechanistically only partly understood. Consistent with these functions clinical associations link the expression of PPARβ/δ and its target genes to an unfavorable outcome of several human cancers. However, the available data do not yield a clear picture of PPARβ/δ's role in cancer-associated processes and are in fact partly controversial. This article provides an overview of this research area and discusses the role of PPARβ/δ in cancer in light of the complex mechanisms of its transcriptional regulation and its potential as a druggable anti-cancer target.
Collapse
Affiliation(s)
- Rolf Müller
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University, Hans-Meerwein-Str. 3, 35043 Marburg, Germany.
| |
Collapse
|
19
|
Parveen M, Azaz S, Ahmad F, Malla AM, Alam M. Silica Bonded N-(Propylcarbamoyl)sulfamic acid (SBPCSA) Mediated Expeditious Approach to C–C Bond Formation: An Innovative Pathway for Acrylonitrile Derivatives. Catal Letters 2016. [DOI: 10.1007/s10562-016-1793-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Wang X, Wang G, Shi Y, Sun L, Gorczynski R, Li YJ, Xu Z, Spaner DE. PPAR-delta promotes survival of breast cancer cells in harsh metabolic conditions. Oncogenesis 2016; 5:e232. [PMID: 27270614 PMCID: PMC4945742 DOI: 10.1038/oncsis.2016.41] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/03/2016] [Indexed: 12/27/2022] Open
Abstract
Expression of the nuclear receptor peroxisome proliferator activated receptor delta (PPARδ) in breast cancer cells is negatively associated with patient survival, but the underlying mechanisms are not clear. High PPARδ protein levels in rat breast adenocarcinomas were found to be associated with increased growth in soft agar and mice. Transgenic expression of PPARδ increased the ability of human breast cancer cell lines to migrate in vitro and form lung metastases in mice. PPARδ also conferred the ability to grow in exhausted tissue culture media and survive in low-glucose and other endoplasmic reticulum stress conditions such as hypoxia. Upregulation of PPARδ by glucocorticoids or synthetic agonists also protected human breast cancer cells from low glucose. Survival in low glucose was related to increased antioxidant defenses mediated in part by catalase and also to late AKT phosphorylation, which is associated with the prolonged glucose-deprivation response. Synthetic antagonists reversed the survival benefits conferred by PPARδ in vitro. These findings suggest that PPARδ conditions breast cancer cells to survive in harsh microenvironmental conditions by reducing oxidative stress and enhancing survival signaling responses. Drugs that target PPARδ may have a role in the treatment of breast cancer.
Collapse
Affiliation(s)
- X Wang
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - G Wang
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Y Shi
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - L Sun
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - R Gorczynski
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Transplant Research Division, Toronto General Hospital, Toronto, Ontario, Canada
| | - Y-J Li
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Anatomy, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Z Xu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - D E Spaner
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Odette Cancer Center, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
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
|
22
|
Toth PM, Lieber S, Scheer FM, Schumann T, Schober Y, Nockher WA, Adhikary T, Müller-Brüsselbach S, Müller R, Diederich WE. Design and Synthesis of Highly Active Peroxisome Proliferator-Activated Receptor (PPAR) β/δ Inverse Agonists with Prolonged Cellular Activity. ChemMedChem 2016; 11:488-96. [DOI: 10.1002/cmdc.201500594] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Philipp M. Toth
- Institut für Pharmazeutische Chemie; Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| | - Sonja Lieber
- Institut für Molekularbiologie und Tumorforschung (IMT); Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| | - Frithjof M. Scheer
- Institut für Pharmazeutische Chemie; Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| | - Tim Schumann
- Institut für Molekularbiologie und Tumorforschung (IMT); Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| | - Yvonne Schober
- Institut für Laboratoriumsmedizin und Pathobiochemie; Molekulare Diagnostik; Philipps-Universität Marburg; Baldingerstraße 35043 Marburg Germany
| | - Wolfgang A. Nockher
- Institut für Laboratoriumsmedizin und Pathobiochemie; Molekulare Diagnostik; Philipps-Universität Marburg; Baldingerstraße 35043 Marburg Germany
- Core Facility Metabolomics; Philipps-Universität Marburg; Baldingerstraße 35043 Marburg Germany
| | - Till Adhikary
- Institut für Molekularbiologie und Tumorforschung (IMT); Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| | - Sabine Müller-Brüsselbach
- Institut für Molekularbiologie und Tumorforschung (IMT); Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| | - Rolf Müller
- Institut für Molekularbiologie und Tumorforschung (IMT); Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| | - Wibke E. Diederich
- Institut für Pharmazeutische Chemie; Zentrum für Tumor- und Immunbiologie (ZTI); Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
- Core Facility Medicinal Chemistry; Philipps-Universität Marburg; Hans-Meerwein-Straße 3 35043 Marburg Germany
| |
Collapse
|
23
|
Synthesis of 5-trifluoromethyl-2-sulfonylpyridine PPARβ/δ antagonists: Effects on the affinity and selectivity towards PPARβ/δ. Bioorg Med Chem 2015; 24:247-60. [PMID: 26707845 DOI: 10.1016/j.bmc.2015.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/05/2015] [Accepted: 12/06/2015] [Indexed: 12/23/2022]
Abstract
The covalent modification of peroxisome-proliferator activated receptor β/δ (PPARβ/δ) is part of the mode of action of 5-trifluoromethyl-2-sulfonylpyridine PPARβ/δ antagonists such as GSK3787 and CC618. Herein, the synthesis and in vitro biological evaluation of a range of structural analogues of the two antagonists are reported. The new ligands demonstrate that an improvement in the selectivity of 5-trifluoromethyl-2-sulfonylpyridine antagonists towards PPARβ/δ is achievable at the expense of their immediate affinity for PPARβ/δ. However, their putatively covalent and irreversible mode of action may ensure their efficacy over time, as observed in time-resolved fluorescence resonance energy transfer (TR-FRET)-based ligand displacement assays.
Collapse
|
24
|
Solano LN, Nelson GL, Ronayne CT, Lueth EA, Foxley MA, Jonnalagadda SK, Gurrapu S, Mereddy VR. Synthesis, in vitro, and in vivo evaluation of novel functionalized quaternary ammonium curcuminoids as potential anti-cancer agents. Bioorg Med Chem Lett 2015; 25:5777-80. [DOI: 10.1016/j.bmcl.2015.10.061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/20/2015] [Indexed: 11/28/2022]
|
25
|
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]
|
26
|
Yao PL, Chen L, Hess RA, Müller R, Gonzalez FJ, Peters JM. Peroxisome Proliferator-activated Receptor-D (PPARD) Coordinates Mouse Spermatogenesis by Modulating Extracellular Signal-regulated Kinase (ERK)-dependent Signaling. J Biol Chem 2015; 290:23416-31. [PMID: 26242735 DOI: 10.1074/jbc.m115.664508] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Indexed: 12/13/2022] Open
Abstract
Ppard(-/-) mice exhibit smaller litter size compared with Ppard(+/+) mice. To determine whether peroxisome proliferator-activated receptor-D (PPARD) could possibly influence this phenotype, the role of PPARD in testicular biology was examined. Atrophic testes and testicular degeneration were observed in Ppard(-/-) mice compared with Ppard(+/+) mice, indicating that PPARD modulates spermatogenesis. Higher expression of p27 and decreased expression of proliferating cellular nuclear antigen in Sertoli cells were observed in Ppard(+/+) mice as compared with Ppard(-/-) mice, and these were associated with decreased Sertoli cell number in Ppard(+/+) mice. Cyclin D1 and cyclin D2 expression was lower in Ppard(+/+) as compared with Ppard(-/-) mice. Ligand activation of PPARD inhibited proliferation of a mouse Sertoli cell line, TM4, and an inverse agonist of PPARD (DG172) rescued this effect. Temporal inhibition of extracellular signal-regulated kinase (ERK) activation by PPARD in the testis was observed in Ppard(+/+) mice and was associated with decreased serum follicle-stimulating hormone and higher claudin-11 expression along the blood-testis barrier. PPARD-dependent ERK activation also altered expression of claudin-11, p27, cyclin D1, and cyclin D2 in TM4 cells, causing inhibition of cell proliferation, maturation, and formation of tight junctions in Sertoli cells, thus confirming a requirement for PPARD in accurate Sertoli cell function. Combined, these results reveal for the first time that PPARD regulates spermatogenesis by modulating the function of Sertoli cells during early testis development.
Collapse
Affiliation(s)
- Pei-Li Yao
- From the Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802,
| | - LiPing Chen
- From the Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Rex A Hess
- Reproductive Biology and Toxicology, Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois, Urbana, Illinois 61802
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research, Center for Tumor and Immunobiology, Philipps University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany, and
| | - Frank J Gonzalez
- Laboratory of Metabolism, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Jeffrey M Peters
- From the Department of Veterinary and Biomedical Sciences and The Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802,
| |
Collapse
|
27
|
Giordano Attianese GMP, Desvergne B. Integrative and systemic approaches for evaluating PPARβ/δ (PPARD) function. NUCLEAR RECEPTOR SIGNALING 2015; 13:e001. [PMID: 25945080 PMCID: PMC4419664 DOI: 10.1621/nrs.13001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/09/2015] [Indexed: 12/13/2022]
Abstract
The peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptors that function as transcription factors regulating the expression of genes involved in cellular differentiation, development, metabolism and also tumorigenesis. Three PPAR isotypes (α, β/δ and γ) have been identified, among which PPARβ/δ is the most difficult to functionally examine due to its tissue-specific diversity in cell fate determination, energy metabolism and housekeeping activities. PPARβ/δ acts both in a ligand-dependent and -independent manner. The specific type of regulation, activation or repression, is determined by many factors, among which the type of ligand, the presence/absence of PPARβ/δ-interacting corepressor or coactivator complexes and PPARβ/δ protein post-translational modifications play major roles. Recently, new global approaches to the study of nuclear receptors have made it possible to evaluate their molecular activity in a more systemic fashion, rather than deeply digging into a single pathway/function. This systemic approach is ideally suited for studying PPARβ/δ, due to its ubiquitous expression in various organs and its overlapping and tissue-specific transcriptomic signatures. The aim of the present review is to present in detail the diversity of PPARβ/δ function, focusing on the different information gained at the systemic level, and describing the global and unbiased approaches that combine a systems view with molecular understanding.
Collapse
|
28
|
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
|
29
|
Kaupang Å, Hildonen S, Halvorsen TG, Mortén M, Vik A, Hansen TV. Involvement of covalent interactions in the mode of action of PPARβ/δ antagonists. RSC Adv 2015. [DOI: 10.1039/c5ra15707b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Investigations on the mode of action of several different chemical modulators of the peroxisome proliferator-activated receptor β/δ (PPARβ/δ) have been reported using MS and NMR experiments.
Collapse
Affiliation(s)
- Åsmund Kaupang
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Siri Hildonen
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Trine G. Halvorsen
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Magnus Mortén
- Department of Chemistry
- University of Oslo
- 0315 Oslo
- Norway
| | - Anders Vik
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Trond Vidar Hansen
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| |
Collapse
|
30
|
Design, synthesis, 3D pharmacophore, QSAR, and docking studies of carboxylic acid derivatives as Histone Deacetylase inhibitors and cytotoxic agents. Bioorg Chem 2014; 57:65-82. [DOI: 10.1016/j.bioorg.2014.08.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 08/12/2014] [Accepted: 08/24/2014] [Indexed: 11/23/2022]
|
31
|
Lieber S, Scheer F, Finkernagel F, Meissner W, Giehl G, Brendel C, Diederich WE, Müller-Brüsselbach S, Müller R. The inverse agonist DG172 triggers a PPARβ/δ-independent myeloid lineage shift and promotes GM-CSF/IL-4-induced dendritic cell differentiation. Mol Pharmacol 2014; 87:162-73. [PMID: 25398837 DOI: 10.1124/mol.114.094672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The stilbene derivative (Z)-2-(2-bromophenyl)-3-{[4-(1-methylpiperazine)amino]phenyl}acrylonitrile (DG172) was developed as a highly selective inhibitory peroxisome proliferator-activated receptor (PPAR)β/δ ligand. Here, we describe a novel PPARβ/δ-independent, yet highly specific, effect of DG172 on the differentiation of bone marrow cells (BMCs). DG172 strongly augmented granulocyte-macrophage-colony-stimulating factor (GM-CSF)-induced differentiation of primary BMCs from Ppard null mice into two specific populations, characterized as mature (CD11c(hi)MHCII(hi)) and immature (CD11c(hi)MHCII(lo)) dendritic cells (DCs). IL-4 synergized with DG172 to shift the differentiation from MHCII(lo) cells to mature DCs in vitro. The promotion of DC differentiation occurred at the expense of differentiation to granulocytic Gr1(+)Ly6B(+) cells. In agreement with these findings, transcriptome analyses showed a strong DG172-mediated repression of genes encoding neutrophilic markers in both differentiating wild-type and Ppard null cells, while macrophage/DC marker genes were up-regulated. DG172 also inhibited the expression of transcription factors driving granulocytic differentiation (Cebpe, Gfi1, and Klf5), and increased the levels of transcription factors promoting macrophage/DC differentiation (Irf4, Irf8, Spib, and Spic). DG172 exerted these effects only at an early stage of BMC differentiation induced by GM-CSF, did not affect macrophage-colony-stimulating factor-triggered differentiation to macrophages and had no detectable PPARβ/δ-independent effect on other cell types tested. Structure-function analyses demonstrated that the 4-methylpiperazine moiety in DG172 is required for its effect on DC differentiation, but is dispensable for PPARβ/δ binding. Based on these data we developed a new compound, (Z)-2-(4-chlorophenyl)-3-[4-(4-methylpiperazine-1-yl)phenyl]acrylonitrile (DG228), which enhances DC differentiation in the absence of significant PPARβ/δ binding.
Collapse
Affiliation(s)
- Sonja Lieber
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Frithjof Scheer
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Wolfgang Meissner
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Gavin Giehl
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Cornelia Brendel
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Wibke E Diederich
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Sabine Müller-Brüsselbach
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research (IMT), Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (S.L., F.F., W.M., S.M.-B., R.M.); Institute of Pharmaceutical Chemistry, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany (F.S., W.E.D.); and Clinic for Hematology, Oncology and Immunology (G.G., C.B.); Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany
| |
Collapse
|
32
|
Mackenzie LS, Lione L. Harnessing the benefits of PPARβ/δ agonists. Life Sci 2013; 93:963-7. [DOI: 10.1016/j.lfs.2013.10.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 10/04/2013] [Accepted: 10/21/2013] [Indexed: 01/03/2023]
|
33
|
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
|
34
|
Abstract
Besides its established functions in intermediary metabolism and developmental processes, the nuclear receptor peroxisome proliferator-activated receptor β/δ (PPARβ/δ) has a less defined role in tumorigenesis. In the present study, we have identified a function for PPARβ/δ in cancer cell invasion. We show that two structurally divergent inhibitory ligands for PPARβ/δ, the inverse agonists ST247 and DG172, strongly inhibit the serum- and transforming growth factor β (TGFβ)-induced invasion of MDA-MB-231 human breast cancer cells into a three-dimensional matrigel matrix. To elucidate the molecular basis of this finding, we performed chromatin immunoprecipitation sequencing (ChIP-Seq) and microarray analyses, which identified the gene encoding angiopoietin-like 4 (ANGPTL4) as the major transcriptional PPARβ/δ target in MDA-MB-231 cells, previously implicated in TGFβ-mediated tumor progression and metastatic dissemination. We show that the induction of ANGPTL4 by TGFβ and other oncogenic signals is strongly repressed by ST247 and DG172 in a PPARβ/δ-dependent fashion, resulting in the inhibition of ANGPTL4 secretion. This effect is attributable to these ligands' ability to induce a dominant transcriptional repressor complex at the site of transcription initiation that blocks preinitiation complex formation through an histone deacetylase-independent, non-canonical mechanism. Repression of ANGPTL4 transcription by inverse PPARβ/δ agonists is functionally linked to the inhibition of cancer cell invasion into a three-dimensional matrix, as (i) invasion of MDA-MB-231 cells is critically dependent on ANGPTL4 expression, (ii) recombinant ANGPTL4 stimulates invasion, and (iii) reverses the inhibitory effect of ST247 and DG172. These findings indicate that a PPARβ/δ-ANGPTL4 pathway is involved in the regulation of tumor cell invasion and that its pharmacological manipulation by inverse PPARβ/δ agonists is feasible.
Collapse
|
35
|
Fibrate-derived N-(methylsulfonyl)amides with antagonistic properties on PPARα. Eur J Med Chem 2012; 58:317-22. [DOI: 10.1016/j.ejmech.2012.10.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 10/08/2012] [Accepted: 10/11/2012] [Indexed: 11/19/2022]
|
36
|
Huang J, Xu W, Xie H, Li S. One-step cyclization: synthesis of N-heteroalkyl-N'-tosylpiperazines. J Org Chem 2012; 77:7506-11. [PMID: 22849619 DOI: 10.1021/jo3012896] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Piperazine derivatives are important intermediates in organic synthesis and useful building blocks in pharmaceutical and fine chemical industries. Currently available synthetic routes for these heterocyclic compounds have limited scope owing to the harsh reaction conditions, low yields, and multistep process. Herein, we reported a practical method for synthesis of alkyl-, alcohol-, amine-, and ester-extended tosylpiperazines under mild conditions with moderate to high yields. This protocol exhibits potential applicability in the synthesis of pharmaceuticals and natural products because of the operational simplicity and the conveniently available reactants. On the basis of the experimental and theoretical results, a plausible mechanism of aliphatic nucleophilic substitution (S(N)) in the cyclization has been postulated and evidence for the formation of a six-membered ring has also been confirmed by means of density functional theory (DFT) calculations.
Collapse
Affiliation(s)
- Jianying Huang
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, P. R. China.
| | | | | | | |
Collapse
|