51
|
Cooper A, Singh S, Hook S, Tyndall JDA, Vernall AJ. Chemical Tools for Studying Lipid-Binding Class A G Protein-Coupled Receptors. Pharmacol Rev 2017; 69:316-353. [PMID: 28655732 DOI: 10.1124/pr.116.013243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 05/15/2017] [Indexed: 12/16/2022] Open
Abstract
Cannabinoid, free fatty acid, lysophosphatidic acid, sphingosine 1-phosphate, prostanoid, leukotriene, bile acid, and platelet-activating factor receptor families are class A G protein-coupled receptors with endogenous lipid ligands. Pharmacological tools are crucial for studying these receptors and addressing the many unanswered questions surrounding expression of these receptors in normal and diseased tissues. An inherent challenge for developing tools for these lipid receptors is balancing the often lipophilic requirements of the receptor-binding pharmacophore with favorable physicochemical properties to optimize highly specific binding. In this study, we review the radioligands, fluorescent ligands, covalent ligands, and antibodies that have been used to study these lipid-binding receptors. For each tool type, the characteristics and design rationale along with in vitro and in vivo applications are detailed.
Collapse
Affiliation(s)
- Anna Cooper
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sameek Singh
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
52
|
Zhang S, Liu B, Mao W, Li Q, Fu C, Zhang N, Zhang Y, Gao L, Shen Y, Cao J. The effect of prostaglandin E 2 receptor (PTGER2) activation on growth factor expression and cell proliferation in bovine endometrial explants. Prostaglandins Leukot Essent Fatty Acids 2017; 122:16-23. [PMID: 28735624 DOI: 10.1016/j.plefa.2017.06.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/18/2017] [Accepted: 06/27/2017] [Indexed: 12/14/2022]
Abstract
The domestic animal endometrium undergoes regular periods of regeneration and degeneration during cycles of oestrus and dioestrus. If blastocyst implantation occurs in the uterus, the endometrium will prepare for pregnancy by changing its pattern of development to build a connection with the embryo to nourish it. Prostaglandin E2 (PGE2) secretion synchronized with endometrial growth in these processes and could be involved in endometrial growth. One of the PGE2 receptors (PTGER2) is present in endometrium and its increased expression accompanies with endometrial growth in above processes. However, the association between PTGER2 and endometrial growth remains unclear. Endometrial growth factors and cell proliferation is the foundation for endometrial growth. Therefore, in this study, the response of growth factors and cell proliferation essential for endometrial growth to PTGER2 activation were investigated in bovine endometrium. The results indicated that mRNA and protein expression of connective tissue growth factor (CTGF), fibroblast growth factor-2 (FGF-2), interleukin-8 (IL-8), transforming growth factor-β1 (TGF-β1), matrix metalloproteinase-2 (MMP-2), and vascular endothelial growth factor A (VEGFA) were up-regulated after PTGER2 activation by corresponding agonist butaprost (P < 0.05), and proliferation of endometrial epithelia and fibroblasts were induced in response to increased levels of proliferating cell nuclear antigen (PCNA), cytokeratin-18 (CK-18) and fibroblast-specific protein 1 (FSP-1) expression in bovine endometrial explants in vitro (P < 0.05).
Collapse
Affiliation(s)
- Shuangyi Zhang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Bo Liu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Wei Mao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Qianru Li
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Changqi Fu
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Nan Zhang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Ying Zhang
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Long Gao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Yuan Shen
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China
| | - Jinshan Cao
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, China.
| |
Collapse
|
53
|
Umei K, Nishigaya Y, Tatani K, Kohno Y, Tanaka N, Seto S. Identification of novel 1,2,3,6-tetrahydropyridyl-substituted benzo[ d ]thiazoles: Lead generation and optimization toward potent and orally active EP 1 receptor antagonists. Bioorg Med Chem 2017; 25:3406-3430. [DOI: 10.1016/j.bmc.2017.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 01/13/2023]
|
54
|
Prostaglandin E 2 Induces Prorenin-Dependent Activation of (Pro)renin Receptor and Upregulation of Cyclooxygenase-2 in Collecting Duct Cells. Am J Med Sci 2017; 354:310-318. [PMID: 28918839 DOI: 10.1016/j.amjms.2017.05.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/26/2017] [Accepted: 05/28/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prostaglandin E2 (PGE2) regulates renin expression in renal juxtaglomerular cells. PGE2 acts through E-prostanoid (EP) receptors in the renal collecting duct (CD) to regulate sodium and water balance. CD cells express EP1 and EP4, which are linked to protein kinase C (PKC) and PKA downstream pathways, respectively. Previous studies showed that the presence of renin in the CD, and that of PKC and PKA pathways, activate its expression. The (pro)renin receptor (PRR) is also expressed in CD cells, and its activation enhances cyclooxygenase-2 (COX-2) through extracellular signal-regulated kinase (ERK). We hypothesized that PGE2 stimulates prorenin and renin synthesis leading to subsequent activation of PRR and upregulation of COX-2. METHODS We used a mouse M-1 CD cell line that expresses EP1, EP3 and EP4 but not EP2. RESULTS PGE2 (10-6M) treatment increased prorenin and renin protein levels at 4 and 8 hours. No differences were found at 12-hour after PGE2 treatment. Phospho-ERK was significantly augmented after 12 hours. COX-2 expression was decreased after 4 hours of PGE2 treatment, but increased after 12 hours. Interestingly, the full-length form of the PRR was upregulated only at 12 hours. PGE2-mediated phospho-ERK and COX-2 upregulation was suppressed by PRR silencing. CONCLUSIONS Our results suggest that PGE2 induces biphasic regulation of COX-2 through renin-dependent PRR activation via EP1 and EP4 receptors. PRR-mediated increases in COX-2 expression may enhance PGE2 synthesis in CD cells serving as a buffer mechanism in conditions of activated renin-angiotensin system.
Collapse
|
55
|
Umei K, Nishigaya Y, Kondo A, Tatani K, Tanaka N, Kohno Y, Seto S. Novel pyrazolo[1,5- a ]pyridines as orally active EP 1 receptor antagonists: Synthesis, structure-activity relationship studies, and biological evaluation. Bioorg Med Chem 2017; 25:2635-2642. [DOI: 10.1016/j.bmc.2017.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/27/2017] [Accepted: 03/03/2017] [Indexed: 10/20/2022]
|
56
|
Park HJ, Baek K, Baek JH, Kim HR. TNFα Increases RANKL Expression via PGE₂-Induced Activation of NFATc1. Int J Mol Sci 2017; 18:ijms18030495. [PMID: 28245593 PMCID: PMC5372511 DOI: 10.3390/ijms18030495] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/12/2017] [Accepted: 02/20/2017] [Indexed: 01/07/2023] Open
Abstract
Tumor necrosis factor α (TNFα) is known to upregulate the expression of receptor activator of NF-κB ligand (RANKL). We investigated the role of the calcineurin/nuclear factor of activated T-cells (NFAT) signaling pathway in TNFα-induced RANKL expression in C2C12 and primary cultured mouse calvarial cells. TNFα-induced RANKL expression was blocked by the calcineurin/NFAT pathway inhibitors. TNFα increased NFAT transcriptional activity and subsequent RANKL promoter binding. Mutations in the NFAT-binding element (MT(N)) suppressed TNFα-induced RANKL promoter activity. TNFα increased prostaglandin E2 (PGE2) production, which in turn enhanced NFAT transcriptional activity and binding to the RANKL promoter. MT(N) suppressed PGE2-induced RANKL promoter activity. TNFα and PGE2 increased the expression of RANKL, NFAT cytoplasmic-1 (NFATc1), cAMP response element-binding protein (CREB), and cyclooxygenase 2 (COX2); which increment was suppressed by indomethacin, a COX inhibitor. Mutations in the CRE-like element blocked PGE2-induced RANKL promoter activity. PGE2 induced the binding of CREB to the RANKL promoter, whereas TNFα increased the binding of both CREB and NFATc1 to this promoter through a process blocked by indomethacin. The PGE2 receptor antagonists AH6809 and AH23848 blocked TNFα-induced expression of RANKL, NFATc1, and CREB; transcriptional activity of NFAT; and binding of NFATc1 or CREB to the RANKL promoter. These results suggest that TNFα-induced RANKL expression depends on PGE2 production and subsequent transcriptional activation/enhanced binding of NFATc1 and CREB to the RANKL promoter.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Korea.
| | - Kyunghwa Baek
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangwon-do 25457, Korea.
| | - Jeong-Hwa Baek
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 08826, Korea.
| | | |
Collapse
|
57
|
Shichiri H, Yamamoto K, Tokura M, Ishida T, Uda A, Bito T, Nishigori C, Nakagawa T, Hirano T, Yano I, Hirai M. Prostaglandin E 1 reduces the keratinocyte toxicity of sorafenib by maintaining signal transducer and activator of transcription 3 (STAT3) activity and enhancing the cAMP response element binding protein (CREB) activity. Biochem Biophys Res Commun 2017; 485:227-233. [PMID: 28237700 DOI: 10.1016/j.bbrc.2017.02.107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/20/2017] [Indexed: 11/25/2022]
Abstract
Hand-foot skin reaction (HFSR) is a common side effect of multiple tyrosine kinase inhibitors (mTKIs). HFSR can necessitate dose reductions or interruption of therapy owing to its negative effect on the quality of life. Therefore, effective use of mTKIs requires measures to prevent HFSR. We evaluated the effect of prostaglandin E1 (PGE1) on HFSR, because PGE1 is already used to treat bed sores and skin ulcers and has established angiogenic and antiproliferative effects in keratinocytes. We found that the pathogenesis of sorafenib-induced HFSR is characterized by a decrease in levels of a phosphorylated signal transducer and activator of transcription 3 (STAT3). We investigated the effect of PGE1 on the sorafenib-mediated reduction in phosphorylated STAT3 levels in HaCaT human epidermal keratinocytes. In cells treated with sorafenib, phosphorylated STAT3 levels decreased in a concentration-dependent manner, and this effect was blocked in cells treated with sorafenib and PGE1. Furthermore, the expression of phosphorylated STAT3, the antiapoptotic proteins myeloid cell leukemia-1 (Mcl-1) and survivin decreased in cells pretreated with an inhibitor of cAMP response element binding protein (CREB). Cell viability increased in cells treated with sorafenib and PGE1 compared with that in cells treated with sorafenib alone, and these effects were not observed in STAT3 knockdown HaCaT cells. Collectively, these findings indicate that PGE1 blocks the inhibitory effects of sorafenib on cell growth by maintaining the activity of STAT3 and enhancing the CREB activity. Therefore, PGE1 might represent an effective treatment for the prevention of sorafenib-induced HFSR.
Collapse
Affiliation(s)
- Hiroaki Shichiri
- Division of Pharmaceutics, Department of Internal Related, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kazuhiro Yamamoto
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan.
| | - Maya Tokura
- Educational Center for Clinical Pharmacy, Kobe Pharmaceutical University, 4-19-1, Motoyamakitamachi, Higashinada-ku, Kobe-shi, Hyogo, 658-8558, Japan
| | - Takahiro Ishida
- R&D Department, Momotani Juntenkan LTD, 1-4-1, Uemachi, Chuo-ku, Osaka, 540-0005, Japan
| | - Atsushi Uda
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Toshinori Bito
- Division of Dermatology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Chikako Nishigori
- Division of Dermatology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tsutomu Nakagawa
- Division of Pharmaceutics, Department of Internal Related, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takeshi Hirano
- Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Ikuko Yano
- Division of Pharmaceutics, Department of Internal Related, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Midori Hirai
- Division of Pharmaceutics, Department of Internal Related, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan; Department of Pharmacy, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
58
|
Akasaka H, Thaliachery N, Zheng X, Blumenthal M, Nikhar S, Murdoch EE, Ling Q, Ruan KH. The key residue within the second extracellular loop of human EP3 involved in selectively turning down PGE 2- and retaining PGE 1-mediated signaling in live cells. Arch Biochem Biophys 2017; 616:20-29. [PMID: 28065721 DOI: 10.1016/j.abb.2016.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/15/2016] [Accepted: 12/02/2016] [Indexed: 10/20/2022]
Abstract
Key residues and binding mechanisms of PGE1 and PGE2 on prostanoid receptors are poorly understood due to the lack of X-ray structures for the receptors. We constructed a human EP3 (hEP3) model through integrative homology modeling using the X-ray structure of the β2-adrenergic receptor transmembrane domain and NMR structures of the thromboxane A2 receptor extracellular loops. PGE1 and PGE2 docking into the hEP3 model showed differing configurations within the extracellular ligand recognition site. While PGE2 could form possible binding contact with S211, PGE1 is unable to form similar contacts. Therefore, S211 could be the critical residue for PGE2 recognition, but is not a significant for PGE1. This prediction was confirmed using HEK293 cells transfected with hEP3 S211L cDNA. The S211L cells lost PGE2 binding and signaling. Interestingly, the S211L cells retained PGE1-mediated signaling. It indicates that S211 within the second extracellular loop is a key residue involved in turning down PGE2 signaling. Our study provided information that S211L within EP3 is the key residue to distinguish PGE1 and PGE2 binding to mediate diverse biological functions at the initial recognition step. The S211L mutant could be used as a model for studying the binding mechanism and signaling pathway specifically mediated by PGE1.
Collapse
Affiliation(s)
- Hironari Akasaka
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| | - Natasha Thaliachery
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| | - Xianghai Zheng
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| | - Marissa Blumenthal
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| | - Sameer Nikhar
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| | - Emma E Murdoch
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| | - Qinglan Ling
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| | - Ke-He Ruan
- Center for Experimental Therapeutics and Pharmacoinformatics and Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204-5037, USA
| |
Collapse
|
59
|
Theiler A, Konya V, Pasterk L, Maric J, Bärnthaler T, Lanz I, Platzer W, Schuligoi R, Heinemann A. The EP1/EP3 receptor agonist 17-pt-PGE 2 acts as an EP4 receptor agonist on endothelial barrier function and in a model of LPS-induced pulmonary inflammation. Vascul Pharmacol 2016; 87:180-189. [PMID: 27664754 DOI: 10.1016/j.vph.2016.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/18/2022]
Abstract
Endothelial dysfunction is a hallmark of inflammatory conditions. We recently demonstrated that prostaglandin (PG)E2 enhances the resistance of pulmonary endothelium in vitro and counteracts lipopolysaccharide (LPS)-induced pulmonary inflammation in vivo via EP4 receptors. The aim of this study was to investigate the role of the EP1/EP3 receptor agonist 17-phenyl-trinor-(pt)-PGE2 on acute lung inflammation in a mouse model. In LPS-induced pulmonary inflammation in mice, 17-pt-PGE2 reduced neutrophil infiltration and inhibited vascular leakage. These effects were unaltered by an EP1 antagonist, but reversed by EP4 receptor antagonists. 17-pt-PGE2 increased the resistance of pulmonary microvascular endothelial cells and prevented thrombin-induced disruption of endothelial junctions. Again, these effects were not mediated via EP1 or EP3 but through activation of the EP4 receptor, as demonstrated by the lack of effect of more selective EP1 and EP3 receptor agonists, prevention of these effects by EP4 antagonists and EP4 receptor knock-down by siRNA. In contrast, the aggregation enhancing effect of 17-pt-PGE2 in human platelets was mediated via EP3 receptors. Our results demonstrate that 17-pt-PGE2 enhances the endothelial barrier in vitro on pulmonary microvascular endothelial cells, and accordingly ameliorates the recruitment of neutrophils, via EP4 receptors in vivo. This suggests a beneficial effect of 17-pt-PGE2 on pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Anna Theiler
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Viktoria Konya
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Lisa Pasterk
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Jovana Maric
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Thomas Bärnthaler
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Ilse Lanz
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Wolfgang Platzer
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Rufina Schuligoi
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| |
Collapse
|
60
|
Schneider G, Reker D, Chen T, Hauenstein K, Schneider P, Altmann KH. Deorphaning the Macromolecular Targets of the Natural Anticancer Compound Doliculide. Angew Chem Int Ed Engl 2016; 55:12408-11. [PMID: 27605391 DOI: 10.1002/anie.201605707] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/11/2016] [Indexed: 11/06/2022]
Abstract
The cyclodepsipeptide doliculide is a marine natural product with strong actin-polymerizing and anticancer activities. Evidence for doliculide acting as a potent and subtype-selective antagonist of prostanoid E receptor 3 (EP3) is presented. Computational target prediction suggested that this membrane receptor is a likely macromolecular target and enabled immediate in vitro validation. This proof-of-concept study demonstrates the in silico deorphanization of phenotypic screening hits as a viable concept for future natural-product-inspired chemical biology and drug discovery efforts.
Collapse
Affiliation(s)
- Gisbert Schneider
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, 8093, Zürich, Switzerland.
| | - Daniel Reker
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, 8093, Zürich, Switzerland
| | - Tao Chen
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, 8093, Zürich, Switzerland
| | - Kurt Hauenstein
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, 8093, Zürich, Switzerland
| | - Petra Schneider
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, 8093, Zürich, Switzerland.,inSili.com LLC, Zürich, Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Wolfgang-Pauli-Strasse 10, 8093, Zürich, Switzerland
| |
Collapse
|
61
|
Schneider G, Reker D, Chen T, Hauenstein K, Schneider P, Altmann KH. Deorphaning the Macromolecular Targets of the Natural Anticancer Compound Doliculide. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201605707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Gisbert Schneider
- Department of Chemistry and Applied Biosciences; Institute of Pharmaceutical Sciences; ETH Zürich; Wolfgang-Pauli-Strasse 10 8093 Zürich Switzerland
| | - Daniel Reker
- Department of Chemistry and Applied Biosciences; Institute of Pharmaceutical Sciences; ETH Zürich; Wolfgang-Pauli-Strasse 10 8093 Zürich Switzerland
| | - Tao Chen
- Department of Chemistry and Applied Biosciences; Institute of Pharmaceutical Sciences; ETH Zürich; Wolfgang-Pauli-Strasse 10 8093 Zürich Switzerland
| | - Kurt Hauenstein
- Department of Chemistry and Applied Biosciences; Institute of Pharmaceutical Sciences; ETH Zürich; Wolfgang-Pauli-Strasse 10 8093 Zürich Switzerland
| | - Petra Schneider
- Department of Chemistry and Applied Biosciences; Institute of Pharmaceutical Sciences; ETH Zürich; Wolfgang-Pauli-Strasse 10 8093 Zürich Switzerland
- inSili.com LLC; Zürich Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences; Institute of Pharmaceutical Sciences; ETH Zürich; Wolfgang-Pauli-Strasse 10 8093 Zürich Switzerland
| |
Collapse
|
62
|
Structural features of subtype-selective EP receptor modulators. Drug Discov Today 2016; 22:57-71. [PMID: 27506873 DOI: 10.1016/j.drudis.2016.08.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/20/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022]
Abstract
Prostaglandin E2 is a potent endogenous molecule that binds to four different G-protein-coupled receptors: EP1-4. Each of these receptors is a valuable drug target, with distinct tissue localisation and signalling pathways. We review the structural features of EP modulators required for subtype-selective activity, as well as the structural requirements for improved pharmacokinetic parameters. Novel EP receptor subtype selective agonists and antagonists appear to be valuable drug candidates in the therapy of many pathophysiological states, including ulcerative colitis, glaucoma, bone healing, B cell lymphoma, neurological diseases, among others, which have been studied in vitro, in vivo and in early phase clinical trials.
Collapse
|
63
|
Kim SO, Duffy DM. Mapping PTGERs to the Ovulatory Follicle: Regional Responses to the Ovulatory PGE2 Signal. Biol Reprod 2016; 95:33. [PMID: 27307073 PMCID: PMC5029471 DOI: 10.1095/biolreprod.116.140574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/27/2016] [Accepted: 06/03/2016] [Indexed: 12/11/2022] Open
Abstract
Prostaglandin E2 (PGE2) is a key intrafollicular mediator of ovulation in many, if not all, mammalian species. PGE2 acts at follicular cells via four distinct PGE2 receptors (PTGERs). Within the ovulatory follicle, each cell type (e.g., oocyte, cumulus granulosa cell, mural granulosa cell, theca cell, endothelial cell) expresses a different subset of the four PTGERs. Expression of a subset of PTGERs has consequences for the generation of intracellular signals and ultimately the unique functions of follicular cells that respond to PGE2. Just as the ovulatory LH surge regulates PGE2 synthesis, the LH surge also regulates expression of the four PTGERs. The pattern of expression of the four PTGERs among follicular cells before and after the LH surge forms a spatial and temporal map of PGE2 responses. Differential PTGER expression, coupled with activation of cell-specific intracellular signals, may explain how a single paracrine mediator can have pleotropic actions within the ovulatory follicle. Understanding the role of each PTGER in ovulation may point to previously unappreciated opportunities to both promote and prevent fertility.
Collapse
Affiliation(s)
- Soon Ok Kim
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
64
|
Hao S, DelliPizzi A, Quiroz-Munoz M, Jiang H, Ferreri NR. The EP3 receptor regulates water excretion in response to high salt intake. Am J Physiol Renal Physiol 2016; 311:F822-F829. [PMID: 27465993 DOI: 10.1152/ajprenal.00589.2015] [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: 12/21/2015] [Accepted: 07/25/2016] [Indexed: 12/31/2022] Open
Abstract
The mechanisms by which prostanoids contribute to the maintenance of whole body water homeostasis are complex and not fully understood. The present study demonstrates that an EP3-dependent feedback mechanism contributes to the regulation of water homeostasis under high-salt conditions. Rats on a normal diet and tap water were placed in metabolic cages and given either sulprostone (20 μg·kg-1·day-1) or vehicle for 3 days to activate EP3 receptors in the thick ascending limb (TAL). Treatment was continued for another 3 days in rats given either 1% NaCl in the drinking water or tap water. Sulprostone decreased expression of cyclooxygenase 2 (COX-2) expression by ∼75% in TAL tubules from rats given 1% NaCl concomitant with a ∼60% inhibition of COX-2-dependent PGE2 levels in the kidney. Urine volume increased after ingestion of 1% NaCl but was reduced ∼40% by sulprostone. In contrast, the highly selective EP3 receptor antagonist L-798106 (100 μg·kg-1·day-1), which increased COX-2 expression and renal PGE2 production, increased urine volume in rats given 1% NaCl. Sulprostone increased expression of aquaporin-2 (AQP2) in the inner medullary collecting duct plasma membrane in association with an increase in phosphorylation at Ser269 and decrease in Ser261 phosphorylation; antagonism of EP3 with L-798106 reduced AQP2 expression. Thus, although acute activation of EP3 by PGE2 in the TAL and collecting duct inhibits the Na-K-2Cl cotransporter and AQP2 activity, respectively, chronic activation of EP3 in vivo limits the extent of COX-2-derived PGE2 synthesis, thereby mitigating the inhibitory effects of PGE2 on these transporters and decreasing urine volume.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | | | - Mariana Quiroz-Munoz
- Department of Physiology, Center for Aging and Regeneration, CARE Chile UC, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Houli Jiang
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York;
| |
Collapse
|
65
|
An EP2 Agonist Facilitates NMDA-Induced Outward Currents and Inhibits Dendritic Beading through Activation of BK Channels in Mouse Cortical Neurons. Mediators Inflamm 2016; 2016:5079597. [PMID: 27298516 PMCID: PMC4889853 DOI: 10.1155/2016/5079597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/19/2016] [Accepted: 05/04/2016] [Indexed: 11/17/2022] Open
Abstract
Prostaglandin E2 (PGE2), a major metabolite of arachidonic acid produced by cyclooxygenase pathways, exerts its bioactive responses by activating four E-prostanoid receptor subtypes, EP1, EP2, EP3, and EP4. PGE2 enables modulating N-methyl-D-aspartate (NMDA) receptor-mediated responses. However, the effect of E-prostanoid receptor agonists on large-conductance Ca2+-activated K+ (BK) channels, which are functionally coupled with NMDA receptors, remains unclear. Here, we showed that EP2 receptor-mediated signaling pathways increased NMDA-induced outward currents (INMDA-OUT), which are associated with the BK channel activation. Patch-clamp recordings from the acutely dissociated mouse cortical neurons revealed that an EP2 receptor agonist activated INMDA-OUT, whereas an EP3 receptor agonist reduced it. Agonists of EP1 or EP4 receptors showed no significant effects on INMDA-OUT. A direct perfusion of 3,5′-cyclic adenosine monophosphate (cAMP) through the patch pipette facilitated INMDA-OUT, which was abolished by the presence of protein kinase A (PKA) inhibitor. Furthermore, facilitation of INMDA-OUT caused by an EP2 receptor agonist was significantly suppressed by PKA inhibitor. Finally, the activation of BK channels through EP2 receptors facilitated the recovery phase of NMDA-induced dendritic beading in the primary cultured cortical neurons. These results suggest that a direct activation of BK channels by EP2 receptor-mediated signaling pathways plays neuroprotective roles in cortical neurons.
Collapse
|
66
|
Abstract
STUDY DESIGN In vitro study using isolated human intervertebral disc (IVD) cells. OBJECTIVE To investigate the effects of prostaglandin (PG)E1 and its orally available derivative limaprost on the regulation of nerve growth factor (NGF) expression and to compare their actions with other prostanoids using interleukin (IL)-1-stimulated human IVD cells. SUMMARY OF BACKGROUND DATA We previously reported that a selective COX-2 inhibitor enhanced, whereas PGE2 suppressed the induction of NGF by IL-1 in human IVD cells, and proposed that PGE2 can suppress NGF expression by a negative feedback mechanism. METHODS Isolated human IVD cells were stimulated with IL-1 in the presence or absence of increasing concentrations of PGE2, PGE1, limaprost, PGI2, PGD2, or PGF2α (10-10,000 nM). For some experiments, an E-series prostanoid receptor (EP)4 antagonist (L-161,982) was added prior to the stimulation. NGF expression was determined by real-time polymerase chain reaction and its protein level was quantified by enzyme-linked immunosorbent assay. RESULTS PGE2, PGE1, and limaprost inhibited the IL-1-mediated induction of NGF in a concentration-dependent manner, with IC50 values of 9.9, 10.6, and 70.9 nM, respectively. PGI2 also suppressed NGF expression but to a much less extent. PGD2, on the other hand, significantly enhanced NGF expression, whereas PGF2α had no effect. Protein expression levels of NGF mirrored its mRNA levels. The suppression of NGF expression by PGE2 and PGE1 was partly reversed by L-161,982. CONCLUSION PGE1 and limaprost exhibited a novel pharmacological action that suppresses NGF expression in human IVD cells, and other prostanoids differentially regulated NGF expression. Limaprost has been used to treat patients with lumbar spinal stenosis in Japan and was proved to be effective in relieving symptoms. Our in vitro results may explain, in part, the mechanism of action of limaprost for low back pain. LEVEL OF EVIDENCE N/A.
Collapse
|
67
|
Santini G, Mores N, Malerba M, Mondino C, Macis G, Montuschi P. Investigational prostaglandin D2 receptor antagonists for airway inflammation. Expert Opin Investig Drugs 2016; 25:639-52. [PMID: 27094922 DOI: 10.1080/13543784.2016.1175434] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION By activating DP1 and DP2 receptors on immune and non-immune cells, prostaglandin D2 (PGD2), a major metabolic product of cyclo-oxygenase pathway released after IgE-mediated mast cell activation, has pro-inflammatory effects, which are relevant to the pathophysiology of allergic airway disease. At least 15 selective, orally active, DP2 receptor antagonists and one DP1 receptor antagonist (asapiprant) are under development for asthma and/or allergic rhinitis. AREAS COVERED In this review, the authors cover the pharmacology of PGD2 and PGD2 receptor antagonists and look at the preclinical, phase I and phase II studies with selective DP1 and DP2 receptor antagonists. EXPERT OPINION Future research should aim to develop once daily compounds and increase the drug clinical potency which, apart from OC000459 and ADC-3680, seems to be relatively low. Further research and development of DP2 receptor antagonists is warranted, particularly in patients with severe uncontrolled asthma, whose management is a top priority. Pediatric studies, which are not available, are required for assessing the efficacy and safety of this novel drug class in children with asthma and allergic rhinitis. Studies on the efficacy of DP2 receptor antagonists in various asthma phenotypes including: smokers, obese subjects, early vs late asthma onset, fixed vs reversible airflow limitation, are required for establishing their pharmacotherapeutic role.
Collapse
Affiliation(s)
- Giuseppe Santini
- a Department of Pharmacology, Faculty of Medicine , Catholic University of the Sacred Heart , Rome , Italy
| | - Nadia Mores
- a Department of Pharmacology, Faculty of Medicine , Catholic University of the Sacred Heart , Rome , Italy
| | - Mario Malerba
- b Department of Internal Medicine , University of Brescia , Brescia , Italy
| | - Chiara Mondino
- c Department of Allergology , 'Bellinzona e Valli' Hospital , Bellinzona , Switzerland
| | - Giuseppe Macis
- d Department of Radiological Sciences, Faculty of Medicine , Catholic University of the Sacred Heart , Rome , Italy
| | - Paolo Montuschi
- a Department of Pharmacology, Faculty of Medicine , Catholic University of the Sacred Heart , Rome , Italy
| |
Collapse
|
68
|
St-Jacques B, Ma W. Preferred recycling pathway by internalized PGE2 EP4 receptor following agonist stimulation in cultured dorsal root ganglion neurons contributes to enhanced EP4 receptor sensitivity. Neuroscience 2016; 326:56-68. [PMID: 27060485 DOI: 10.1016/j.neuroscience.2016.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 04/01/2016] [Accepted: 04/02/2016] [Indexed: 10/22/2022]
Abstract
Prostaglandin E2 (PGE2), a well-known pain mediator abundantly produced in injured tissues, sensitizes nociceptive dorsal root ganglion (DRG) neurons (nociceptors) through its four EP receptors (EP1-4). Our prior study showed that PGE2 or EP4 agonist stimulates EP4 externalization and this event was not only suppressed by the inhibitor of anterograde export, but also by the recycling inhibitor (St-Jacques and Ma, 2013). These data suggest that EP4 recycling also contributes to agonist-enhanced EP4 surface abundance. In the current study, we tested this hypothesis using antibody-feeding-based internalization assay, recycling assay and FITC-PGE2 binding assay. We observed that selective EP4 agonist 1-hydroxy-PGE1 (1-OH-PGE1) or CAY10850 time- and concentration-dependently increased EP4 internalization in cultured DRG neuron. Internalized EP4 was predominantly localized in the early endosomes and recycling endosomes, but rarely in the late endosomes and lysosomes. These observations were confirmed by FITC-PGE2 binding assay. We further revealed that 1-OH-PGE1 or CAY10850 time- and concentration-dependently increased EP4 recycling. Double exposures to 1-OH-PGE1 induced a greater increase in calcitonin gene-related peptide (CGRP) release than a single exposure or vehicle exposure, an event blocked by pre-treatment with the recycling inhibitor monensin. Our data suggest that EP4 recycling contributes to agonist-induced cell surface abundance and consequently enhanced receptor sensitivity. Facilitating EP4 externalization and recycling is a novel mechanism underlying PGE2-induced nociceptor sensitization.
Collapse
Affiliation(s)
- Bruno St-Jacques
- Douglas Mental Health University Institute, McGill University, Montréal, Québec H4H 1R3, Canada
| | - Weiya Ma
- Douglas Mental Health University Institute, McGill University, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H4H 1R3, Canada.
| |
Collapse
|
69
|
Tse KH, Chow KBS, Wise H. PGE2 released by primary sensory neurons modulates Toll-like receptor 4 activities through an EP4 receptor-dependent process. J Neuroimmunol 2016; 293:8-16. [PMID: 27049555 DOI: 10.1016/j.jneuroim.2016.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/28/2016] [Accepted: 02/08/2016] [Indexed: 11/20/2022]
Abstract
Exogenous prostaglandin E2 (PGE2) displays mixed regulatory properties with regard to inflammatory gene expression in dorsal root ganglion (DRG) cells. We show here that endogenously-produced nanomolar concentrations of PGE2, such as that generated in response to Toll-like receptor 4 (TLR4) stimulation, inhibits both cyclooxygenase-2 (COX-2) and tumour necrosis factor alpha (TNFα) mRNA expression in DRG cells in an EP4 receptor-dependent manner. DRG neurons appear to be the major source of PGE2 in the DRG and likely serve as both an autocrine and paracrine system for limiting over-activation of both DRG neurons and glial cells in response to TLR4 stimulation.
Collapse
Affiliation(s)
- Kai-Hei Tse
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Kevin B S Chow
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Helen Wise
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
70
|
Molecular mechanisms of target recognition by lipid GPCRs: relevance for cancer. Oncogene 2015; 35:4021-35. [PMID: 26640151 DOI: 10.1038/onc.2015.467] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 12/18/2022]
|
71
|
Eichner C, Øvergård AC, Nilsen F, Dalvin S. Molecular characterization and knock-down of salmon louse (Lepeophtheirus salmonis) prostaglandin E synthase. Exp Parasitol 2015; 159:79-93. [DOI: 10.1016/j.exppara.2015.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/23/2015] [Accepted: 09/03/2015] [Indexed: 11/29/2022]
|
72
|
Dissecting fibrosis: therapeutic insights from the small-molecule toolbox. Nat Rev Drug Discov 2015; 14:693-720. [PMID: 26338155 DOI: 10.1038/nrd4592] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrosis, which leads to progressive loss of tissue function and eventual organ failure, has been estimated to contribute to ~45% of deaths in the developed world, and so new therapeutics to modulate fibrosis are urgently needed. Major advances in our understanding of the mechanisms underlying pathological fibrosis are supporting the search for such therapeutics, and the recent approval of two anti-fibrotic drugs for idiopathic pulmonary fibrosis has demonstrated the tractability of this area for drug discovery. This Review examines the pharmacology and structural information for small molecules being evaluated for lung, liver, kidney and skin fibrosis. In particular, we discuss the insights gained from the use of these pharmacological tools, and how these entities can inform, and probe, emerging insights into disease mechanisms, including the potential for future drug combinations.
Collapse
|
73
|
Prostanoids regulate angiogenesis acting primarily on IP and EP4 receptors. Microvasc Res 2015; 101:127-34. [DOI: 10.1016/j.mvr.2015.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 07/02/2015] [Accepted: 07/15/2015] [Indexed: 02/02/2023]
|
74
|
Manieri NA, Mack MR, Himmelrich MD, Worthley DL, Hanson EM, Eckmann L, Wang TC, Stappenbeck TS. Mucosally transplanted mesenchymal stem cells stimulate intestinal healing by promoting angiogenesis. J Clin Invest 2015; 125:3606-18. [PMID: 26280574 DOI: 10.1172/jci81423] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/08/2015] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cell (MSC) therapy is an emerging field of regenerative medicine; however, it is often unclear how these cells mediate repair. Here, we investigated the use of MSCs in the treatment of intestinal disease and modeled abnormal repair by creating focal wounds in the colonic mucosa of prostaglandin-deficient mice. These wounds developed into ulcers that infiltrated the outer intestinal wall. We determined that penetrating ulcer formation in this model resulted from increased hypoxia and smooth muscle wall necrosis. Prostaglandin I₂ (PGI₂) stimulated VEGF-dependent angiogenesis to prevent penetrating ulcers. Treatment of mucosally injured WT mice with a VEGFR inhibitor resulted in the development of penetrating ulcers, further demonstrating that VEGF is critical for mucosal repair. We next used this model to address the role of transplanted colonic MSCs (cMSCs) in intestinal repair. Compared with intravenously injected cMSCs, mucosally injected cMSCs more effectively prevented the development of penetrating ulcers, as they were more efficiently recruited to colonic wounds. Importantly, mucosally injected cMSCs stimulated angiogenesis in a VEGF-dependent manner. Together, our results reveal that penetrating ulcer formation results from a reduction of local angiogenesis and targeted injection of MSCs can optimize transplantation therapy. Moreover, local MSC injection has potential for treating diseases with features of abnormal angiogenesis and repair.
Collapse
|
75
|
Leclerc JL, Ahmad AS, Singh N, Soshnik-Schierling L, Greene E, Dang A, Doré S. Intracerebral hemorrhage outcomes following selective blockade or stimulation of the PGE2 EP1 receptor. BMC Neurosci 2015; 16:48. [PMID: 26232001 PMCID: PMC4521449 DOI: 10.1186/s12868-015-0182-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 07/13/2015] [Indexed: 01/18/2023] Open
Abstract
Background Inflammation following intracerebral hemorrhage (ICH) significantly contributes to secondary brain damage and poor outcomes. Prostaglandin E2 (PGE2) is known to modulate neuroinflammatory responses and is upregulated in response to brain injury as a result of changes in inducible cyclooxygenase 2 (COX-2) and the membrane-bound type of PGE synthase. Inhibition of COX-2 activity has been reported to attenuate ICH-induced brain injury; however, the clinical utility of such drugs is limited due to the potential for severe side effects. Therefore, it is now important to search for downstream targets capable of preferentially modulating PGE2 signaling, and the four E prostanoid receptors, EP1-4, which are the main targets of PGE2, remain a viable therapeutic option. We have previously shown that EP1 receptor deletion aggravates ICH-induced brain injury and impairs functional recovery, thus the current study aimed to elaborate on these results by including a pharmacologic approach targeting the EP1 receptor. Results Chronic post-treatment with the selective EP1 receptor antagonist, SC-51089, increased lesion volume by 30.1 ± 14.5% (p < 0.05) and treatment with the EP1 agonist, 17-pt-PGE2, improved neuromuscular functional recovery on grip strength (p < 0.01) and hanging wire (p < 0.05) behavioral testing. To begin identifying the mechanisms involved in EP1-mediated neuroprotection after ICH, histology was performed to assess ferric iron content, neuroinflammation, leukocyte transendothelial migratory potential, and peripheral neutrophil and immunoglobulin infiltration. Following ICH, mice treated with the antagonist displayed increased ferric iron (p < 0.05) and cortical microgliosis (p < 0.05), whereas treatment with the agonist decreased cortical (p < 0.01) and striatal (p < 0.001) astrogliosis, leukocyte transendothelial migratory potential (p < 0.01), neutrophil infiltration (p < 0.05), and blood brain barrier breakdown (p < 0.05). Conclusions In agreement with our previous results, selective antagonism of the EP1 receptor aggravated ICH-induced brain injury. Furthermore, EP1 receptor agonism improved anatomical outcomes and functional recovery. Thus, the present data continues to reinforce a putative role for EP1 as a new and more selective therapeutic target for the treatment of ICH that could reduce the side effects associated with COX-2 inhibition while still exploiting the beneficial effects.
Collapse
Affiliation(s)
- Jenna L Leclerc
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA. .,Department of Neuroscience, University of Florida, Gainesville, FL, USA.
| | - Abdullah S Ahmad
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA.
| | - Nilendra Singh
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA.
| | | | - Ellis Greene
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA.
| | - Alex Dang
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA.
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA. .,Department of Neuroscience, University of Florida, Gainesville, FL, USA. .,Departments of Neurology, Psychiatry, Psychology and Pharmaceutics, University of Florida, Gainesville, FL, USA. .,University of Florida College of Medicine, 1275 Center Drive, Gainesville, FL, 32610-0159, USA.
| |
Collapse
|
76
|
Iresjö BM, Wang W, Nilsberth C, Andersson M, Lönnroth C, Smedh U. Food intake, tumor growth, and weight loss in EP2 receptor subtype knockout mice bearing PGE2-producing tumors. Physiol Rep 2015. [PMID: 26197930 PMCID: PMC4552524 DOI: 10.14814/phy2.12441] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Previous studies in our laboratory have demonstrated that prostaglandin (PG) E2 is involved in anorexia/cachexia development in MCG 101 tumor-bearing mice. In the present study, we investigate the role of PGE receptor subtype EP2 in the development of anorexia after MCG 101 implantation in wild-type (EP2+/+) or EP2-receptor knockout (EP2−/−) mice. Our results showed that host absence of EP2 receptors attenuated tumor growth and development of anorexia in tumor-bearing EP2 knockout mice compared to tumor-bearing wild-type animals. Microarray profiling of the hypothalamus revealed a relative twofold change in expression of around 35 genes including mRNA transcripts coding for Phospholipase A2 and Prostaglandin D2 synthase (Ptgds) in EP2 receptor knockout mice compared to wild-type mice. Prostaglandin D2 synthase levels were increased significantly in EP2 receptor knockouts, suggesting that improved food intake may depend on altered balance of prostaglandin production in hypothalamus since PGE2 and PGD2 display opposing effects in feeding control.
Collapse
Affiliation(s)
- Britt-Marie Iresjö
- Surgical Metabolic Research Laboratory, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Wenhua Wang
- Surgical Metabolic Research Laboratory, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Camilla Nilsberth
- Department of Geriatric Medicine and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Marianne Andersson
- Surgical Metabolic Research Laboratory, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christina Lönnroth
- Surgical Metabolic Research Laboratory, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ulrika Smedh
- Surgical Metabolic Research Laboratory, Department of Surgery, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
77
|
Fu Y, Yang MS, Jiang J, Ganesh T, Joe E, Dingledine R. EP2 Receptor Signaling Regulates Microglia Death. Mol Pharmacol 2015; 88:161-70. [PMID: 25715797 PMCID: PMC4468645 DOI: 10.1124/mol.115.098202] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 02/20/2015] [Indexed: 11/22/2022] Open
Abstract
The timely resolution of inflammation prevents continued tissue damage after an initial insult. In the brain, the death of activated microglia by apoptosis has been proposed as one mechanism to resolve brain inflammation. How microglial death is regulated after activation is still unclear. We reported that exposure to lipopolysaccharide (LPS) and interleukin (IL)-13 together initially activates and then kills rat microglia in culture by a mechanism dependent on cyclooxygenase-2 (COX-2). We show here that activation of the E prostanoid receptor 2 (EP2, PTGER2) for prostaglandin E2 mediates microglial death induced by LPS/IL-13, and that EP2 activation by agonist alone kills microglia. Both EP2 antagonists and reactive oxygen scavengers block microglial death induced by either LPS/IL-13 or EP2 activation. By contrast, the homeostatic induction of heme oxygenase 1 (Hmox1) by LPS/IL-13 or EP2 activation protects microglia. Both the Hmox1 inducer cobalt protoporphyrin and a compound that releases the Hmox1 product carbon monoxide (CO) attenuated microglial death produced by LPS/IL-13. Whereas CO reduced COX-2 protein expression, EP2 activation increased Hmox1 and COX-2 expression at both the mRNA and protein level. Interestingly, caspase-1 inhibition prevented microglial death induced by either LPS/IL-13 or low (but not high) concentrations of butaprost, suggestive of a predominantly pyroptotic mode of death. Butaprost also caused the expression of activated caspase-3 in microglia, pointing to apoptosis. These results indicate that EP2 activation, which initially promotes microglial activation, later causes delayed death of activated microglia, potentially contributing to the resolution phase of neuroinflammation.
Collapse
Affiliation(s)
- Yujiao Fu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Myung-Soon Yang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Jianxiong Jiang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Thota Ganesh
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Eunhye Joe
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| | - Raymond Dingledine
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia (Y.F., M.-S.Y., J.J., T.G., R.D.); Neurology Department, Xiangya Hospital, Hunan, China (Y.F.); and Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea (M.-S.Y., E.J.)
| |
Collapse
|
78
|
Guan NN, Svennersten K, de Verdier PJ, Wiklund NP, Gustafsson LE. Receptors involved in the modulation of guinea pig urinary bladder motility by prostaglandin D2. Br J Pharmacol 2015; 172:4024-37. [PMID: 25917171 DOI: 10.1111/bph.13174] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 04/17/2015] [Accepted: 04/22/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE We have described a urothelium-dependent release of PGD2 -like activity which had inhibitory effects on the motility of guinea pig urinary bladder. Here, we have pharmacologically characterized the receptors involved and localized the sites of PGD2 formation and of its receptors. EXPERIMENTAL APPROACH In the presence of selective DP and TP receptor antagonists alone or combined, PGD2 was applied to urothelium-denuded diclofenac-treated urinary bladder strips mounted in organ baths. Antibodies against PGD2 synthase and DP1 receptors were used with Western blots and for histochemistry. KEY RESULTS PGD2 inhibited nerve stimulation -induced contractions in strips of guinea pig urinary bladder with estimated pIC50 of 7.55 ± 0.15 (n = 13), an effect blocked by the DP1 receptor antagonist BW-A868C. After blockade of DP1 receptors, PGD2 enhanced the contractions, an effect abolished by the TP receptor antagonist SQ-29548. Histochemistry revealed strong immunoreactivity for PGD synthase in the urothelium/suburothelium with strongest reaction in the suburothelium. Immunoreactive DP1 receptors were found in the smooth muscle of the bladder wall with a dominant localization to smooth muscle membranes. CONCLUSIONS AND IMPLICATIONS In guinea pig urinary bladder, the main effect of PGD2 is an inhibitory action via DP1 receptors localized to the smooth muscle, but an excitatory effect via TP receptors can also be evoked. The urothelium with its suburothelium might signal to the smooth muscle which is rich in PGD2 receptors of the DP1 type. The results are important for our understanding of regulation of bladder motility.
Collapse
Affiliation(s)
- Na N Guan
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Svennersten
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Petra J de Verdier
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - N Peter Wiklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lars E Gustafsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
79
|
Clapp LH, Gurung R. The mechanistic basis of prostacyclin and its stable analogues in pulmonary arterial hypertension: Role of membrane versus nuclear receptors. Prostaglandins Other Lipid Mediat 2015; 120:56-71. [PMID: 25917921 DOI: 10.1016/j.prostaglandins.2015.04.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/13/2015] [Indexed: 12/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease of distal pulmonary arteries in which patients suffer from elevated pulmonary arterial pressure, extensive vascular remodelling and right ventricular failure. To date prostacyclin (PGI2) therapy remains the most efficacious treatment for PAH and is the only approved monotherapy to have a positive impact on long-term survival. A key thing to note is that improvement exceeds that predicted from vasodilator testing strongly suggesting that additional mechanisms contribute to the therapeutic benefit of prostacyclins in PAH. Given these agents have potent antiproliferative, anti-inflammatory and endothelial regenerating properties suggests therapeutic benefit might result from a slowing, stabilization or even some reversal of vascular remodelling in vivo. This review discusses evidence that the pharmacology of each prostacyclin (IP) receptor agonist so far developed is distinct, with non-IP receptor targets clearly contributing to the therapeutic and side effect profile of PGI2 (EP3), iloprost (EP1), treprostinil (EP2, DP1) along with a family of nuclear receptors known as peroxisome proliferator-activated receptors (PPARs), to which PGI2 and some analogues directly bind. These targets are functionally expressed to varying degrees in arteries, veins, platelets, fibroblasts and inflammatory cells and are likely to be involved in the biological actions of prostacylins. Recently, a highly selective IP agonist, selexipag has been developed for PAH. This agent should prove useful in distinguishing IP from other prostanoid receptors or PPAR binding effects in human tissue. It remains to be determined whether selectivity for the IP receptor gives rise to a superior or inferior clinical benefit in PAH.
Collapse
Affiliation(s)
- Lucie H Clapp
- Department of Medicine, UCL, Rayne Building, London WC1E 6JF, UK.
| | - Rijan Gurung
- Department of Medicine, UCL, Rayne Building, London WC1E 6JF, UK
| |
Collapse
|
80
|
Lu A, Zuo C, He Y, Chen G, Piao L, Zhang J, Xiao B, Shen Y, Tang J, Kong D, Alberti S, Chen D, Zuo S, Zhang Q, Yan S, Fei X, Yuan F, Zhou B, Duan S, Yu Y, Lazarus M, Su Y, Breyer RM, Funk CD, Yu Y. EP3 receptor deficiency attenuates pulmonary hypertension through suppression of Rho/TGF-β1 signaling. J Clin Invest 2015; 125:1228-42. [PMID: 25664856 DOI: 10.1172/jci77656] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 01/05/2015] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is commonly associated with chronic hypoxemia in disorders such as chronic obstructive pulmonary disease (COPD). Prostacyclin analogs are widely used in the management of PAH patients; however, clinical efficacy and long-term tolerability of some prostacyclin analogs may be compromised by concomitant activation of the E-prostanoid 3 (EP3) receptor. Here, we found that EP3 expression is upregulated in pulmonary arterial smooth muscle cells (PASMCs) and human distal pulmonary arteries (PAs) in response to hypoxia. Either pharmacological inhibition of EP3 or Ep3 deletion attenuated both hypoxia and monocrotaline-induced pulmonary hypertension and restrained extracellular matrix accumulation in PAs in rodent models. In a murine PAH model, Ep3 deletion in SMCs, but not endothelial cells, retarded PA medial thickness. Knockdown of EP3α and EP3β, but not EP3γ, isoforms diminished hypoxia-induced TGF-β1 activation. Expression of either EP3α or EP3β in EP3-deficient PASMCs restored TGF-β1 activation in response to hypoxia. EP3α/β activation in PASMCs increased RhoA-dependent membrane type 1 extracellular matrix metalloproteinase (MMP) translocation to the cell surface, subsequently activating pro-MMP-2 and promoting TGF-β1 signaling. Activation or disruption of EP3 did not influence PASMC proliferation. Together, our results indicate that EP3 activation facilitates hypoxia-induced vascular remodeling and pulmonary hypertension in mice and suggest EP3 inhibition as a potential therapeutic strategy for pulmonary hypertension.
Collapse
MESH Headings
- Animals
- Cell Hypoxia
- Cells, Cultured
- Extracellular Matrix/metabolism
- Extracellular Matrix Proteins/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Pulmonary Artery/metabolism
- Rats, Sprague-Dawley
- Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP3 Subtype/genetics
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Signal Transduction
- Sulfonamides/pharmacology
- Transforming Growth Factor beta1/physiology
- Vascular Remodeling
- rho GTP-Binding Proteins/metabolism
- rhoA GTP-Binding Protein
Collapse
|
81
|
Abstract
Prostaglandins, particularly PGE2, are important to adult bone and joint health, but how prostaglandins act on growth plate cartilage to affect bone growth is unclear. We show that growth plate cartilage is distinct from articular cartilage with respect to cyclooxygenase (COX)-2 mRNA expression; although articular chondrocytes express very little COX-2, COX-2 expression is high in growth plate chondrocytes and is increased by IGF-I. In bovine primary growth plate chondrocytes, ATDC5 cells, and human metatarsal explants, inhibition of COX activity with nonsteroidal antiinflammatory drugs (NSAIDs) inhibits chondrocyte proliferation and ERK activation by IGF-I. This inhibition is reversed by prostaglandin E2 and prostacyclin (PGI2) but not by prostaglandin D2 or thromboxane B2. Inhibition of COX activity in young mice by ip injections of NSAIDs causes dwarfism. In growth plate chondrocytes, inhibition of proliferation and ERK activation by NSAIDs is reversed by forskolin, 8-bromoadenosine, 3',5'-cAMP and a prostacyclin analog, iloprost. The inhibition of proliferation and ERK activation by celecoxib is also reversed by 8CPT-2Me-cAMP, an activator of Epac, implicating the small G protein Rap1 in the pathway activated by iloprost. These results imply that prostacyclin is required for proper growth plate development and bone growth.
Collapse
Affiliation(s)
- Michele R Hutchison
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | | |
Collapse
|
82
|
Tang EHC, Cai Y, Wong CK, Rocha VZ, Sukhova GK, Shimizu K, Xuan G, Vanhoutte PM, Libby P, Xu A. Activation of prostaglandin E2-EP4 signaling reduces chemokine production in adipose tissue. J Lipid Res 2014; 56:358-68. [PMID: 25510249 DOI: 10.1194/jlr.m054817] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inflammation of adipose tissue induces metabolic derangements associated with obesity. Thus, determining ways to control or inhibit inflammation in adipose tissue is of clinical interest. The present study tested the hypothesis that in mouse adipose tissue, endogenous prostaglandin E2 (PGE2) negatively regulates inflammation via activation of prostaglandin E receptor 4 (EP4). PGE2 (5-500 nM) attenuated lipopolysaccharide-induced mRNA and protein expression of chemokines, including interferon-γ-inducible protein 10 and macrophage-inflammatory protein-1α in mouse adipose tissue. A selective EP4 antagonist (L161,982) reversed, and two structurally different selective EP4 agonists [CAY10580 and CAY10598] mimicked these actions of PGE2. Adipose tissue derived from EP4-deficient mice did not display this response. These findings establish the involvement of EP4 receptors in this anti-inflammatory response. Experiments performed on adipose tissue from high-fat-fed mice demonstrated EP4-dependent attenuation of chemokine production during diet-induced obesity. The anti-inflammatory actions of EP4 became more important on a high-fat diet, in that EP4 activation suppressed a greater variety of chemokines. Furthermore, adipose tissue and systemic inflammation was enhanced in high-fat-fed EP4-deficient mice compared with wild-type littermates, and in high-fat-fed untreated C57BL/6 mice compared with mice treated with EP4 agonist. These findings provide in vivo evidence that PGE2-EP4 signaling limits inflammation. In conclusion, PGE2, via activation of EP4 receptors, functions as an endogenous anti-inflammatory mediator in mouse adipose tissue, and targeting EP4 may mitigate adipose tissue inflammation.
Collapse
Affiliation(s)
- Eva H C Tang
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong The Research Centre of Heart, Brain, Hormone & Healthy Aging, The University of Hong Kong, Hong Kong
| | - Yin Cai
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
| | - Chi Kin Wong
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong
| | - Viviane Z Rocha
- Lipid Clinic, Heart Institute (InCor), University of Sao Paulo, Sao Paulo - SP, Brazil
| | - Galina K Sukhova
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Koichi Shimizu
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ge Xuan
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Paul M Vanhoutte
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong Department of Clinical Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Aimin Xu
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong Department of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
83
|
von der Emde L, Goltz D, Latz S, Müller SC, Kristiansen G, Ellinger J, Syring I. Prostaglandin receptors EP1-4 as a potential marker for clinical outcome in urothelial bladder cancer. Am J Cancer Res 2014; 4:952-962. [PMID: 25520883 PMCID: PMC4266727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/15/2014] [Indexed: 06/04/2023] Open
Abstract
Prostaglandins, especially prostaglandin E2 (PGE2), and COX-2 play an important role in carcinogenesis of many tumors including bladder cancer (BCA). The PGE2 receptors EP1-4 regulate tumor cell growth, invasion and migration in different tumor entities but EP expression in BCA remains to be determined. In the present study we examined the expression of EP1-4 in non-muscle invasive bladder cancer (NMIBC), muscle invasive bladder cancer (MIBC) and normal urothelial tissue (NU) using immunohistochemistry. Nuclear and cytoplasmic EP1-4 expression was correlated with clinicopathological parameters and survival of BCA patients. EP1, EP2 and EP3 were significantly less expressed in the cytoplasm und nucleus of NMIBC and MIBC than in NU; EP4 cytoplasmic staining in MIBC was significantly higher compared to NU. The cytoplasmic staining was significantly more abundant in MIBC than in NMIBC in all investigated receptors except EP2. The level of EP staining in NMIBC was correlated with staging and grading, especially cytoplasmic EP1. Nuclear staining of EP1 was an independent predictor of BCA recurrence-free survival in NMIBC patients. EP receptors are dysregulated in BCA. The increase of EP1 may be used as prognostic parameter in NMIBC patients and its dysregulation could be targeted by specific EP1 inhibitors.
Collapse
Affiliation(s)
- Laura von der Emde
- Clinic for Urology and Pediatric Urology, University Hospital of BonnGermany
| | - Diane Goltz
- Institute of Pathology, University Hospital of BonnGermany
| | - Stefan Latz
- Clinic for Urology and Pediatric Urology, University Hospital of BonnGermany
| | - Stefan C Müller
- Clinic for Urology and Pediatric Urology, University Hospital of BonnGermany
| | | | - Jörg Ellinger
- Clinic for Urology and Pediatric Urology, University Hospital of BonnGermany
| | - Isabella Syring
- Clinic for Urology and Pediatric Urology, University Hospital of BonnGermany
| |
Collapse
|
84
|
Li X, Melief E, Postupna N, Montine KS, Keene CD, Montine TJ. Prostaglandin E2 receptor subtype 2 regulation of scavenger receptor CD36 modulates microglial Aβ42 phagocytosis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:230-9. [PMID: 25452117 DOI: 10.1016/j.ajpath.2014.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 09/14/2014] [Accepted: 09/18/2014] [Indexed: 12/18/2022]
Abstract
Recent studies underline the potential relevance of microglial innate immune activation in Alzheimer disease. Primary mouse microglia that lack prostaglandin E2 receptor subtype 2 (EP2) show decreased innate immune-mediated neurotoxicity and increased amyloid β (Aβ) peptide phagocytosis, features that were replicated in vivo. Here, we tested the hypothesis that scavenger receptor CD36 is an effector of EP2-regulated Aβ phagocytosis. CD36 expression was 143-fold greater in mouse primary microglia than in primary astrocytes. Three different means of suppressing EP2 signaling increased and an agonist of EP2 decreased CD36 expression in primary wild-type microglia. Activation of Toll-like receptor (TLR) 3, TLR4, and TLR7, but not TLR2 or TLR9, reduced primary microglial CD36 transcription and cell surface CD36 protein and reduced Aβ42 phagocytosis as well. At each step, the effects of innate immune activation on CD36 were reversed by at least 50% by an EP2 antagonist, and this partial rescue of microglia Aβ42 phagocytosis was largely mediated by CD36 activity. Finally, we showed in hippocampus of wild-type mice that innate immune activation suppressed CD36 expression by an EP2-dependent mechanism. Taken together with results of others that found brain clearance of Aβ peptides and behavioral improvements mediated by CD36 in mice, regulation of CD36-mediated Aβ phagocytosis by suppression of EP2 signaling may provide a new approach to suppressing some aspects of Alzheimer disease pathogenesis.
Collapse
Affiliation(s)
- Xianwu Li
- Department of Pathology, University of Washington, Seattle, Washington.
| | - Erica Melief
- Department of Pathology, University of Washington, Seattle, Washington
| | - Nadia Postupna
- Department of Pathology, University of Washington, Seattle, Washington
| | | | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, Washington
| |
Collapse
|
85
|
Nikolopoulou E, Papacleovoulou G, Jean-Alphonse F, Grimaldi G, Parker MG, Hanyaloglu AC, Christian M. Arachidonic acid-dependent gene regulation during preadipocyte differentiation controls adipocyte potential. J Lipid Res 2014; 55:2479-90. [PMID: 25325755 PMCID: PMC4242441 DOI: 10.1194/jlr.m049551] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Arachidonic acid (AA) is a major PUFA that has been implicated in the regulation of adipogenesis. We examined the effect of a short exposure to AA at different stages of 3T3-L1 adipocyte differentiation. AA caused the upregulation of fatty acid binding protein 4 (FABP4/aP2) following 24 h of differentiation. This was mediated by the prostaglandin F2α (PGF2α), as inhibition of cyclooxygenases or PGF2α receptor signaling counteracted the AA-mediated aP2 induction. In addition, calcium, protein kinase C, and ERK are all key elements of the pathway through which AA induces the expression of aP2. We also show that treatment with AA during the first 24 h of differentiation upregulates the expression of the transcription factor Fos-related antigen 1 (Fra-1) via the same pathway. Finally, treatment with AA for 24 h at the beginning of the adipocyte differentiation is sufficient to inhibit the late stages of adipogenesis through a Fra-1-dependent pathway, as Fra-1 knockdown rescued adipogenesis. Our data show that AA is able to program the differentiation potential of preadipocytes by regulating gene expression at the early stages of adipogenesis.
Collapse
Affiliation(s)
- Evanthia Nikolopoulou
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | | | - Frederic Jean-Alphonse
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Giulia Grimaldi
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Malcolm G Parker
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Mark Christian
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
86
|
PGD2 DP1 receptor stimulation following stroke ameliorates cerebral blood flow and outcomes. Neuroscience 2014; 279:260-8. [DOI: 10.1016/j.neuroscience.2014.08.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/18/2014] [Accepted: 08/29/2014] [Indexed: 01/05/2023]
|
87
|
Jain PP, Leber R, Nagaraj C, Leitinger G, Lehofer B, Olschewski H, Olschewski A, Prassl R, Marsh LM. Liposomal nanoparticles encapsulating iloprost exhibit enhanced vasodilation in pulmonary arteries. Int J Nanomedicine 2014; 9:3249-61. [PMID: 25045260 PMCID: PMC4094575 DOI: 10.2147/ijn.s63190] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Prostacyclin analogues are standard therapeutic options for vasoconstrictive diseases, including pulmonary hypertension and Raynaud’s phenomenon. Although effective, these treatment strategies are expensive and have several side effects. To improve drug efficiency, we tested liposomal nanoparticles as carrier systems. In this study, we synthesized liposomal nanoparticles tailored for the prostacyclin analogue iloprost and evaluated their pharmacologic efficacy on mouse intrapulmonary arteries, using a wire myograph. The use of cationic lipids, stearylamine, or 1,2-di-(9Z-octadecenoyl)-3-trimethylammonium-propane (DOTAP) in liposomes promoted iloprost encapsulation to at least 50%. The addition of cholesterol modestly reduced iloprost encapsulation. The liposomal nanoparticle formulations were tested for toxicity and pharmacologic efficacy in vivo and ex vivo, respectively. The liposomes did not affect the viability of human pulmonary artery smooth muscle cells. Compared with an equivalent concentration of free iloprost, four out of the six polymer-coated liposomal formulations exhibited significantly enhanced vasodilation of mouse pulmonary arteries. Iloprost that was encapsulated in liposomes containing the polymer polyethylene glycol exhibited concentration-dependent relaxation of arteries. Strikingly, half the concentration of iloprost in liposomes elicited similar pharmacologic efficacy as nonencapsulated iloprost. Cationic liposomes can encapsulate iloprost with high efficacy and can serve as potential iloprost carriers to improve its therapeutic efficacy.
Collapse
Affiliation(s)
- Pritesh P Jain
- Ludwig Boltzmann Institute for Lung Vascular Research, University of Graz, Graz, Austria
| | - Regina Leber
- Ludwig Boltzmann Institute for Lung Vascular Research, University of Graz, Graz, Austria ; Biophysics Division, Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, University of Graz, Graz, Austria
| | - Gerd Leitinger
- Research Unit Electron Microscopic Techniques, Institute of Cell Biology, Histology, and Embryology, Medical University of Graz, Graz, Austria
| | - Bernhard Lehofer
- Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, University of Graz, Graz, Austria ; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, University of Graz, Graz, Austria ; Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Ruth Prassl
- Ludwig Boltzmann Institute for Lung Vascular Research, University of Graz, Graz, Austria ; Institute of Biophysics, Medical University of Graz, Graz, Austria
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, University of Graz, Graz, Austria
| |
Collapse
|
88
|
Commentary on Kaushik et al.: Prostaglandin D2 is crucial for seizure suppression and postictal sleep. Novel evidence supporting a role for prostanoid receptors in seizure control. Exp Neurol 2014; 257:157-61. [DOI: 10.1016/j.expneurol.2014.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/26/2014] [Accepted: 05/02/2014] [Indexed: 11/22/2022]
|
89
|
Shimizu T, Tanaka K, Nakamura K, Taniuchi K, Yawata T, Higashi Y, Ueba T, Dimitriadis F, Shimizu S, Yokotani K, Saito M. Possible involvement of brain prostaglandin E2 and prostanoid EP3 receptors in prostaglandin E2 glycerol ester-induced activation of central sympathetic outflow in the rat. Neuropharmacology 2014; 82:19-27. [PMID: 24657150 DOI: 10.1016/j.neuropharm.2014.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 03/07/2014] [Accepted: 03/11/2014] [Indexed: 12/21/2022]
|
90
|
Abdel-Salam OME, Mohammed NA, Morsy SMY, Youness ER, Omara EA, Sleem AA. Misoprostol decreases oxidative stress and liver injury in bacterial lipopolysaccharide-induced endotoxemia in mice. COMPARATIVE CLINICAL PATHOLOGY 2014; 23:589-601. [DOI: 10.1007/s00580-012-1656-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2025]
|
91
|
Yang P, Jiang Y, Fischer SM. Prostaglandin E3 metabolism and cancer. Cancer Lett 2014; 348:1-11. [PMID: 24657656 DOI: 10.1016/j.canlet.2014.03.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 03/03/2014] [Accepted: 03/07/2014] [Indexed: 01/13/2023]
Abstract
The anticancer activity of n-3 fatty acids, especially those derived from fish, such as eicosapentaenoic acid (EPA) and docosahexaenoic acid) (DHA), has been studied for centuries. While there is a growing body of evidence that EPA and DHA may influence cancer initiation and development through targeting multiple events of tumor development, the underlying mechanisms responsible for these activities are still not fully understood. A number of studies have suggested that the anticancer activities of EPA and DHA are associated with their effects on eicosanoid metabolism by which they inhibit prostaglandin E2 (PGE2) production. In contrast to DHA, EPA can function as a substrate for cyclooxygenases (COXs) to synthesize unique 3-series prostaglandin compounds, especially PGE3. With advance technology in mass spectrometry, there is renewed interest in studying the role of PGE3 in EPA elicited anti-proliferative activity in various cancers, with some promising results. Here, we summarize the regulation of PGE3 synthesis in cancer cells and its role in EPA elicited anticancer activity. The development of PGE3 and its metabolites as potential biomarkers for future clinical evaluation of EPA and fish oil in cancer care is discussed.
Collapse
Affiliation(s)
- Peiying Yang
- Department of General Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States.
| | - Yan Jiang
- Department of General Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Susan M Fischer
- Department of Molecular Carcinogenesis, The University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
92
|
De Caterina R. Inhibiting thrombosis without causing bleeding: can EP3 blockers fulfil the dream? Cardiovasc Res 2014; 101:335-8. [DOI: 10.1093/cvr/cvu020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
93
|
Kay LJ, Gilbert M, Pullen N, Skerratt S, Farrington J, Seward EP, Peachell PT. Characterization of the EP receptor subtype that mediates the inhibitory effects of prostaglandin E2 on IgE-dependent secretion from human lung mast cells. Clin Exp Allergy 2014; 43:741-51. [PMID: 23786281 DOI: 10.1111/cea.12142] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/16/2013] [Accepted: 04/18/2013] [Indexed: 01/11/2023]
Abstract
BACKGROUND Prostaglandin E2 (PGE2 ) has been shown to inhibit IgE-dependent histamine release from human lung mast cells. This effect of PGE2 is believed to be mediated by EP2 receptors. However, definitive evidence that this is the case has been lacking in the absence of EP2 -selective antagonists. Moreover, recent evidence has suggested that PGE2 activates EP4 receptors to inhibit respiratory cell function. OBJECTIVE The aim of this study was to determine the receptor by which PGE2 inhibits human lung mast cell responses by using recently developed potent and selective EP2 and EP4 receptor antagonists alongside other established EP receptor ligands. METHODS The effects of non-selective (PGE2 , misoprostol), EP2 -selective (ONO-AE1-259, AH13205, butaprost-free acid) and EP4 -selective (L-902,688, TCS251) agonists on IgE-dependent histamine release and cyclic-AMP generation in mast cells were determined. The effects of EP2 -selective (PF-04418948, PF-04852946) and EP4 -selective (CJ-042794, L-161,982) antagonists on PGE2 responses of mast cells were studied. The expression of EP receptor subtypes was determined by RT-PCR. RESULTS Prostaglandin E2 , EP2 agonists and EP4 agonists inhibited IgE-dependent histamine release from mast cells. PGE2 and EP2 agonists, but not EP4 agonists, increased cyclic-AMP levels in mast cells. EP4 -selective antagonists did not affect the PGE2 inhibition of histamine release, whereas EP2 -selective antagonists caused rightward shifts in the PGE2 concentration-response curves. RT-PCR studies indicated that mast cells expressed EP2 and EP4 receptors. CONCLUSIONS AND CLINICAL RELEVANCE Although human lung mast cells may express both EP2 and EP4 receptors, the principal mechanism by which PGE2 inhibits mediator release in mast cells is by activating EP2 receptors.
Collapse
Affiliation(s)
- L J Kay
- Academic Unit of Respiratory Medicine, The Medical School, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | |
Collapse
|
94
|
Baretella O, Chung SK, Barton M, Xu A, Vanhoutte PM. Obesity and heterozygous endothelial overexpression of prepro-endothelin-1 modulate responsiveness of mouse main and segmental renal arteries to vasoconstrictor agents. Life Sci 2014; 118:206-12. [PMID: 24412387 DOI: 10.1016/j.lfs.2013.12.214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/11/2013] [Accepted: 12/27/2013] [Indexed: 11/19/2022]
Abstract
AIMS Levels of the endothelium-derived peptide endothelin-1 (ET-1) are elevated in obese humans, and ET-1 mediated vascular tone is increased. Renal arterial smooth muscle is highly responsive to ET-1. Whether or not endothelium-derived ET-1 affects contractions of the renal artery under normal conditions or in obesity is unknown. The present study was designed to investigate whether or not overexpression of endogenous ET-1 in the endothelium affects the responsiveness of the main and segmental renal arteries differently in obesity. MAIN METHODS Mice with tie-1 promoter-driven endothelium-restricted heterozygous overexpression of preproendothelin-1 were used (TET(het)). Obesity was induced in TET(het) mice and wild-type (WT) littermates by feeding a high fat diet for 30 weeks; lean controls were kept on standard chow. The renal arteries were studied in wire myographs testing contractions (in the presence of l-NAME) to ET-1, serotonin, and U46619. KEY FINDINGS Contractions to ET-1 were comparable between groups in main renal arteries, but augmented in segmental preparations from obese mice. Serotonin-induced responses were enhanced in obese TET(het) mice renal arteries compared to lean controls. Concentration-contraction curves to U46619 were shifted significantly to the left in main renal arteries of obese animals, and the maximal response was significantly increased between lean and obese TET(het) mice. SIGNIFICANCE These results indicate an augmented responsiveness of main renal arteries in obesity particularly to TP receptor activation. When combined with endothelial ET-1 overexpression this effect is even more pronounced, which may help to gain further insights into the mechanisms of hypertension in obesity.
Collapse
Affiliation(s)
- Oliver Baretella
- Department of Pharmacology & Pharmacy, and State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong,China
| | - Sookja K Chung
- Department of Anatomy, The University of Hong Kong, Hong Kong,China; Research Centre of Heart, Brain, Hormone & Healthy Aging, The University of Hong Kong, Hong Kong,China
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, 8057 Zürich, Switzerland
| | - Aimin Xu
- Department of Pharmacology & Pharmacy, and State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong,China; Research Centre of Heart, Brain, Hormone & Healthy Aging, The University of Hong Kong, Hong Kong,China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Paul M Vanhoutte
- Department of Pharmacology & Pharmacy, and State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong,China; Research Centre of Heart, Brain, Hormone & Healthy Aging, The University of Hong Kong, Hong Kong,China.
| |
Collapse
|
95
|
Abstract
In the mammalian kidney, prostaglandins (PGs) are important mediators of physiologic processes, including modulation of vascular tone and salt and water. PGs arise from enzymatic metabolism of free arachidonic acid (AA), which is cleaved from membrane phospholipids by phospholipase A2 activity. The cyclooxygenase (COX) enzyme system is a major pathway for metabolism of AA in the kidney. COX are the enzymes responsible for the initial conversion of AA to PGG2 and subsequently to PGH2, which serves as the precursor for subsequent metabolism by PG and thromboxane synthases. In addition to high levels of expression of the "constitutive" rate-limiting enzyme responsible for prostanoid production, COX-1, the "inducible" isoform of cyclooxygenase, COX-2, is also constitutively expressed in the kidney and is highly regulated in response to alterations in intravascular volume. PGs and thromboxane A2 exert their biological functions predominantly through activation of specific 7-transmembrane G-protein-coupled receptors. COX metabolites have been shown to exert important physiologic functions in maintenance of renal blood flow, mediation of renin release and regulation of sodium excretion. In addition to physiologic regulation of prostanoid production in the kidney, increases in prostanoid production are also seen in a variety of inflammatory renal injuries, and COX metabolites may serve as mediators of inflammatory injury in renal disease.
Collapse
Affiliation(s)
- Raymond C Harris
- George M. O'Brien Kidney and Urologic Diseases Center and Division of Nephrology, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee, USA.
| | | |
Collapse
|
96
|
Tilly P, Charles AL, Ludwig S, Slimani F, Gross S, Meilhac O, Geny B, Stefansson K, Gurney ME, Fabre JE. Blocking the EP3 receptor for PGE2 with DG-041 decreases thrombosis without impairing haemostatic competence. Cardiovasc Res 2013; 101:482-91. [PMID: 24323317 DOI: 10.1093/cvr/cvt276] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Haemostasis interrupts bleeding from disrupted blood vessels by activating platelet aggregation and coagulation. A similar mechanism termed thrombosis generates obstructive thrombi inside diseased arteries. As a consequence of this similarity, current anti-thrombotic agents increase the risk of bleeding. Atherosclerotic plaques produce significant amounts of prostaglandin E2 (PGE2), which activates its receptor EP3 on platelets and aggravates atherothrombosis. We investigated whether blocking EP3 could dissociate atherothrombosis from haemostasis. METHODS AND RESULTS Inhibiting in vivo the receptor EP3 for PGE2 with the blocking agent DG-041 reduced murine thrombosis triggered by local delivery of arachidonic acid or ferric chloride on healthy arteries. Importantly, it also reduced thrombosis triggered by scratching murine atherosclerotic plaques. PGE2 was not produced at the bleeding site after tail clipping. Consistently, blocking EP3 did not alter murine tail, liver, or cerebral haemostasis. Furthermore, blocking EP3 reduced murine pulmonary embolism and intensified platelet inhibition by clopidogrel leaving tail bleeding times unchanged. Human atherosclerotic plaques produced PGE2, which facilitated platelet aggregation in human blood and rescued the function of P2Y12-blocked platelets. Finally, in healthy patients, DG-041 reduced platelet aggregation, but did not significantly alter the cutaneous bleeding time at doses up to eight times the dose that inhibited the facilitating effect of PGE2 on platelets. CONCLUSION In mice, blocking EP3 inhibited atherothrombosis without affecting haemostasis and intensified efficiency of conventional anti-platelet treatment without aggravating the bleeding risk. In patients, blocking EP3 should improve the prevention of cardiovascular diseases, which is currently limited by the risk of bleeding.
Collapse
Affiliation(s)
- Peggy Tilly
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut National de la Santé et de la Recherche Médicale U596, Centre National de la Recherche Scientifique UMR7104, Université Louis Pasteur, 67400 Illkirch, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
A comparative study of PGI2 mimetics used clinically on the vasorelaxation of human pulmonary arteries and veins, role of the DP-receptor. Prostaglandins Other Lipid Mediat 2013; 107:48-55. [DOI: 10.1016/j.prostaglandins.2013.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/18/2013] [Accepted: 07/02/2013] [Indexed: 01/11/2023]
|
98
|
Uchida T, Hazekawa M, Yoshida M, Matsumoto K, Sakai Y. Novel long-acting prostacyclin agonist (ONO-1301) with an angiogenic effect: promoting synthesis of hepatocyte growth factor and increasing cyclic AMP concentration via IP-receptor signaling. J Pharmacol Sci 2013; 123:392-401. [PMID: 24292413 DOI: 10.1254/jphs.13073fp] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The purpose of this study was to evaluate the angiogenic potency of ONO-1301, a novel prostacyclin agonist, using a murine sponge model. Solutions of ONO-1301 or hepatocyte growth factor (HGF), as a positive control, were injected into sponges in the backs of mice, daily for 14 days. Hemoglobin and HGF levels in the sponge were increased for up to 14 days on daily treatment with ONO-1301 while on HGF treatment, they peaked on day 7 and had decreased again by day 14. ONO-1301 also upregulated c-Met expression for 14 days in a dose-dependent manner. When the mice were pretreated with an antibody to HGF or the prostaglandin I (IP)-receptor antagonist CAY10441, the angiogenic effect of ONO-1301 was dramatically reduced. Plasma concentrations of cyclic adenosine monophosphate (cAMP) were increased in a dose-dependent manner by once daily treatment with ONO-1301 for 14 days. This effect was reduced by pretreatment with the IP-receptor antagonist. In conclusion, hemoglobin level was increased by repeated treatment with ONO-1301 for 14 days. It is suggested that ONO-1301 induced angiogenesis by promoting the synthesis of HGF and upregulated c-Met expression, followed by an increase in cAMP concentrations mediated by IP-receptor signaling.
Collapse
Affiliation(s)
- Takahiro Uchida
- Department of Clinical Pharmaceutics, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Japan
| | | | | | | | | |
Collapse
|
99
|
de Luca B, Xavier-Elsas P, Barradas M, Luz RA, Queto T, Jones C, Arruda MA, Cunha TM, Cunha FQ, Gaspar-Elsas MI. Essential roles of PKA, iNOS, CD95/CD95L, and terminal caspases in suppression of eosinopoiesis by PGE2 and other cAMP-elevating agents. ScientificWorldJournal 2013; 2013:208705. [PMID: 24376378 PMCID: PMC3859205 DOI: 10.1155/2013/208705] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 09/12/2013] [Indexed: 02/08/2023] Open
Abstract
Up- and downregulation of eosinopoiesis control pulmonary eosinophilia in human asthma. In mice, eosinopoiesis is suppressed in vitro by prostaglandin E2 (PGE2) and in vivo by diethylcarbamazine, through a proapoptotic mechanism sequentially requiring inducible NO synthase (iNOS) and the ligand for death receptor CD95 (CD95L). We examined the roles of iNOS, cAMP-mediated signaling, caspases, and CD95L/CD95 in suppression of eosinopoiesis by PGE2 and other agents signaling through cAMP. Bone-marrow collected from BALB/c mice, or from iNOS-, CD95-, or CD95L-deficient mutants (and wild-type controls), was cultured with interleukin-5 (IL-5), alone or associated with PGE2, cAMP-inducing/mimetic agents, caspase inhibitor zVAD-fmk, iNOS inhibitor aminoguanidine, or combinations thereof, and eosinopoiesis was evaluated at various times. PGE2, added up to 24 hours of culture, dose-dependently suppressed eosinopoiesis, by inducing apoptosis. This effect was (a) paralleled by induction of iNOS in eosinophils; (b) duplicated by sodium nitroprusside, isoproterenol, and cAMP-inducing/mimetic agents; (c) prevented by protein kinase A inhibition. NO was produced through iNOS by dibutyryl-cAMP-stimulated bone-marrow. Overall, PGE2 and isoproterenol shared a requirement for four effector elements (iNOS, CD95L, CD95, and terminal caspases), which together define a pathway targeted by several soluble up- and downmodulators of eosinopoiesis, including drugs, mediators of inflammation, and cytokines.
Collapse
Affiliation(s)
- Bianca de Luca
- Department of Pediatrics, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, FIOCRUZ, Avenue Rui Barbosa 716, 22250-020 Rio de Janeiro, RJ, Brazil
| | - Pedro Xavier-Elsas
- Department of Immunology, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, CCS, Bloco I, Room I-2-066, 22205-020, Rio de Janeiro, Brazil
| | - Mônica Barradas
- Department of Immunology, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, CCS, Bloco I, Room I-2-066, 22205-020, Rio de Janeiro, Brazil
| | - Ricardo A. Luz
- Department of Immunology, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, CCS, Bloco I, Room I-2-066, 22205-020, Rio de Janeiro, Brazil
| | - Túlio Queto
- Department of Pediatrics, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, FIOCRUZ, Avenue Rui Barbosa 716, 22250-020 Rio de Janeiro, RJ, Brazil
| | - Carla Jones
- Department of Immunology, Instituto de Microbiologia Professor Paulo de Góes, Universidade Federal do Rio de Janeiro, CCS, Bloco I, Room I-2-066, 22205-020, Rio de Janeiro, Brazil
| | - Maria Augusta Arruda
- Farmanguinhos, FIOCRUZ, Avenue Comandante Guaranys No. 447, Jacarepaguá, 22775-903 Rio de Janeiro, RJ, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Faculdade de Medicina da USP, Avenue Bandeirantes 3900, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Faculdade de Medicina da USP, Avenue Bandeirantes 3900, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Maria Ignez Gaspar-Elsas
- Department of Pediatrics, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, FIOCRUZ, Avenue Rui Barbosa 716, 22250-020 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
100
|
Shiraishi N, Nomura T, Tanizaki H, Nakajima S, Narumiya S, Miyachi Y, Tokura Y, Kabashima K. Prostaglandin E2-EP3 axis in fine-tuning excessive skin inflammation by restricting dendritic cell functions. PLoS One 2013; 8:e69599. [PMID: 23922752 PMCID: PMC3726673 DOI: 10.1371/journal.pone.0069599] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 06/11/2013] [Indexed: 11/29/2022] Open
Abstract
Prostaglandin E2 (PGE2) is produced in the skin and is suggested to play a role in the regulation of cutaneous immune homeostasis and responses. However, the multifaceted functions of PGE2 continue to elude our understanding, especially because of the multiplicity of PGE2 receptors—EP1, EP2, EP3, and EP4. While cAMP-elevating EP4 is known to activate the functions of cutaneous dendritic cells (DCs), including Langerhans cells (LCs) and dermal DCs, the role of cAMP-suppressing EP3 in this process remains unknown. Here we demonstrated that an EP3 receptor selective agonist, ONO-AE-248, inhibited chemotaxis and co-stimulatory molecule expressions of DCs in vitro. A suboptimal dose of antigen was sufficient to induce contact hypersensitivity in EP3-deficient mice. Intriguingly, EP3 deficiency did not impair skin inflammation at all when the antigen dose was sufficiently high. EP3 limited the functions of cutaneous DCs only when the antigen dose was low. In contrast to EP4, the observed unappreciated function of EP3 may stabilize the cutaneous DCs to halt the impetuous response to a suboptimal dose of antigen. Taken together, PGE2-EP3 signaling is essential for fine-tuning excessive skin inflammation by restricting DC functions.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Movement/drug effects
- Dendritic Cells/cytology
- Dendritic Cells/drug effects
- Dendritic Cells/metabolism
- Dinoprostone/pharmacology
- Flow Cytometry
- Inflammation/metabolism
- Mice
- Mice, Inbred BALB C
- Receptors, Prostaglandin E, EP1 Subtype/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP3 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Skin/drug effects
- Skin/immunology
Collapse
Affiliation(s)
- Noriko Shiraishi
- Department of Dermatology, University of Environmental and Occupational Health, Kitakyushu, Japan
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideaki Tanizaki
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuh Narumiya
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshiki Miyachi
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshiki Tokura
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- * E-mail:
| |
Collapse
|