Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Redox Biol ; 73: 103186, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38744193

ABSTRACT

Recent studies have highlighted the indispensable role of oxidized lipids in inflammatory responses, cell death, and disease pathogenesis. Consequently, inhibitors targeting oxidized lipids, particularly lipid-derived radicals critical in lipid peroxidation, which are known as radical-trapping antioxidants (RTAs), have been actively pursued. We focused our investigation on nitroxide compounds that have rapid second-order reaction rate constants for reaction with lipid-derived radicals. A novel screening system was developed by employing competitive reactions between library compounds and a newly developed profluorescence nitroxide probe with lipid-derived radicals to identify RTA compounds. A PubMed search of the top hit compounds revealed their wide application as repositioned drugs. Notably, the inhibitory efficacy of methyldopa, selected from these compounds, against retinal damage and bilateral common carotid artery stenosis was confirmed in animal models. These findings underscore the efficacy of our screening system and suggest that it is an effective approach for the discovery of RTA compounds.

2.
Nat Commun ; 15(1): 2195, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38472233

ABSTRACT

Recent evidence indicates ferroptosis is implicated in the pathophysiology of various liver diseases; however, the organ-specific regulation mechanism is poorly understood. Here, we demonstrate 7-dehydrocholesterol reductase (DHCR7), the terminal enzyme of cholesterol biosynthesis, as a regulator of ferroptosis in hepatocytes. Genetic and pharmacological inhibition (with AY9944) of DHCR7 suppress ferroptosis in human hepatocellular carcinoma Huh-7 cells. DHCR7 inhibition increases its substrate, 7-dehydrocholesterol (7-DHC). Furthermore, exogenous 7-DHC supplementation using hydroxypropyl ß-cyclodextrin suppresses ferroptosis. A 7-DHC-derived oxysterol metabolite, 3ß,5α-dihydroxycholest-7-en-6-one (DHCEO), is increased by the ferroptosis-inducer RSL-3 in DHCR7-deficient cells, suggesting that the ferroptosis-suppressive effect of DHCR7 inhibition is associated with the oxidation of 7-DHC. Electron spin resonance analysis reveals that 7-DHC functions as a radical trapping agent, thus protecting cells from ferroptosis. We further show that AY9944 inhibits hepatic ischemia-reperfusion injury, and genetic ablation of Dhcr7 prevents acetaminophen-induced acute liver failure in mice. These findings provide new insights into the regulatory mechanism of liver ferroptosis and suggest a potential therapeutic option for ferroptosis-related liver diseases.


Subject(s)
Ferroptosis , Liver Diseases , Oxidoreductases Acting on CH-CH Group Donors , Mice , Animals , Humans , trans-1,4-Bis(2-chlorobenzaminomethyl)cyclohexane Dihydrochloride , Oxidoreductases Acting on CH-CH Group Donors/metabolism
3.
Biol Pharm Bull ; 47(3): 641-651, 2024.
Article in English | MEDLINE | ID: mdl-38508744

ABSTRACT

Recently, mitochondrial dysfunction has gained attention as a causative factor in the pathogenesis and progression of age-related macular degeneration (AMD). Mitochondrial damage plays a key role in metabolism and disrupts the balance of intracellular metabolic pathways, such as oxidative phosphorylation (OXPHOS) and glycolysis. In this study, we focused on oxidized low-density lipoprotein (ox-LDL), a major constituent of drusen that accumulates in the retina of patients with AMD, and investigated whether it could be a causative factor for metabolic alterations in retinal pigment epithelial (RPE) cells. We found that prolonged exposure to ox-LDL induced changes in fatty acid ß-oxidation (FAO), OXPHOS, and glycolytic activity and increased the mitochondrial reactive oxygen species production in RPE cells. Notably, the effects on metabolic alterations varied with the concentration and duration of ox-LDL treatment. In addition, we addressed the limitations of using ARPE-19 cells for retinal disease research by highlighting their lower barrier function and FAO activity compared to those of induced pluripotent stem cell-derived RPE cells. Our findings can aid in the elucidation of mechanisms underlying the metabolic alterations in AMD.


Subject(s)
Macular Degeneration , Retinal Pigment Epithelium , Humans , Retinal Pigment Epithelium/metabolism , Lipoproteins, LDL/metabolism , Oxidative Stress , Epithelial Cells , Retinal Pigments/metabolism , Retinal Pigments/pharmacology
4.
Biol Pharm Bull ; 47(1): 104-111, 2024.
Article in English | MEDLINE | ID: mdl-38171771

ABSTRACT

White matter lesions induced by chronic cerebral hypoperfusion can cause vascular dementia; however, no appropriate treatments are currently available for these diseases. In this study, we investigated lipid peroxidation, which has recently been pointed out to be associated with cerebrovascular disease and vascular dementia, as a therapeutic target for chronic cerebral hypoperfusion. We used ethoxyquin, a lipid-soluble antioxidant, in a neuronal cell line and mouse model of the disease. The cytoprotective effect of ethoxyquin on glutamate-stimulated HT-22 cells, a mouse hippocampal cell line, was comparable to that of a ferroptosis inhibitor. In addition, the administration of ethoxyquin to bilateral common carotid artery stenosis model mice suppressed white matter lesions, blood-brain barrier disruption, and glial cell activation. Taken together, we propose that the inhibition of lipid peroxidation may be a useful therapeutic approach for chronic cerebrovascular disease and the resulting white matter lesions.


Subject(s)
Brain Ischemia , Carotid Stenosis , Cerebrovascular Disorders , Dementia, Vascular , White Matter , Animals , Mice , Dementia, Vascular/complications , Ethoxyquin/metabolism , Ethoxyquin/pharmacology , Ethoxyquin/therapeutic use , White Matter/metabolism , White Matter/pathology , Brain Ischemia/pathology , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/complications , Cerebrovascular Disorders/metabolism , Disease Models, Animal , Carotid Stenosis/complications , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Mice, Inbred C57BL
5.
Cell Rep ; 40(11): 111323, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36103815

ABSTRACT

Prostaglandin receptors have been implicated in a wide range of functions, including inflammation, immune response, reproduction, and cancer. Our group has previously determined the crystal structure of the active-like EP3 bound to its endogenous agonist, prostaglandin E2. Here, we present the single-particle cryoelectron microscopy (cryo-EM) structure of the human EP3-Gi signaling complex at a resolution of 3.4 Å. The structure reveals the binding mode of Gi to EP3 and the structural changes induced in EP3 by Gi binding. In addition, we compare the structure of the EP3-Gi complex with other subtypes of prostaglandin receptors (EP2 and EP4) bound to Gs that have been previously reported and examine the differences in amino acid composition at the receptor-G protein interface. Mutational analysis reveals that the selectivity of the G protein depends on specific amino acid residues in the second intracellular loop and TM5.


Subject(s)
Dinoprostone , Receptors, Prostaglandin E , Amino Acids , Cryoelectron Microscopy , Dinoprostone/pharmacology , Humans , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism
6.
Yakugaku Zasshi ; 141(4): 473-479, 2021.
Article in Japanese | MEDLINE | ID: mdl-33790113

ABSTRACT

Prostanoids [prostaglandins (PGs) and thromboxanes (TXs)] are a series of bioactive lipid metabolites that function in an autacoid manner via activation of cognate G protein-coupled receptors (GPCRs). The nine subtypes of prostanoid receptors (DP1, DP2, EP1, EP2, EP3, EP4, FP, IP, TP) are involved in a wide range of functions, including inflammation, immune response, reproduction, and homeostasis of the intestinal mucosa and cardiovascular system. Among the prostanoid receptors, the structure of antagonist-bound DP2, which belongs to the chemoattractant receptor family, was previously determined. However, the mechanisms of prostanoid recognition and receptor activation remained elusive. To address this issue, we determined the crystal structures of antagonist-bound EP4 and PGE2-bound EP3. The EP3-PGE2 complex exhibits an active-like conformation, including outward movement of the cytoplasmic end of transmembrane (TM) 6 relative to the cytoplasmic end of TM6 of the EP4 complex. The carboxyl moiety of PGE2 is recognized through three hydrogen bonds formed by highly conserved residues: Y1142.65, T206Extracelluar loop 2 (ECL2), and R3337.40 (superscripts denote Ballesteros-Weinstein numbering). In addition, the ω-chain of PGE2 orients toward TM6, which appears to contribute to receptor activation. The structure reveals important insights into the activation mechanism of prostanoid receptors and provides a molecular basis for the binding modes of endogenous ligands. These findings should facilitate the development of subtype-selective and non-PG-like ligands.


Subject(s)
Receptors, Prostaglandin/chemistry , Receptors, Prostaglandin/metabolism , Crystallography, X-Ray , Dinoprostone/chemistry , Dinoprostone/metabolism , Ligands , Molecular Conformation , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Prostaglandin/physiology , Receptors, Prostaglandin E, EP3 Subtype/chemistry , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/chemistry , Receptors, Prostaglandin E, EP4 Subtype/metabolism
7.
Structure ; 29(3): 252-260.e6, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33264604

ABSTRACT

Prostaglandin E receptor EP4, a class A G protein-coupled receptor (GPCR), is a common drug target in various disorders, such as acute decompensated heart failure and ulcerative colitis. Here, we report the cryoelectron microscopy (cryo-EM) structure of the EP4-heterotrimeric G protein (Gs) complex with the endogenous ligand at a global resolution of 3.3 Å. In this structure, compared with that in the inactive EP4 structure, the sixth transmembrane domain is shifted outward on the intracellular side, although the shift is smaller than that in other class A GPCRs bound to Gs. Instead, the C-terminal helix of Gs is inserted toward TM2 of EP4, and the conserved C-terminal hook structure formsthe extended state. These structural features are formed by the conserved residues in prostanoid receptors (Phe542.39 and Trp3277.51). These findings may be important for the thorough understanding of the G protein-binding mechanism of EP4 and other prostanoid receptors.


Subject(s)
GTP-Binding Proteins/chemistry , Receptors, Prostaglandin E, EP4 Subtype/chemistry , Animals , Binding Sites , Cryoelectron Microscopy , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Molecular Docking Simulation , Protein Binding , Protein Conformation, alpha-Helical , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sf9 Cells , Spodoptera
8.
Nat Chem Biol ; 15(1): 8-10, 2019 01.
Article in English | MEDLINE | ID: mdl-30510192

ABSTRACT

Prostanoids are a series of bioactive lipid metabolites that function in an autacoid manner via activation of cognate G-protein-coupled receptors (GPCRs). Here, we report the crystal structure of human prostaglandin (PG) E receptor subtype EP3 bound to endogenous ligand PGE2 at 2.90 Å resolution. The structure reveals important insights into the activation mechanism of prostanoid receptors and provides a molecular basis for the binding modes of endogenous ligands.


Subject(s)
Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/chemistry , Binding Sites , Crystallography, X-Ray , Dinoprostone/chemistry , Dinoprostone/metabolism , Humans , Models, Molecular , Protein Conformation , Receptors, Prostaglandin E, EP3 Subtype/genetics , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Transforming Growth Factor alpha/metabolism
9.
Nat Chem Biol ; 15(1): 18-26, 2019 01.
Article in English | MEDLINE | ID: mdl-30510193

ABSTRACT

Prostaglandin E receptor EP4, a G-protein-coupled receptor, is involved in disorders such as cancer and autoimmune disease. Here, we report the crystal structure of human EP4 in complex with its antagonist ONO-AE3-208 and an inhibitory antibody at 3.2 Å resolution. The structure reveals that the extracellular surface is occluded by the extracellular loops and that the antagonist lies at the interface with the lipid bilayer, proximal to the highly conserved Arg316 residue in the seventh transmembrane domain. Functional and docking studies demonstrate that the natural agonist PGE2 binds in a similar manner. This structural information also provides insight into the ligand entry pathway from the membrane bilayer to the EP4 binding pocket. Furthermore, the structure reveals that the antibody allosterically affects the ligand binding of EP4. These results should facilitate the design of new therapeutic drugs targeting both orthosteric and allosteric sites in this receptor family.


Subject(s)
Receptors, Prostaglandin E, EP4 Subtype/chemistry , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Binding Sites , Caprylates/chemistry , Caprylates/metabolism , Crystallography, X-Ray , Epoprostenol/analogs & derivatives , Epoprostenol/chemistry , Epoprostenol/metabolism , Humans , Ligands , Lipid Bilayers , Molecular Docking Simulation , Naphthalenes/chemistry , Naphthalenes/metabolism , Phenyl Ethers/chemistry , Phenyl Ethers/metabolism , Phenylbutyrates/chemistry , Phenylbutyrates/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics , Spodoptera/genetics
10.
RSC Adv ; 8(39): 21754-21758, 2018 Jun 13.
Article in English | MEDLINE | ID: mdl-35541723

ABSTRACT

Various 2,3-disubstituted 6,7-thienobenzynes have been efficiently generated from the corresponding o-silylaryl triflate-type precursors by activation with fluoride ions. The method has expanded the scope of synthesizable multisubstituted benzothiophenes, including those with various heteroatom substituents, and can be applied to the synthesis of EP4 antagonist analogs.

11.
J Phys Chem B ; 121(26): 6341-6350, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28644022

ABSTRACT

G protein-coupled receptors (GPCRs), which are indispensable to life and also implicated in a number of diseases, construct important drug targets. For the efficient structure-guided drug design, however, their structural stabilities must be enhanced. An amino-acid mutation is known to possibly lead to the enhancement, but currently available experimental and theoretical methods for identifying stabilizing mutations suffer such drawbacks as the incapability of exploring the whole mutational space with minor effort and the unambiguous physical origin of the enhanced or lowered stability. In general, after the identification is successfully made for a GPCR, the whole procedure must be followed all over again for the identification for another GPCR. Here we report a theoretical strategy by which many different GPCRs can be considered at the same time. The strategy is illustrated for three GPCRs of Class A in the inactive state. We argue that a mutation of the residue at a position of NBW = 3.39 (NBW is the Ballesteros-Weinstein number), a hot-spot residue, leads to substantially higher stability for significantly many GPCRs of Class A in the inactive state. The most stabilizing mutations of the residues with NBW = 3.39 are then identified for two of the three GPCRs, using the improved version of our free-energy function. These identifications are experimentally corroborated, which is followed by the determination of new three-dimensional (3D) structures for the two GPCRs. We expect that on the basis of the strategy, the 3D structures of many GPCRs of Class A can be solved for the first time in succession.


Subject(s)
Models, Molecular , Mutation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Protein Folding , Protein Stability , Quantum Theory , Receptors, G-Protein-Coupled/classification , Thermodynamics
12.
J Immunol ; 192(3): 1130-7, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24342806

ABSTRACT

PGE2 has long been known as a potentiator of acute inflammation, but its mechanisms of action still remain to be defined. In this study, we employed inflammatory swelling induced in mice by arachidonate and PGE2 as models and dissected the role and mechanisms of action of each EP receptor at the molecular level. Arachidonate- or PGE2-induced vascular permeability was significantly reduced in EP3-deficient mice. Intriguingly, the PGE2-induced response was suppressed by histamine H1 antagonist treatment, histidine decarboxylase deficiency, and mast cell deficiency. The impaired PGE2-induced response in mast cell-deficient mice was rescued upon reconstitution with wild-type mast cells but not with EP3-deficient mast cells. Although the number of mast cells, protease activity, and histamine contents in ear tissues in EP3-deficient mice were comparable to those in wild-type mice, the histamine contents in ear tissues were attenuated upon PGE2 treatment in wild-type but not in EP3-deficient mice. Consistently, PGE2-EP3 signaling elicited histamine release in mouse peritoneal and bone marrow-derived mast cells, and it exerted degranulation and IL-6 production in a manner sensitive to pertussis toxin and a PI3K inhibitor and dependent on extracellular Ca(2+) ions. These results demonstrate that PGE2 triggers mast cell activation via an EP3-Gi/o-Ca(2+) influx/PI3K pathway, and this mechanism underlies PGE2-induced vascular permeability and consequent edema formation.


Subject(s)
Dinoprostone/physiology , Edema/physiopathology , Inflammation/physiopathology , Mast Cells/physiology , Receptors, Prostaglandin E, EP3 Subtype/physiology , Animals , Arachidonic Acid/toxicity , Calcium/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Degranulation , Edema/etiology , Histamine Release/drug effects , Inflammation/complications , Interleukin-6/physiology , Mast Cells/drug effects , Mast Cells/enzymology , Mice , Mice, Inbred C57BL , Neutrophil Activation , Peroxidase/analysis , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Receptors, Prostaglandin E, EP3 Subtype/deficiency , Signal Transduction/drug effects , Signal Transduction/physiology , Specific Pathogen-Free Organisms
13.
Biochem Biophys Res Commun ; 438(2): 353-8, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23892039

ABSTRACT

Prostanoids comprising prostaglandins (PGs) and thromboxanes (TXs) have been shown to play physiological and pathological roles in zebrafish. However, the molecular basis of zebrafish prostanoid receptors has not been established. Here, we demonstrate that there exist at least five 'contractile' (Ca(2+)-mobilizing) and one 'inhibitory' (Gi-coupled) prostanoid receptors in zebrafish; five 'contractile' receptors consisting of two PGE2 receptors (EP1a and EP1b), two PGF2α receptors (FP1 and FP2), and one TXA2 receptor TP, and one 'inhibitory' receptor, the PGE2 receptor EP3. [(3)H]PGE2 specifically bound to the membranes of cells expressing zebrafish EP1a, EP1b and EP3 with a Kd of 4.8, 1.8 and 13.6nM, respectively, and [(3)H]PGF2α specifically bound to the membranes of cells expressing zebrafish FP1 and FP2, with a Kd of 6.5 and 1.6nM, respectively. U-46619, a stable agonist for human and mouse TP receptors, significantly increased the specific binding of [(35)S]GTPγS to membranes expressing the zebrafish TP receptor. Upon agonist stimulation, all six receptors showed an increase in intracellular Ca(2+) levels, although the increase was very weak in EP1b, and pertussis toxin abolished only the EP3-mediated response. Zebrafish EP3 receptor also suppressed forskolin-induced cAMP formation in a pertussis toxin-sensitive manner. In association with the low structural conservation with mammalian receptors, most agonists and antagonists specific for mammalian EP1, EP3 and TP failed to work on each corresponding zebrafish receptor. This work provides further insights into the diverse prostanoid actions mediated by their receptors in zebrafish.


Subject(s)
Receptors, Prostaglandin/metabolism , Zebrafish Proteins/metabolism , Zebrafish/physiology , Animals , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Cloning, Molecular , Colforsin/pharmacology , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Muscle Contraction/drug effects , Pertussis Toxin/chemistry , Phylogeny , Receptors, G-Protein-Coupled/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Thromboxane/metabolism , Signal Transduction , Tissue Distribution
14.
Biochem Biophys Res Commun ; 436(4): 685-90, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23770421

ABSTRACT

Prostanoids comprising prostaglandins (PGs) and thromboxanes have been shown to play physiological and pathological roles in zebrafish. However, the molecular basis of zebrafish prostanoid receptors has not been characterized to date. Here, we demonstrate that there exist at least six 'relaxant' (Gs-coupled) prostanoid receptors in zebrafish; one PGI2 receptor IP and five PGE2 receptors comprising two EP2 (EP2a and EP2b), and three EP4 receptors (EP4a, EP4b and EP4c). In contrast, we failed to find a zebrafish PGD2 receptor with any structure and/or character similarities to the mammalian DP1 receptor. [(3)H]iloprost, a stable IP radioligand, specifically bound to the membrane of cells expressing zebrafish IP with a Kd of 42nM, and [(3)H]PGE2 specifically bound to the membranes of cells expressing zebrafish EP2a, EP2b, EP4a, EP4b and EP4c with a Kd of 6.9, 6.0, 1.4, 3.3 and 1.2nM, respectively. Upon agonist stimulation, the 'relaxant' prostanoid receptors showed intracellular cAMP accumulation. The responsiveness of these zebrafish receptors to subtype-specific agonists correlated with their structural conservation to the corresponding receptor in mammals. RT-PCR analysis revealed that the six zebrafish prostanoid receptors show unique tissue distribution patterns; each receptor gene may hence be under unique transcriptional regulation. This work provides further insights into the diverse functions of prostanoids in zebrafish.


Subject(s)
Receptors, Prostaglandin/metabolism , Animals , COS Cells , Chlorocebus aethiops , Gene Expression Profiling , Ligands , Polymerase Chain Reaction , Protein Binding , Signal Transduction , Zebrafish
15.
Nat Immunol ; 14(6): 554-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624557

ABSTRACT

Microenvironment-based alterations in phenotypes of mast cells influence the susceptibility to anaphylaxis, yet the mechanisms underlying proper maturation of mast cells toward an anaphylaxis-sensitive phenotype are incompletely understood. Here we report that PLA2G3, a mammalian homolog of anaphylactic bee venom phospholipase A2, regulates this process. PLA2G3 secreted from mast cells is coupled with fibroblastic lipocalin-type PGD2 synthase (L-PGDS) to provide PGD2, which facilitates mast-cell maturation via PGD2 receptor DP1. Mice lacking PLA2G3, L-PGDS or DP1, mast cell-deficient mice reconstituted with PLA2G3-null or DP1-null mast cells, or mast cells cultured with L-PGDS-ablated fibroblasts exhibited impaired maturation and anaphylaxis of mast cells. Thus, we describe a lipid-driven PLA2G3-L-PGDS-DP1 loop that drives mast cell maturation.


Subject(s)
Group III Phospholipases A2/immunology , Mast Cells/immunology , Paracrine Communication/immunology , Prostaglandin D2/immunology , Receptors, Prostaglandin/immunology , Animals , Blotting, Western , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression Profiling , Group III Phospholipases A2/genetics , Group III Phospholipases A2/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/immunology , Lipocalins/metabolism , Mast Cells/metabolism , Mast Cells/ultrastructure , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Paracrine Communication/genetics , Prostaglandin D2/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Yakugaku Zasshi ; 132(11): 1217-23, 2012.
Article in Japanese | MEDLINE | ID: mdl-23123710

ABSTRACT

Contact dermatitis is an inflammatory skin disease caused by toxic factors that activate the skin innate immunity (irritant contact dermatitis) or by a T cell-mediated hypersensitivity reaction (allergic contact dermatitis). These inflammatory skin diseases are sometimes still not easy to control. Therefore, the development of new effective drugs with fewer side effects is anticipated. In the skin under pathophysiological conditions, multiple prostaglandins are produced and their receptors are expressed in time- and/or cell-dependent manners. However, the precise role of prostaglandins and their receptors in contact dermatitis has not been fully understood. Recently, studies using mice with a disruption of each prostaglandin receptor gene, as well as receptor-selective compounds revealed that prostaglandin receptors have manifold functions, sometimes resulting in opposite outcomes. Here, we review new advances in the roles of prostaglandin receptors in contact hypersensitivity as a cutaneous immune response model, and also discuss the clinical potentials of receptor-selective drugs.


Subject(s)
Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Drug Discovery , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/immunology , Skin/immunology , Animals , Dendritic Cells/immunology , Dermatitis, Contact/drug therapy , Disease Models, Animal , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Keratinocytes/immunology , Molecular Targeted Therapy , Prostaglandins/metabolism , Prostaglandins/physiology , Receptors, Prostaglandin/metabolism , T-Lymphocytes/immunology
17.
J Lipid Res ; 52(8): 1500-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21646392

ABSTRACT

The prostaglandin (PG) receptors EP4 and FP have the potential to exert negative effects on adipogenesis, but the exact contribution of endogenous PG-driven receptor signaling to this process is not fully understood. In this study, we employed an adipocyte differentiation system from mouse embryonic fibroblasts (MEF) and compared the effects of each PG receptor-deficiency on adipocyte differentiation. In wild-type (WT) MEF cells, inhibition of endogenous PG synthesis by indomethacin augmented the differentiation, whereas exogenous PGE2, as well as an FP agonist, reversed the effect of indomethacin. In EP4-deficient cells, basal differentiation was upregulated to the levels in indomethacin-treated WT cells, and indomethacin did not further enhance differentiation. Differentiation in FP-deficient cells was equivalent to WT and was still sensitive to indomethacin. PGE2 or indomethacin treatment of WT MEF cells for the first two days was enough to suppress or enhance transcription of the Pparg2 gene as well as the subsequent differentiation, respectively. Differentiation stimuli induced COX-2 gene and protein expression, as well as PGE2 production, in WT MEF cells. These results suggest that PGE2-EP4 signaling suppresses adipocyte differentiation by affecting Pparg2 expression in an autocrine manner and that FP-mediated inhibition is not directly involved in adipocyte differentiation in the MEF system.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Autocrine Communication , Cell Differentiation/drug effects , Fibroblasts/metabolism , Receptors, Prostaglandin E, EP4 Subtype/deficiency , Receptors, Prostaglandin E/deficiency , Adipocytes/cytology , Adipocytes/drug effects , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Dinoprostone/pharmacology , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Indomethacin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/analysis , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation
18.
Biochimie ; 92(6): 665-75, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20399827

ABSTRACT

To understand the role of prostaglandin (PG) receptor EP2 (Ptger2) signaling in ovulation and fertilization, we investigated time-dependent expression profiles in wild-type (WT) and Ptger2(-/-) cumuli before and after ovulation by using microarrays. We prepared cumulus cells from mice just before and 3, 9 and 14 h after human chorionic gonadotropin injection. Key genes including cAMP-related and epidermal growth factor (EGF) genes, as well as extracellular matrix- (ECM-) related and chemokine genes were up-regulated in WT cumuli at 3 h and 14 h, respectively. Ptger2 deficiency differently affected the expression of many of the key genes at 3 h and 14 h. These results indicate that the gene expression profile of cumulus cells greatly differs before and after ovulation, and in each situation, PGE(2)-EP2 signaling plays a critical role in cAMP-regulated gene expression in the cumulus cells under physiological conditions.


Subject(s)
Cumulus Cells/metabolism , Cyclic AMP/metabolism , Gene Expression Profiling , Ovulation/genetics , Receptors, Prostaglandin E/metabolism , Signal Transduction , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovulation/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype
19.
Biochem Biophys Res Commun ; 389(4): 678-82, 2009 Nov 27.
Article in English | MEDLINE | ID: mdl-19769944

ABSTRACT

We previously demonstrated that prostaglandin EP3 receptor augments EP2-elicited cAMP formation in COS-7 cells in a G(i/o)-insensitive manner. The purpose of our current study was to identify the signaling pathways involved in EP3-induced augmentation of receptor-stimulated cAMP formation. The enhancing effect of EP3 receptor was irrespective of the C-terminal structure of the EP3 isoform. This EP3 action was abolished by treatment with inhibitors for phospholipase C and intracellular Ca(2+)-related signaling molecules such as U73122, staurosporine, 2-APB and SK&F 96365. Indeed, an EP3 agonist stimulated IP(3) formation and intracellular Ca(2+) mobilization, which was blocked by U73122, but not by pertussis toxin. The enhancing effect by EP3 on cAMP formation was mimicked by both a Ca(2+) ionophore and the activation of a typical G(q)-coupled receptor. Moreover, EP3 was exclusively localized to the raft fraction in COS-7 cells and EP3-elicited augmentation of cAMP formation was abolished by cholesterol depletion and introduction of a dominant negative caveolin-1 mutant. These results suggest that EP3 elicits adenylyl cyclase superactivation via G(q)/phospholipase C activation and intracellular Ca(2+) mobilization in a lipid raft microdomain-dependent manner.


Subject(s)
Adenylyl Cyclases/biosynthesis , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Membrane Microdomains/metabolism , Receptors, Prostaglandin E/metabolism , Type C Phospholipases/metabolism , Animals , Boron Compounds/pharmacology , COS Cells , Caveolin 1/metabolism , Chlorocebus aethiops , Enzyme Activation , Estrenes/pharmacology , Humans , Imidazoles/pharmacology , Ionophores/pharmacology , Pyrrolidinones/pharmacology , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype , Staurosporine/pharmacology , Type C Phospholipases/antagonists & inhibitors
20.
Endocrinology ; 150(7): 3345-52, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19342461

ABSTRACT

Cumulus cells surround the oocyte and regulate the production and assembly of the extracellular matrix (ECM) around the cumulus-oocyte complex for its timely interaction with sperm in the oviduct. We recently found that C-C chemokines such as CCL2, CCL7, and CCL9 are produced and stimulate integrin-mediated ECM assembly in the postovulatory cumulus to protect eggs and that prostaglandin E(2)-EP2 signaling in the cumulus cells facilitates fertilization by suppressing this chemokine signaling, which otherwise results in fertilization failure by preventing sperm penetration through the cumulus ECM. However, it remains unknown as to what mechanisms underlie chemokine-induced cumulus ECM assembly. Here we report that inhibition of EP2 signaling or addition of CCL7 augments RhoA activation and induces the surface accumulation of integrin and the contraction of cumulus cells. Enhanced surface accumulation of integrin then stimulates the formation and assembly of fibronectin fibrils as well as induces cumulus ECM resistance to hyaluronidase and sperm penetration. These changes in the cumulus ECM as well as cell contraction are relieved by the addition of Y27632 or blebbistatin. These results suggest that chemokines induce integrin engagement to the ECM and consequent ECM remodeling through the RhoA/Rho kinase/actomyosin pathway, making the cumulus ECM barrier resistant to sperm penetration. Based on these results, we propose that prostaglandin E(2)-EP2 signaling negatively regulates chemokine-induced Rho/ROCK signaling in cumulus cells for successful fertilization.


Subject(s)
Cumulus Cells/physiology , Receptors, Prostaglandin E/physiology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Chemokine CCL7/pharmacology , Chemokines, CC/metabolism , Cumulus Cells/ultrastructure , Extracellular Matrix/metabolism , Female , Fertilization , Humans , Male , Mice , Receptors, Prostaglandin E, EP1 Subtype , Signal Transduction , Sperm-Ovum Interactions/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...