Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2780: 345-359, 2024.
Article in English | MEDLINE | ID: mdl-38987477

ABSTRACT

Chemical protein knockdown technology using proteolysis-targeting chimeras (PROTACs) to hijack the endogenous ubiquitin-proteasome system is a powerful strategy to degrade disease-related proteins. This chapter describes in silico design of a hematopoietic prostaglandin D synthase (H-PGDS) degrader, PROTAC(H-PGDS), using a docking simulation of the ternary complex of H-PGDS/PROTAC/E3 ligase as well as the synthesis of the designed PROTAC(H-PGDS)s and evaluation of their H-PGDS degradation activity.


Subject(s)
Intramolecular Oxidoreductases , Lipocalins , Molecular Docking Simulation , Proteolysis , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/antagonists & inhibitors , Humans , Lipocalins/metabolism , Lipocalins/chemistry , Computer Simulation , Drug Design , Ubiquitin-Protein Ligases/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/chemistry
2.
Int J Mol Sci ; 25(3)2024 Feb 03.
Article in English | MEDLINE | ID: mdl-38339125

ABSTRACT

The leading cause of death for patients with Duchenne muscular dystrophy (DMD), a progressive muscle disease, is heart failure. Prostaglandin (PG) D2, a physiologically active fatty acid, is synthesized from the precursor PGH2 by hematopoietic prostaglandin D synthase (HPGDS). Using a DMD animal model (mdx mice), we previously found that HPGDS expression is increased not only in injured muscle but also in the heart. Moreover, HPGDS inhibitors can slow the progression of muscle injury and cardiomyopathy. However, the location of HPGDS in the heart is still unknown. Thus, this study investigated HPGDS expression in autopsy myocardial samples from DMD patients. We confirmed the presence of fibrosis, a characteristic phenotype of DMD, in the autopsy myocardial sections. Additionally, HPGDS was expressed in mast cells, pericytes, and myeloid cells of the myocardial specimens but not in the myocardium. Compared with the non-DMD group, the DMD group showed increased HPGDS expression in mast cells and pericytes. Our findings confirm the possibility of using HPGDS inhibitor therapy to suppress PGD2 production to treat skeletal muscle disorders and cardiomyopathy. It thus provides significant insights for developing therapeutic drugs for DMD.


Subject(s)
Cardiomyopathies , Intramolecular Oxidoreductases , Lipocalins , Muscular Dystrophy, Duchenne , Animals , Humans , Mice , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Disease Models, Animal , Mast Cells/metabolism , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myocardium/metabolism , Pericytes/metabolism
3.
Bioorg Med Chem ; 28(23): 115791, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33059303

ABSTRACT

GlaxoSmithKline and Astex Pharmaceuticals recently disclosed the discovery of the potent H-PGDS inhibitor GSK2894631A 1a (IC50 = 9.9 nM) as part of a fragment-based drug discovery collaboration with Astex Pharmaceuticals. This molecule exhibited good murine pharmacokinetics, allowing it to be utilized to explore H-PGDS pharmacology in vivo. Yet, with prolonged dosing at higher concentrations, 1a induced CNS toxicity. Looking to attenuate brain penetration in this series, aza-quinolines, were prepared with the intent of increasing polar surface area. Nitrogen substitutions at the 6- and 8-positions of the quinoline were discovered to be tolerated by the enzyme. Subsequent structure activity studies in these aza-quinoline scaffolds led to the identification of 1,8-naphthyridine 1y (IC50 = 9.4 nM) as a potent peripherally restricted H-PGDS inhibitor. Compound 1y is efficacious in four in vivo inflammatory models and exhibits no CNS toxicity.


Subject(s)
Aza Compounds/chemistry , Enzyme Inhibitors/chemistry , Quinolines/chemistry , Animals , Binding Sites , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Drug Stability , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Kinetics , Male , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Rats , Structure-Activity Relationship
4.
Eur J Pharmacol ; 875: 173030, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32084417

ABSTRACT

Nasal obstruction is one of the most bothersome symptoms of allergic rhinitis (AR) affecting sleep-related quality of life in AR patients. Although several treatments were tested to control nasal obstruction, some patients with moderate to severe AR do not respond to current treatments, including the combined administration of different types of anti-allergic medicine. Thus, new options for AR treatment are needed. This study aimed to evaluate the effects of combined treatment with a novel inhibitor of hematopoietic prostaglandin D synthase (HPGDS), TAS-205, and different types of anti-allergic medicine on nasal obstruction in AR. Firstly, we demonstrated that TAS-205 selectively inhibited prostaglandin D2 (PGD2) synthesis in an enzymatic assay in a cell-based assay and in vivo models of AR. Moreover, treatment with TAS-205 alone suppressed eosinophil infiltration into the nasal cavity and late phase nasal obstruction. The combined administration of TAS-205 with montelukast, a cysteinyl leukotriene receptor 1 antagonist, showed significant additive inhibitory effects on eosinophil infiltration and late phase nasal obstruction compared to treatment with each agent alone. In contrast, concomitant treatment with TAS-205 and fexofenadine, a histamine H1 blocker, showed inhibitory effects on late phase and early phase nasal obstruction, although the magnitude of the inhibitory effects upon combined administration was comparable to that of each single treatment. These results suggest that combined treatment with an HPGDS inhibitor and different types of anti-allergic medicine may be a promising strategy to control nasal obstruction in AR patients.


Subject(s)
Anti-Allergic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Lipocalins/antagonists & inhibitors , Morpholines/pharmacology , Nasal Obstruction/drug therapy , Piperidines/pharmacology , Pyrroles/pharmacology , Rhinitis, Allergic/drug therapy , Acetates/pharmacology , Acetates/therapeutic use , Animals , Anti-Allergic Agents/therapeutic use , Cell Line , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination/methods , Enzyme Inhibitors/therapeutic use , Guinea Pigs , Humans , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Male , Morpholines/therapeutic use , Nasal Mucosa/drug effects , Nasal Mucosa/immunology , Nasal Obstruction/immunology , Ovalbumin/administration & dosage , Ovalbumin/immunology , Piperidines/therapeutic use , Prostaglandin D2/metabolism , Pyrroles/therapeutic use , Quality of Life , Quinolines/pharmacology , Quinolines/therapeutic use , Rats , Rhinitis, Allergic/complications , Rhinitis, Allergic/immunology , Sulfides/pharmacology , Sulfides/therapeutic use , Terfenadine/analogs & derivatives , Terfenadine/pharmacology , Terfenadine/therapeutic use
5.
Bioorg Med Chem ; 27(8): 1456-1478, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30858025

ABSTRACT

With the goal of discovering more selective anti-inflammatory drugs, than COX inhibitors, to attenuate prostaglandin signaling, a fragment-based screen of hematopoietic prostaglandin D synthase was performed. The 76 crystallographic hits were sorted into similar groups, with the 3-cyano-quinoline 1a (FP IC50 = 220,000 nM, LE = 0.43) being a potent member of the 6,6-fused heterocyclic cluster. Employing SAR insights gained from structural comparisons of other H-PGDS fragment binding mode clusters, the initial hit 1a was converted into the 70-fold more potent quinoline 1d (IC50 = 3,100 nM, LE = 0.49). A systematic substitution of the amine moiety of 1d, utilizing structural information and array chemistry, with modifications to improve inhibitor stability, resulted in the identification of the 300-fold more active H-PGDS inhibitor tool compound 1bv (IC50 = 9.9 nM, LE = 0.42). This selective inhibitor exhibited good murine pharmacokinetics, dose-dependently attenuated PGD2 production in a mast cell degranulation assay and should be suitable to further explore H-PGDS biology.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Lipocalins/antagonists & inhibitors , Quinolines/chemistry , Quinolines/pharmacology , Animals , Drug Discovery , Enzyme Inhibitors/pharmacokinetics , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/metabolism , Lipocalins/chemistry , Lipocalins/metabolism , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Quinolines/pharmacokinetics
6.
Bioorg Med Chem ; 26(16): 4726-4734, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30121213

ABSTRACT

Hematopoietic prostaglandin D synthase (H-PGDS) is one of the two enzymes that catalyze prostaglandin D2 synthesis and a potential therapeutic target of allergic and inflammatory responses. To reveal key molecular interactions between a high-affinity ligand and H-PGDS, we designed and synthesized a potent new inhibitor (KD: 0.14 nM), determined the crystal structure in complex with human H-PGDS, and quantitatively analyzed the ligand-protein interactions by the fragment molecular orbital calculation method. In the cavity, 10 water molecules were identified, and the interaction energy calculation indicated their stable binding to the surface amino acids in the cavity. Among them, 6 water molecules locating from the deep inner cavity to the peripheral part of the cavity contributed directly to the ligand binding by forming hydrogen bonding interactions. Arg12, Gly13, Gln36, Asp96, Trp104, Lys112 and an essential co-factor glutathione also had strong interactions with the ligand. A strong repulsive interaction between Leu199 and the ligand was canceled out by forming a hydrogen bonding network with the adjacent conserved water molecule. Our quantitative studies including crystal water molecules explained that compounds with an elongated backbone structure to fit from the deep inner cavity to the peripheral part of the cavity would have strong affinity to human H-PGDS.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Water/chemistry , Binding Sites , Crystallography, X-Ray , Drug Design , Humans , Hydrogen Bonding , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/genetics , Ligands , Lipocalins/antagonists & inhibitors , Lipocalins/genetics , Molecular Dynamics Simulation , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Surface Plasmon Resonance , Thermodynamics , Water/metabolism
7.
Front Med (Lausanne) ; 4: 165, 2017.
Article in English | MEDLINE | ID: mdl-29057225

ABSTRACT

There is an absolute requirement for Th2 cells in the pathogenesis of allergen-driven eosinophil-rich type 2 inflammation. Although Th2 cells are generally regarded as a homogeneous population, in the past decade there has been increasing evidence for a minority subpopulation of IL-5+ Th2 cells that have enhanced effector function. This IL-5+ Th2 subpopulation has been termed pathogenic effector Th2 (peTh2), as it exhibits greater effector function and disease association than conventional Th2 cells. peTh2 cells have a different expression profile, differentially express transcription factors, and preferentially use specific signaling pathways. As such, peTh2 cells are a potential target in the treatment of allergic eosinophilic inflammation. This review examines peTh2 cells, both in mouse models and human disease, with an emphasis on their role in the pathogenesis of allergic eosinophilic inflammation.

8.
J Allergy Clin Immunol ; 137(3): 833-43, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26792210

ABSTRACT

BACKGROUND: Prostaglandin (PG) D2 is an early-phase mediator in inflammation, but its action and the roles of the 2 D-type prostanoid receptors (DPs) DP1 and DP2 (also called chemoattractant receptor-homologous molecule expressed on T(H)2 cells) in regulating macrophages have not been elucidated to date. OBJECTIVE: We investigated the role of PGD2 receptors on primary human macrophages, as well as primary murine lung macrophages, and their ability to influence neutrophil action in vitro and in vivo. METHODS: In vitro studies, including migration, Ca(2+) flux, and cytokine secretion, were conducted with primary human monocyte-derived macrophages and neutrophils and freshly isolated murine alveolar and pulmonary interstitial macrophages. In vivo pulmonary inflammation was assessed in male BALB/c mice. RESULTS: Activation of DP1, DP2, or both receptors on human macrophages induced strong intracellular Ca(2+) flux, cytokine release, and migration of macrophages. In a murine model of LPS-induced pulmonary inflammation, activation of each PGD2 receptor resulted in aggravated airway neutrophilia, tissue myeloperoxidase activity, cytokine contents, and decreased lung compliance. Selective depletion of alveolar macrophages abolished the PGD2-enhanced inflammatory response. Activation of PGD2 receptors on human macrophages enhanced the migratory capacity and prolonged the survival of neutrophils in vitro. In human lung tissue specimens both DP1 and DP2 receptors were located on alveolar macrophages along with hematopoietic PGD synthase, the rate-limiting enzyme of PGD2 synthesis. CONCLUSION: For the first time, our results show that PGD2 markedly augments disease activity through its ability to enhance the proinflammatory actions of macrophages and subsequent neutrophil activation.


Subject(s)
Lung/immunology , Lung/metabolism , Macrophages/immunology , Macrophages/metabolism , Neutrophil Infiltration/immunology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Animals , Calcium Signaling , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Endotoxins/adverse effects , Endotoxins/immunology , Gene Expression , Humans , Inflammation Mediators/metabolism , Lung/pathology , Lung Injury/etiology , Lung Injury/metabolism , Lung Injury/pathology , Macrophages/drug effects , Mice , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Prostaglandin D2/pharmacology , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Tumor Necrosis Factor-alpha/biosynthesis
9.
J Allergy Clin Immunol ; 137(3): 907-18.e9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26431580

ABSTRACT

BACKGROUND: IL-5(+) pathogenic effector T(H)2 (peT(H)2) cells are a T(H)2 cell subpopulation with enhanced proinflammatory function that has largely been characterized in murine models of allergic inflammation. OBJECTIVE: We sought to identify phenotype markers for human peT(H)2 cells and characterize their function in patients with allergic eosinophilic inflammatory diseases. METHODS: Patients with eosinophilic gastrointestinal disease (EGID), patients with atopic dermatitis (AD), and nonatopic healthy control (NA) subjects were enrolled. peT(H)2 and conventional T(H)2 (cT(H)2) cell phenotype, function, and cytokine production were analyzed by using flow cytometry. Confirmatory gene expression was measured by using quantitative RT-PCR. Prostaglandin D2 levels were measured with ELISA. Gut T(H)2 cells were obtained by means of esophagogastroduodenoscopy. RESULTS: peT(H)2 cells were identified as chemoattractant receptor-homologous molecule expressed on T(H)2 cells-positive (CRTH2(+)), hematopoietic prostaglandin D synthase-positive CD161(hi) CD4 T cells. peT(H)2 cells expressed significantly greater IL-5 and IL-13 than did hematopoietic prostaglandin D synthase-negative and CD161(-) cT(H)2 cells. peT(H)2 cells were highly correlated with blood eosinophilia (r = 0.78-0.98) and were present in 30- to 40-fold greater numbers in subjects with EGID and those with AD versus NA subjects. Relative to cT(H)2 cells, peT(H)2 cells preferentially expressed receptors for thymic stromal lymphopoietin, IL-25, and IL-33 and demonstrated greater responsiveness to these innate pro-TH2 cytokines. peT(H)2 but not cT(H)2 cells produced prostaglandin D2. In patients with EGID and those with AD, peT(H)2 cells expressed gut- and skin-homing receptors, respectively. There were significantly greater numbers of peT(H)2 cells in gut tissue from patients with EGID versus NA subjects. CONCLUSION: peT(H)2 cells are the primary functional proinflammatory human T(H)2 cell subpopulation underlying allergic eosinophilic inflammation. The unambiguous phenotypic identification of human peT(H)2 cells provides a powerful tool to track these cells in future pathogenesis studies and clinical trials.


Subject(s)
Eosinophils/immunology , Eosinophils/metabolism , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Line , Cytokines/metabolism , Disease Models, Animal , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunity, Innate , Immunologic Memory , Immunophenotyping , Interleukin-5/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Phenotype , Receptors, CCR/metabolism , Receptors, Lymphocyte Homing/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th2 Cells/cytology
10.
J Dermatol Sci ; 71(3): 174-83, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23688403

ABSTRACT

BACKGROUND: Thymic stromal lymphopoietin (TSLP) is produced by epidermal keratinocytes, and it induces Th2-mediated inflammation. TSLP expression is enhanced in lesions with atopic dermatitis, and is a therapeutic target. Antimycotic agents improve the symptoms of atopic dermatitis. OBJECTIVE: The objective of this study was to examine whether antimycotics suppress TSLP expression in human keratinocytes. METHODS: Normal human keratinocytes were incubated with polyinosinic-polycytidylic acid (poly I:C) plus IL-4 in the presence of antimycotics. TSLP expression was analyzed by ELISA and real time PCR. Luciferase assays were performed to analyze NF-κB activity. IκBα degradation was analyzed by Western blot analysis. RESULTS: Poly I:C plus IL-4 increased the secretion and mRNA levels of TSLP, which was suppressed by an NF-κB inhibitor, and also enhanced NF-κB transcriptional activities and induced the degradation of IκBα in keratinocytes. The antimycotics itraconazole, ketoconazole, luliconazole, terbinafine, butenafine, and amorolfine suppressed the secretion and mRNA expression of TSLP, NF-κB activity, and IκBα degradation induced by poly I:C plus IL-4. These suppressive effects were similarly manifested by 15-deoxy-Δ-(12,14)-PGJ2 (15d-PGJ2), a prostaglandin D2 metabolite. Antimycotics increased the release of 15d-PGJ2 from keratinocytes and decreased the release of thromboxane B2, a thromboxane A2 metabolite. Antimycotic-induced suppression of TSLP production and NF-κB activity was counteracted by an inhibitor of lipocalin type-prostaglandin D synthase. CONCLUSIONS: Antimycotics itraconazole, ketoconazole, luliconazole, terbinafine, butenafine, and amorolfine may suppress poly I:C plus IL-4-induced production of TSLP by inhibiting NF-κB via increasing 15d-PGJ2 production in keratinocytes. These antimycotics may block the overexpression of TSLP in lesions with atopic dermatitis.


Subject(s)
Antifungal Agents/pharmacology , Cytokines/biosynthesis , Keratinocytes/drug effects , Keratinocytes/metabolism , Cells, Cultured , Cytokines/genetics , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Dermatitis, Atopic/immunology , Humans , Interleukin-4/pharmacology , NF-kappa B/metabolism , Poly I-C/pharmacology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Prostaglandin D2/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Thymic Stromal Lymphopoietin
11.
ACS Med Chem Lett ; 1(2): 59-63, 2010 May 13.
Article in English | MEDLINE | ID: mdl-24900177

ABSTRACT

Hematopoietic prostaglandin D synthase (HPGDS) is primarly expressed in mast cells, antigen-presenting cells, and Th-2 cells. HPGDS converts PGH2 into PGD2, a mediator thought to play a pivotal role in airway allergy and inflammatory processes. In this letter, we report the discovery of an orally potent and selective inhibitor of HPGDS that reduces the antigen-induced response in allergic sheep.

SELECTION OF CITATIONS
SEARCH DETAIL