Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Neuroinflammation ; 18(1): 304, 2021 Dec 27.
Article in English | MEDLINE | ID: mdl-34961526

ABSTRACT

BACKGROUND: Neuroinflammation is a key pathological component of neurodegenerative disease and is characterized by microglial activation and the secretion of proinflammatory mediators. We previously reported that a surge in prostaglandin D2 (PGD2) production and PGD2-induced microglial activation could provoke neuroinflammation. We also reported that a lipid sensor GPR120 (free fatty acid receptor 4), which is expressed in intestine, could be activated by polyunsaturated fatty acids (PUFA), thereby mediating secretion of glucagon-like peptide-1 (GLP-1). Dysfunction of GPR120 results in obesity in both mice and humans. METHODS: To reveal the relationship between PGD2-microglia-provoked neuroinflammation and intestinal PUFA/GPR120 signaling, we investigated neuroinflammation and neuronal function with gene and protein expression, histological, and behavioral analysis in GPR120 knockout (KO) mice. RESULTS: In the current study, we discovered notable neuroinflammation (increased PGD2 production and microglial activation) and neurodegeneration (declines in neurogenesis, hippocampal volume, and cognitive function) in GPR120 KO mice. We also found that Hematopoietic-prostaglandin D synthase (H-PGDS) was expressed in microglia, microglia were activated by PGD2, H-PGDS expression was upregulated in GPR120 KO hippocampus, and inhibition of PGD2 production attenuated this neuroinflammation. GPR120 KO mice exhibited reduced intestinal, plasma, and intracerebral GLP-1 contents. Peripheral administration of a GLP-1 analogue, liraglutide, reduced PGD2-microglia-provoked neuroinflammation and further neurodegeneration in GPR120 KO mice. CONCLUSIONS: Our results suggest that neurological phenotypes in GPR120 KO mice are probably caused by dysfunction of intestinal GPR120. These observations raise the possibility that intestinal GLP-1 secretion, stimulated by intestinal GPR120, may remotely contributed to suppress PGD2-microglia-provoked neuroinflammation in the hippocampus.


Subject(s)
Hippocampus/pathology , Microglia/pathology , Neurodegenerative Diseases/genetics , Neuroinflammatory Diseases/genetics , Prostaglandin D2/genetics , Receptors, G-Protein-Coupled/genetics , Suppression, Genetic/genetics , Animals , Behavior, Animal , Fatty Acids, Unsaturated/metabolism , Glucagon-Like Peptide 1/metabolism , Liraglutide/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/psychology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/psychology , Prostaglandin D2/biosynthesis
2.
Mol Hum Reprod ; 27(5)2021 05 08.
Article in English | MEDLINE | ID: mdl-33851217

ABSTRACT

Adenomyosis is characterised by epithelial gland and mesenchymal stroma invasion of the uterine myometrium. Adenomyosis is an oestrogen-dependent gynaecological disease in which a number of factors, such as inflammatory molecules, prostaglandins (PGs), angiogenic factors, cell proliferation and extracellular matrix remodelling proteins, also play a role as key disease mediators. In this study, we used mice lacking both lipocalin and hematopoietic-PG D synthase (L- and H-Pgds) genes in which PGD2 is not produced to elucidate PGD2 roles in the uterus. Gene expression studied by real-time PCR and hormone dosages performed by ELISA or liquid chromatography tandem mass spectroscopy in mouse uterus samples showed that components of the PGD2 signalling pathway, both PGDS and PGD2-receptors, are expressed in the mouse endometrium throughout the oestrus cycle with some differences among uterine compartments. We showed that PGE2 production and the steroidogenic pathway are dysregulated in the absence of PGD2. Histological analysis of L/H-Pgds-/- uteri, and immunohistochemistry and immunofluorescence analyses of proliferation (Ki67), endothelial cell (CD31), epithelial cell (pan-cytokeratin), myofibroblast (α-SMA) and mesenchymal cell (vimentin) markers, identify that 6-month-old L/H-Pgds-/- animals developed adenomyotic lesions, and that disease severity increased with age. In conclusion, this study suggests that the PGD2 pathway has major roles in the uterus by protecting the endometrium against adenomyosis development. Additional experiments, using for instance transcriptomic approaches, are necessary to fully determine the molecular mechanisms that lead to adenomyosis in L/H-Pgds-/- mice and to confirm whether this strain is an appropriate model for studying the human disease.


Subject(s)
Adenomyosis/metabolism , Prostaglandin D2/physiology , Signal Transduction , Uterus/metabolism , Animals , Dinoprostone/metabolism , Female , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Mice , Prostaglandin D2/genetics , Prostaglandin D2/metabolism , Real-Time Polymerase Chain Reaction , Steroids/biosynthesis , Uterus/physiology
3.
Clin Exp Nephrol ; 25(5): 445-455, 2021 May.
Article in English | MEDLINE | ID: mdl-33595729

ABSTRACT

BACKGROUND: Lipid-metabolizing enzymes and their metabolites affect inflammation and fibrosis, but their roles in chronic kidney disease (CKD) have not been completely understood. METHODS: To clarify their role in CKD, we measured the mRNA levels of major lipid-metabolizing enzymes in 5/6 nephrectomized (Nx) kidneys of C57BL/6 J mice. Mediator lipidomics was performed to reveal lipid profiles of CKD kidneys. RESULTS: In 5/6 Nx kidneys, both mRNA and protein levels of Alox15 were higher when compared with those in sham kidneys. With respect to in situ hybridization, the mRNA level of Alox15 was higher in renal tubules of 5/6 Nx kidneys. To examine the role of Alox15 in CKD pathogenesis, we performed 5/6 Nx on Alox15-/- mice. Alox15-/- CKD mice exhibited better renal functions than wild-type mice. Interstitial fibrosis was also inhibited in Alox15-/- CKD mice. Mediator lipidomics revealed that Alox15-/- CKD mouse kidneys had significantly higher levels of PGD2 than the control. To investigate the effects of PGD2 on renal fibrosis, we administered PGD2 to TGF-ß1-stimulated NRK-52E cells and HK-2 cells, which lead to a dose-dependent suppression of type I collagen and αSMA in both cell lines. CONCLUSION: Increased PGD2 in Alox15-/- CKD mouse kidneys could inhibit fibrosis, thereby resulting in CKD improvement. Thus, Alox15 inhibition and PGD2 administration may be novel therapeutic targets for CKD.


Subject(s)
Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Kidney/pathology , Lipid Metabolism/genetics , Prostaglandin D2/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Actins/genetics , Actins/metabolism , Animals , Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Cell Line , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Fibrosis , Humans , Intramolecular Oxidoreductases/genetics , Kidney/metabolism , Kidney Tubules, Proximal/metabolism , Lipocalins/genetics , Male , Mice, Inbred C57BL , Nephrectomy , Prostaglandin D2/pharmacology , RNA, Messenger/metabolism , Renal Insufficiency, Chronic/pathology
4.
Article in English | MEDLINE | ID: mdl-32389118

ABSTRACT

BACKGROUND: The vessel restenosis is related to the inflammatory events in subendothelial space. It is proposed that the inflammatory agents affect the prostaglandin synthesis pathway. In this study, we investigated the COX-1, COX-2, PTGDS and miRNA-520a-5p expression levels and the serum 15-Deoxy-Δ12,14-PGJ2 metabolite values in patients with the stenosed and re-stenosed vessels. Furthermore, the associations between genes and miR-520 were evaluated in the monocyte transfection studies. METHODS: The subjects (n=60) were included three groups; healthy subjects (control (stenosis < 5%), stent no restenosis (SNR, restenosis < 5%) and in-stent restenosis (ISR, restenosis > 70%)). The miRNA and gene expression levels were measured by RT-qPCR technique. 15-Deoxy-Δ12,14-PGJ2 values were measured by the ELISA technique. The miR-520 was transfected into myocytes using PEI polymer. RESULTS: The monocyte COX-1, COX-2 and PTGDS gene expression levels and the serum 15-Deoxy- Δ12,14-PGJ2 values increased significantly in the patients. Furthermore, the miR-520 correlated conversely with the COX-1, and PTGDS gene expression levels. CONCLUSION: The results showed that the PGD2 synthesis pathway is active in the patients and, miR- 520 may be involved in the function of this pathway.


Subject(s)
Coronary Restenosis/metabolism , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Intramolecular Oxidoreductases/biosynthesis , MicroRNAs/biosynthesis , Prostaglandin D2/biosynthesis , Aged , Coronary Restenosis/diagnosis , Coronary Restenosis/genetics , Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Female , Gene Expression , Humans , Intramolecular Oxidoreductases/genetics , Male , MicroRNAs/genetics , Middle Aged , Prostaglandin D2/genetics , Signal Transduction/physiology
5.
Blood ; 136(2): 235-246, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32350523

ABSTRACT

Anemic stress induces stress erythropoiesis, which rapidly generates new erythrocytes to restore tissue oxygenation. Stress erythropoiesis is best understood in mice where it is extramedullary and occurs primarily in the spleen. However, both human and mouse stress erythropoiesis use signals and progenitor cells that are distinct from steady-state erythropoiesis. Immature stress erythroid progenitors (SEPs) are derived from short-term hematopoietic stem cells. Although the SEPs are capable of self-renewal, they are erythroid restricted. Inflammation and anemic stress induce the rapid proliferation of SEPs, but they do not differentiate until serum erythropoietin (Epo) levels increase. Here we show that rather than directly regulating SEPs, Epo promotes this transition from proliferation to differentiation by acting on macrophages in the splenic niche. During the proliferative stage, macrophages produce canonical Wnt ligands that promote proliferation and inhibit differentiation. Epo/Stat5-dependent signaling induces the production of bioactive lipid mediators in macrophages. Increased production of prostaglandin J2 (PGJ2) activates peroxisome proliferator-activated receptor γ (PPARγ)-dependent repression of Wnt expression, whereas increased production of prostaglandin E2 (PGE2) promotes the differentiation of SEPs.


Subject(s)
Cell Differentiation , Erythroid Cells/metabolism , Macrophages/metabolism , Receptors, Erythropoietin/metabolism , Signal Transduction , Spleen/metabolism , Stem Cell Niche , Animals , Dinoprostone/genetics , Dinoprostone/metabolism , Erythroid Cells/cytology , Humans , Macrophages/cytology , Mice , Mice, Transgenic , PPAR gamma/genetics , PPAR gamma/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/genetics , Prostaglandin D2/metabolism , Receptors, Erythropoietin/genetics , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Spleen/cytology
6.
Peptides ; 122: 170150, 2019 12.
Article in English | MEDLINE | ID: mdl-31541683

ABSTRACT

Endothelin-1 (ET-1) is a potent stimulus for the secretion of atrial natriuretic peptide (ANP) and hypoxia stimulates the release of ET-1, which is involved in the regulation of atrial ANP secretion. However, the precise mechanism of endogenous ET-1 in the regulation of hypoxia-induced ANP secretion is unclear. Therefore, this study aimed to investigate the mechanism of hypoxia-induced endogenous ET-1 regulation of ANP secretion in isolated perfused hypoxic beating rat atria. The results of this study showed that acute hypoxia significantly stimulated ET-1 release and upregulated the expression of its type A as well as type B receptors (ETA and ETB receptors). Endogenous ET-1 induced by hypoxia markedly upregulated the expression of cyclooxygenase 2 (COX2) through activation of its two receptors, leading to an increase in lipocalin-type prostaglandin D synthase (L-PGDS) expression and prostaglandin D2 (PGD2) production. L-PGDS-derived PGD2 activated peroxisome proliferator-activated receptor γ (PPARγ), ultimately promoting hypoxia-induced ANP secretion. Conversely, L-PGDS-derived PGD2 may in turn regulate L-PGDS expression by a nuclear factor erythroid-2-related factor 2 (NRF2)-mediated feedback mechanism. These results indicate that endogenous ET-1 induced by hypoxia promotes hypoxia-induced ANP secretion by activation of COX2-L-PGDS-PPARγ signaling in beating rat atria. In addition, the positive feedback loop between L-PGDS-derived PGD2 and L-PGDS expression induced by hypoxia is part of the mechanism of hypoxia-induced ANP secretion by endogenous ET-1.


Subject(s)
Atrial Natriuretic Factor/genetics , Endothelin-1/genetics , Heart Atria/metabolism , Hypoxia/genetics , NF-E2-Related Factor 2/genetics , Animals , Cyclooxygenase 2/genetics , Gene Expression Regulation/genetics , Heart Atria/pathology , Intramolecular Oxidoreductases/genetics , Isolated Heart Preparation , Lipocalins/genetics , PPAR gamma/genetics , Prostaglandin D2/genetics , Rats , Signal Transduction
7.
Neuron ; 103(2): 309-322.e7, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31151773

ABSTRACT

Body temperature control is essential for survival. In mammals, thermoregulation is mediated by the preoptic area of anterior hypothalamus (POA), with ∼30% of its neurons sensitive to brain temperature change. It is still unknown whether and how these temperature-sensitive neurons are involved in thermoregulation, because for eight decades they have only been identified via electrophysiological recording. By combining single-cell RNA-seq with whole-cell patch-clamp recordings, we identified Ptgds as a genetic marker for temperature-sensitive POA neurons. Then, we demonstrated these neurons' role in thermoregulation via chemogenetics. Given that Ptgds encodes the enzyme that synthesizes prostaglandin D2 (PGD2), we further explored its role in thermoregulation. Our study revealed that rising temperature of POA alters the activity of Ptgds-expressing neurons so as to increase PGD2 production. PGD2 activates its receptor DP1 and excites downstream neurons in the ventral medial preoptic area (vMPO) that mediates body temperature decrease, a negative feedback loop for thermoregulation.


Subject(s)
Body Temperature Regulation/physiology , Neurons/physiology , Preoptic Area/cytology , Preoptic Area/physiology , Prostaglandin D2/metabolism , Temperature , Action Potentials/drug effects , Action Potentials/physiology , Animals , Body Temperature/drug effects , Body Temperature/physiology , Body Temperature Regulation/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Clozapine/pharmacology , Dinoprostone/genetics , Dinoprostone/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Locomotion/drug effects , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Preoptic Area/drug effects , Prostaglandin D2/genetics
8.
FASEB J ; 33(7): 8202-8210, 2019 07.
Article in English | MEDLINE | ID: mdl-31018708

ABSTRACT

The precise role of prostaglandin D (PGD)2 in allergic lung inflammation remains controversial. Here, we aimed to clarify the role of PGD2 in chronic allergic lung inflammation using hematopoietic PGD synthase (H-PGDS)-deficient mice. Repeated intranasal administration of ovalbumin (OVA) resulted in eosinophilic infiltration and mucin production in the lungs of wild type (WT) mice, leading to respiratory dysfunction. H-PGDS deficiency exacerbated these effects, which were accompanied by increased mRNA expression of TNF-α and eosinophil chemoattractants. The bronchial epithelium expressed both H-PGDS and TNF-α in the inflamed WT lung, and H-PGDS deficiency increased TNF-α expression further. In cultured bronchial tissue of WT mice, treatment with LPS elevated mRNA expression of TNF-α and eosinophil chemoattractants. H-PGDS deficiency promoted the expression of these factors, which was inhibited by treatment with PGD2 receptor, D prostanoid (DP) receptor agonist, or PGD2 metabolite 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2). Treatment with TNF-α receptor antibody inhibited eosinophil chemoattractant expression. In vivo, administration of DP agonist or 15d-PGJ2 inhibited OVA-induced allergic lung inflammation. Bronchial epithelial cell-derived PGD2 attenuated lung eosinophilic infiltration with chronic allergic inflammation; these phenomena are at least partly attributed to the inhibition of TNF-α production via DP activation or 15-deoxy-Δ12,14-PGJ2 signaling.-Maehara, T., Nakamura, T., Maeda, S., Aritake, K., Nakamura, M., Murata, T. Epithelial cell-derived prostaglandin D2 inhibits chronic allergic lung inflammation in mice.


Subject(s)
Asthma/metabolism , Epithelial Cells/metabolism , Lung/metabolism , Pneumonia/metabolism , Prostaglandin D2/metabolism , Signal Transduction , Animals , Asthma/chemically induced , Asthma/genetics , Chronic Disease , Epithelial Cells/pathology , Gene Expression Regulation , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipopolysaccharides/toxicity , Lung/pathology , Mice , Mice, Knockout , Pneumonia/chemically induced , Pneumonia/genetics , Prostaglandin D2/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Sleep ; 42(6)2019 06 11.
Article in English | MEDLINE | ID: mdl-30893431

ABSTRACT

To meet the new challenges of modern lifestyles, we often compromise a good night's sleep. In preclinical models as well as in humans, a chronic lack of sleep is reported to be among the leading causes of various physiologic, psychologic, and neurocognitive deficits. Thus far, various endogenous mediators have been implicated in inter-regulatory networks that collectively influence the sleep-wake cycle. One such mediator is the lipocalin-type prostaglandin D2 synthase (L-PGDS)-Prostaglandin D2 (PGD2)-DP1 receptor (L-PGDS-PGD2-DP1R) axis. Findings in preclinical models confirm that DP1R are predominantly expressed in the sleep-regulating centers. This finding led to the discovery that the L-PGDS-PGD2-DP1R axis is involved in sleep regulation. Furthermore, we showed that the L-PGDS-PGD2-DP1R axis is beneficial in protecting the brain from ischemic stroke. Protein sequence homology was also performed, and it was found that L-PGDS and DP1R share a high degree of homology between humans and rodents. Based on the preclinical and clinical data thus far pertaining to the role of the L-PGDS-PGD2-DP1R axis in sleep regulation and neurologic conditions, there is optimism that this axis may have a high translational potential in human therapeutics. Therefore, here the focus is to review the regulation of the homeostatic component of the sleep process with a special focus on the L-PGDS-PGD2-DP1R axis and the consequences of sleep deprivation on health outcomes. Furthermore, we discuss whether the pharmacological regulation of this axis could represent a tool to prevent sleep disturbances and potentially improve outcomes, especially in patients with acute brain injuries.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Prostaglandin D2/metabolism , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Sleep/physiology , Amino Acid Sequence , Animals , Brain/metabolism , Humans , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Male , Prostaglandin D2/genetics , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Stroke/prevention & control
10.
Sci Rep ; 9(1): 1931, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760783

ABSTRACT

Lipocalin-type prostaglandin (PG) D synthase (L-PGDS) is responsible for the production of PGD2 in adipocytes and is selectively induced by a high-fat diet (HFD) in adipose tissue. In this study, we investigated the effects of HFD on obesity and insulin resistance in two distinct types of adipose-specific L-PGDS gene knockout (KO) mice: fatty acid binding protein 4 (fabp4, aP2)-Cre/L-PGDS flox/flox and adiponectin (AdipoQ)-Cre/L-PGDS flox/flox mice. The L-PGDS gene was deleted in adipocytes in the premature stage of the former strain and after maturation of the latter strain. The L-PGDS expression and PGD2 production levels decreased in white adipose tissue (WAT) under HFD conditions only in the aP2-Cre/L-PGDS flox/flox mice, but were unchanged in the AdipoQ-Cre/L-PGDS flox/flox mice. When fed an HFD, aP2-Cre/L-PGDS flox/flox mice significantly reduced body weight gain, adipocyte size, and serum cholesterol and triglyceride levels. In WAT of the HFD-fed aP2-Cre/L-PGDS flox/flox mice, the expression levels of the adipogenic, lipogenic, and M1 macrophage marker genes were decreased, whereas those of the lipolytic and M2 macrophage marker genes were enhanced or unchanged. Insulin sensitivity was improved in the HFD-fed aP2-Cre/L-PGDS flox/flox mice. These results indicate that PGD2 produced by L-PGDS in premature adipocytes is involved in the regulation of body weight gain and insulin resistance under nutrient-dense conditions.


Subject(s)
Adipocytes/metabolism , Insulin Resistance , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Obesity/metabolism , Prostaglandin D2/biosynthesis , Adipocytes/pathology , Animals , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Mice , Mice, Transgenic , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Prostaglandin D2/genetics
11.
J Pharmacol Exp Ther ; 367(3): 425-432, 2018 12.
Article in English | MEDLINE | ID: mdl-30305427

ABSTRACT

Prostaglandin (PG) D2 is formed by two distinct PGD synthases (PGDS): lipocalin-type PGDS (L-PGDS), which acts as a PGD2-producing enzyme and as extracellular lipophilic transporter, and hematopoietic PGDS (H-PGDS), a σ glutathione-S-transferase. PGD2 plays an important role in the maintenance of vascular function; however, the relative contribution of L-PGDS- and H-PGDS-dependent formation of PGD2 in this setting is unknown. To gain insight into the function played by these distinct PGDS, we assessed systemic blood pressure (BP) and thrombogenesis in L-Pgds and H-Pgds knockout (KO) mice. Deletion of L-Pgds depresses urinary PGD2 metabolite (PGDM) by ∼35%, whereas deletion of H-Pgds does so by ∼90%. Deletion of L-Pgds, but not H-Pgds, elevates BP and accelerates the thrombogenic occlusive response to a photochemical injury to the carotid artery. HQL-79, a H-PGDS inhibitor, further depresses PGDM in L-Pgds KO mice, but has no effect on BP or on the thrombogenic response. Gene expression profiling reveals that pathways relevant to vascular function are dysregulated in the aorta of L-Pgds KOs. These results indicate that the functional impact of L-Pgds deletion on vascular homeostasis may result from an autocrine effect of L-PGDS-dependent PGD2 on the vasculature and/or the L-PGDS function as lipophilic carrier protein.


Subject(s)
Hypertension/genetics , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Prostaglandin D2/genetics , Sequence Deletion/genetics , Animals , Carotid Arteries/pathology , Glutathione Transferase/genetics , Male , Mice , Mice, Knockout
12.
Mol Cell ; 72(1): 48-59.e4, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30220562

ABSTRACT

The signaling of prostaglandin D2 (PGD2) through G-protein-coupled receptor (GPCR) CRTH2 is a major pathway in type 2 inflammation. Compelling evidence suggests the therapeutic benefits of blocking CRTH2 signaling in many inflammatory disorders. Currently, a number of CRTH2 antagonists are under clinical investigation, and one compound, fevipiprant, has advanced to phase 3 clinical trials for asthma. Here, we present the crystal structures of human CRTH2 with two antagonists, fevipiprant and CAY10471. The structures, together with docking and ligand-binding data, reveal a semi-occluded pocket covered by a well-structured amino terminus and different binding modes of chemically diverse CRTH2 antagonists. Structural analysis suggests a ligand entry port and a binding process that is facilitated by opposite charge attraction for PGD2, which differs significantly from the binding pose and binding environment of lysophospholipids and endocannabinoids, revealing a new mechanism for lipid recognition by GPCRs.


Subject(s)
Prostaglandin D2/chemistry , Receptors, G-Protein-Coupled/chemistry , Receptors, Immunologic/chemistry , Receptors, Prostaglandin/chemistry , Carbazoles/chemistry , Humans , Indoleacetic Acids/chemistry , Ligands , Molecular Docking Simulation , Prostaglandin D2/genetics , Protein Binding , Pyridines/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/genetics , Signal Transduction , Sulfonamides/chemistry
13.
Stem Cells ; 36(7): 990-1003, 2018 07.
Article in English | MEDLINE | ID: mdl-29604141

ABSTRACT

The antitumor effect of prostaglandin D2 (PGD2) on gastric cancer (GC) has been known for decades. However, the mechanism of PGD2's control of GC growth is unclear. Cancer stem cells (CSCs) are implicated in tumor neovascularization, invasiveness, and therapeutic resistance. Herein, we discovered that signaling between PGD2 and its receptor (PTGDR2) has the ability to restrict the self-renewal of GC cells in vitro and suppress tumor growth and metastasis in vivo. To obtain these findings, we first determined that PGD2 synthase (L-PTGDS) and PTGDR2 expression were lower in GC tissues than adjacent tissues and was associated with the patients' prognosis. Moreover, the expression of L-PTGDS and PTGDR2 was negatively correlated with the GC-CSC markers Sall4 and Lgr5 in GC tissues. Second, L-PTGDS and PTGDR2 expression were knocked down in CSC-like cells, resulting in enhanced expression of CSC markers and self-renewal ability. Direct PGD2 stimulation and L-PTGDS overexpression produced the opposite effect. Thirdly, PGD2 inhibited tumor growth and incidence rate in a subcutaneous tumor model and suppressed liver and mesenteric metastasis in a peritoneal metastasis model. Interfering with the expression of PTGDR2 reversed these effects in vivo. Last, a mechanistic study found that PGD2 inhibited STAT3 phosphorylation and nuclear expression. Further experiments revealed that the inhibitory effect of PGD2 on the expression of CSC markers disappeared after mutations were introduced into STAT3 phosphorylation (Thr705) site. In short, this study reveals a novel function of PGD2/PTGDR2 signaling on CSC regulation and provides a new way to control the development of GC. Stem Cells 2018;36:990-1003.


Subject(s)
Carcinogenesis/genetics , Immunohistochemistry/methods , Neoplastic Stem Cells/metabolism , Prostaglandin D2/genetics , Stomach Neoplasms/genetics , Animals , Carcinogenesis/metabolism , Humans , Mice , Signal Transduction , Stomach Neoplasms/pathology , Transfection
14.
J Allergy Clin Immunol ; 138(4): 1089-1097.e3, 2016 10.
Article in English | MEDLINE | ID: mdl-27423494

ABSTRACT

BACKGROUND: Aspirin-exacerbated respiratory disease (AERD) differs from aspirin-tolerant disease in part because of eosinophilic tissue infiltration and overexpression of arachidonic acid metabolic pathway components that lead to enhanced secretion of cysteinyl leukotrienes and prostaglandin (PG) D2 observed constitutively and paradoxically in response to aspirin and other COX inhibitors. We have previously demonstrated the capacity of IFN-γ to drive cysteinyl leukotriene expression and response. OBJECTIVE: We investigated eosinophils as a source of PGD2 production in patients with AERD. METHODS: Eosinophils were enriched from tissue and peripheral blood obtained from control subjects, patients with aspirin-tolerant disease, and patients with AERD. mRNA was extracted and evaluated for expression of hematopoietic prostaglandin D synthase (hPGDS). Expression of hPGDS protein was confirmed with Western hybridization and immunofluorescence staining. Cells were stimulated with aspirin, and secretion of PGD2 was quantified. CD34+ progenitor cells were isolated and matured into eosinophils in the presence or absence of IFN-γ and hPGDS mRNA, and PGD2 release was measured. RESULTS: Gene expression analysis revealed that eosinophils from tissue and blood of patients with AERD display increased levels of hPGDS compared with asthmatic and control samples. Western hybridization confirmed the increase in hPGDS mRNA translated to increased protein expression. Immunofluorescence confirmed mast cells and eosinophils from tissue of patients with AERD and asthma demonstrated hPGDS expression, with higher levels in eosinophils from patients with AERD. Incubation of eosinophils from blood and tissue with aspirin stimulated PGD2 release. IFN-γ-matured eosinophil progenitors showed enhanced hPGDS expression and increased levels of PGD2 release at baseline and after aspirin stimulation. CONCLUSIONS: In addition to mast cells, eosinophils represent an important source of PGD2 in patients with AERD and identify a new target for therapeutic intervention.


Subject(s)
Asthma, Aspirin-Induced , Eosinophils/immunology , Prostaglandin D2/immunology , Aspirin/pharmacology , Cells, Cultured , Cyclooxygenase Inhibitors/pharmacology , Eosinophils/cytology , Eosinophils/drug effects , Gene Expression Regulation/drug effects , Humans , Paranasal Sinuses/cytology , Paranasal Sinuses/immunology , Prostaglandin D2/genetics
15.
Sci Rep ; 6: 27615, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27271931

ABSTRACT

Eosinophils play a major pathologic role in the pathogenesis of diverse inflammatory diseases including chronic rhinosinusitis (CRS). Dysregulated production of prostaglandin (PG), particularly PGD2, is considered to be an important contributing factor to eosinophilic inflammation in CRS primarily through proinflammatory and chemotactic effects on eosinophils. Here, we provide evidence that PGD2 can promote eosinophilic inflammation through a suppression of Natural killer (NK) cell effector function and NK cell-mediated eosinophil regulation. Eosinophil apoptosis mediated by NK cells was significantly decreased in CRS patients compared with healthy controls. This decrease was associated with NK cell dysfunction and eosinophilic inflammation. Tissue eosinophils were positively correlated with blood eosinophils in CRS patients. In a murine model of CRS, NK cell depletion caused an exacerbation of blood eosinophilia and eosinophilic inflammation in the sinonasal tissue. PGD2 and its metabolite, but not PGE2 and a panel of cytokines including TGF-ß, were increased in CRS patients compared with controls. Effector functions of NK cells were potently suppressed by PGD2-dependent, rather than PGE2-dependent, pathway in controls and CRS patients. Thus, our results suggest decreased NK cell-mediated eosinophil regulation, possibly through an increased level of PGD2, as a previously unrecognized link between PG dysregulation and eosinophilic inflammation in CRS.


Subject(s)
Eosinophils/metabolism , Inflammation/genetics , Prostaglandin D2/genetics , Sinusitis/genetics , Adult , Aged , Animals , Chronic Disease , Eosinophils/pathology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Killer Cells, Natural/metabolism , Leukocyte Count , Male , Middle Aged , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Sinusitis/metabolism , Sinusitis/pathology
16.
Sci Rep ; 6: 20992, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26875548

ABSTRACT

Apoptotic cell clearance results in the release of growth factors and the action of signaling molecules involved in tissue homeostasis maintenance. Here, we investigated whether and how macrophages programmed by apoptotic cells inhibit the TGF-ß1-induced Epithelial-mesenchymal transition (EMT) process in lung alveolar epithelial cells. Treatment with conditioned medium derived from macrophages exposed to apoptotic cells, but not viable or necrotic cells, inhibited TGF-ß1-induced EMT, including loss of E-cadherin, synthesis of N-cadherin and α-smooth muscle actin, and induction of EMT-activating transcription factors, such as Snail1/2, Zeb1/2, and Twist1. Exposure of macrophages to cyclooxygenase (COX-2) inhibitors (NS-398 and COX-2 siRNA) or RhoA/Rho kinase inhibitors (Y-27632 and RhoA siRNA) and LA-4 cells to antagonists of prostaglandin E2 (PGE2) receptor (EP4 [AH-23848]), PGD2 receptors (DP1 [BW-A868C] and DP2 [BAY-u3405]), or the hepatocyte growth factor (HGF) receptor c-Met (PHA-665752), reversed EMT inhibition by the conditioned medium. Additionally, we found that apoptotic cell instillation inhibited bleomycin-mediated EMT in primary mouse alveolar type II epithelial cells in vivo. Our data suggest a new model for epithelial cell homeostasis, by which the anti-EMT programming of macrophages by apoptotic cells may control the progressive fibrotic reaction via the production of potent paracrine EMT inhibitors.


Subject(s)
Alveolar Epithelial Cells/drug effects , Apoptosis/genetics , Dinoprostone/biosynthesis , Hepatocyte Growth Factor/biosynthesis , Prostaglandin D2/biosynthesis , Alveolar Epithelial Cells/metabolism , Amides/administration & dosage , Animals , Apoptosis/drug effects , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/administration & dosage , Dinoprostone/antagonists & inhibitors , Dinoprostone/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation/drug effects , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/genetics , Macrophages/metabolism , Mice , Nitrobenzenes/administration & dosage , Prostaglandin D2/antagonists & inhibitors , Prostaglandin D2/genetics , Pyridines/administration & dosage , Sulfonamides/administration & dosage , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
17.
BMC Gastroenterol ; 15: 112, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26338799

ABSTRACT

BACKGROUND: Recent works provide evidence of the importance of the prostaglandin D2 (PGD2) metabolic pathway in inflammatory bowel diseases. We investigated the expression of PGD2 metabolic pathway actors in Crohn's disease (CD) and the ability of the enteric nervous system (ENS) to produce PGD2 in inflammatory conditions. METHODS: Expression of key actors involved in the PGD2 metabolic pathway and its receptors was analyzed using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) in colonic mucosal biopsies of patients from three groups: controls, quiescent and active CD patients. To determine the ability of the ENS to secrete PGD2 in proinflammatory conditions, Lipocalin-type prostaglandin D synthase (L-PGDS) expression by neurons and glial cells was analyzed by immunostaining. PGD2 levels were determined in a medium of primary culture of ENS and neuro-glial coculture model treated by lipopolysaccharide (LPS). RESULTS: In patients with active CD, inflamed colonic mucosa showed significantly higher COX2 and L-PGDS mRNA expression, and significantly higher PGD2 levels than healthy colonic mucosa. On the contrary, peroxysome proliferator-activated receptor Gamma (PPARG) expression was reduced in inflamed colonic mucosa of CD patients with active disease. Immunostaining showed that L-PGDS was expressed in the neurons of human myenteric and submucosal plexi. A rat ENS primary culture model confirmed this expression. PGD2 levels were significantly increased on primary culture of ENS treated with LPS. This production was abolished by AT-56, a specific competitive L-PGDS inhibitor. The neuro-glial coculture model revealed that each component of the ENS, ECG and neurons, could contribute to PGD2 production. CONCLUSIONS: Our results highlight the activation of the PGD2 metabolic pathway in Crohn's disease. This study supports the hypothesis that in Crohn's disease, enteric neurons and glial cells form a functional unit reacting to inflammation by producing PGD2.


Subject(s)
Crohn Disease/metabolism , Intramolecular Oxidoreductases/metabolism , Lipocalins/metabolism , Myenteric Plexus/metabolism , Neuroglia/metabolism , Neurons/metabolism , Prostaglandin D2/metabolism , Submucous Plexus/metabolism , Adolescent , Adult , Aged , Animals , Cells, Cultured , Coculture Techniques , Crohn Disease/pathology , Cyclooxygenase 2/genetics , Cytokines/genetics , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Intramolecular Oxidoreductases/genetics , Lipocalins/genetics , Male , Middle Aged , PPAR gamma/metabolism , Prostaglandin D2/genetics , RNA, Messenger/metabolism , Rats , Severity of Illness Index , Young Adult
18.
Biol Reprod ; 91(2): 33, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24920039

ABSTRACT

The sex differentiation mechanisms in zebrafish (Danio rerio) remains elusive, partly because of the absence of sex chromosomes but also because the process appears to depend on the synchrony of multiple genes and possibly environmental factors. Zebrafish gonadal development is initiated through the development of immature oocytes. Depending on multiple signaling cues, in about half of the individuals, the juvenile ovaries degenerate or undergo apoptosis to initiate testes development while the other half maintains the oogenic pathway. We have previously shown that activation of NFκB and prostaglandin synthase 2 (ptgs2) results in female-biased sex ratios. Prostaglandin synthase and prostaglandins are involved in multiple physiological functions, including cell survival and apoptosis. In the present study, we show that inhibition of ptgs2 by meloxicam results in male-biased sex ratios. On further evaluation, we observed that exposure with the prostaglandin D2 (PGD2) analogue BW-245C induced SRY-box containing gene 9a (sox9a) and resulted in male-biased sex ratios. On the other hand, prostaglandin E2 (PGE2) treatment resulted in female-biased sex ratios and involved activation of NFκB and the ß-catenin pathway as well as inhibition of sox9. Exposure to the ß-catenin inhibitor PNU-74654 resulted in up-regulation of ptgds and male-biased sex ratios, further confirming the involvement of ß-catenin in the female differentiation pathway. In this study, we show that PGD2 and PGE2 can program the gonads to either the testis or the ovary differentiation pathways, indicating that prostaglandins are involved in the regulation of zebrafish gonadal differentiation.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Ovary/physiology , Sex Determination Processes/physiology , Testis/growth & development , Zebrafish/growth & development , Animals , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/genetics , Dinoprostone/metabolism , Embryo, Nonmammalian/enzymology , Female , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic , Intramolecular Oxidoreductases/genetics , Male , Meloxicam , Ovary/enzymology , Prostaglandin D2/genetics , Prostaglandin D2/metabolism , Prostaglandin-E Synthases , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sex Ratio , Signal Transduction/physiology , Testis/enzymology , Thiazines/pharmacology , Thiazoles/pharmacology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , beta Catenin/metabolism
19.
Cancer Res ; 74(11): 3011-9, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24879565

ABSTRACT

Compared with prostaglandin E2, which has an established role in cancer, the role of the COX metabolite prostaglandin D2 (PGD2) in chronic inflammation leading to tumorigenesis is uncertain. In this study, we investigated the role of PGD2 in colitis and colitis-associated colon cancer (CAC) using genetically modified mice and an established model of inflammatory colon carcinogenesis. Systemic genetic deficiency in hematopoietic PGD synthase (H-PGDS) aggravated colitis and accelerated tumor formation in a manner associated with increased TNFα expression. Treatment with a TNFα receptor antagonist attenuated colitis regardless of genotype. Histologic analysis revealed that infiltrated mast cells strongly expressed H-PGDS in inflamed colons. Mast cell-specific H-PGDS deficiency also aggravated colitis and accelerated CAC. In contrast, treatment with a PGD2 receptor agonist inhibited colitis and CAC. Together, our results identified mast cell-derived PGD2 as an inhibitor of colitis and CAC, with implications for its potential use in preventing or treating colon cancer.


Subject(s)
Colitis/metabolism , Colonic Neoplasms/metabolism , Mast Cells/metabolism , Prostaglandin D2/metabolism , Animals , Colitis/drug therapy , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Genotype , Intramolecular Oxidoreductases/deficiency , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prostaglandin D2/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Biomed Res Int ; 2013: 683405, 2013.
Article in English | MEDLINE | ID: mdl-24024207

ABSTRACT

Prostanoids, including prostaglandins (PGs), thromboxanes (TXs), and prostacyclins, are synthesized from arachidonic acid (AA) by the action of Cyclooxygenase (COX) enzymes. They are bioactive inflammatory lipid mediators that play a key role in immunity and immunopathology. Prostanoids exert their effects on immune and inflammatory cells by binding to membrane receptors that are widely expressed throughout the immune system and act at multiple levels in innate and adaptive immunity. The immunoregulatory role of prostanoids results from their ability to regulate cell-cell interaction, antigen presentation, cytokine production, cytokine receptor expression, differentiation, survival, apoptosis, cell-surface molecule levels, and cell migration in both autocrine and paracrine manners. By acting on immune cells of both systems, prostanoids and their receptors have great impact on immune regulation and play a pivotal role in connecting innate and adaptive immunity. This paper focuses on the immunobiology of prostanoid receptor signaling because of their potential clinical relevance for various disorders including inflammation, autoimmunity, and tumorigenesis. We mainly discuss the effects of major COX metabolites, PGD2, PGE2, their signaling during dendritic cell (DC)-natural killer (NK) reciprocal crosstalk, DC-T cell interaction, and subsequent consequences on determining crucial aspects of innate and adaptive immunity in normal and pathological settings.


Subject(s)
Adaptive Immunity/genetics , Dinoprostone/immunology , Immunity, Innate/genetics , Prostaglandin D2/immunology , Receptors, Prostaglandin/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dinoprostone/genetics , Humans , Inflammation/immunology , Inflammation/metabolism , Killer Cells, Natural/immunology , Prostaglandin D2/genetics , Prostaglandins , Receptors, Prostaglandin/metabolism , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...