151
|
Crystal structure of a Bombyx mori sigma-class glutathione transferase exhibiting prostaglandin E synthase activity. Biochim Biophys Acta Gen Subj 2013; 1830:3711-8. [DOI: 10.1016/j.bbagen.2013.02.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 02/13/2013] [Accepted: 02/22/2013] [Indexed: 11/17/2022]
|
152
|
Zhou X, Li D, Resnick MB, Wands J, Cao W. NADPH oxidase NOX5-S and nuclear factor κB1 mediate acid-induced microsomal prostaglandin E synthase-1 expression in Barrett's esophageal adenocarcinoma cells. Mol Pharmacol 2013; 83:978-90. [PMID: 23439561 PMCID: PMC3629825 DOI: 10.1124/mol.112.083287] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 02/25/2013] [Indexed: 12/20/2022] Open
Abstract
The mechanisms of progression from Barrett's esophagus (BE) to esophageal adenocarcinoma (EA) are not known. Cycloxygenase-2 (COX-2)-derived prostaglandin E₂ (PGE₂) has been shown to be important in esophageal tumorigenesis. We have shown that COX-2 mediates acid-induced PGE₂ production. The prostaglandin E synthase (PGES) responsible for acid-induced PGE2 production in BE, however, is not known. We found that microsomal PGES1 (mPGES1), mPGES2, and cytosolic PGES (cPGES) were present in FLO EA cells. Pulsed acid treatment significantly increased mPGES1 mRNA and protein levels but had little or no effect on mPGES2 or cPGES mRNA. Knockdown of mPGES1 by mPGES1 small interfering RNA (siRNA) blocked acid-induced increase in PGE2 production and thymidine incorporation. Knockdown of NADPH oxidase, NOX5-S, a variant lacking calcium-binding domains, by NOX5 siRNA significantly inhibited acid-induced increase in mPGES1 expression, thymidine incorporation, and PGE2 production. Overexpression of NOX5-S significantly increased the luciferase activity in FLO cells transfected with a nuclear factor κB (NF-κB) in vivo activation reporter plasmid pNF-κB-Luc. Knockdown of NF-κB1 p50 by p50 siRNA significantly decreased acid-induced increase in mPGES1 expression, thymidine incorporation, and PGE₂ production. Two novel NF-κB binding elements, GGAGTCTCCC and CGGGACACCC, were identified in the mPGES1 gene promoter. We conclude that mPGES1 mediates acid-induced increase in PGE₂ production and cell proliferation. Acid-induced mPGES1 expression depends on activation of NOX5-S and NF-κB1 p50. Microsomal PGES1 may be a potential target to prevent or treat EA.
Collapse
Affiliation(s)
- Xiaoxu Zhou
- Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | | | | | | | | |
Collapse
|
153
|
Matysiak M, Fortak-Michalska M, Szymanska B, Orlowski W, Jurewicz A, Selmaj K. MicroRNA-146a Negatively Regulates the Immunoregulatory Activity of Bone Marrow Stem Cells by Targeting Prostaglandin E2 Synthase-2. THE JOURNAL OF IMMUNOLOGY 2013; 190:5102-9. [DOI: 10.4049/jimmunol.1202397] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
154
|
Shiro T, Kakiguchi K, Takahashi H, Nagata H, Tobe M. Synthesis and biological evaluation of substituted imidazoquinoline derivatives as mPGES-1 inhibitors. Bioorg Med Chem 2013; 21:2068-78. [DOI: 10.1016/j.bmc.2013.01.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 12/28/2012] [Accepted: 01/06/2013] [Indexed: 11/29/2022]
|
155
|
Effect of Diethylcarbamazine (DEC) on prostaglandin levels in Wuchereria bancrofti infected microfilaraemics. Parasitol Res 2013; 112:2353-9. [PMID: 23525692 DOI: 10.1007/s00436-013-3399-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 03/12/2013] [Indexed: 10/27/2022]
Abstract
Diethylcarbamazine (DEC) interferes with arachidonic acid metabolism for the clearance of microfilariae in Wuchereria bancrofti infected individuals. In this study, we have quantified the plasma concentrations of prostaglandin E2 (PGE2) and 6-keto-PGF1α, the end products of arachidonic acid metabolic pathway in microfilaraemics (DEC treated and untreated), and normal healthy individuals at pre- and 3,9,12,36, and 72 h of post-DEC treatment. We have also determined the microfilariae counts at pre and post day 2 (36 h) and day 3 (72 h) of DEC treatment by membrane filtration technique. Significant reduction in PGE2 and 6-keto-PGF1α concentrations was found at 12 h of DEC treatment. Rapid reduction in microfilarial counts was observed at 36 h of post-DEC treatment. Higher levels of prostaglandins were found at pre-treatment hours in microfilaraemics compared to normal healthy individuals (P < 0.05). Our findings indicate that DEC inhibits prostaglandins for the clearance of microfilariae, and increased levels of prostaglandins in microfilaraemics may be contributed by the parasite or host upon stimulation.
Collapse
|
156
|
Kats A, Båge T, Georgsson P, Jönsson J, Quezada HC, Gustafsson A, Jansson L, Lindberg C, Näsström K, Yucel-Lindberg T. Inhibition of microsomal prostaglandin E synthase-1 by aminothiazoles decreases prostaglandin E2 synthesis in vitro and ameliorates experimental periodontitis in vivo. FASEB J 2013; 27:2328-41. [PMID: 23447581 PMCID: PMC3659347 DOI: 10.1096/fj.12-214445] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The potent inflammatory mediator prostaglandin E2 (PGE2) is implicated in the pathogenesis of several chronic inflammatory conditions, including periodontitis. The inducible enzyme microsomal prostaglandin E synthase-1 (mPGES-1), catalyzing the terminal step of PGE2 biosynthesis, is an attractive target for selective PGE2 inhibition. To identify mPGES-1 inhibitors, we investigated the effect of aminothiazoles on inflammation-induced PGE2 synthesis in vitro, using human gingival fibroblasts stimulated with the cytokine IL-1β and a cell-free mPGES-1 activity assay, as well as on inflammation-induced bone resorption in vivo, using ligature-induced experimental periodontitis in Sprague-Dawley rats. Aminothiazoles 4-([4-(2-naphthyl)-1,3-thiazol-2-yl]amino)phenol (TH-848) and 4-(3-fluoro-4-methoxyphenyl)-N-(4-phenoxyphenyl)-1,3-thiazol-2-amine (TH-644) reduced IL-1β-induced PGE2 production in fibroblasts (IC50 1.1 and 1.5 μM, respectively) as well as recombinant mPGES-1 activity, without affecting activity or expression of the upstream enzyme cyclooxygenase-2. In ligature-induced experimental periodontitis, alveolar bone loss, assessed by X-ray imaging, was reduced by 46% by local treatment with TH-848, compared to vehicle, without any systemic effects on PGE2, 6-keto PGF1α, LTB4 or cytokine levels. In summary, these results demonstrate that the aminothiazoles represent novel mPGES-1 inhibitors for inhibition of PGE2 production and reduction of bone resorption in experimental periodontitis, and may be used as potential anti-inflammatory drugs for treatment of chronic inflammatory diseases, including periodontitis.
Collapse
Affiliation(s)
- Anna Kats
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Takusagawa F. Microsomal prostaglandin E synthase type 2 (mPGES2) is a glutathione-dependent heme protein, and dithiothreitol dissociates the bound heme to produce active prostaglandin E2 synthase in vitro. J Biol Chem 2013; 288:10166-10175. [PMID: 23426368 DOI: 10.1074/jbc.m112.418475] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
An x-ray study indicated that microsomal prostaglandin E synthase type 2 (mPGES2) is a heme-bound protein and catalyzes prostaglandin (PG) H2 degradation, but not PGE2 formation (Yamada, T., and Takusagawa, F. (2007) Biochemistry 46, 8414-8424). In response to the x-ray study, Watanabe et al. claimed that mPGES2 is a heme-free protein and that both the heme-free and heme-bound proteins have PGE2 synthesis activity in the presence of dithiothreitol (Watanabe, K., Ito, S., and Yamamoto, S. (2008) Biochem. Biophys. Res. Commun. 367, 782-786). To resolve the contradictory results, the heme-binding scheme of mPGES2 was further characterized in vivo and in vitro by absorption and fluorescence spectroscopies. A substantial amount of heme-bound mPGES2 was detected in cell extracts. The heme content in mPGES2 was increased along with an increase in Fe(3+) in the culture medium. Heme-free mPGES2 was converted to the heme-bound form by mixing it with pig liver extract, indicating that mPGES2 is capable of forming a complex with heme in mammalian cells. Heme binds to mPGES2 only in the presence of glutathione. The newly determined heme dissociation constant (2.9 nM) supports strongly that mPGES2 is a heme-bound protein in vivo. The bound heme was not dissociated by oxidation by H2O2 or reduction by glutathione or 2-mercaptoethanol. However, reduction by dithiothreitol (an artificial reducing compound) induced the bound heme to dissociate from mPGES2 and released heme-free mPGES2, which exhibited PGE2 synthesis activity in vitro. Imidazole bound to mPGES2 by stacking on the bound heme and inhibited heme oxidation by H2O2 and reduction by dithiothreitol.
Collapse
Affiliation(s)
- Fusao Takusagawa
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66045-7534.
| |
Collapse
|
158
|
Quan Y, Jiang J, Dingledine R. EP2 receptor signaling pathways regulate classical activation of microglia. J Biol Chem 2013; 288:9293-302. [PMID: 23404506 DOI: 10.1074/jbc.m113.455816] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of EP2 receptors by prostaglandin E2 (PGE2) promotes brain inflammation in neurodegenerative diseases, but the pathways responsible are unclear. EP2 receptors couple to Gαs and increase cAMP, which associates with protein kinase A (PKA) and cAMP-regulated guanine nucleotide exchange factors (Epacs). Here, we studied EP2 function and its signaling pathways in rat microglia in their resting state or undergoing classical activation in vitro following treatment with low concentrations of lipopolysaccharide and interferon-γ. Real time PCR showed that PGE2 had no effect on expression of CXCL10, TGF-β1, and IL-11 and exacerbated the rapid up-regulation of mRNAs encoding cyclooxygenase-2, inducible NOS, IL-6, and IL-1β but blunted the production of mRNAs encoding TNF-α, IL-10, CCL3, and CCL4. These effects were mimicked fully by the EP2 agonist butaprost but only weakly by the EP1/EP3 agonist 17-phenyl trinor PGE2 or the EP4 agonist CAY10598 and not at all by the EP3/EP1 agonist sulprostone and confirmed by protein measurements of cyclooxygenase-2, IL-6, IL-10, and TNF-α. In resting microglia, butaprost induced cAMP formation and altered the mRNA expression of inflammatory mediators, but protein expression was unchanged. The PKA inhibitor H89 had little or no effect on inflammatory mediators modulated by EP2, whereas the Epac activator 8-(4-chlorophenylthio)-2'-O-methyladenosine 3',5'-cyclic monophosphate acetoxymethyl ester mimicked all butaprost effects. These results indicate that EP2 activation plays a complex immune regulatory role during classical activation of microglia and that Epac pathways are prominent in this role.
Collapse
Affiliation(s)
- Yi Quan
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
159
|
Kageyama H, Endo K, Osaka T, Watanabe J, Wang LH, Ito K, Suzuki M, Sakagami J, Takenoya F, Shioda S. Galanin-like peptide (GALP) facilitates thermogenesis via synthesis of prostaglandin E2 by astrocytes in the periventricular zone of the third ventricle. J Mol Neurosci 2013; 50:443-52. [PMID: 23354880 DOI: 10.1007/s12031-013-9952-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 01/02/2013] [Indexed: 11/25/2022]
Abstract
Administration of galanin-like peptide (GALP) leads to a decrease in both total food intake and body weight 24 h after injection, compared to controls. Moreover, GALP induces an increase in core body temperature. To elucidate the mechanism by which GALP exerts its effect on energy homeostasis, urethane-anesthetized rats were intracerebroventricularly injected with GALP or saline, after which oxygen consumption, heart rate, and body temperature were monitored for 4 h. In some cases, animals were also pretreated with the cyclooxygenase (COX) inhibitor, diclofenac, via intracerebroventricular (i.c.v.) or intravenous (i.v.) injection. c-Fos expression in the brain was also examined after injection of GALP, and the levels of COX and prostaglandin E(2) synthetase (PGES) mRNA in primary cultured astrocytes treated with GALP were analyzed by using qPCR. The i.c.v. injection of GALP caused biphasic thermogenesis, an effect which could be blocked by pretreatment with centrally (i.c.v.), but not peripherally (i.v.) administered diclofenac. c-Fos immunoreactivity was observed in astrocytes in the periventricular zone of the third ventricle. GALP treatment also increased COX-2 and cytosolic PGES, but not COX-1, microsomal PGES-1, or microsomal PGES-2 mRNA levels in cultured astrocytes. We, therefore, suggest that GALP elicits thermogenesis via a prostaglandin E(2)-mediated pathway in astrocytes of the central nervous system.
Collapse
Affiliation(s)
- Haruaki Kageyama
- Faculty of Health Care, Kiryu University, Midori City, Gunma, 379-2392, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Prostaglandins as PPARγ Modulators in Adipogenesis. PPAR Res 2012; 2012:527607. [PMID: 23319937 PMCID: PMC3540890 DOI: 10.1155/2012/527607] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 11/20/2012] [Indexed: 02/01/2023] Open
Abstract
Adipocytes and fat cells play critical roles in the regulation of energy homeostasis. Adipogenesis (adipocyte differentiation) is regulated via a complex process including coordinated changes in hormone sensitivity and gene expression. PPARγ is a ligand-dependent transcription factor and important in adipogenesis, as it enhances the expression of numerous adipogenic and lipogenic genes in adipocytes. Prostaglandins (PGs), which are lipid mediators, are associated with the regulation of PPARγ function in adipocytes. Prostacyclin promotes the differentiation of adipocyte-precursor cells to adipose cells via activation of the expression of C/EBPβ and δ. These proteins are important transcription factors in the activation of the early phase of adipogenesis, and they activate the expression of PPARγ, which event precedes the maturation of adipocytes. PGE2 and PGF2α strongly suppress the early phase of adipocyte differentiation by enhancing their own production via receptor-mediated elevation of the expression of cycloxygenase-2, and they also suppress the function of PPARγ. In contrast, PGD2 and its non-enzymatic metabolite, Δ12-PGJ2, activate the middle-late phase of adipocyte differentiation through both DP2 receptors and PPARγ. This paper focuses on potential roles of PGs as PPARγ modulators in adipogenesis and regulators of obesity.
Collapse
|
161
|
Takeuchi C, Matsumoto Y, Kohyama K, Uematsu S, Akira S, Yamagata K, Takemiya T. Microsomal prostaglandin E synthase-1 aggravates inflammation and demyelination in a mouse model of multiple sclerosis. Neurochem Int 2012; 62:271-80. [PMID: 23266396 DOI: 10.1016/j.neuint.2012.12.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 11/02/2012] [Accepted: 12/13/2012] [Indexed: 12/15/2022]
Abstract
Microsomal prostaglandin synthetase-1 (mPGES-1) is an inducible terminal enzyme required for prostaglandin E(2) (PGE(2)) biosynthesis. In this study, we examined the role of mPGES-1 in the inflammation and demyelination observed in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We induced EAE with myelin oligodendrocyte glycoprotein(35-55) peptide in mPGES-1-deficient (mPGES-1(-/-)) and wild-type (WT) mice. First, we examined the histopathology in the early and late phases of EAE progression. Next, we measured the concentration of PGE(2) in the spinal cord and investigated the expression of mPGES-1 using immunohistochemistry. In addition, we examined the progression of the severity of EAE using an EAE score to investigate a correlation between pathological features and paralysis. In this paper, we demonstrate that WT mice showed extensive inflammation and demyelination, whereas mPGES-1(-/-) mice exhibited significantly smaller and more localized changes in the perivascular area. The mPGES-1 protein was induced in vascular endothelial cells and microglia around inflammatory foci, and PGE(2) production was increased in WT mice but not mPGES-1(-/-) mice. Furthermore, mPGES-1(-/-) mice showed a significant reduction in the maximum EAE score and improved locomotor activity. These results suggest that central PGE(2) derived from non-neuronal mPGES-1 aggravates the disruption of the vessel structure, leading to the spread of inflammation and local demyelination in the spinal cord, which corresponds to the symptoms of EAE. The inhibition of mPGES-1 may be useful for the treatment of human MS.
Collapse
Affiliation(s)
- Chisen Takeuchi
- Medical Research Institute, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
162
|
Lipopolysaccharide-induced expression of microsomal prostaglandin E synthase-1 mediates late-phase PGE2 production in bone marrow derived macrophages. PLoS One 2012; 7:e50244. [PMID: 23226252 PMCID: PMC3511480 DOI: 10.1371/journal.pone.0050244] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 10/22/2012] [Indexed: 11/24/2022] Open
Abstract
Cyclooxygenase (COX)-2 expression and release of prostaglandins (PGs) by macrophages are consistent features of lipopolysaccharide (LPS)-induced macrophage inflammation. The two major PGs, PGE2 and PGD2, are synthesized by the prostanoid isomerases, PGE synthases (PGES) and PGD synthases (PGDS), respectively. Since the expression profile and the individual role of these prostanoid isomerases-mediated inflammation in macrophages has not been defined, we examined the LPS-stimulated PGs production pattern and the expression profile of their synthases in the primary cultured mouse bone marrow derived macrophages (BMDM). Our data show that LPS induced both PGE2 and PGD2 production, which was evident by ∼8 hrs and remained at a similar ratio (∼1∶1) in the early phase (≤12 hrs) of LPS treatment. However, PGE2 production continued increase further in the late phase (16–24 hrs); whereas the production of PGD2 remained at a stable level from 12 to 24 hrs post-treatment. In response to LPS-treatment, the expression of both COX-2 and inducible nitric oxide synthase (iNOS) was detected within 2 to 4 hrs; whereas the increased expression of microsomal PGES (mPGES)-1 and a myeloid cell transcription factor PU.1 did not appear until later phase (≥12 hrs). In contrast, the expression of COX-1, hematopoietic-PGDS (H-PGDS), cytosolic-PGES (c-PGES), or mPGES-2 in BMDM was not affected by LPS treatment. Selective inhibition of mPGES-1 with either siRNA or isoform-selective inhibitor CAY10526, but not mPGES-2, c-PGES or PU.1, attenuated LPS-induced burst of PGE2 production indicating that mPGES-1 mediates LPS-induced PGE2 production in BMDM. Interestingly, selective inhibition of mPGES-1 was also associated with a decrease in LPS-induced iNOS expression. In summary, our data show that mPGES-1, but not mPGES-2 or c-PGES isomerase, mediates LPS-induced late-phase burst of PGE2 generation, and regulates LPS-induced iNOS expression in BMDM.
Collapse
|
163
|
Abstract
Rheumatoid arthritis (RA) is a chronic, autoimmune, and complex inflammatory disease leading to bone and cartilage destruction, whose cause remains obscure. Accumulation of genetic susceptibility, environmental factors, and dysregulated immune responses are necessary for mounting this self-reacting disease. Inflamed joints are infiltrated by a heterogeneous population of cellular and soluble mediators of the immune system, such as T cells, B cells, macrophages, cytokines, and prostaglandins (PGs). Prostaglandins are lipid inflammatory mediators derived from the arachidonic acid by multienzymatic reactions. They both sustain homeostatic mechanisms and mediate pathogenic processes, including the inflammatory reaction. They play both beneficial and harmful roles during inflammation, according to their site of action and the etiology of the inflammatory response. With respect to the role of PGs in inflammation, they can be effective mediators in the pathophysiology of RA. Thus the use of agonists or antagonists of PG receptors may be considered as a new therapeutic protocol in RA. In this paper, we try to elucidate the role of PGs in the immunopathology of RA.
Collapse
|
164
|
Matsumoto C, Oda T, Yokoyama S, Tominari T, Hirata M, Miyaura C, Inada M. Toll-like receptor 2 heterodimers, TLR2/6 and TLR2/1 induce prostaglandin E production by osteoblasts, osteoclast formation and inflammatory periodontitis. Biochem Biophys Res Commun 2012; 428:110-5. [PMID: 23063683 DOI: 10.1016/j.bbrc.2012.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 10/03/2012] [Indexed: 10/27/2022]
Abstract
TLR2 forms heterodimers with TLR1 and TLR6, and regulates host defense mechanisms against pathogens. We examined the role of TLR2 heterodimer signaling in osteoclast formation and inflammatory periodontitis. In co-cultures of mouse bone marrow cells and osteoblasts, a TLR2/6 ligand (diacylated lipopeptide designed from Gram-positive bacteria) markedly induced osteoclast formation. A TLR2/1 ligand (triacylated lipopeptide designed from Gram-negative bacteria) also induced osteoclast formation. The osteoclast formation induced by TLR2/6 and TLR2/1 ligands was completely suppressed by indomethacin. Osteoblasts expressed TLR1, 2, 4, and 6 mRNAs, and both TLR2/6 and TLR2/1 ligands induced the expression of COX-2, mPGES-1, and RANKL mRNA, as well as PGE production in osteoblasts. Both TLR2/6 and TLR2/1 ligands induced the resorption of mandibular alveolar bone in organ cultures, and elicited inflammatory periodontitis in vivo. Therefore, TLR2 heterodimer signaling may play a key role in PGE-mediated inflammatory bone loss in periodontal disease.
Collapse
Affiliation(s)
- Chiho Matsumoto
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | | | | | | | | | | | | |
Collapse
|
165
|
Fujimori K, Yano M, Ueno T. Synergistic suppression of early phase of adipogenesis by microsomal PGE synthase-1 (PTGES1)-produced PGE2 and aldo-keto reductase 1B3-produced PGF2α. PLoS One 2012; 7:e44698. [PMID: 22970288 PMCID: PMC3436788 DOI: 10.1371/journal.pone.0044698] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 08/09/2012] [Indexed: 12/30/2022] Open
Abstract
We recently reported that aldo-keto reductase 1B3-produced prostaglandin (PG) F2α suppressed the early phase of adipogenesis. PGE2 is also known to suppress adipogenesis. In this study, we found that microsomal PGE2 synthase (PGES)-1 (mPGES-1; PTGES1) acted as the PGES in adipocytes and that PGE2 and PGF2α synergistically suppressed the early phase of adipogenesis. PGE2 production was detected in preadipocytes and transiently enhanced at 3 h after the initiation of adipogenesis of mouse adipocytic 3T3-L1 cells, followed by a quick decrease; and its production profile was similar to the expression of the cyclooxygenase-2 (PTGS2) gene. When 3T3-L1 cells were transfected with siRNAs for any one of the three major PTGESs, i.e., PTGES1, PTGES2 (mPGES-2), and PTGES3 (cytosolic PGES), only PTGES1 siRNA suppressed PGE2 production and enhanced the expression of adipogenic genes. AE1-329, a PTGER4 (EP4) receptor agonist, increased the expression of the Ptgs2 gene with a peak at 1 h after the initiation of adipogenesis. PGE2-mediated enhancement of the PTGS2 expression was suppressed by the co-treatment with L-161982, a PTGER4 receptor antagonist. Moreover, AE1-329 enhanced the expression of the Ptgs2 gene by binding of the cyclic AMP response element (CRE)-binding protein to the CRE of the Ptgs2 promoter; and its binding was suppressed by co-treatment with L-161982, which was demonstrated by promoter luciferase and chromatin immunoprecipitation assays. Furthermore, when 3T3-L1 cells were caused to differentiate into adipocytes in medium containing both PGE2 and PGF2α, the expression of the adipogenic genes and the intracellular triglyceride level were decreased to a greater extent than in medium containing either of them, revealing that PGE2 and PGF2α independently suppressed adipogenesis. These results indicate that PGE2 was synthesized by PTGES1 in adipocytes and synergistically suppressed the early phase of adipogenesis of 3T3-L1 cells in cooperation with PGF2α through receptor-mediated activation of PTGS2 expression.
Collapse
Affiliation(s)
- Ko Fujimori
- Laboratory of Biodefense and Regulation, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan
- * E-mail:
| | - Mutsumi Yano
- Laboratory of Biodefense and Regulation, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan
| | - Toshiyuki Ueno
- Laboratory of Biodefense and Regulation, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan
| |
Collapse
|
166
|
Reebye V, Querol Cano L, Lavery DN, Brooke GN, Powell SM, Chotai D, Walker MM, Whitaker HC, Wait R, Hurst HC, Bevan CL. Role of the HSP90-associated cochaperone p23 in enhancing activity of the androgen receptor and significance for prostate cancer. Mol Endocrinol 2012; 26:1694-706. [PMID: 22899854 DOI: 10.1210/me.2012-1056] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Prostate tumor growth initially depends on androgens, which act via the androgen receptor (AR). Despite androgen ablation therapy, tumors eventually progress to a castrate-resistant stage in which the AR remains active. The mechanisms are poorly understood but it may be that changes in levels or activity of AR coregulators affect trafficking and activation of the receptor. A key stage in AR signaling occurs in the cytoplasm, where unliganded receptor is associated with the heat shock protein (HSP)90 foldosome complex. p23, a key component of this complex, is best characterized as a cochaperone for HSP90 but also has HSP90-independent activity and has been reported as having differential effects on the activity of different steroid receptors. Here we report that p23 increases activity of the AR, and this appears to involve steps both in the cytoplasm (increasing ligand-binding capacity, possibly via direct interaction with AR) and the nucleus (enhancing AR occupancy at target promoters). We show, for the first time, that AR and p23 can interact, perhaps directly, when HSP90 is not present in the same complex. The effects of p23 on AR activity are at least partly HSP90 independent because a mutant form of p23, unable to bind HSP90, nevertheless increases AR activity. In human prostate tumors, nuclear p23 was higher in malignant prostate cells compared with benign/normal cells, supporting the utility of p23 as a therapeutic target in prostate cancer.
Collapse
Affiliation(s)
- Vikash Reebye
- Androgen Signaling Laboratory, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Parazzoli S, Harmon JS, Vallerie SN, Zhang T, Zhou H, Robertson RP. Cyclooxygenase-2, not microsomal prostaglandin E synthase-1, is the mechanism for interleukin-1β-induced prostaglandin E2 production and inhibition of insulin secretion in pancreatic islets. J Biol Chem 2012; 287:32246-53. [PMID: 22822059 DOI: 10.1074/jbc.m112.364612] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Arachidonic acid is converted to prostaglandin E(2) (PGE(2)) by a sequential enzymatic reaction performed by two isoenzyme groups, cyclooxygenases (COX-1 and COX-2) and terminal prostaglandin E synthases (cPGES, mPGES-1, and mPGES-2). mPGES-1 is widely considered to be the final enzyme regulating COX-2-dependent PGE(2) synthesis. These generalizations have been based in most part on experiments utilizing gene expression analyses of cell lines and tumor tissue. To assess the relevance of these generalizations to a native mammalian tissue, we used isolated human and rodent pancreatic islets to examine interleukin (IL)-1β-induced PGE(2) production, because PGE(2) has been shown to mediate IL-1β inhibition of islet function. Rat islets constitutively expressed mRNAs of COX-1, COX-2, cPGES, and mPGES-1. As expected, IL-1β increased mRNA levels for COX-2 and mPGES-1, but not for COX-1 or cPGES. Basal protein levels of COX-1, cPGES, and mPGES-2 were readily detected in whole cell extracts but were not regulated by IL-1β. IL-1β increased protein levels of COX-2, but unexpectedly mPGES-1 protein levels were low and unaffected. In microsomal extracts, mPGES-1 protein was barely detectable in rat islets but clearly present in human islets; however, in neither case did IL-1β increase mPGES-1 protein levels. To further assess the importance of mPGES-1 to IL-1β regulation of an islet physiologic response, glucose-stimulated insulin secretion was examined in isolated islets of WT and mPGES-1-deficient mice. IL-1β inhibited glucose-stimulated insulin secretion equally in both WT and mPGES-1(-/-) islets, indicating that COX-2, not mPGES-1, mediates IL-1β-induced PGE(2) production and subsequent inhibition of insulin secretion.
Collapse
Affiliation(s)
- Susan Parazzoli
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington, Seattle, Washington 98122, USA
| | | | | | | | | | | |
Collapse
|
168
|
KOEBERLE ANDREAS, WERZ OLIVER. Microsomal Prostaglandin E2 Synthase-1. ANTI-INFLAMMATORY DRUG DISCOVERY 2012. [DOI: 10.1039/9781849735346-00001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The prostanoids and leukotrienes (LTs) formed from arachidonic acid (AA) via the cyclooxygenase (COX)-1/2 and 5-lipoxygenase (5-LO) pathway, respectively, mediate inflammatory responses, chronic tissue remodelling, cancer, asthma and autoimmune disorders, but also possess homeostatic functions in the gastrointestinal tract, uterus, brain, kidney, vasculature and host defence. Based on the manifold functions of these eicosanoids, the clinical use of non-steroidal anti-inflammatory drugs (NSAIDs), a class of drugs that block formation of all prostanoids, is hampered by severe side-effects including gastrointestinal injury, renal irritations and cardiovascular risks. Therefore, anti-inflammatory agents interfering with eicosanoid biosynthesis require a well-balanced pharmacological profile to minimize these on-target side-effects. Current anti-inflammatory research aims at identifying compounds that can suppress the massive formation of pro-inflammatory prostaglandin (PG)E2 without affecting homeostatic PGE2 and PGI2 synthesis. The inducible microsomal prostaglandin E2 synthase-1 (mPGES-1) is one promising target enzyme. We will give an overview about the structure, regulation and function of mPGES-1 and then present novel inhibitors of mPGES-1 that may possess a promising pharmacological profile.
Collapse
Affiliation(s)
- ANDREAS KOEBERLE
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy University Jena Philosophenweg 14, D-07743 Jena Germany
| | - OLIVER WERZ
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy University Jena Philosophenweg 14, D-07743 Jena Germany
| |
Collapse
|
169
|
Prostaglandin E2 deficiency uncovers a dominant role for thromboxane A2 in house dust mite-induced allergic pulmonary inflammation. Proc Natl Acad Sci U S A 2012; 109:12692-7. [PMID: 22802632 DOI: 10.1073/pnas.1207816109] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Prostaglandin E(2) (PGE(2)) is an abundant lipid inflammatory mediator with potent but incompletely understood anti-inflammatory actions in the lung. Deficient PGE(2) generation in the lung predisposes to airway hyperresponsiveness and aspirin intolerance in asthmatic individuals. PGE(2)-deficient ptges(-/-) mice develop exaggerated pulmonary eosinophilia and pulmonary arteriolar smooth-muscle hyperplasia compared with PGE(2)-sufficient controls when challenged intranasally with a house dust mite extract. We now demonstrate that both pulmonary eosinophilia and vascular remodeling in the setting of PGE(2) deficiency depend on thromboxane A(2) and signaling through the T prostanoid (TP) receptor. Deletion of TP receptors from ptges(-/-) mice reduces inflammation, vascular remodeling, cytokine generation, and airway reactivity to wild-type levels, with contributions from TP receptors localized to both hematopoietic cells and tissue. TP receptor signaling ex vivo is controlled heterologously by E prostanoid (EP)(1) and EP(2) receptor-dependent signaling pathways coupling to protein kinases C and A, respectively. TP-dependent up-regulation of intracellular adhesion molecule-1 expression is essential for the effects of PGE(2) deficiency. Thus, PGE(2) controls the strength of TP receptor signaling as a major bronchoprotective mechanism, carrying implications for the pathobiology and therapy of asthma.
Collapse
|
170
|
Lima IVDA, Bastos LFS, Limborço-Filho M, Fiebich BL, de Oliveira ACP. Role of prostaglandins in neuroinflammatory and neurodegenerative diseases. Mediators Inflamm 2012; 2012:946813. [PMID: 22778499 PMCID: PMC3385693 DOI: 10.1155/2012/946813] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/05/2012] [Indexed: 11/17/2022] Open
Abstract
Increasing data demonstrates that inflammation participates in the pathophysiology of neurodegenerative diseases. Among the different inflammatory mediators involved, prostaglandins play an important role. The effects induced by prostaglandins might be mediated by activation of their known receptors or by nonclassical mechanisms. In the present paper, we discuss the evidences that link prostaglandins, as well as the enzymes that produce them, to some neurological diseases.
Collapse
Affiliation(s)
- Isabel Vieira de Assis Lima
- Department of Pharmacology, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - Leandro Francisco Silva Bastos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, Brazil
- Department of Psychology and Neuroscience, Muenzinger Building, Colorado University of Colorado Boulder, Avenida, Boulder, CO 80309-0354, USA
| | - Marcelo Limborço-Filho
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, Brazil
| | - Bernd L. Fiebich
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Hauptstraße 5, 79104 Freiburg, Germany
- VivaCell Biotechnology GmbH, Ferdinand-Porsche-Straße 5, 79211 Denzlingen, Germany
| | - Antonio Carlos Pinheiro de Oliveira
- Department of Pharmacology, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
- Department of Psychiatry and Psychotherapy, University of Freiburg Medical School, Hauptstraße 5, 79104 Freiburg, Germany
| |
Collapse
|
171
|
Degousee N, Simpson J, Fazel S, Scholich K, Angoulvant D, Angioni C, Schmidt H, Korotkova M, Stefanski E, Wang XH, Lindsay TF, Ofek E, Pierre S, Butany J, Jakobsson PJ, Keating A, Li RK, Nahrendorf M, Geisslinger G, Backx PH, Rubin BB. Lack of Microsomal Prostaglandin E
2
Synthase-1 in Bone Marrow–Derived Myeloid Cells Impairs Left Ventricular Function and Increases Mortality After Acute Myocardial Infarction. Circulation 2012; 125:2904-13. [DOI: 10.1161/circulationaha.112.099754] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background—
Microsomal prostaglandin E
2
synthase-1 (mPGES-1), encoded by the
Ptges
gene, catalyzes prostaglandin E
2
biosynthesis and is expressed by leukocytes, cardiac myocytes, and cardiac fibroblasts.
Ptges
−/−
mice develop more left ventricle (LV) dilation, worse LV contractile function, and higher LV end-diastolic pressure than
Ptges
+/+
mice after myocardial infarction. In this study, we define the role of mPGES-1 in bone marrow–derived leukocytes in the recovery of LV function after coronary ligation.
Methods and Results—
Cardiac structure and function in
Ptges
+/+
mice with
Ptges
+/+
bone marrow (
BM
+/+
) and
Ptges
+/+
mice with
Ptges
−/−
BM (
BM
−/−
) were assessed by morphometric analysis, echocardiography, and invasive hemodynamics before and 7 and 28 days after myocardial infarction. Prostaglandin levels and prostaglandin biosynthetic enzyme gene expression were measured by liquid chromatography–tandem mass spectrometry and real-time polymerase chain reaction, immunoblotting, immunohistochemistry, and immunofluorescence microscopy, respectively. After myocardial infarction,
BM
−/−
mice had more LV dilation, worse LV systolic and diastolic function, higher LV end-diastolic pressure, more cardiomyocyte hypertrophy, and higher mortality but similar infarct size and pulmonary edema compared with
BM
+/+
mice.
BM
−/−
mice also had higher levels of COX-1 protein and more leukocytes in the infarct, but not the viable LV, than
BM
+/+
mice. Levels of prostaglandin E
2
were higher in the infarct and viable myocardium of
BM
−/−
mice than in
BM
+/+
mice.
Conclusions—
Lack of mPGES-1 in bone marrow–derived leukocytes negatively regulates COX-1 expression, prostaglandin E
2
biosynthesis, and inflammation in the infarct and leads to impaired LV function, adverse LV remodeling, and decreased survival after acute myocardial infarction.
Collapse
Affiliation(s)
- Norbert Degousee
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Jeremy Simpson
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Shafie Fazel
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Klaus Scholich
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Denis Angoulvant
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Carlo Angioni
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Helmut Schmidt
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Marina Korotkova
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Eva Stefanski
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Xing-Hua Wang
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Thomas F. Lindsay
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Efrat Ofek
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Sandra Pierre
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Jagdish Butany
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Per-Johan Jakobsson
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Armand Keating
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Ren-Ke Li
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Matthias Nahrendorf
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Gerd Geisslinger
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Peter H. Backx
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| | - Barry B. Rubin
- From the Divisions of Vascular Surgery (N.D., E.S., T.F.L., B.B.R.), Cardiac Surgery (S.F., R.-K.L.), Cardiology (P.H.B.), and Pathology (E.O., J.B.), Peter Munk Cardiac Centre, and the Department of Medical Oncology & Hematology (X.-H.W., A.K.), Toronto General Hospital, University Health Network, Toronto, Canada; Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada (J.S., P.H.B.); Institut für Klinische Pharmakologie, Frankfurt am Main, Germany (K.S., C.A., H
| |
Collapse
|
172
|
Liu X, Zou L, Zhu L, Zhang H, Du C, Li Z, Gao C, Zhao X, Bao S, Zheng H. miRNA mediated up-regulation of cochaperone p23 acts as an anti-apoptotic factor in childhood acute lymphoblastic leukemia. Leuk Res 2012; 36:1098-104. [PMID: 22677230 DOI: 10.1016/j.leukres.2012.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 04/18/2012] [Accepted: 05/08/2012] [Indexed: 01/24/2023]
Abstract
p23 is a heat shock protein 90 (Hsp90) cochaperone that plays a significant role in estrogen receptor (ER) alpha signal transduction and telomerase activity; it is up-regulated in several cancers. Recent studies have found that high level of p23 may promote tumor progression and poor prognosis in breast cancer patients. p23 was found to be overexpressed in our previous microarray assay of 100 childhood acute lymphoblastic leukemia (ALL) bone marrow (BM) samples. In the present study, we verified the upregulation of p23 in clinical ALL samples, and identified p23 to be an anti-apoptotic factor in the process of chemotherapy. We also found that p23 was regulated by hsa-miR-101 which was down-regulated in childhood ALL cases. Altogether these data demonstrate that the misregulation of hsa-miR-101 contributes partly to the overexpression of p23 in childhood ALL. As an anti-apoptotic factor, p23 is able to be a potential target for anti-leukemic therapy.
Collapse
Affiliation(s)
- Xiao Liu
- Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Sha W, Brüne B, Weigert A. The multi-faceted roles of prostaglandin E2 in cancer-infiltrating mononuclear phagocyte biology. Immunobiology 2012; 217:1225-32. [PMID: 22727331 DOI: 10.1016/j.imbio.2012.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/07/2012] [Indexed: 12/14/2022]
Abstract
Extensive research in the last two decades implemented that the inflammatory cell infiltrate, especially in solid tumors, is a major determinant for patient prognosis. Mononuclear phagocytes, i.e. monocytes/macrophages, dendritic cells and myeloid-derived suppressor cells, constitute the majority of tumor-associated immune cells. Instead of inducing anti-tumor immunity, mononuclear phagocytes are functionally subverted by tumor microenvironmental factors to support each stage of oncogenesis. Although mechanisms how tumors program their inflammatory infiltrate to support tumor development are ill-defined, few master regulators are beginning to emerge. One of them is the inflammatory eicosanoid prostaglandin E(2) (PGE(2)), produced by tumor cells or the infiltrating immune cells. In this review we summarize the impact of PGE(2) on mononuclear phagocytes in inflammation and cancer and discuss potential implications for cancer therapy.
Collapse
Affiliation(s)
- Weixiao Sha
- Institute of Biochemistry I/ZAFES, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | | | | |
Collapse
|
174
|
Frölich S, Olliges A, Kern N, Schreiber Y, Narumiya S, Nüsing RM. Temporal expression of the PGE2 synthetic system in the kidney is associated with the time frame of renal developmental vulnerability to cyclooxygenase-2 inhibition. Am J Physiol Renal Physiol 2012; 303:F209-19. [PMID: 22573380 DOI: 10.1152/ajprenal.00418.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pharmacological blockade of cyclooxygenase-2 (COX-2) causes impairment of kidney development. The present study was aimed at determining temporal expression pattern and activity of the PGE(2) synthetic pathway during postnatal nephrogenesis in mice and its association to the time window sensitive to COX-2 inhibition. During the first 10 days after birth, we observed transient induction of mRNA and protein for microsomal PGE synthase (mPGES)-1 between postnatal days 4 (P4) and P8, but not for mPGES-2 or cytosolic PGE synthase (cPGES). PGE(2) synthetic activity using arachidonic acid and PGH(2) as substrates and also urinary excretion of PGE(2) were enhanced during this time frame. In parallel to the PGE(2) system, COX-2 but not COX-1 expression was also transiently induced. Studying glomerulogenesis in EP receptor knockout mice revealed a reduction in glomerular size in EP1(-/-), EP2(-/-), and EP4(-/-) mice, supporting the developmental role of PGE(2). The most vulnerable time window to COX-2 inhibition by SC-236 was found closely related to the temporal expression of COX-2 and mPGES-1. The strongest effects of COX-2 inhibition were achieved following 8 days of drug administration. Similar developmental damage was caused by application of rofecoxib, but not by the COX-1-selective inhibitor SC-560. COX-2 inhibition starting after P10 has had no effect on the size of glomeruli or on the relative number of superficial glomeruli; however, growth of the renal cortex was significantly diminished, indicating the requirement of COX-2 activity after P10. Effects of COX-2 inhibition on renal cell differentiation and on renal fibrosis needed a prolonged time of exposition of at least 10 days. In conclusion, temporal expression of the PGE(2) synthetic system coincides with the most vulnerable age interval for the induction of irreversible renal abnormalities. We assume that mPGES-1 is coregulated with COX-2 for PGE(2) synthesis to orchestrate postnatal kidney development and growth.
Collapse
Affiliation(s)
- Stefanie Frölich
- Institute of Clinical Pharmacology, Johann Wolfgang Goethe-University, Theodor Stern Kai 7, Frankfurt, Germany
| | | | | | | | | | | |
Collapse
|
175
|
Wang M, FitzGerald GA. Cardiovascular biology of microsomal prostaglandin E synthase-1. Trends Cardiovasc Med 2012; 20:189-95. [PMID: 22137640 DOI: 10.1016/j.tcm.2011.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 04/13/2011] [Indexed: 10/14/2022]
Abstract
Both traditional and purpose-designed nonsteroidal anti-inflammatory drugs, selective for inhibition of cyclooxygenase (COX)-2, alleviate pain and inflammation but confer a cardiovascular hazard attributable to inhibition of COX-2-derived prostacyclin (PGI(2)). Deletion of microsomal PGE synthase-1 (mPGES-1), the dominant enzyme that converts the COX-derived intermediate product PGH(2) to PGE(2), modulates inflammatory pain in rodents. In contrast with COX-2 deletion or inhibition, PGI(2) formation is augmented in mPGES-1(-/-) mice-an effect that may confer cardiovascular benefit but may undermine the analgesic potential of inhibitors of this enzyme. This review considers the cardiovascular biology of mPGES1 and the complex challenge of developing inhibitors of this enzyme.
Collapse
Affiliation(s)
- Miao Wang
- Institute for Translational Medicine and Therapeutics, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA
| | | |
Collapse
|
176
|
Abstract
Polyunsaturated fatty acids (PUFA) play important roles in the normal physiology and in pathological states including inflammation and cancer. While much is known about the biosynthesis and biological activities of eicosanoids derived from ω6 PUFA, our understanding of the corresponding ω3 series lipid mediators is still rudimentary. The purpose of this review is not to offer a comprehensive summary of the literature on fatty acids in prostate cancer but rather to highlight some of the areas where key questions remain to be addressed. These include substrate preference and polymorphic variants of enzymes involved in the metabolism of PUFA, the relationship between de novo lipid synthesis and dietary lipid metabolism pathways, the contribution of cyclooxygenases and lipoxygenases as well as terminal synthases and prostanoid receptors in prostate cancer, and the potential role of PUFA in angiogenesis and cell surface receptor signaling.
Collapse
|
177
|
Regulation of human microsomal prostaglandin E synthase-1 by IL-1β requires a distal enhancer element with a unique role for C/EBPβ. Biochem J 2012; 443:561-71. [DOI: 10.1042/bj20111801] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The studies of PGE2 (prostaglandin E2) biosynthesis have focused primarily on the role of cyclo-oxygenases. Efforts have shifted towards the specific PGE2 terminal synthases, particularly mPGES-1 (microsomal PGE synthase 1), which has emerged as the crucial inducible synthase with roles in pain, cancer and inflammation. mPGES-1 is induced by pro-inflammatory cytokines with studies focusing on the proximal promoter, mediated specifically through Egr-1 (early growth-response factor 1). Numerous studies demonstrate that the mPGES-1 promoter (PTGES) alone cannot account for the level of IL-1β (interleukin 1β) induction. We identified two DNase I-hypersensitive sites within the proximal promoter near the Egr-1 element and a novel distal site near −8.6 kb. Functional analysis of the distal site revealed two elements that co-operate with basal promoter expression and a stimulus-dependent enhancer. A specific binding site for C/EBPβ (CCAAT/enhancer-binding protein β) in the enhancer was directly responsible for inducible enhancer activity. ChIP (chromatin immunoprecipitation) analysis demonstrated constitutive Egr-1 binding to the promoter and induced RNA polymerase II and C/EBPβ binding to the promoter and enhancer respectively. Knockout/knockdown studies established a functional role for C/EBPβ in mPGES-1 gene regulation and the documented interaction between Egr-1 and C/EBPβ highlights the proximal promoter co-operation with a novel distal enhancer element in regulating inducible mPGES-1 expression.
Collapse
|
178
|
Role of PGE2 in asthma and nonasthmatic eosinophilic bronchitis. Mediators Inflamm 2012; 2012:645383. [PMID: 22529528 PMCID: PMC3316983 DOI: 10.1155/2012/645383] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 01/09/2012] [Accepted: 01/09/2012] [Indexed: 11/17/2022] Open
Abstract
Eosinophilic bronchitis is a common cause of chronic cough, which like asthma is characterized by sputum eosinophilia, but unlike asthma there is no variable airflow obstruction or airway hyperresponsiveness. Several studies suggest that prostaglandins may play an important role in orchestrating interactions between different cells in several inflammatory diseases such as asthma. PGE2 is important because of the multiplicity of its effects on immune response in respiratory diseases; however, respiratory system appears to be unique in that PGE2 has beneficial effects. We described that the difference in airway function observed in patients with eosinophilic bronchitis and asthma could be due to differences in PGE2 production. PGE2 present in induced sputum supernatant from NAEB patients decreases BSMC proliferation, probably due to simultaneous stimulation of EP2 and EP4 receptors with inhibitory activity. This protective effect of PGE2 may not only be the result of a direct action exerted on airway smooth-muscle proliferation but may also be attributable to the other anti-inflammatory actions.
Collapse
|
179
|
Church RJ, Jania LA, Koller BH. Prostaglandin E(2) produced by the lung augments the effector phase of allergic inflammation. THE JOURNAL OF IMMUNOLOGY 2012; 188:4093-102. [PMID: 22412193 DOI: 10.4049/jimmunol.1101873] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Elevated PGE(2) is a hallmark of most inflammatory lesions. This lipid mediator can induce the cardinal signs of inflammation, and the beneficial actions of nonsteroidal anti-inflammatory drugs are attributed to inhibition of cyclooxygenase (COX)-1 and COX-2, enzymes essential in the biosynthesis of PGE(2) from arachidonic acid. However, both clinical studies and rodent models suggest that, in the asthmatic lung, PGE(2) acts to restrain the immune response and limit physiological change secondary to inflammation. To directly address the role of PGE(2) in the lung, we examined the development of disease in mice lacking microsomal PGE(2) synthase-1 (mPGES1), which converts COX-1/COX-2-derived PGH(2) to PGE(2). We show that mPGES1 determines PGE(2) levels in the naive lung and is required for increases in PGE(2) after OVA-induced allergy. Although loss of either COX-1 or COX-2 increases the disease severity, surprisingly, mPGES1(-/-) mice show reduced inflammation. However, an increase in serum IgE is still observed in the mPGES1(-/-) mice, suggesting that loss of PGE(2) does not impair induction of a Th2 response. Furthermore, mPGES1(-/-) mice expressing a transgenic OVA-specific TCR are also protected, indicating that PGE(2) acts primarily after challenge with inhaled Ag. PGE(2) produced by the lung plays the critical role in this response, as loss of lung mPGES1 is sufficient to protect against disease. Together, this supports a model in which mPGES1-dependent PGE(2) produced by populations of cells native to the lung contributes to the effector phase of some allergic responses.
Collapse
Affiliation(s)
- Rachel J Church
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
180
|
Cao X, Peterson JR, Wang G, Anrather J, Young CN, Guruju MR, Burmeister MA, Iadecola C, Davisson RL. Angiotensin II-dependent hypertension requires cyclooxygenase 1-derived prostaglandin E2 and EP1 receptor signaling in the subfornical organ of the brain. Hypertension 2012; 59:869-76. [PMID: 22371360 DOI: 10.1161/hypertensionaha.111.182071] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cyclooxygenase (COX)-derived prostanoids have long been implicated in blood pressure (BP) regulation. Recently prostaglandin E(2) (PGE(2)) and its receptor EP(1) (EP(1)R) have emerged as key players in angiotensin II (Ang II)-dependent hypertension (HTN) and related end-organ damage. However, the enzymatic source of PGE(2,) that is, COX-1 or COX-2, and its site(s) of action are not known. The subfornical organ (SFO) is a key forebrain region that mediates systemic Ang II-dependent HTN via reactive oxygen species (ROS). We tested the hypothesis that cross-talk between PGE(2)/EP(1)R and ROS signaling in the SFO is required for Ang II HTN. Radiotelemetric assessment of blood pressure revealed that HTN induced by infusion of systemic "slow-pressor" doses of Ang II was abolished in mice with null mutations in EP(1)R or COX-1 but not COX-2. Slow-pressor Ang II-evoked HTN and ROS formation in the SFO were prevented when the EP(1)R antagonist SC-51089 was infused directly into brains of wild-type mice, and Ang-II-induced ROS production was blunted in cells dissociated from SFO of EP(1)R(-/-) and COX-1(-/-) but not COX-2(-/-) mice. In addition, slow-pressor Ang II infusion caused a ≈3-fold increase in PGE(2) levels in the SFO but not in other brain regions. Finally, genetic reconstitution of EP(1)R selectively in the SFO of EP(1)R-null mice was sufficient to rescue slow-pressor Ang II-elicited HTN and ROS formation in the SFO of this model. Thus, COX 1-derived PGE(2) signaling through EP(1)R in the SFO is required for the ROS-mediated HTN induced by systemic infusion of Ang II and suggests that EP(1)R in the SFO may provide a novel target for antihypertensive therapy.
Collapse
Affiliation(s)
- Xian Cao
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Abstract
Microsomal prostaglandin E synthase-1 (mPGES-1) is the terminal synthase responsible for the synthesis of the pro-tumorigenic prostaglandin E(2) (PGE(2)). mPGES-1 is overexpressed in a wide variety of cancers. Since its discovery in 1997 by Bengt Samuelsson and collaborators, the enzyme has been the object of over 200 peer-reviewed articles. Although today mPGES-1 is considered a validated and promising therapeutic target for anticancer drug discovery, challenges in inhibitor design and selectivity are such that up to this date there are only a few published records of small-molecule inhibitors targeting the enzyme and exhibiting some in vivo anticancer activity. This review summarizes the structures, and the in vitro and in vivo activities of these novel mPGES-1 inhibitors. Challenges that have been encountered are also discussed.
Collapse
|
182
|
Miyagishi H, Kosuge Y, Ishige K, Ito Y. Expression of microsomal prostaglandin E synthase-1 in the spinal cord in a transgenic mouse model of amyotrophic lateral sclerosis. J Pharmacol Sci 2012; 118:225-36. [PMID: 22302024 DOI: 10.1254/jphs.11221fp] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Prostaglandin E(2) (PGE(2)) is a key molecule involved in the neuroinflammatory processes that characterize amyotrophic lateral sclerosis (ALS). Although PGE(2) synthesis is regulated by PGE(2) synthases (PGESs), the pathological role of PGESs in ALS still remains unknown. Experiments were performed to elucidate the expression of PGESs and the localization of microsomal PGES-1 (mPGES-1) in neurons and glial cells in the spinal cord of ALS model (G93A) mice. Neurological symptom was observed in G93A mice from 14 weeks by the tail suspension test, and rotarod performances were decreased at 16 weeks and older. Western blotting revealed that the level of mPGES-1 was increased in G93A mice at 15 weeks and older. In contrast, the levels of cytosolic PGES and mPGES-2 did not change at any age. Immunohistochemical analysis demonstrated that age-dependent expression of mPGES-1 was found in motor neurons in G93A mice at 11 and 15 weeks. Immunoreactivity of mPGES-1 was also co-localized in Iba1-positive microglia in G93A mice at 15 weeks. These results suggest that mPGES-1 in motor neurons may play a role in the pathogenesis of ALS and that mPGES-1 may work sequentially in motor neurons and activated microglia to produce ALS symptoms in G93A mice.
Collapse
Affiliation(s)
- Hiroko Miyagishi
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, Japan
| | | | | | | |
Collapse
|
183
|
Wiegard A, Hanekamp W, Griessbach K, Fabian J, Lehr M. Pyrrole alkanoic acid derivatives as nuisance inhibitors of microsomal prostaglandin E2 synthase-1. Eur J Med Chem 2012; 48:153-63. [DOI: 10.1016/j.ejmech.2011.12.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 11/29/2011] [Accepted: 12/04/2011] [Indexed: 02/02/2023]
|
184
|
Synthesis and SAR study of imidazoquinolines as a novel structural class of microsomal prostaglandin E2 synthase-1 inhibitors. Bioorg Med Chem Lett 2012; 22:285-8. [DOI: 10.1016/j.bmcl.2011.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 11/04/2011] [Accepted: 11/05/2011] [Indexed: 11/19/2022]
|
185
|
Novel selective COX-1 inhibitors suppress neuroinflammatory mediators in LPS-stimulated N13 microglial cells. Pharmacol Res 2012; 65:137-48. [DOI: 10.1016/j.phrs.2011.09.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 09/26/2011] [Accepted: 09/26/2011] [Indexed: 11/21/2022]
|
186
|
Calcium signaling in cerebral vasoregulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:833-58. [PMID: 22453972 DOI: 10.1007/978-94-007-2888-2_37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The tight coupling of regional neurometabolic activity with synaptic activity and regional cerebral blood perfusion constitutes a single functional unit, described generally as a neurovascular unit. This is central to any discussion of haemodynamic response linked to any neuronal activation. In normal as well as in pathologic conditions, neurons, astrocytes and endothelial cells of the vasculature interact to generate the complex activity-induced cerebral haemodynamic responses, with astrocytes not only partaking in the signaling but actually controlling it in many cases. Neurons and astrocytes have highly integrated signaling mechanisms, yet they form two separate networks. Bidirectional neuron-astrocyte interactions are crucial for the function and survival of the central nervous system. The primary purpose of such regulation is the homeostasis of the brain's microenvironment. In the maintenance of such homeostasis, astrocytic calcium response is a crucial variable in determining neurovascular control. Future work will be directed towards resolving the nature and extent of astrocytic calcium-mediated mechanisms for gene transcription, in modelling neurovascular control, and in determining calcium sensitive imaging assays that can capture disease variables.
Collapse
|
187
|
Menicali E, Moretti S, Voce P, Romagnoli S, Avenia N, Puxeddu E. Intracellular signal transduction and modification of the tumor microenvironment induced by RET/PTCs in papillary thyroid carcinoma. Front Endocrinol (Lausanne) 2012; 3:67. [PMID: 22661970 PMCID: PMC3357465 DOI: 10.3389/fendo.2012.00067] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 04/30/2012] [Indexed: 01/06/2023] Open
Abstract
RET gene rearrangements (RET/PTCs) represent together with BRAF point mutations the two major groups of mutations involved in papillary thyroid carcinoma (PTC) initiation and progression. In this review, we will examine the mechanisms involved in RET/PTC-induced thyroid cell transformation. In detail, we will summarize the data on the molecular mechanisms involved in RET/PTC formation and in its function as a dominant oncogene, on the activated signal transduction pathways and on the induced gene expression modifications. Moreover, we will report on the effects of RET/PTCs on the tumor microenvironment. Finally, a short review of the literature on RET/PTC prognostic significance will be presented.
Collapse
Affiliation(s)
- Elisa Menicali
- Dipartimento di Medicina, University of PerugiaPerugia, Italy
- Centro di Proteomica e Genomica della Tiroide, University of PerugiaPerugia and Terni, Italy
| | - Sonia Moretti
- Dipartimento di Medicina, University of PerugiaPerugia, Italy
- Centro di Proteomica e Genomica della Tiroide, University of PerugiaPerugia and Terni, Italy
| | - Pasquale Voce
- Dipartimento di Medicina, University of PerugiaPerugia, Italy
- Centro di Proteomica e Genomica della Tiroide, University of PerugiaPerugia and Terni, Italy
| | | | - Nicola Avenia
- Centro di Proteomica e Genomica della Tiroide, University of PerugiaPerugia and Terni, Italy
- Dipartimento di Chirurgia, University of PerugiaPerugia, Italy
| | - Efisio Puxeddu
- Dipartimento di Medicina, University of PerugiaPerugia, Italy
- Centro di Proteomica e Genomica della Tiroide, University of PerugiaPerugia and Terni, Italy
- *Correspondence: Efisio Puxeddu, Dipartimento di Medicina, Sezione MIENDO, Via Enrico dal Pozzo – Padiglione X, 06126 Perugia, Italy. e-mail:
| |
Collapse
|
188
|
Abstract
Hsp90 is a highly abundant and ubiquitous molecular chaperone which plays an essential role in many cellular processes including cell cycle control, cell survival, hormone and other signalling pathways. It is important for the cell's response to stress and is a key player in maintaining cellular homeostasis. In the last ten years, it has become a major therapeutic target for cancer, and there has also been increasing interest in it as a therapeutic target in neurodegenerative disorders, and in the development of anti-virals and anti-protozoan infections. The focus of this review is the structural and mechanistic studies which have been performed in order to understand how this important chaperone acts on a wide variety of different proteins (its client proteins) and cellular processes. As with many of the other classes of molecular chaperone, Hsp90 has a critical ATPase activity, and ATP binding and hydrolysis known to modulate the conformational dynamics of the protein. It also uses a host of cochaperones which not only regulate the ATPase activity and conformational dynamics but which also mediate interactions with Hsp90 client proteins. The system is also regulated by post-translational modifications including phosphorylation and acetylation. This review discusses all these aspects of Hsp90 structure and function.
Collapse
|
189
|
Sanchez ER. Chaperoning steroidal physiology: lessons from mouse genetic models of Hsp90 and its cochaperones. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1823:722-9. [PMID: 22155719 DOI: 10.1016/j.bbamcr.2011.11.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 11/17/2011] [Accepted: 11/18/2011] [Indexed: 02/06/2023]
Abstract
The molecular chaperone Hsp90 is abundant, ubiquitous, and catholic to biological processes in eukaryotes, controlling phosphorylation cascades, protein stability and turnover, client localization and trafficking, and ligand-receptor interactions. Not surprisingly, Hsp90 does not accomplish these activities alone. Instead, an ever-growing number of cochaperones have been identified, leading to an explosion of reports on their molecular and cellular effects on Hsp90 chaperoning of client substrates. Notable among these clients are many members of the steroid receptor family, such as glucocorticoid, androgen, estrogen and progesterone receptors. Cochaperones typically associated with the mature, hormone-competent states of these receptors include p23, the FK506-binding protein 52 (FKBP52), FKBP51, protein phosphatase 5 (PP5) and cyclophilin 40 (Cyp40). The ultimate relevance of these cochaperones to steroid receptor action depends on their physiological effects. In recent years, the first mouse genetic models of these cochaperones have been developed. This work will review the complex and intriguing phenotypes so far obtained in genetically-altered mice and compare them to the known molecular and cellular impacts of cochaperones on steroid receptors. This article is part of a Special Issue entitled: Heat Shock Protein 90 (HSP90).
Collapse
Affiliation(s)
- Edwin R Sanchez
- Department of Physiologyand Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA.
| |
Collapse
|
190
|
Abstract
Many lines of evidence demonstrate that prostaglandins play an important role in cancer, and enhanced synthesis of prostaglandin E(2) (PGE(2)) is often observed in various human malignancies often associated with poor prognosis. PGE(2) synthesis is initiated with the release of arachidonic acid by phospholipase enzymes, where it is then converted into the intermediate prostaglandin prostaglandin H(2) (PGH(2)) by members of the cyclooxygenase family. The synthesis of PGE(2) from PGH(2) is facilitated by three different PGE synthases, and functional PGE(2) can promote tumor growth by binding to four EP receptors to activate signaling pathways that control cell proliferation, migration, apoptosis, and angiogenesis. An integral method of controlling gene expression is by posttranscriptional mechanisms that regulate mRNA stability and protein translation. Messenger RNA regulatory elements typically reside within the 3' untranslated region (3'UTR) of the transcript and play a critical role in targeting specific mRNAs for posttranscriptional regulation through microRNA (miRNA) binding and adenylate- and uridylate-rich element RNA-binding proteins. In this review, we highlight the current advances in our understanding of the impact these RNA sequence elements have upon regulating PGE(2) levels. We also identify various RNA sequence elements consistently observed within the 3'UTRs of the genes involved in the PGE(2) pathway, indicating these binding sites for miRNAs and RNA-binding proteins to be central regulators of PGE(2) synthesis and function. These findings may provide a rationale for the development of new therapeutic approaches to control tumor growth and metastasis promoted by elevated PGE(2) levels.
Collapse
Affiliation(s)
- Ashleigh E. Moore
- Department of Biological Sciences and Center for Colon Cancer Research, University of South Carolina, Columbia, SC, USA
| | - Lisa E. Young
- Novartis Institutes for Biomedical Research, RNAi Therapeutics, Cambridge, MA, USA
| | - Dan A. Dixon
- Department of Biological Sciences and Center for Colon Cancer Research, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
191
|
Smith WL, Urade Y, Jakobsson PJ. Enzymes of the cyclooxygenase pathways of prostanoid biosynthesis. Chem Rev 2011; 111:5821-65. [PMID: 21942677 PMCID: PMC3285496 DOI: 10.1021/cr2002992] [Citation(s) in RCA: 355] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- William L Smith
- Department of Biological Chemistry, University of Michigan Medical School, 1150 West Medical Center Drive, 5301 MSRB III, Ann Arbor, Michigan 48109-5606, USA.
| | | | | |
Collapse
|
192
|
Spahiu L, Stenberg P, Larsson C, Wannberg J, Alterman M, Kull B, Nekhotiaeva N, Morgenstern R. A Facilitated Approach to Evaluate the Inhibitor Mode and Potency of Compounds Targeting Microsomal Prostaglandin E Synthase-1. Assay Drug Dev Technol 2011; 9:487-95. [DOI: 10.1089/adt.2010.0350] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | - Ralf Morgenstern
- Actar AB, Solna, Sweden
- NovaSAID AB, Solna, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
193
|
Inoue N, Nogawa M, Ito S, Tajima K, Kume S, Kyoi T. The enantiomers of etodolac, a racemic anti-inflammatory agent, play different roles in efficacy and gastrointestinal safety. Biol Pharm Bull 2011; 34:655-9. [PMID: 21532152 DOI: 10.1248/bpb.34.655] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The anti-inflammatory agent etodolac is used worldwide and it has a good gastrointestinal safety profile. Etodolac consists of two enantiomers, S- and R-etodolac. Here, we investigated the beneficial activities of racemic etodolac and its enantiomers. First, we compared S- and R-etodolac in terms of their inhibition of cyclooxygenase (COX) activity in vitro and their suppression of paw swelling in adjuvant-induced arthritic rats. The COX-2 inhibitory and anti-inflammatory effects of etodolac were found to be due to the S-enantiomer. We previously reported that etodolac attenuates allodynia in a mouse model of neuropathic pain by a COX-2-independent mechanism [N. Inoue et al., J. Pharmacol. Sci., 109, 600-605 (2009)]. In the present study, we showed that the anti-allodynic effects of etodolac in mice were also due to the S-enantiomer. In addition, we investigated the ulcerogenic activity of racemic etodolac and its enantiomers. At high doses, racemic etodolac showed a lower gastric lesion index in rats than the equivalent dose of S-etodolac. In contrast, R-etodolac showed no ulcerogenic activity and even showed protection against HCl/ethanol-induced gastric damage in rats. In conclusion, S-etodolac exhibited anti-inflammatory effects mediated by COX-2 inhibition and anti-allodynic effects that were independent of COX-2 inhibition, while R-etodolac showed gastroprotective effects that may contribute to the low gastrointestinal toxicity of racemic etodolac. Our results show that each enantiomer plays a different role in the efficacy and gastrointestinal safety of etodolac.
Collapse
Affiliation(s)
- Naoki Inoue
- Pharmacology Department, Discovery Research Laboratories, Nippon Shinyaku Co., Ltd. Kisshoin, Minami-ku, Kyoto 601–8550, Japan.
| | | | | | | | | | | |
Collapse
|
194
|
Chang HH, Song Z, Wisner L, Tripp T, Gokhale V, Meuillet EJ. Identification of a novel class of anti-inflammatory compounds with anti-tumor activity in colorectal and lung cancers. Invest New Drugs 2011; 30:1865-77. [PMID: 21931968 DOI: 10.1007/s10637-011-9748-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 08/30/2011] [Indexed: 12/13/2022]
Abstract
Chronic inflammation is associated with 25% of all cancers. In the inflammation-cancer axis, prostaglandin E(2) (PGE(2)) is one of the major players. PGE(2) synthases (PGES) are the enzymes downstream of the cyclooxygenases (COXs) in the PGE(2) biosynthesis pathway. Microsomal prostaglandin E(2) synthase 1 (mPGES-1) is inducible by pro-inflammatory stimuli and constitutively expressed in a variety of cancers. The potential role for this enzyme in tumorigenesis has been reported and mPGES-1 represents a novel therapeutic target for cancers. In order to identify novel small molecule inhibitors of mPGES-1, we screened the ChemBridge library and identified 13 compounds as potential hits. These compounds were tested for their ability to bind directly to the enzyme using surface plasmon resonance spectroscopy and to decrease cytokine-stimulated PGE(2) production in various cancer cell lines. We demonstrate that the compound PGE0001 (ChemBridge ID number 5654455) binds to human mPGES-1 recombinant protein with good affinity (K(D) = 21.3 ± 7.8 μM). PGE0001 reduces IL-1β-induced PGE(2) release in human HCA-7 colon and A549 lung cancer cell lines with EC(50) in the sub-micromolar range. Although PGE0001 may have alternative targets based on the results from in vitro assays, it shows promising effects in vivo. PGE0001 exhibits significant anti-tumor activity in SW837 rectum and A549 lung cancer xenografts in SCID mice. Single injection i.p. of PGE0001 at 100 mg/kg decreases serum PGE(2) levels in mice within 5 h. In summary, our data suggest that the identified compound PGE0001 exerts anti-tumor activity via the inhibition of the PGE(2) synthesis pathway.
Collapse
Affiliation(s)
- Hui-Hua Chang
- Arizona Cancer Center, The University of Arizona, Tucson, AZ 85724, USA
| | | | | | | | | | | |
Collapse
|
195
|
Inflammatory effect of advanced glycation end products on human meniscal cells from osteoarthritic knees. Inflamm Res 2011; 60:1039-48. [DOI: 10.1007/s00011-011-0365-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 07/21/2011] [Accepted: 07/23/2011] [Indexed: 02/07/2023] Open
|
196
|
Management of cytoskeleton architecture by molecular chaperones and immunophilins. Cell Signal 2011; 23:1907-20. [PMID: 21864675 DOI: 10.1016/j.cellsig.2011.07.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/22/2011] [Accepted: 07/26/2011] [Indexed: 11/20/2022]
Abstract
Cytoskeletal structure is continually remodeled to accommodate normal cell growth and to respond to pathophysiological cues. As a consequence, several cytoskeleton-interacting proteins become involved in a variety of cellular processes such as cell growth and division, cell movement, vesicle transportation, cellular organelle location and function, localization and distribution of membrane receptors, and cell-cell communication. Molecular chaperones and immunophilins are counted among the most important proteins that interact closely with the cytoskeleton network, in particular with microtubules and microtubule-associated factors. In several situations, heat-shock proteins and immunophilins work together as a functionally active heterocomplex, although both types of proteins also show independent actions. In circumstances where homeostasis is affected by environmental stresses or due to genetic alterations, chaperone proteins help to stabilize the system. Molecular chaperones facilitate the assembly, disassembly and/or folding/refolding of cytoskeletal proteins, so they prevent aberrant protein aggregation. Nonetheless, the roles of heat-shock proteins and immunophilins are not only limited to solve abnormal situations, but they also have an active participation during the normal differentiation process of the cell and are key factors for many structural and functional rearrangements during this course of action. Cytoskeleton modifications leading to altered localization of nuclear factors may result in loss- or gain-of-function of such factors, which affects the cell cycle and cell development. Therefore, cytoskeletal components are attractive therapeutic targets, particularly microtubules, to prevent pathological situations such as rapidly dividing tumor cells or to favor the process of cell differentiation in other cases. In this review we will address some classical and novel aspects of key regulatory functions of heat-shock proteins and immunophilins as housekeeping factors of the cytoskeletal network.
Collapse
|
197
|
Prage EB, Pawelzik SC, Busenlehner LS, Kim K, Morgenstern R, Jakobsson PJ, Armstrong RN. Location of inhibitor binding sites in the human inducible prostaglandin E synthase, MPGES1. Biochemistry 2011; 50:7684-93. [PMID: 21805999 DOI: 10.1021/bi2010448] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The inducible microsomal prostaglandin E(2) synthase 1 (MPGES1) is an integral membrane protein coexpressed with and functionally coupled to cyclooxygenase 2 (COX-2) generating the pro-inflammatory molecule PGE(2). The development of effective inhibitors of MPGES1 holds promise as a highly selective route for controlling inflammation. In this paper, we describe the use of backbone amide H/D exchange mass spectrometry to map the binding sites of different types of inhibitors of MPGES1. The results reveal the locations of specific inhibitor binding sites that include the GSH binding site and a hydrophobic cleft in the protein thought to accommodate the prostaglandin H(2) substrate. In the absence of three-dimensional crystal structures of the enzyme-bound inhibitors, the results provide clear physical evidence that three pharmacologically active inhibitors bind in a hydrophobic cleft composed of sections of transmembrane helices Ia, IIb, IIIb, and IVb at the interface of subunits in the trimer. In principle, the H/D exchange behavior of the protein can be used as a preliminary guide for optimization of inhibitor efficacy. Finally, a comparison of the structures and H/D exchange behavior of MPGES1 and the related enzyme MGST1 in the presence of glutathione and the inhibitor glutathione sulfonate confirms the unusual observation that two proteins from the same superfamily harbor GSH binding sites in different locations.
Collapse
Affiliation(s)
- Edward B Prage
- Departments of Chemistry and Biochemistry, Center in Molecular Toxicology, and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232-0146, United States
| | | | | | | | | | | | | |
Collapse
|
198
|
Ugajin T, Satoh T, Kanamori T, Aritake K, Urade Y, Yokozeki H. FcεRI, but not FcγR, signals induce prostaglandin D2 and E2 production from basophils. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:775-82. [PMID: 21712025 DOI: 10.1016/j.ajpath.2011.04.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 03/21/2011] [Accepted: 04/25/2011] [Indexed: 10/18/2022]
Abstract
Prostaglandin (PG) D2 and PGE2 are arachidonic acid metabolites that are generated though an isomerization reaction catalyzed by PG synthases. PGs have been implicated in immunologic reactions in addition to a wide range of physiological functions. It has long been thought that basophils, in contrast to mast cells, do not synthesize PGs, although they do release leukotrienes and platelet-activating factor. Here, we show that basophils function as a source of PGD2 and PGE2. In vitro-cultured basophils from mouse bone marrow produced both PGD2 and PGE2 in response to IgE + antigen (Ag), but not to IgG + Ag. Release of PGs was almost completely abrogated in cultured basophils from FcRγ-chain(-/-) mice, indicating the involvement of FcεRI. Basophils freshly isolated from bone marrow cells (primary basophils) were also capable of secreting PGD2 and PGE2. Although the amount of PGD2 released from primary basophils was lower than that from mast cells, the capability of primary basophils to generate PGE2 was more potent than that of mast cells. Transcripts and proteins for both hematopoietic-type PGD synthase and PGE synthase were detected in basophils. In addition, human basophils, like mouse basophils, also produced PGD2 through IgE-mediated stimulation. Thus, basophils could be an important source of PGD2/PGE2 and may contribute to allergic inflammation and immune responses.
Collapse
Affiliation(s)
- Tsukasa Ugajin
- Department of Dermatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
199
|
Abstract
Prostaglandins are lipid autacoids derived from arachidonic acid. They both sustain homeostatic functions and mediate pathogenic mechanisms, including the inflammatory response. They are generated from arachidonate by the action of cyclooxygenase isoenzymes, and their biosynthesis is blocked by nonsteroidal antiinflammatory drugs, including those selective for inhibition of cyclooxygenase-2. Despite the clinical efficacy of nonsteroidal antiinflammatory drugs, prostaglandins may function in both the promotion and resolution of inflammation. This review summarizes insights into the mechanisms of prostaglandin generation and the roles of individual mediators and their receptors in modulating the inflammatory response. Prostaglandin biology has potential clinical relevance for atherosclerosis, the response to vascular injury and aortic aneurysm.
Collapse
Affiliation(s)
- Emanuela Ricciotti
- Institute for Translational Medicine and Therapeutics, 153 Johnson Pavilion, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
200
|
Norris PC, Reichart D, Dumlao DS, Glass CK, Dennis EA. Specificity of eicosanoid production depends on the TLR-4-stimulated macrophage phenotype. J Leukoc Biol 2011; 90:563-74. [PMID: 21653236 DOI: 10.1189/jlb.0311153] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Eicosanoid metabolism differs in profile and quantity between macrophages of different tissue origin and method of elicitation, as well as between primary and immortalized macrophages after activation with inflammatory stimuli. Using a lipidomic approach, we comprehensively analyzed the eicosanoids made by murine RPMs, TGEMs, BMDM, and the macrophage-like cell line RAW after stimulation with the TLR-4-specific agonist KLA. Direct correlation among total COX metabolites, COX side-products (11-HETE, 15-HETE), COX-2 mRNA, and protein at 8 h was found when comparing each cell type. Comprehensive qPCR analysis was used to compare relative transcript levels between the terminal prostanoid synthases themselves as well as between each cell type. Levels of PGE(2), PGD(2), and TxB(2) generally correlated with enzyme transcript expression of PGES, PGDS, and TBXS, providing evidence of comparable enzyme activities. PGIS transcript was expressed only in RPM and TGEM macrophages and at an exceptionally low level, despite high metabolite production compared with other synthases. Presence of PGIS in RPM and TGEM also lowered the production of PGE(2) versus PGD(2) by approximately tenfold relative to BMDM and RAW cells, which lacked this enzyme. Our results demonstrate that delayed PG production depends on the maximal level of COX-2 expression in different macrophages after TLR-4 stimulation. Also, the same enzymes in each cell largely dictate the profile of eicosanoids produced depending on the ratios of expression between them, with the exception of PGIS, which appears to have much greater synthetic capacity and competes selectively with mPGES-1.
Collapse
Affiliation(s)
- Paul C Norris
- Department of Chemistry, University of California, San Diego, La Jolla, CA 92093-0601, USA
| | | | | | | | | |
Collapse
|