Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 920: 174863, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35240193

ABSTRACT

Chronic inflammatory bowel disease (IBD), which is characterized by prolonged inflammation of the gastrointestinal tract is associated with an increased risk of colorectal cancer. Recent studies revealed that the pathology of IBD is caused by hyperactivated immune responses mediated by differentiated CD4+ naïve helper T cells, such as Th1 and Th17 cells, but not Th2 cells. The human E-type prostanoid 4 (EP4) receptor and its pathways have also been implicated in and/or associated with the early developmental stages of colorectal cancer along with increases in the levels of prostaglandin E2 (PGE2) and cyclooxygenase-2 (COX-2), the hallmarks of colorectal carcinogenesis. In the present study, using an in silico analysis and pharmacological experiments, we demonstrated that interleukin (IL)-4, a signature cytokine of Th2 cells, down-regulated the expression of COX-2 and PGE2 in the human colon cancer cell line, HCA-7. This result may be attributed to a reduction in the expression of prostanoid EP4 receptors through the induction of hypoxia inducible factor-1α via the interleukin-4 receptor-stimulated activation of signal transducer and activator of transcription 6. However, another major Th2 cytokine IL-13 had no effect on the expression of COX-2 or prostanoid EP4 receptors in HCA-7 cells. Therefore, instead of the hyperactivation of Th1/Th17 cells, the deactivation/down-regulation of Th2 cells followed by a decrease in the production of IL-4 in IBD may play a role in the cancerous transformation of cells, at least in prostanoid EP4 receptor-overactivated tumorigenesis.


Subject(s)
Colonic Neoplasms , Interleukin-4 , Colonic Neoplasms/pathology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Down-Regulation , Humans , Interleukin-4/metabolism , Interleukin-4/pharmacology , Prostaglandins E , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism
2.
FEBS Open Bio ; 12(4): 775-783, 2022 04.
Article in English | MEDLINE | ID: mdl-35124898

ABSTRACT

EP2 and EP4 prostanoid receptors have long been considered to have similar roles, since they are known to couple with Gαs-protein and activate cAMP-mediated signaling pathways. In this study, we re-evaluated the results of cAMP assays with or without phosphodiesterase (PDE) inhibitor pretreatment. Here, we show that in the absence of PDE inhibitor pretreatment, prostaglandin E2 causes accumulation of cAMP in EP2 receptors, whereas markedly low levels of cAMP accumulated in EP4 receptors. By applying the Black/Leff operational model calculation, we found that EP2 receptors have a biased ability to intrinsically activate the Gαs-protein-mediated pathway, whereas EP4 receptors have strong biased activity for the Gαi-protein-mediated pathway. Thus, EP2 and EP4 receptors may not be similar Gαs-coupled receptors but instead substantially different receptors with distinct roles.


Subject(s)
Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Prostaglandins , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction/physiology
3.
Pharmacol Rep ; 73(3): 946-953, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33786738

ABSTRACT

BACKGROUND: Human DP and EP2 receptors are two of the most homologically related receptors coupling with Gαs-protein, which stimulate adenylyl cyclase to produce cAMP. Indeed, both receptors are considered to be generated by tandem duplication. It has been reported that other highly homologous and closely related ß1- and ß2-adrenergic receptors interact distinctly with and differentially regulate cAMP-specific phosphodiesterase (PDE) 4 recruitment. METHODS: First, we focused on the cAMP degradation pathways of DP and EP2 receptors stimulated by prostaglandin (PG) D2 or PGE2 using HEK cells stably expressing either human DP receptors or EP2 receptors. Then, distances between ligands and amino acids of the receptors were evaluated by molecular dynamics (MD) analysis. RESULTS: We found that PGD2/EP2 receptors exerted a greater effect on PDE4 activity than PGE2/EP2 receptors. Moreover, by MD analysis, either the PGD2 or EP2 receptor was moved and the distance was shortened between them. According to the results, DP receptors retain reactivity for PGE2, but EP2 receptors may be activated only by PGE2, at least in terms of cAMP formation, through the differential functional coupling of PDE4 probably with ß-arrestin. CONCLUSION: Since DP receptors and EP2 receptors are considered to be duplicated genes, DP receptors may still be in a rapid evolutionary stage as a duplicated copy of EP2 receptors and have not yet sufficient selectivity for their cognate ligand, PGD2.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dinoprostone/metabolism , Prostaglandin D2/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Adenylyl Cyclases/metabolism , Cell Line , Cyclic AMP/metabolism , HEK293 Cells , Humans , Ligands , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Signal Transduction/physiology
4.
J Biol Chem ; 295(38): 13338-13352, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32727851

ABSTRACT

Prostaglandin E2 (PGE2) is well-known as an endogenous proinflammatory prostanoid synthesized from arachidonic acid by the activation of cyclooxygenase-2. E type prostanoid (EP) receptors are cognates for PGE2 that have four main subtypes: EP1 to EP4. Of these, the EP2 and EP4 prostanoid receptors have been shown to couple to Gαs-protein and can activate adenylyl cyclase to form cAMP. Studies suggest that EP4 receptors are involved in colorectal homeostasis and cancer development, but further work is needed to identify the roles of EP2 receptors in these functions. After sufficient inflammation has been evoked by PGE2, it is metabolized to 15-keto-PGE2 Thus, 15-keto-PGE2 has long been considered an inactive metabolite of PGE2 However, it may have an additional role as a biased and/or partial agonist capable of taking over the actions of PGE2 to gradually terminate reactions. Here, using cell-based experiments and in silico simulations, we show that PGE2-activated EP4 receptor-mediated signaling may evoke the primary initiating reaction of the cells, which would take over the 15-keto-PGE2-activated EP2 receptor-mediated signaling after PGE2 is metabolized to 15-keto-PGE2 The present results shed light on new aspects of 15-keto-PGE2, which may have important roles in passing on activities to EP2 receptors from PGE2-stimulated EP4 receptors as a "switched agonist." This novel mechanism may be significant for gradually terminating PGE2-evoked inflammation and/or maintaining homeostasis of colorectal tissues/cells functions.


Subject(s)
Computer Simulation , Dinoprostone/analogs & derivatives , Models, Biological , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Signal Transduction , Dinoprostone/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Receptors, Prostaglandin E, EP4 Subtype/metabolism
5.
Eur J Pharmacol ; 853: 308-315, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30980797

ABSTRACT

Microbiota produce short chain fatty acids (SCFAs), which are known to maintain gut homeostasis, by the fermentation of dietary fiber in the human colon. Among SCFAs, butyrate has been considered as the most physiologically effective SCFA in colorectal epithelial cells for growth and differentiation. Here we show that the E-type prostanoid 4 (EP4) receptor expression level is regulated by different concentrations of butyrate, but not by other SCFAs, in human colon cancer HCA-7 cells, through sodium-coupled monocarboxylate transporter-1 (SMCT-1)-mediated uptake followed by the activation of histone acetyltransferase: cAMP response element binding protein-binding protein/p300. Of particular interest, the prostanoid EP4 receptors are known to be expressed in normal colorectal crypt epithelial cells and maintain intestinal homeostasis by preserving mucosal integrity, while they are also known to be involved in the early stage of carcinogenesis. Thus, the links between butyrate and the expression of prostanoid EP4 receptors are both important factors for maintaining homeostasis. Based on in silico analysis, almost half of colorectal cancer tissues have lost the expression of SMCT-1 mRNA when compared with healthy corresponding tissues. Therefore, with the collapse of homeostasis systems such as a decrease in the concentration of butyrate in colorectal tissues, or reduced butyrate uptake, there is a possibility of early stage colorectal cancer development; the transformation of normal cells to the cancerous phenotype may be due to the overexpression of prostanoid EP4 receptors followed by excessive cyclooxygenase-2 induction, which are caused by a reduced amount of butyrate and/or its uptake, in/around colorectal epithelial cells.


Subject(s)
Butyrates/metabolism , Colonic Neoplasms/pathology , Cyclooxygenase 2/biosynthesis , Gene Expression Regulation, Neoplastic , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Acetylation , Animals , Cell Line, Tumor , Cell Proliferation , Cyclic AMP/biosynthesis , E1A-Associated p300 Protein/metabolism , Enzyme Induction , Histone Acetyltransferases/metabolism , Histones/metabolism , Humans , Monocarboxylic Acid Transporters/metabolism , Peptide Fragments/metabolism , Sialoglycoproteins/metabolism
6.
Pharmacol Res Perspect ; 6(6): e00441, 2018 12.
Article in English | MEDLINE | ID: mdl-30455960

ABSTRACT

The up-regulated expression of E-type prostanoid (EP) 4 receptors has been implicated in carcinogenesis; however, the expression of EP4 receptors has also been reported to be weaker in tumor tissues than in normal tissues. Indeed, EP4 receptors have been suggested to play a role in the maintenance of colorectal homeostasis. This study aimed to examine the underlying mechanisms/reasons for why inconsistent findings have been reported regarding EP4 receptor expression levels in homeostasis and carcinogenesis by focusing on cellular densities. Thus, the human colon cancer HCA-7 cells, which retain some functional features of normal epithelia, and luciferase reporter genes containing wild-type or mutated EP4 receptor promoters were used for elucidating the cellular density-dependent mechanisms about the regulation of EP4 receptor expression. In silico analysis was also utilized for confirming the relevance of the findings with respect to colon cancer development. We here demonstrated that the expression of EP4 receptors was up-regulated by c-Myc by binding to Sp-1 under low cellular density conditions, but was down-regulated under high cellular density conditions via the increase in the expression levels of HIF-1α protein, which may pull out c-Myc and Sp-1 from DNA-binding. The tightly regulated EP4 receptor expression mechanism may be a critical system for maintaining homeostasis in normal colorectal epithelial cells. Therefore, once the system is altered, possibly due to the transient overexpression of EP4 receptors, it may result in aberrant cellular proliferation and transformation to cancerous phenotypes. However, at the point, EP4 receptors themselves and their mediated homeostasis would be no longer required.


Subject(s)
Colonic Neoplasms/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Carcinogenesis/genetics , Cell Count , Cell Line, Tumor , Colonic Neoplasms/pathology , Computational Biology , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sp1 Transcription Factor/metabolism , Up-Regulation
7.
FEBS Lett ; 591(22): 3771-3780, 2017 11.
Article in English | MEDLINE | ID: mdl-28986997

ABSTRACT

The 2-series of prostaglandin E (PGE2 ) is regarded as a pro-cancer prostanoid, whereas the 1-series (PGE1 ) and the 3-series (PGE3 ) are considered to act as anti-cancer prostanoids. In the present study, we provide possible reasons why PGE1 and PGE3 , but not PGE2 , exert anti-cancer effects by focusing on each diverged E-type prostanoid (EP)4 receptor-mediated signaling pathway. PGE1 , PGE2 and PGE3 function as full agonists in terms of Gαs - and Gαi -protein-mediated signaling. However, PGE1 and PGE3 function as partial agonists of T-cell factor (TCF)/ß-catenin (ß-cat)-mediated activity, the well-known cancer-related signaling pathway. Furthermore, pretreatment with PGE1 or PGE3 almost completely reduces PGE2 -induced TCF/ß-cat activity. These results provide a plausible reason why PGE1 and PGE3 function as anti-cancer prostanoids as a result of novel biased activity for EP4 receptors.


Subject(s)
Alprostadil/analogs & derivatives , Alprostadil/metabolism , Dinoprostone/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , beta Catenin/metabolism , Binding Sites , Crystallography, X-Ray , Gene Expression Regulation , HEK293 Cells , Humans , Hydrogen Bonding , Ligands , Models, Molecular , Protein Binding , Signal Transduction
8.
Biol Pharm Bull ; 40(10): 1806-1812, 2017.
Article in English | MEDLINE | ID: mdl-28966256

ABSTRACT

Increases in the expression of prostaglandin E2 (PGE2) are widely known to be involved in aberrant growth in the early stage of colon cancer development. We herein demonstrated that the novel indole compound MW-03 reduced PGE2-induced cAMP formation by catalization to an inactive metabolite by inducing 15-hydroxyprostaglandin dehydrogenase through the activation of peroxisome proliferator-activated receptor-γ. MW-03 also inhibited colon cancer cell growth by arresting the cell cycle at the S phase. Although the target of MW-03 for cell cycle inhibition has not yet been identified, these dual anti-cancer effects of MW-03 itself and/or its leading compound(s) on colon cancer cells may reduce colon cancer development and, thus, have potential as a novel treatment for the early stage of this disease.


Subject(s)
Antineoplastic Agents/pharmacology , Hydroxyprostaglandin Dehydrogenases/metabolism , Indoles/pharmacology , PPAR gamma/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Cyclic AMP/metabolism , Dinoprostone/pharmacology , Humans
9.
FEBS J ; 283(21): 3931-3940, 2016 11.
Article in English | MEDLINE | ID: mdl-27636113

ABSTRACT

Human D-type prostanoid (DP) and E-type prostanoid 2 (EP2) receptors are G protein-coupled receptors and are regarded as the most closely related receptors among prostanoid receptors because they are generated by tandem duplication. The DP receptor-cognate ligand, prostaglandin D2 (PGD2 ) has the ability to activate not only DP receptors but also EP2 receptors. Likewise, the EP2 receptor-cognate ligand, prostaglandin E2 (PGE2 ) has the ability to activate DP receptors in addition to EP receptors in order to stimulate cAMP formation. However, since PGD2 and/or PGE2 activate DP and EP2 receptors to similar maximal levels, that is, their similar efficacies, differences between the ligands in each receptor have not yet been determined in detail except for their different affinities. Herein we demonstrated, using an in silico simulation to predict binding patterns among DP or EP2 receptors and PGD2 , PGE2 , or prostaglandin F2α as the reference prostanoid, that DP and EP2 receptors plausibly take on distinct forms depending on the diverse binding of different ligands. Since these ligands have the potential to make these receptors form distinct conformations with discrete signaling pathways, they are consequently regarded as endogenous biased ligands. Moreover, by using functional assays, the susceptibilities of the DP receptors to the noncognate ligands were approximately 10 times lower than those of EP2 receptors. Thus, EP2 receptors seem to be able to distinguish endogenous ligands better than DP receptors, thereby both receptors are plausibly gaining role-sharing functions with respect to one another as the copies of duplicated gene.


Subject(s)
Dinoprostone/metabolism , Prostaglandin D2/metabolism , Receptors, Immunologic/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin/metabolism , Amino Acid Sequence , Binding, Competitive , Computer Simulation , Cyclic AMP/metabolism , Dinoprost/chemistry , Dinoprost/metabolism , Dinoprostone/chemistry , HEK293 Cells , Humans , Ligands , Models, Molecular , Molecular Structure , Prostaglandin D2/chemistry , Protein Domains , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics , Receptors, Prostaglandin/chemistry , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin E, EP2 Subtype/chemistry , Receptors, Prostaglandin E, EP2 Subtype/genetics , Sequence Homology, Amino Acid
10.
Eur J Pharmacol ; 768: 149-59, 2015 Dec 05.
Article in English | MEDLINE | ID: mdl-26518053

ABSTRACT

Approximately two decades have passed since E-type prostanoid 4 (EP4) receptors were cloned, and the signaling pathways mediated by these receptors have since been implicated in cancer development through the alliance of Gαi-protein/phosphatidylinositol 3-kinase (PI3K)/extracellular signal-regulated kinases (ERKs) activation. Although prostanoid EP4 receptors were initially identified as Gαs-coupled receptors, the specific/distinctive role(s) of prostanoid EP4 receptor-induced cAMP/protein kinase A (PKA) pathways in cancer development have not yet been elucidated in detail. We previously reported using HCA-7 human colon cancer cells that prostaglandin E2 (PGE2)-stimulated prostanoid EP4 receptors induced cyclooxygenase-2 (COX-2) as an initiating event in development of colon cancer. Moreover, this induction of COX-2 was mediated by transactivation of epidermal growth factor (EGF) receptors. However, direct activation of EGF receptors by EGF also induced similar amounts of COX-2 in this cell line. Thus, the emergence of unique role(s) for prostanoid EP4 receptors is expected by clarifying the different signaling mechanisms between PGE2-stimulated prostanoid EP4 receptors and EGF-stimulated EGF receptors to induce COX-2 and produce PGE2. We here demonstrated that prostanoid EP4 receptor activation by PGE2 in HCA-7 cells led to PKA-dependent re-activation of ERKs, which resulted in prolonged de novo synthesis of PGE2. Although EGF-stimulated EGF receptors in cells also induced COX-2 and the de novo synthesis of PGE2, the activation of this pathway was transient and not mediated by PKA. Therefore, the novel mechanism underlying prolonged de novo synthesis of PGE2 has provided an insight into the importance of prostanoid EP4 receptor-mediated Gαs-protein/cAMP/PKA pathway in development of colon cancer.


Subject(s)
Colonic Neoplasms/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/biosynthesis , Dinoprostone/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Androstadienes/pharmacology , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclooxygenase 2/biosynthesis , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Humans , Isoquinolines/pharmacology , Phosphorylation/drug effects , Sulfonamides/pharmacology , Time Factors , Wortmannin
11.
Pharmacol Res Perspect ; 3(1): e00083, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25692008

ABSTRACT

Increases in prostaglandin E2 (PGE2) and cyclooxygenase-2 (COX-2) levels are features of colon cancer. Among the different E-type prostanoid receptor subtypes, EP4 receptors are considered to play a crucial role in carcinogenesis by, for example, inducing COX-2 when stimulated with PGE2. However, EP4 receptor levels and PGE2-induced cellular responses are inconsistent among the cellular conditions. Therefore, the connections responsible for the expression of EP4 receptors were investigated in the present study by focusing on cell density-induced hypoxia-inducible factor-1α (HIF-1α). The expression of EP4 receptors was examined using immunoblot analysis, quantitative polymerase chain reaction, and reporter gene assays in HCA-7 human colon cancer cells with different cellular densities. The involvement of HIF-1α and its signaling pathways were also examined by immunoblot analysis, reporter gene assays, and with siRNA. We here demonstrated that EP4 receptors as well as EP4 receptor-mediated COX-2 expression levels decreased with an increase in cellular density. In contrast, HIF-1α levels increased in a cellular density-dependent manner. The knockdown of HIF-1α by siRNA restored the expression of EP4 receptors and EP4 receptor-mediated COX-2 in cells at a high density. Thus, the cellular density-dependent increase observed in HIF-1α expression levels reduced the expression of COX-2 by decreasing EP4 receptor levels. This novel regulation mechanism for the expression of EP4 receptors by HIF-1α may provide an explanation for the inconsistent actions of PGE2. The expression levels of EP4 receptors may vary depending on cellular density, which may lead to the differential activation of their signaling pathways by PGE2. Thus, cellular density-dependent PGE2-mediated signaling may determine the fate/stage of cancer cells, i.e., the surrounding environments could define the fate/stage of malignancies associated with colon cancer.

12.
Arch Biochem Biophys ; 541: 21-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24239863

ABSTRACT

Indoles are composed of a common core structure, the indole ring, and are widely used as pharmaceuticals and their precursors. In this study, a newly composed relatively small indole compound, AWT-489 was examined to find a novel specific antagonist for DP receptors; the cognate receptors for prostaglandin D2 (PGD2), to prevent colon cancer malignancy. Here we showed that AWT-489 antagonized DP receptor-mediated cyclic AMP formation, and expression of CD55, an inhibitor of the complement system that correlates with poor survival in patients with colorectal cancer, in LS174T human colon cancer cells. Interestingly, unlike a popular indole compound, indomethacin, AWT-489 did not act on the cyclooxygenases as a non-steroidal anti-inflammatory drug. Moreover, AWT-489 exhibited a better inhibitory effect than that of the well-used DP receptor antagonist, BWA868C when a dose close to the physiological concentration of PGD2 was used. These results suggest that AWT-489 can act as a novel human DP receptor antagonist to reduce the expression of CD55 in LS174T human colon cancer cells. We believe that AWT-489 has potential as a lead compound for designing a new DP receptor antagonist that may help improve PGD2-related diseases, especially colon cancer in the near future.


Subject(s)
CD55 Antigens/genetics , CD55 Antigens/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Indoles/pharmacology , Prostaglandin D2/pharmacology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Carcinogenesis/drug effects , Cell Line, Tumor , Cyclic AMP/biosynthesis , Dinoprostone/pharmacology , Drug Design , Humans , Hydantoins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Eur J Pharmacol ; 718(1-3): 408-17, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23973650

ABSTRACT

Increased expressions of cyclooxygenase-2 (COX-2) and its downstream metabolite, prostaglandin E2 (PGE2), are well documented events in the development of colorectal cancer. Interestingly, PGE2 itself can induce the expression of COX-2 thereby creating the potential for positive feedback. Although evidence for such a positive feedback has been previously described, the specific E-type prostanoid (EP) receptor subtype that mediates this response, as well as the relevant signaling pathways, remain unclear. We now report that the PGE2 stimulated induction of COX-2 expression in human colon cancer HCA-7 cells is mediated by activation of the prostanoid EP4 receptor subtype and is followed by coupling of the receptor to Gαi and the activation of phosphatidylinositol 3-kinase. Subsequent activation of metalloproteinases releases membrane bound heparin-binding epidermal growth factor-like growth factor resulting in the transactivation of epidermal growth factor receptors and the activation of the extracellular signal-regulated kinases and induction of COX-2 expression. This induction of COX-2 expression by PGE2 stimulation of the prostanoid EP4 receptor may underlie the upregulation of COX-2 during colorectal cancer and appears to be an early event in the process of tumorigenesis.


Subject(s)
Colonic Neoplasms/pathology , Cyclooxygenase 2/metabolism , Dinoprostone/pharmacology , ErbB Receptors/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Transcriptional Activation/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Enzyme Activation/drug effects , ErbB Receptors/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Metalloproteases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Tyrosine/metabolism
14.
J Nutr Biochem ; 22(7): 681-90, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21036588

ABSTRACT

Lipoic acid (LA) is a naturally occurring fatty acid that exhibits anti-oxidant and anti-inflammatory properties and is being pursued as a therapeutic for many diseases including multiple sclerosis, diabetic polyneuropathy and Alzheimer's disease. We previously reported on the novel finding that racemic LA (50:50 mixture of R-LA and S-LA) stimulates cAMP production, activates prostanoid EP2 and EP4 receptors and adenylyl cyclases (AC), and suppresses activation and cytotoxicity in NK cells. In this study, we present evidence that furthers our understanding of the mechanisms of action of LA. Using various LA derivatives, such as dihydrolipoic acid (DHLA), S,S-dimethyl lipoic acid (DMLA) and lipoamide (LPM), we discovered that only LA is capable of stimulating cAMP production in NK cells. Furthermore, there is no difference in cAMP production after stimulation with either R-LA, S-LA or racemic LA. Competition and synergistic studies indicate that LA may also activate AC independent of the EP2 and EP4 receptors. Pretreatment of PBMCs with KH7 (a specific peptide inhibitor of soluble AC) and the calcium inhibitor (Bapta) prior to LA treatment resulted in reduced cAMP levels, suggesting that soluble AC and calcium signaling mediate LA stimulation of cAMP production. In addition, pharmacological inhibitor studies demonstrate that LA also activates other G protein-coupled receptors, including histamine and adenosine but not the ß-adrenergic receptors. These novel findings provide information to better understand the mechanisms of action of LA, which can help facilitate the use of LA as a therapeutic for various diseases.


Subject(s)
Cyclic AMP/biosynthesis , Receptors, G-Protein-Coupled/physiology , Thioctic Acid/pharmacology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Calcium Signaling/drug effects , Dinoprostone/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , HEK293 Cells , Humans , Killer Cells, Natural , Leukocytes, Mononuclear/drug effects , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Thioctic Acid/analogs & derivatives
15.
Biochem Pharmacol ; 81(3): 379-87, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21070749

ABSTRACT

An important event in the development of tumors is angiogenesis, or the formation of new blood vessels. Angiogenesis is also known to be involved in tumor cell metastasis and is dependent upon the activity of the vascular endothelial growth factor (VEGF) signaling pathway. Studies of mice in which the EP3 prostanoid receptors have been genetically deleted have shown a role for these receptors in cancer growth and angiogenesis. In the present study, human colon cancer HCA-7 cells were used as a model system to understand the potential role of EP3 receptors in tumor cell migration. We now show that stimulation of HCA-7 cells with PGE2 enhanced the up-regulation of VEGF receptor-1 (VEGFR-1) expression by a mechanism involving EP3 receptor-mediated activation of phosphatidylinositol 3-kinase and the extracellular signal-regulated kinases. Moreover, the PGE2 stimulated increase in VEGFR-1 expression was accompanied by an increase in the cellular migration of HCA-7 cells. Given the known involvement of VEGFR-1 in cellular migration, our results suggest that EP3 receptors may contribute to tumor cell metastasis by increasing cellular migration through the up-regulation of VEGFR-1 signaling.


Subject(s)
Cell Movement/drug effects , Colonic Neoplasms/metabolism , Dinoprostone/physiology , Up-Regulation , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Cell Line, Tumor , Colon/blood supply , Enzyme Induction/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , RNA, Messenger/metabolism , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-1/genetics
16.
Methods Enzymol ; 484: 95-107, 2010.
Article in English | MEDLINE | ID: mdl-21036228

ABSTRACT

The potential for G-protein-coupled receptors (GPCRs) to show constitutive activity is emerging as one of the fundamental properties of GPCRs signal transduction. Indeed, of the four subtypes of E-type prostanoid (EP) receptors, the EP3 and EP4 subtypes show constitutive activity in addition to their innate ligand-dependent activation of signaling pathways. The constitutive activity of the EP3 and EP4 receptor subtypes was discovered during the initial characterizations of these receptors and may be important for setting the basal level of cellular tone in the given signaling pathway. This chapter introduces some of the methods that can be used to study the constitutive activity of the EP receptors.


Subject(s)
Biological Assay/methods , Receptors, Prostaglandin E/metabolism , Animals , Cyclic AMP/metabolism , Humans , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism
17.
Cancer Biol Ther ; 10(10): 1056-66, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20890108

ABSTRACT

BACKGROUND: Prostaglandin E2 (PGE2) levels are frequently elevated in colorectal carcinomas. PGE2 is perceived via four transmembrane G protein coupled receptors (EP1-4), among which the EP4 receptor is most relevant. PGE2/EP4-receptor interaction activates CREB via the ERK/MEK pathway. However, the downstream target genes activated by this pathway remained to be investigated. METHODOLOGY/PRINICIPAL FINDINGS: Here, we have identified S100P (an EF-hand calcium binding protein) as a novel downstream target. We show by realtime RT-PCR that S100P mRNA levels are elevated in 14/17 (82%) colon tumor tissues as compared to paired adjacent normal colonic tissues. S100P expression is stimulated in the presence of PGE2 in a time dependent manner at mRNA and protein levels in colon, breast and pancreatic cancer cells. Pharmacological and RNAi-mediated inhibition of the EP4 receptor attenuates PGE2-dependent S100P mRNA induction. RNA(i)-mediated knockdown of CREB inhibits endogenous S100P expression. Furthermore, using luciferase reporter analysis and EMSA we show that mutation and/or deletion of the CRE sequence within the S100P promoter abolished PGE2-mediated transcriptional induction. Finally, we demonstrate that RNA(i)-mediated knockdown of S100P compromised invadopodia formation, colony growth and motility of colon cancer cells. Interestingly, endogenous knock down of S100P decreases ERK expression levels, suggesting a role for ERK in regulating S100P mediated cell growth and motility. CONCLUSIONS/SIGNIFICANCE: Together, our findings show for the first time that S100P expression is regulated by PGE2/EP4-receptor signaling and may participate in a feedback signaling that perpetuates tumor cell growth and migration. Therefore, our data suggest that dysregulated S100P expression resulting from aberrant PGE2/EP4 receptor signaling may have important consequences relevant to colon cancer pathogenesis.


Subject(s)
Calcium-Binding Proteins/metabolism , Colonic Neoplasms/metabolism , Dinoprostone/metabolism , Neoplasm Proteins/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction , Blotting, Western , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colony-Forming Units Assay , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Dinoprostone/genetics , Electrophoretic Mobility Shift Assay , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutagenesis, Site-Directed , Mutation/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Curr Eye Res ; 35(5): 408-18, 2010 May.
Article in English | MEDLINE | ID: mdl-20450254

ABSTRACT

PURPOSE: Agonists of the F prostanoid receptor for prostaglandin F2alpha exert. exert an ocular hypotensive effect that has been attributed to increased aqueous humor outflow through the uveoscleral pathway. Although tissue remodeling of the ciliary muscle has been described, the signaling mechanisms that link activation of the FP receptor to remodeling of the ciliary muscle are poorly understood. Herein, we describe the identification of novel signaling mechanisms that may contribute to this process. MATERIALS AND METHODS: Cultures of human ciliary smooth muscle cells were established from fetal eye tissue explants. The cells were validated by their expression of alpha-smooth muscle-actin and functional FP receptors. Cultures were treated with prostaglandin F(2 alpha) and examined for the induction of three immediate early genes related to tissue remodeling using Western blot analysis, quantitative real-time polymerase chain reaction, and reporter gene assays. RESULTS: Human ciliary smooth muscle cells express functional FP receptors whose activation up-regulates the expression of early growth response factor-1 and connective tissue growth factor at the mRNA and protein levels. Prostaglandin F(2 alpha) stimulation also increases the protein expression of hypoxia-inducible factor-1 alpha and activates luciferase reporter plasmids under the control of the hypoxia response element. CONCLUSIONS: Early growth response factor-1 and hypoxia-inducible factor-1 alpha are important transcriptional activators of downstream genes involved in tissue remodeling and angiogenesis, whereas connective tissue growth factor is a secreted growth factor that also contributes to these processes. Thus, stimulation of FP receptors in human ciliary smooth muscle cells up-regulates the expression of immediate early genes that may coordinate the remodeling of the ciliary muscle, thereby facilitating aqueous outflow.


Subject(s)
Ciliary Body/drug effects , Dinoprost/pharmacology , Early Growth Response Protein 1/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Muscle, Smooth/drug effects , Neovascularization, Physiologic , Receptors, Prostaglandin/metabolism , Actins/metabolism , Blotting, Western , Cells, Cultured , Ciliary Body/embryology , Ciliary Body/metabolism , Connective Tissue Growth Factor/genetics , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/drug effects , Genes, Immediate-Early/physiology , Humans , Inositol Phosphates/metabolism , Muscle, Smooth/embryology , Muscle, Smooth/metabolism , RNA, Messenger/metabolism , Receptors, Prostaglandin/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
Mol Pharmacol ; 77(6): 1025-36, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20335389

ABSTRACT

The EP1 prostanoid receptor is one of four subtypes whose cognate physiological ligand is prostaglandin-E2 (PGE(2)). It is in the family of G-protein-coupled receptors and is known to activate Ca(2+) signaling, although relatively little is known about other aspects of E-type prostanoid receptor (EP) 1 receptor signaling. In human embryonic kidney (HEK) cells expressing human EP1 receptors, we now show that PGE(2) stimulation of the EP1 receptor up-regulates the expression of hypoxia-inducible factor-1 alpha (HIF-1 alpha), which can be completely blocked by pertussis toxin, indicating coupling to G(i/o). This up-regulation of HIF-1 alpha occurs under normoxic conditions and could be inhibited with wortmannin, Akt inhibitor, and rapamycin, consistent with the activation of a phosphoinositide-3 kinase/Akt/mammalian target of rapamycin (mTOR) signaling pathway, respectively. In contrast to the hypoxia-induced up-regulation of HIF-1 alpha, which involves decreased protein degradation, the up-regulation of HIF-1 alpha by the EP1 receptor was associated with the phosphorylation of ribosomal protein S6 (rpS6), suggesting activation of the ribosomal S6 kinases and increased translation. Stimulation of endogenous EP1 receptors in human HepG2 hepatocellular carcinoma cells recapitulated the normoxic up-regulation of HIF-1 alpha observed in HEK cells, was sensitive to pertussis toxin, and involved the activation of mTOR signaling and phosphorylation of rpS6. In addition, treatment of HepG2 cells with sulprostone, an EP1-selective agonist, up-regulated the mRNA expression of vascular endothelial growth factor-C, a HIF-regulated gene. HIF-1 alpha is known to promote tumor growth and metastasis and is often up-regulated in cancer. Our findings provide a potential mechanism by which increased PGE(2) biosynthesis could up-regulate the expression of HIF-1 alpha and promote tumorigenesis.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Prostaglandin E/metabolism , Signal Transduction , Up-Regulation , Androstadienes/pharmacology , Cell Line , Dinoprostone/pharmacology , Genes, Reporter , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Phosphorylation , Polymerase Chain Reaction , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E, EP1 Subtype , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Up-Regulation/drug effects , Wortmannin
20.
Biochim Biophys Acta ; 1791(4): 238-45, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19416642

ABSTRACT

Prostaglandin-E(2) (PGE(2)) is a hormone derived from the metabolism of arachidonic acid whose functions include regulation of platelet aggregation, fever and smooth muscle contraction/relaxation. PGE(2) mediates its physiological and pathophysiological effects through its binding to four G-protein coupled receptor subtypes, named EP(1), EP(2), EP(3) and EP(4). The EP(3) prostanoid receptor is unique in that it has multiple isoforms generated by alternative mRNA splicing. These splice variants display differences in tissue expression, constitutive activity and regulation of signaling molecules. To date there are few reports identifying differential activities of EP(3) receptor isoforms and their effects on gene regulation. We generated HEK cell lines expressing the human EP(3-Ia), EP(3-II) or EP(3-III) isoforms. Using immunoblot analysis we found that nM concentrations of PGE(2) strongly stimulated the phosphorylation of ERK 1/2 by the EP(3-II) and EP(3-III) isoforms; whereas, ERK 1/2 phosphorylation by the EP(3-Ia) isoform was minimal and only occurred at muM concentrations of PGE(2). Furthermore, the mechanisms of the PGE(2) mediated phosphorylation of ERK 1/2 by the EP(3-II) and EP(3-III) isoforms were different. Thus, PGE(2) stimulation of ERK 1/2 phosphorylation by the EP(3-III) isoform involves activation of a Galpha(i)/PI3K/PKC/Src and EGFR-dependent pathway; while for the EP(3-II) isoform it involves activation of a Galpha(i)/Src and EGFR-dependent pathway. These differences result in unique differences in the regulation of reporter plasmid activity for the downstream effectors ELK1 and AP-1 by the EP(3-II) and EP(3-III) prostanoid receptor isoforms.


Subject(s)
Gene Expression Regulation, Enzymologic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Prostaglandin E/metabolism , Cells, Cultured , Cyclic AMP , Enzyme Activation , Humans , Immunoblotting , Kidney/cytology , Kidney/metabolism , Luciferases/metabolism , MAP Kinase Signaling System , Phosphorylation , Protein Isoforms , Radioligand Assay , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...