Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
J Clin Endocrinol Metab ; 100(9): E1197-205, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26192875

ABSTRACT

CONTEXT: Fetal ovarian development and primordial follicle formation underpin future female fertility. Prokineticin (PROK) ligands regulate cell survival, proliferation, and angiogenesis in adult reproductive tissues including the ovary. However, their expression and function during fetal ovarian development remains unclear. OBJECTIVE: This study aimed to investigate expression and localization of the PROK ligands, receptors, and their downstream transcriptional targets in the human fetal ovary. SETTING: This study was conducted at the University of Edinburgh. PARTICIPANTS: Ovaries were collected from 37 morphologically normal human fetuses. DESIGN AND MAIN OUTCOME MEASURES: mRNA and protein expression of PROK ligands and receptors was determined in human fetal ovaries using qRT-PCR, immunoblotting, and immunohistochemistry. Functional studies were performed using a human germ cell tumor line (TCam-2) stably transfected with Prokineticin receptor 1 (PROKR1). RESULTS: Expression of PROK1 and PROKR1 was significantly higher in mid-gestation ovaries (17-20 wk) than at earlier gestations (8-11 and 14-16 wk). PROK2 significantly increased across the gestations examined. PROKR2 expression remained unchanged. PROK ligand and receptor proteins were predominantly localized to germ cells (including oocytes within primordial follicles) and endothelial cells, indicating these cell types to be the targets of PROK signaling in the human fetal ovary. PROK1 treatment of a germ cell line stably expressing PROKR1 resulted in ERK phosphorylation and elevated COX2 expression. CONCLUSIONS: Developmental changes in expression and regulation of COX2 and phosphorylated ERK (pERK) by PROK1 suggest that PROK ligands may be novel regulators of germ cell development in the human fetal ovary, interacting within a network of growth and survival factors prior to primordial follicle formation.


Subject(s)
Cyclooxygenase 2/metabolism , Gastrointestinal Hormones/metabolism , Germ Cells/metabolism , Neuropeptides/metabolism , Ovary/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Cyclooxygenase 2/genetics , Female , Fetal Development , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/pharmacology , Germ Cells/drug effects , Humans , Neuropeptides/genetics , Ovary/embryology , Phosphorylation/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Signal Transduction/drug effects , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/pharmacology
2.
Reproduction ; 146(6): 581-91, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24051059

ABSTRACT

The mechanisms that regulate the induction of term or preterm delivery (PTD) are not fully understood. Infection is known to play a role in the induction of pro-inflammatory cascades in uteroplacental tissues associated with preterm pathological parturition. Similar but not identical cascades are evident in term labour. In the current study, we used a mouse model to evaluate the role of prokineticins in term and preterm parturition. Prokineticins are multi-functioning secreted proteins that signal through G-protein-coupled receptors to induce gene expression, including genes important in inflammatory responses. Expression of prokineticins (Prok1 and Prok2) was quantified in murine uteroplacental tissues by QPCR in the days preceding labour (days 16-19). Prok1 mRNA expression increased significantly on D18 in fetal membranes (compared with D16) but not in uterus or placenta. Intrauterine injection of PROK1 on D17 induced fetal membrane mRNA expression of the pro-inflammatory mediators Il6, Il1b, Tnf, Cxcl2 and Cxcl5, which are not normally up-regulated until D19 of pregnancy. However, intrauterine injection of PROK1 did not result in PTD. As expected, injection of lipopolysaccharide (LPS) induced PTD, but this was not associated with changes in expression of Prok1 or its receptor (Prokr1) in fetal membranes. These results suggest that although Prok1 exhibits dynamic mRNA regulation in fetal membranes preceding labour and induces a pro-inflammatory response when injected into the uterus on D17, it is insufficient to induce PTD. Additionally, prokineticin up-regulation appears not to be part of the LPS-induced inflammatory response in mouse fetal membranes.


Subject(s)
Extraembryonic Membranes/drug effects , Inflammation/chemically induced , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/pharmacology , Animals , Extraembryonic Membranes/immunology , Extraembryonic Membranes/metabolism , Female , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Injections , Lipopolysaccharides/pharmacology , Male , Mice , Pregnancy , Premature Birth , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Up-Regulation/drug effects , Uterus , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism
3.
Endocrinology ; 154(10): 3864-76, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23861370

ABSTRACT

The conceptus and endometrium secrete large amounts of prostaglandin E2 (PGE2) into the porcine uterine lumen during the periimplantation period. We hypothesized that PGE2 acts on conceptus/trophoblast cells through auto- and paracrine mechanisms. Real-time RT-PCR analysis revealed that PGE2 receptor (PTGER)2 mRNA was 14-fold greater in conceptuses/trophoblasts on days 14-25 (implantation and early placentation period) vs preimplantation day 10-13 conceptuses (P < .05). Similarly, expression of PTGER2 protein increased during implantation. Conceptus expression of PTGER4 mRNA and protein did not differ on days 10-19. PGE2 stimulated PTGER2 mRNA expression in day 15 trophoblast cells through PTGER2 receptor signaling. PGE2 elevated aromatase expression and estradiol-17ß secretion by trophoblast cells. Moreover, PGE2 and the PTGER2 agonist, butaprost, increased the adhesive capacity of both human HTR-8/SVneo trophoblast and primary porcine trophoblast cells to extracellular matrix. This PGE2-induced alteration in trophoblast cell adhesion to extracellular matrix was abolished by incubation of these cells with AH6809 (PTGER2 antagonist), ITGAVB3-directed tetrapeptide arg-gly-asp-ser or integrin ITGAVB3 antibody. PGE2 stimulated adhesion of porcine trophoblast cells via the estrogen receptor and MEK/MAPK signaling pathway. PGE2 induced phosphorylation of MAPK1/MAPK3 through PTGER2 and up-regulated expression of cell adhesion proteins such as focal adhesion kinase and intercellular adhesion molecule-1. Our study indicates that elevated PGE2 in the periimplantation uterine lumen stimulates conceptus PTGER2 expression, which in turn promotes trophoblast adhesion via integrins, and synthesis and secretion of the porcine embryonic signal estradiol-17ß. Moreover, the mechanism through which PGE2 increases trophoblast adhesion is not species specific because it is PTGER2- and integrin-dependent in both porcine and human trophoblast cells.


Subject(s)
Autocrine Communication , Dinoprostone/metabolism , Embryonic Development , Paracrine Communication , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Trophoblasts/metabolism , Animals , Autocrine Communication/drug effects , Cell Adhesion/drug effects , Cell Line , Cells, Cultured , Crosses, Genetic , Dinoprostone/agonists , Dinoprostone/antagonists & inhibitors , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Development/drug effects , Estradiol/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Developmental/drug effects , Humans , Integrins/antagonists & inhibitors , Integrins/metabolism , MAP Kinase Signaling System/drug effects , Paracrine Communication/drug effects , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/genetics , Sus scrofa , Trophoblasts/cytology , Trophoblasts/drug effects
4.
Biochim Biophys Acta ; 1823(10): 1789-95, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22732298

ABSTRACT

Cervical cancer is one of the leading gynecological malignancies in women. We have recently shown that seminal plasma (SP) can regulate the inflammatory cyclooxygenase-prostaglandin pathway and enhance the growth of cervical epithelial tumours in vivo by promoting cellular proliferation and alteration of vascular function. This study investigated the molecular mechanism whereby SP regulates vascular function using an in vitro model system of HeLa cervical adenocarcinoma cells and human umbilical vein endothelial cells (HUVECs). We found that SP rapidly enhanced the expression of the angiogenic chemokines, interleukin (IL)-8 and growth regulated oncogene alpha (GRO) in HeLa cells in a time-dependent manner. We investigated the molecular mechanism of SP-mediated regulation of IL-8 and GRO using a panel of chemical inhibitors of cell signalling. We found that treatment of HeLa cells with SP elevated expression of IL-8 and GRO by transactivation of the epidermal growth factor receptor, activation of extracellular signal-regulated kinase and induction of cyclooxygenase enzymes and nuclear factor kappa B. We investigated the impact of IL-8 and GRO, released from HeLa cells after treatment with SP, on vascular function using a co-culture model system of conditioned medium (CM) from HeLa cells, treated with or without SP, and HUVECs. We found that CM from HeLa cells induced the arrangement of endothelial cells into a network of tube-like structures via the CXCR2 receptor on HUVECs. Taken together our data outline a molecular mechanism whereby SP can alter vascular function in cervical cancers via the pro-angiogenic chemokines, IL-8 and GRO.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Blood Vessels/physiopathology , Chemokine CXCL1/genetics , Interleukin-8/genetics , Semen/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/physiopathology , Blood Vessels/drug effects , Blood Vessels/metabolism , Chemokine CXCL1/metabolism , Culture Media, Conditioned/pharmacology , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-8/metabolism , Male , Models, Biological , Phosphorylation/drug effects , Signal Transduction , Up-Regulation/drug effects , Up-Regulation/genetics , Uterine Cervical Neoplasms/blood supply , Uterine Cervical Neoplasms/enzymology
5.
J Immunol ; 188(11): 5303-10, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22544928

ABSTRACT

The ubiquitous protein CD46, a regulator of complement activity, promotes T cell activation and differentiation toward a regulatory Tr1-like phenotype. The CD46-mediated differentiation pathway is defective in several chronic inflammatory diseases, underlying the importance of CD46 in controlling T cell function and the need to understand its regulatory mechanisms. Using an RNA interference-based screening approach in primary T cells, we have identified that two members of the G protein-coupled receptor kinases were involved in regulating CD46 expression at the surface of activated cells. We have investigated the role of PGE(2), which binds to the E-prostanoid family of G protein-coupled receptors through four subtypes of receptors called EP 1-4, in the regulation of CD46 expression and function. Conflicting roles of PGE(2) in T cell functions have been reported, and the reasons for these apparent discrepancies are not well understood. We show that addition of PGE(2) strongly downregulates CD46 expression in activated T cells. Moreover, PGE(2) differentially affects T cell activation, cytokine production, and phenotype depending on the activation signals received by the T cells. This was correlated with a distinct pattern of the PGE(2) receptors expressed, with EP4 being preferentially induced by CD46 activation. Indeed, addition of an EP4 antagonist could reverse the effects observed on cytokine production after CD46 costimulation. These data demonstrate a novel role of the PGE(2)-EP4 axis in CD46 functions, which might at least partly explain the diverse roles of PGE(2) in T cell functions.


Subject(s)
Dinoprostone/physiology , Lymphocyte Activation/immunology , Membrane Cofactor Protein/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Cell Proliferation , Cells, Cultured , Dinoprostone/metabolism , G-Protein-Coupled Receptor Kinase 1/physiology , Gene Expression Regulation/immunology , Humans , Membrane Cofactor Protein/antagonists & inhibitors , Membrane Cofactor Protein/biosynthesis , RNA Interference/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism
6.
PLoS One ; 7(3): e33848, 2012.
Article in English | MEDLINE | ID: mdl-22442729

ABSTRACT

Cervical cancer is one of the leading causes of cancer-related death in women in sub-Saharan Africa. Extensive evidence has shown that cervical cancer and its precursor lesions are caused by Human papillomavirus (HPV) infection. Although the vast majority of HPV infections are naturally resolved, failure to eradicate infected cells has been shown to promote viral persistence and tumorigenesis. Furthermore, following neoplastic transformation, exposure of cervical epithelial cells to inflammatory mediators either directly or via the systemic circulation may enhance progression of the disease. It is well recognised that seminal plasma contains an abundance of inflammatory mediators, which are identified as regulators of tumour growth. Here we investigated the role of seminal plasma in regulating neoplastic cervical epithelial cell growth and tumorigenesis. Using HeLa cervical adenocarcinoma cells, we found that seminal plasma (SP) induced the expression of the inflammatory enzymes, prostaglandin endoperoxide synthase (PTGS1 and PTGS2), cytokines interleukin (IL) -6, and -11 and vascular endothelial growth factor-A (VEGF-A). To investigate the role of SP on tumour cell growth in vivo, we xenografted HeLa cells subcutaneously into the dorsal flank of nude mice. Intra-peritoneal administration of SP rapidly and significantly enhanced the tumour growth rate and size of HeLa cell xenografts in nude mice. As observed in vitro, we found that SP induced expression of inflammatory PTGS enzymes, cytokines and VEGF-A in vivo. Furthermore we found that SP enhances blood vessel size in HeLa cell xenografts. Finally we show that SP-induced cytokine production, VEGF-A expression and cell proliferation are mediated via the induction of the inflammatory PTGS pathway.


Subject(s)
Adenocarcinoma/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neovascularization, Pathologic/metabolism , Semen , Uterine Cervical Neoplasms/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Africa South of the Sahara/epidemiology , Animals , Female , HeLa Cells , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/mortality , Neovascularization, Pathologic/pathology , Transplantation, Heterologous , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology
7.
Mol Hum Reprod ; 18(2): 88-95, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21965347

ABSTRACT

Fetal ovarian development and primordial follicle formation are imperative for adult fertility in the female. Data suggest the interleukin (IL)6-type cytokines, leukaemia inhibitory factor (LIF), IL6, oncostatin M (OSM) and ciliary neurotrophic factor (CNTF), are able to regulate the survival, proliferation and differentiation of fetal murine germ cells (GCs) in vivo and in vitro. We postulated that these factors may play a similar role during early human GC development and primordial follicle formation. To test this hypothesis, we have investigated the expression and regulation of IL6-type cytokines, using quantitative reverse transcription polymerase chain reaction and immunohistochemistry. Expression of transcripts encoding OSM increased significantly across the gestational range examined (8-20 weeks), while expression of IL6 increased specifically between the first (8-11 weeks) and early second (12-16 weeks) trimesters, co-incident with the initiation of meiosis. LIF and CNTF expression remained unchanged. Expression of the genes encoding the LIF and IL6 receptors, and their common signalling subunit gp130, was also found to be developmentally regulated, with expression increasing significantly with increasing gestation. LIF receptor and gp130 proteins localized exclusively to GCs, including oocytes in primordial follicles, indicating this cell type to be the sole target of IL6-type cytokine signalling in the human fetal ovary. These data establish that IL6-type cytokines and their receptors are expressed in the human fetal ovary and may directly influence GC development at multiple stages of maturation.


Subject(s)
Gene Expression Regulation, Developmental , Oocytes/metabolism , Ovarian Follicle/metabolism , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Adult , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Female , Fetus , Gestational Age , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Oncostatin M/genetics , Oncostatin M/metabolism , Oocytes/growth & development , Ovarian Follicle/growth & development , Pregnancy , Pregnancy Trimesters , Real-Time Polymerase Chain Reaction , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
8.
Endocrinology ; 153(1): 395-403, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22186417

ABSTRACT

Human parturition is an inflammatory process that can be activated prematurely by pathological stimuli. This study investigated the expression of G protein-coupled receptors GPR43 and GPR41 receptors in human uteroplacental tissues and the role of short-chain fatty acids (SCFA) in modulating inflammatory pathways in fetal membranes. Expression of GPR43 and GPR41 was investigated in uteroplacental tissues collected from women delivering at term or preterm after ethical approval and patient informed consent. The effect of SCFA on expression of inflammatory genes was assessed in amnion explants after culture with a mimetic of infection (lipopolysaccharide, LPS). Sodium propionate effect on LPS-induced neutrophil chemotaxis was evaluated by transwell assay. GPR43 and GPR41 mRNA expression was higher in myometrium and fetal membranes collected from women after the onset of labor. GPR43 protein expression localized to immune cells and vascular endothelium in the myometrium and epithelium of fetal membranes. Treatment with LPS significantly increased mRNA expression of GPR43 and inflammatory genes. Cotreatment with LPS and sodium propionate decreased LPS-induced expression of inflammatory genes including IL-6, IL-8, cyclooxygenase-2, IL-1α, intercellular adhesion molecule-1, and platelet endothelial cell adhesion molecule-1 but not IL-1ß or lymphocyte function-associated antigen-1. Sodium propionate reduced LPS-induced neutrophil chemotaxis and protein secretion of the neutrophil chemoattractant IL-8. Finally, fetal membrane expression of GPR43 was significantly higher in women delivering preterm with evidence of infection. GPR43-SCFA interactions may represent novel pathways that regulate inflammatory processes involved in human labor. Suppression of inflammatory pathways by SCFA may be therapeutically beneficial for pregnant women at risk of pathogen-induced preterm delivery.


Subject(s)
Fatty Acids, Volatile/physiology , Inflammation Mediators/physiology , Labor, Obstetric/physiology , Cell Adhesion Molecules/genetics , Chemotaxis, Leukocyte/drug effects , Cytokines/genetics , Extraembryonic Membranes/drug effects , Extraembryonic Membranes/metabolism , Female , Gene Expression/drug effects , Humans , Infant, Newborn , Interleukin-8/biosynthesis , Interleukin-8/genetics , Labor, Obstetric/genetics , Lipopolysaccharides/pharmacology , Myometrium/drug effects , Myometrium/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Obstetric Labor, Premature/genetics , Obstetric Labor, Premature/metabolism , Placenta/drug effects , Placenta/metabolism , Pregnancy , Propionates/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Tissue Culture Techniques
9.
Am J Pathol ; 179(6): 2709-19, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21983634

ABSTRACT

The infiltration of human myometrium and cervix with leukocytes and the formation of a pro-inflammatory environment within the uterus have been associated with the initiation of both term and preterm parturition. The mechanism regulating the onset of this pro-inflammatory cascade is not fully elucidated. We demonstrate that prokineticin 1 (PROK1) is up-regulated in human myometrium and placenta during labor. The expression of PROK1 receptor remains unchanged during labor and is abundantly expressed in the myometrium. Gene array analysis identified 65 genes up-regulated by PROK1 in human myometrium, mainly cytokines and chemokines, including IL-1ß, chemokine C-C motif ligand 3, and colony-stimulating factor 3. In addition, we demonstrate that PROK1 increases the expression of chemokine C-C motif ligand 20, IL-6, IL-8, prostaglandin synthase 2, and prostaglandin E(2) and F(2α) secretion. The treatment of myometrial explants with 100 ng/mL of lipopolysaccharide up-regulates the expression of PROK1, PROK1 receptor, and inflammatory mediators. The infection of myometrial explants with lentiviral microRNA targeting PROK1, preceding treatment with lipopolysaccharide, reduces the expression of inflammatory genes. We propose that PROK1 is a novel inflammatory mediator that can contribute to the onset of human parturition at term and partially mediate premature onset of inflammatory pathways during bacterial infection.


Subject(s)
Endometritis/etiology , Gastrointestinal Hormones/physiology , Myometrium/metabolism , Premature Birth/etiology , Term Birth/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/physiology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Endometritis/metabolism , Female , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Gene Expression , Humans , Lipopolysaccharides/pharmacology , MicroRNAs/pharmacology , Pregnancy , Premature Birth/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Up-Regulation , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism
10.
J Clin Endocrinol Metab ; 96(8): 2475-83, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21677035

ABSTRACT

CONTEXT: The human endometrium has an exceptional capacity for repeated repair after menses, but its regulation remains undefined. Premenstrually, progesterone levels fall and prostaglandin (PG) F2α synthesis increases, causing spiral arteriole constriction. We hypothesized that progesterone withdrawal, PGF2α, and hypoxia increase vascular endothelial growth factor (VEGF), an endometrial repair factor. DESIGN AND RESULTS: Endometrial biopsies were collected (n = 47) with ethical approval and consent. VEGF mRNA, quantified by quantitative RT-PCR, was increased during menstruation (P < 0.01).VEGF protein was maximally secreted from proliferative endometrial explants. Treatment of an endometrial epithelial cell line and primary human endometrial stromal cells with 100 nm PGF2α or hypoxia (0.5% O2) resulted in significant increases in VEGF mRNA and protein. VEGF was maximal when cells were cotreated with PGF(2α) and hypoxia simultaneously (P < 0.05-0.001). Secretory-phase endometrial explants also showed an increase in VEGF with cotreatment (P < 0.05). However, proliferative-phase explants showed no increase in VEGF on treatment with PGF2α and/or hypoxia. Proliferative tissue was induced to increase VEGF mRNA expression when exposed to progesterone and its withdrawal in vitro but only in the presence of hypoxia and PG. Hypoxia-inducible factor-1α (HIF-1α) silencing with RNA interference suppressed hypoxia-induced VEGF expression in endometrial cells but did not alter PGF2α-induced VEGF expression. CONCLUSIONS: Endometrial VEGF is increased at the time of endometrial repair. Progesterone withdrawal, PGF2α, and hypoxia are necessary for this perimenstrual VEGF expression. Hypoxia acts via HIF-1α to increase VEGF, whereas PGF2α acts in a HIF-1α-independent manner. Hence, two pathways regulate the expression of VEGF during endometrial repair.


Subject(s)
Dinoprost/metabolism , Endometrium/physiology , Hypoxia/metabolism , Hypoxia/physiopathology , Menstrual Cycle/physiology , Vascular Endothelial Growth Factor A/genetics , Adult , Biopsy , Cells, Cultured , Cyclooxygenase Inhibitors/pharmacology , Endometrium/cytology , Endometrium/drug effects , Estradiol/blood , Female , Hormone Antagonists/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indomethacin/pharmacology , Middle Aged , Mifepristone/pharmacology , Progesterone/antagonists & inhibitors , Progesterone/blood , RNA, Messenger/metabolism , Regeneration/physiology , Young Adult
11.
Reproduction ; 142(2): 345-52, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21555360

ABSTRACT

Lipoxin A(4) is a lipid mediator that elicits anti-inflammatory and pro-resolution actions via its receptor, formyl peptide receptor 2 (FPR2/ALX). In this study, we aimed to investigate the expression and potential role of lipoxin A(4) and FPR2/ALX in the regulation of inflammation associated with cyclical remodeling of the human endometrium across the menstrual cycle and during early pregnancy. Using quantitative RT-PCR analysis, we found that FPR2/ALX expression is upregulated during the menstrual phase of the cycle and in decidua tissue from the first trimester of pregnancy. We localized the site of expression of FPR2/ALX in menstrual phase endometrium and first-trimester decidua tissue to glandular epithelial cells and cells within the stromal compartment, including cells lining the blood vessels and immune cells. Measurement of serum lipoxin A(4) by ELISA revealed no difference in its levels across the menstrual cycle but an elevation in early pregnancy (P<0.001). We found that lipoxin A(4) was regulated by human chorionic gonadotrophin (hCG) during early pregnancy, because treatment of human decidua tissue with hCG increased lipoxin A(4) release (P<0.01). Finally, we have shown that lipoxin A(4) can suppress phorbol myristate acetate-induced expression of the inflammatory cytokines interleukin 6 and 8 in human endometrium and decidua tissue. These results demonstrate for the first time that lipoxin A(4) and its receptor FPR2/ALX can regulate inflammatory events in the human endometrium and decidua of early pregnancy.


Subject(s)
Endometrium/immunology , Inflammation Mediators/antagonists & inhibitors , Lipoxins/metabolism , Menstrual Cycle/metabolism , Adult , Chorionic Gonadotropin/metabolism , Decidua/cytology , Decidua/drug effects , Decidua/immunology , Decidua/metabolism , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Interleukins/genetics , Interleukins/metabolism , Lipoxins/blood , Menstrual Cycle/blood , Pregnancy , Pregnancy Trimester, First/blood , Pregnancy Trimester, First/metabolism , RNA, Messenger/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/toxicity , Tissue Culture Techniques , Young Adult
12.
Endocrinology ; 152(7): 2845-56, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21558311

ABSTRACT

After menstruation, the endometrium has a remarkable capacity for repair, but the factors involved remain undefined. We hypothesize adrenomedullin (AM) plays a role in this process. Premenstrually progesterone levels decline, stimulating prostaglandin (PG) synthesis, vasoconstriction, and hypoxia. This study aimed to determine 1) AM expression throughout the menstrual (M) cycle and 2) its regulation by PG and hypoxia. Human endometrial biopsies (n = 51) were collected with ethical approval and consent. AM mRNA expression was examined by quantitative RT-PCR and was found to be selectively elevated in endometrium from the menstrual (M) phase (P < 0.001). AM immunohistochemical staining was maximal in M and proliferative (P) endometrium. Culture of secretory, but not P, explants with 100 nm PGF(2α) or hypoxia (0.5% O2) increased AM mRNA (P < 0.05). P explants were induced to increase AM expression using in vitro progesterone withdrawal but required the presence of hypoxia (P < 0.05). Short hairpin sequences against hypoxia-inducible factor-1α (HIF-1α) inhibited AM hypoxic up-regulation but did not alter PGF(2α)-induced expression. The AM receptor was immunolocalized to endothelial cells in both lymphatic and blood vessels. Conditioned medium from PGF(2α)-treated cells increased endothelial cell proliferation and branching (P < 0.05). This was abolished by AM receptor antagonists. In conclusion, AM is elevated at the time of endometrial repair and induces both angiogenesis and lymphangiogenesis by stimulating endothelial cell proliferation and tube formation. In the human endometrium, AM expression is up-regulated by two mechanisms: a HIF-1α-mediated hypoxic induction and a HIF-1α-independent PGF(2α) pathway. These physiological mechanisms may provide novel therapeutic targets for disorders such as heavy menstrual bleeding.


Subject(s)
Adrenomedullin/metabolism , Endometrium/physiology , Gene Expression Regulation , Menstrual Cycle/metabolism , Adrenomedullin/genetics , Adult , Angiogenesis Inducing Agents/metabolism , Cell Hypoxia , Cell Line , Cell Proliferation , Dinoprost/metabolism , Endometrium/blood supply , Endometrium/cytology , Endothelium, Vascular/physiology , Female , Gene Silencing , Humans , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Hypoxia-Inducible Factor 1/genetics , Lymphangiogenesis , Middle Aged , Organ Culture Techniques , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Adrenomedullin/antagonists & inhibitors , Receptors, Adrenomedullin/metabolism , Receptors, Prostaglandin/metabolism
13.
Mol Hum Reprod ; 17(10): 626-36, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21546446

ABSTRACT

Prokineticin 1 (PROK1) signalling via prokineticin receptor 1 (PROKR1) regulates the expression of several genes with important roles in endometrial receptivity and implantation. This study investigated PROK1 regulation of Dickkopf 1 (DKK1) expression, a negative regulator of canonical Wnt signalling, and its function in the non-pregnant endometrium and first trimester decidua. DKK1 mRNA expression is elevated during the mid-secretory phase of the menstrual cycle and expression increases further in first trimester decidua. DKK1 protein expression is localized to glandular epithelial and stromal cells during the proliferative, early- and mid-secretory phases, whereas expression is confined to the stroma in the late-secretory phase and first trimester decidua. PROK1 induces the expression of DKK1 in endometrial epithelial cells stably expressing PROKR1 and in first trimester decidua explants, via a Gq-calcium-calcineurin-nuclear factor of activated T-cells-mediated pathway. Endometrial epithelial cell proliferation is negatively regulated by PROK1-PROKR1 signalling. We demonstrate that this effect on cell proliferation occurs via DKK1 expression, as siRNA targeted against DKK1 reduces the PROK1-induced decrease in proliferation. Furthermore, decidualization of primary human endometrial stromal cells with progesterone and cyclic adenosine monophosphate is inhibited by miRNA knock down of PROK1 or DKK1. These data demonstrate important roles for PROK1 and DKK1 during endometrial receptivity and early pregnancy, which include regulation of endometrial cell proliferation and decidualization.


Subject(s)
Decidua/physiology , Gastrointestinal Hormones/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Adult , Cell Proliferation , Cells, Cultured , Cyclic AMP/pharmacology , Decidua/drug effects , Embryo Implantation , Epithelial Cells/physiology , Female , Gastrointestinal Hormones/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Luteal Phase/metabolism , Placentation/physiology , Pregnancy , Progesterone/pharmacology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Signal Transduction , Stromal Cells/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics
14.
PLoS One ; 6(5): e19209, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21589857

ABSTRACT

The prostaglandin endoperoxide synthase (PTGS) pathway is a potent driver of tumour development in humans by enhancing the biosynthesis and signalling of prostaglandin (PG) E(2). PTGS2 expression and PGE(2) biosynthesis is elevated in endometrial adenocarcinoma, however the mechanism whereby PTGS and PGE(2) regulate endometrial tumour growth is unknown. Here we investigated (a) the expression profile of the PGE synthase enzymes (PTGES, PTGES-2, PTGES-3) and PGE receptors (PTGER1-4) in endometrial adenocarcinomas compared with normal endometrium and (b) the role of PTGER4 in endometrial tumorigenesis in vivo. We found elevated expression of PTGES2 and PTGER4 and suppression of PTGER1 and PTGER3 in endometrial adenocarcinomas compared with normal endometrium. Using WT Ishikawa endometrial adenocarcinoma cells and Ishikawa cells stably transfected with the full length PTGER4 cDNA (PTGER4 cells) xenografted in the dorsal flanks of nude mice, we show that PTGER4 rapidly and significantly enhances tumour growth rate. Coincident with enhanced PTGER4-mediated tumour growth we found elevated expression of PTGS2 in PTGER4 xenografts compared with WT xenografts. Furthermore we found that the augmented growth rate of the PTGER4 xenografts was not due to enhanced angiogenesis, but regulated by an increased proliferation index and hypoxia. In vitro, we found that PGE(2) and hypoxia independently induce expression of PTGER4 indicating two independent pathways regulating prostanoid receptor expression. Finally we have shown that PGE(2) and hypoxia synergise to promote cellular proliferation of endometrial adenocarcinoma cells.


Subject(s)
Adenocarcinoma/metabolism , Endometrial Neoplasms/metabolism , Hypoxia/physiopathology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction , Adenocarcinoma/pathology , Cell Line, Tumor , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Microscopy, Confocal , Receptors, Prostaglandin E, EP4 Subtype/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
Reproduction ; 142(1): 15-28, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490125

ABSTRACT

Inflammatory processes are central to reproductive events including ovulation, menstruation, implantation and labour, while inflammatory dysregulation is a feature of numerous reproductive pathologies. In recent years, there has been much research into the endogenous mechanisms by which inflammatory reactions are terminated and tissue homoeostasis is restored, a process termed resolution. The identification and characterisation of naturally occurring pro-resolution mediators including lipoxins and annexin A1 has prompted a shift in the field of anti-inflammation whereby resolution is now observed as an active process, triggered as part of a normal inflammatory response. This review will address the process of resolution, discuss available evidence for expression of pro-resolution factors in the reproductive tract and explore possible roles for resolution in physiological reproductive processes and associated pathologies.


Subject(s)
Genital Diseases, Female/immunology , Genitalia, Female/immunology , Inflammation/metabolism , Reproduction , Animals , Annexin A1/metabolism , Anti-Inflammatory Agents/therapeutic use , Eicosanoids/metabolism , Fatty Acids, Omega-3/metabolism , Female , Genital Diseases, Female/drug therapy , Genital Diseases, Female/metabolism , Genitalia, Female/drug effects , Genitalia, Female/metabolism , Glucocorticoids/metabolism , Homeostasis , Humans , Inflammation/drug therapy , Inflammation/immunology , Molecular Targeted Therapy , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/metabolism , Signal Transduction/drug effects
16.
Am J Pathol ; 178(3): 1245-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21356375

ABSTRACT

The endometrium has a remarkable capacity for efficient repair; however, factors involved remain undefined. Premenstrual progesterone withdrawal leads to increased prostaglandin (PG) production and local hypoxia. Here we determined human endometrial expression of interleukin-8 (IL-8) and the roles of PGE(2) and hypoxia in its regulation. Endometrial biopsy specimens (n = 51) were collected. Endometrial cells and explants were exposed to 100 nmol/L of PGE(2) or 0.5% O(2). The endometrial IL-8 concentration peaked during menstruation (P < 0.001) and had a significant proangiogenic effect. IL-8 was increased by PGE(2) and hypoxia in secretory but not proliferative explants, which suggests that exposure to progesterone is essential. In vitro progesterone withdrawal induced significant IL-8 up-regulation in proliferative explants primed with progestins, but only in the presence of hypoxia. Epithelial cells treated simultaneously with PGE(2) and hypoxia demonstrated synergistic increases in IL-8. Inhibition of HIF-1 by short hairpin RNA abolished hypoxic IL-8 induction, and inhibition of NF-κB by an adenoviral dominant negative inhibitor decreased PGE(2)-induced IL-8 expression (P > 0.05). Increased menstrual IL-8 is consistent with a role in repair. Progesterone withdrawal, hypoxia, and PGE(2) regulate endometrial IL-8 by acting via HIF-1 and NF-κB. Hence, progesterone withdrawal may activate two distinct pathways to initiate endometrial repair.


Subject(s)
Dinoprostone/pharmacology , Endometrium/metabolism , Endometrium/pathology , Interleukin-8/metabolism , Wound Healing/drug effects , Adult , Cell Hypoxia/drug effects , Echinomycin/pharmacology , Endometrium/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Silencing/drug effects , Genes, Dominant/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-8/genetics , Menstruation/drug effects , Middle Aged , NF-kappa B/metabolism , Neovascularization, Physiologic/drug effects , Progesterone/pharmacology , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Reproducibility of Results , Up-Regulation/drug effects
17.
Am J Pathol ; 178(1): 253-60, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224062

ABSTRACT

Chlamydia trachomatis and smoking are major risk factors for tubal ectopic pregnancy (EP), but the underlying mechanisms of these associations are not completely understood. Fallopian tube (FT) from women with EP exhibit altered expression of prokineticin receptors 1 and 2 (PROKR1 and PROKR2); smoking increases FT PROKR1, resulting in a microenvironment predisposed to EP. We hypothesize that C. trachomatis also predisposes to EP by altering FT PROKR expression and have investigated this by examining NFκB activation via ligation of the Toll-like receptor (TLR) family of cell-surface pattern recognition receptors. PROKR2 mRNA was higher in FT from women with evidence of past C. trachomatis infection than in those without (P < 0.05), and was also increased in FT explants and in oviductal epithelial cell line OE-E6/E7 infected with C. trachomatis (P < 0.01) or exposed to UV-killed organisms (P < 0.05). The ability of both live and dead organisms to induce this effect suggests ligation of a cell-surface-expressed receptor. FT epithelium and OE-E6/E7 were both found to express TLR2 and TLR4 by immunohistochemistry. Transfection of OE-E6/E7 cells with dominant-negative TLR2 or IκBα abrogated the C. trachomatis-induced PROKR2 expression. We propose that ligation of tubal TLR2 and activation of NFκB by C. trachomatis leads to increased tubal PROKR2, thereby predisposing the tubal microenvironment to ectopic implantation.


Subject(s)
Chlamydia Infections/complications , Chlamydia Infections/pathology , Chlamydia trachomatis , Fallopian Tubes/pathology , NF-kappa B/metabolism , Pregnancy, Ectopic/microbiology , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Toll-Like Receptor 2/metabolism , Adult , Cell Line , Fallopian Tubes/metabolism , Fallopian Tubes/microbiology , Female , Humans , I-kappa B Proteins/metabolism , Middle Aged , NF-KappaB Inhibitor alpha , Pregnancy , Pregnancy, Ectopic/metabolism , Pregnancy, Ectopic/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Toll-Like Receptor 4/metabolism
18.
Mol Hum Reprod ; 17(1): 22-32, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20705717

ABSTRACT

Murine knock-out models and blastocyst co-culture studies have identified prostaglandin-endoperoxide synthase (PTGS) 2, prostaglandin (PG) E receptor 2 (PTGER2) and the chemokine receptor CXCR4 as important regulators of early pregnancy events. In vitro studies and studies in non-human primates have shown that these proteins are regulated in the endometrium by the early embryonic signal, chorionic gonadotrophin (CG). Here we show that expressions of PTGER2 and CXCR4 are elevated during the mid-secretory phase of the menstrual cycle and decidua of early pregnancy in humans. Using first trimester decidua explants, we show that CG induces expression of PTGS2 and biosynthesis of PGE2, and expression of PTGER2. Subsequently, PGE2via PTGER2 induces expression of CXCR4. Using an in vitro model system of Ishikawa endometrial epithelial cells stably expressing PTGER2 and human first trimester decidua explants, we demonstrate that CXCR4 expression is regulated by PTGER2 via the epidermal growth factor receptor (EGFR)-phosphatidylinositol-3-kinase (PI3K)-extracellular signal-regulated kinase (ERK1/2) pathway.Taken together, our data suggest that early embryonic signals may regulate fetal-maternal crosstalk in the human endometrium by inducing CXCR4 expression via the PGE2-PTGER2-mediated induction of the EGFR, PI3K and ERK1/2 pathways.


Subject(s)
Chorionic Gonadotropin/pharmacology , Embryo Implantation/physiology , Endometrium/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, CXCR4/metabolism , Blotting, Western , Cell Line , Decidua/drug effects , Decidua/metabolism , Embryo Implantation/genetics , Endometrium/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , In Vitro Techniques , Menstrual Cycle/metabolism , Pregnancy , Pregnancy Trimester, First/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
19.
Mol Cell Endocrinol ; 331(1): 129-35, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20816914

ABSTRACT

Prostaglandin F(2α) (PGF(2α)) is an inflammatory mediator which signals through a G-protein coupled receptor, the F-prostanoid (FP) receptor. We have previously shown elevated FP receptor expression in endometrial adenocarcinoma, a common gynaecological malignancy in Western countries. In this study, the expression of the chemokine CC motif Ligand 20 (CCL20) was determined to be regulated by PGF(2α)-FP receptor signalling in endometrial adenocarcinoma explants and cell line, and expression of CCL20 and its receptor CCR6 was elevated in endometrial adenocarcinoma compared to non-malignant endometrium. Both CCL20 and CCR6 were localised to neoplastic endometrial epithelial cells. The induction of CCL20 expression by PGF(2α)-FP signalling in an endometrial adenocarcinoma cell line stably expressing the FP receptor (FPS cells) was found to be dependent on the intracellular signalling of Gq, EGFR, ERK, calcineurin and nuclear factor of activated T-cells (NFAT) proteins. The treatment of FPS cells with recombinant CCL20 caused a significant increase in proliferation. Therefore these data demonstrate a role for the FP receptor in regulation of the chemokine CCL20, which can mediate proliferation of endometrial adenocarcinoma epithelial cells.


Subject(s)
Adenocarcinoma/pathology , Chemokine CCL20/genetics , Endometrial Neoplasms/pathology , Receptors, Prostaglandin/metabolism , Signal Transduction , Adenocarcinoma/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CCL20/metabolism , Dinoprost/pharmacology , Endometrial Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Models, Biological , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR6/genetics , Receptors, CCR6/metabolism , Signal Transduction/drug effects
20.
FASEB J ; 25(2): 569-75, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20959513

ABSTRACT

The purpose of this study was to investigate the role of lipoxin A(4), an anti-inflammatory and proresolution modulator, during human parturition. We measured serum levels of lipoxin A(4) and myometrial protein release using ELISA, quantified lipoxin receptor (FPR2/ALX) mRNA expression using qRT-PCR, and localized protein expression using immunohistochemstry in myometrial biopsies from pregnant women. In addition, we compared the effects of lipoxin A(4) (100 nM) with vehicle on basal and LPS-stimulated expression of proinflammatory cytokines from samples of myometrium from pregnant women. Mean ± SE circulating level of lipoxin A(4) was 5.89 ± 0.63 nM at 24-wk gestation, with a further modest increase during pregnancy (P<0.05), but no differences in gestation matched women before and after labor (P>0.05). Levels of lipoxin A(4) in nonpregnant women were 0.48 ± 0.04 nM, significantly lower than in pregnant women (P<0.001). FPR2/ALX localized to myocytes and neutrophils, with a 9-fold increase in mRNA expression in labor (P<0.001). Lipoxin A(4) significantly reduced LPS-induced but not basal expression of the proinflammatory cytokines IL-6 and IL-8 in cultured myometrium (P<0.05), compared to vehicle-treated controls. We demonstrate for the first time a potential role for lipoxin A(4) and its receptor in the resolution of the inflammatory events of both physiological and pathological labor.


Subject(s)
Labor, Obstetric/physiology , Lipoxins/blood , Lipoxins/metabolism , Parturition/physiology , Adult , Female , Gene Expression Regulation/physiology , Humans , Lipopolysaccharides , Myometrium/drug effects , Myometrium/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/genetics , Receptors, Lipoxin/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...