Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Hum Reprod ; 20(11): 1144-57, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25103627

ABSTRACT

The interplay between inflammation, cervical cancer and HIV acquisition in women is poorly understood. We have previously shown that seminal plasma (SP) can promote cervical tumour cell growth in vitro and in vivo via the activation of potent inflammatory pathways. In this study, we investigated whether SP could regulate expression of chemokine receptors with known roles in HIV infection, in the cervix and in cervical cancer. The expression of CD4 and CCR5 was investigated by RT-PCR analysis and immunohistochemistry. CD4 and CCR5 expression was elevated in cervical cancer tissue compared with normal cervix. Ex vivo studies conducted on cervical tissues and HeLa cells showed that SP significantly increases the expression of CD4 and CCR5 transcripts. Furthermore, it was found that SP also up-regulates CCR5 protein in HeLa cells. The regulation of CCR5 expression was investigated following treatment of HeLa cells with SP in the presence/absence of chemical inhibitors of intracellular signalling, EP2 and EP4 antagonists, prostaglandin (PG) E2 and a cyclooxygenase (COX)-1 doxycycline-inducible expression system. These experiments demonstrated that the regulation of CCR5 expression by SP occurs via the epidermal growth factor receptor (EGFR)-COX-1-PGE2 pathway. This study provides a link between activation of inflammatory pathways and regulation of HIV receptor expression in cervical cancer cells.


Subject(s)
Receptors, CCR5/metabolism , Semen/metabolism , Up-Regulation , Uterine Cervical Neoplasms/genetics , Adult , CD4 Antigens/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 1/physiology , Female , HeLa Cells , Humans , Immunohistochemistry , Male , RNA, Messenger/metabolism , Receptors, CCR5/genetics , Uterine Cervical Neoplasms/metabolism
2.
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
3.
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
4.
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
5.
Am J Pathol ; 176(1): 435-45, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008143

ABSTRACT

Interleukin-11 (IL-11) up-regulates the proliferative and invasive capacity of many cancers. Coexpression of glycoprotein 130 (GP130) and IL-11 receptor alpha (IL-11Ralpha) is necessary for high-affinity binding of IL-11 to IL-11Ralpha. This study investigated the expression of IL-11 and role of prostaglandin F(2alpha)-F-prostanoid receptor (FP receptor) signaling in the modulation of IL-11 expression in endometrial adenocarcinoma cells. Localization of IL-11, IL-11Ralpha, and GP130 expression was performed by immunohistochemistry. IL-11 and regulator of calcineurin 1 isoform 4 (RCAN1-4) mRNA and protein expression were determined by real-time RT-PCR and/or enzyme-linked immunosorbent assay/Western blot analysis using Ishikawa endometrial adenocarcinoma cells stably expressing the FP receptor (FPS cells) and endometrial adenocarcinoma explants. IL-11 mRNA expression was significantly elevated in endometrial adenocarcinoma samples compared with normal endometrium and increased with tumor grade. IL-11 protein expression localized with FP receptor, IL-11Ralpha, and GP130 in the neoplastic glandular epithelium of endometrial adenocarcinomas. Prostaglandin F(2alpha)-FP receptor signaling significantly elevated the expression of IL-11 mRNA and protein in a Gq-protein kinase C-calcium-calcineurin-nuclear factor of activated T cells-dependent manner in FPS cells. The calcineurin signaling pathway is known to be controlled by the RCAN (RCAN1-4). Indeed, RCAN1-4 expression was significantly elevated in well-differentiated endometrial adenocarcinoma compared with normal endometrium and was found to decrease with tumor grade and negatively regulate IL-11 expression in vitro. This study has highlighted a new mechanism regulating IL-11 expression in endometrial adenocarcinoma cells by the FP receptor via the calcium-calcineurin-nuclear factor of activated T cells pathway.


Subject(s)
Calcineurin/metabolism , Calcium/metabolism , Endometrial Neoplasms/genetics , Interleukin-11/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , NFATC Transcription Factors/metabolism , Receptors, Prostaglandin/metabolism , Aged , Cell Differentiation , Cell Proliferation , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , DNA-Binding Proteins , Endometrial Neoplasms/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Interleukin-11/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Middle Aged , Models, Biological , Muscle Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-11/genetics , Receptors, Interleukin-11/metabolism
6.
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
7.
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
8.
Biochim Biophys Acta ; 1793(12): 1917-28, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19819266

ABSTRACT

Pro-inflammatory mediators, like prostaglandin (PG) and chemokines, promote tumourigenesis by enhancing cell proliferation, migration of immune cells and recruitment of blood vessels. Recently we showed elevated expression of the chemokine (C-X-C motif) receptor 2 (CXCR2) in endometrial adenocarcinomas localized to neutrophils and neoplastic epithelial and vascular cells. Furthermore we found that PGF(2alpha)-F-prostanoid (FP) receptor regulates the expression of the CXCR2 ligand CXCL1, to promote neutrophil chemotaxis in endometrial adenocarcinomas. In the present study we identified another CXCR2 ligand, CXCL8 as a target for PGF(2alpha)-FP receptor signalling which enhances epithelial cell proliferation in endometrial adenocarcinoma cells in vitro and in nude mice in vivo. We found that PGF(2alpha)-FP receptor interaction induces CXCL8 expression in endometrial adenocarcinoma cells via the protein kinase C-calcium-calcineurin-NFAT signaling pathway. Promoter analysis revealed that CXCL8 transcriptional activation by PGF(2alpha) signaling is mediated by cooperative interactions between the AP1 and NFAT binding sites. Furthermore, PGF(2alpha) via the FP receptor induced the expression of the regulator of calcineurin 1 isoform 4 (RCAN1-4) via the calcineurin/NFAT pathway in a reciprocal manner to CXCL8. Using an adenovirus to overexpress RCAN1-4, we found that RCAN1-4 is a negative regulator of CXCL8 expression in endometrial adenocarcinoma cells. Taken together our data have elucidated the molecular and cellular mechanism whereby PGF(2alpha) regulates CXCL8 expression via the FP receptor in endometrial adenocarcinomas and have highlighted RCAN1-4 as a negative regulator of CXCL8 expression which may be exploited therapeutically to inhibit CXCL8-mediated tumour development.


Subject(s)
Adenocarcinoma/metabolism , Calcineurin/metabolism , Calcium/metabolism , Endometrial Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Interleukin-8/biosynthesis , NFATC Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Receptors, Prostaglandin/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Dinoprost/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Protein Kinase C/metabolism , Response Elements , Transplantation, Heterologous
9.
BMC Cell Biol ; 11: 8, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-20092633

ABSTRACT

BACKGROUND: Prostaglandin (PG) F(2alpha) is a key regulator of endometrial function and exerts its biological action after coupling with its heptahelical G protein-coupled receptor (FP receptor). In endometrial adenocarcinoma the FP receptor expression is elevated. We have shown previously that PGF(2alpha)-FP receptor signalling in endometrial adenocarcinoma cells can upregulate several angiogenic factors including fibroblast growth factor-2 (FGF2). In the present study, we investigated the paracrine effect of conditioned medium produced via PGF(2alpha)-FP receptor signalling in endometrial adenocarcinoma cells stably expressing the FP receptor (Ishikawa FPS cells), on endothelial cell function. RESULTS: Conditioned medium (CM) was collected from FPS cells after 24 hrs treatment with either vehicle (V CM) or 100 nM PGF(2alpha) (P CM). Treatment of human umbilical vein endothelial cells (HUVECs) with P CM significantly enhanced endothelial cell differentiation (network formation) and proliferation. Using chemical inhibitors of intracellular signalling, we found that P CM-stimulated endothelial cell network formation was mediated by secretion of endothelial PGF(2alpha) and activation of endothelial FP receptors, following FGF2-FGFR1 signalling, phosphorylation of ERK1/2 and induction of COX-2. Whereas, P CM stimulation of endothelial cell proliferation occurred independently of PGF(2alpha) secretion via an FGF2-FGFR1-ERK1/2 dependent mechanism involving activation of the mTOR pathway. CONCLUSIONS: Taken together, we have shown a novel mechanism whereby epithelial prostaglandin F(2alpha)-FP signalling regulates endothelial cell network formation and proliferation. In addition we provide novel in vitro evidence to suggest that prostaglandin F(2alpha) can directly regulate endothelial cell network formation but not endothelial cell proliferation. These findings have relevance for pathologies where the FP receptor is aberrantly expressed, such as endometrial adenocarcinoma, and provide in vitro evidence to suggest that targeting the FP receptor could provide an anti-angiogenic approach to reducing tumour vasculature and growth.


Subject(s)
Endothelial Cells/cytology , Fibroblast Growth Factor 2/metabolism , Receptors, Prostaglandin/metabolism , Cell Differentiation , Cyclooxygenase 2/metabolism , Endothelial Cells/physiology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
10.
Mol Hum Reprod ; 16(3): 158-69, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19801577

ABSTRACT

Prokineticin-1 (PROK1) is a multifunctional secreted protein which signals via the G-protein coupled receptor, PROKR1. Previous data from our laboratory using a human genome survey microarray showed that PROK1-prokineticin receptor 1 (PROKR1) signalling regulates numerous genes important for establishment of early pregnancy, including the cytokine interleukin (IL)-11. Here, we have shown that PROK1-PROKR1 induces the expression of IL-11 in PROKR1 Ishikawa cells and first trimester decidua via the calcium-calcineurin signalling pathway in a guanine nucleotide-binding protein (G(q/11)), extracellular signal-regulated kinases, Ca(2+) and calcineurin-nuclear factor of activated T cells dependent manner. Conversely, treatment of human decidua with a lentiviral miRNA to abolish endogenous PROK1 expression results in a significant reduction in IL-11 expression and secretion. Importantly, we have also shown a regulatory role for the regulator of calcineurin 1 isoform 4 (RCAN1-4). Overexpression of RCAN1-4 in PROKR1 Ishikawa cells using an adenovirus leads to a reduction in PROK1 induced IL-11 indicating that RCAN1-4 is a negative regulator in the calcineurin-mediated signalling to IL-11. Finally, we have shown the potential for both autocrine and paracrine signalling in the human endometrium by co-localizing IL-11, IL-11Ralpha and PROKR1 within the stromal and glandular epithelial cells of non-pregnant endometrium and first trimester decidua. Overall we have identified and characterized the signalling components of a novel PROK1-PROKR1 signalling pathway regulating IL-11.


Subject(s)
Calcineurin/physiology , Gene Expression Regulation/drug effects , Interleukin-11/metabolism , NFATC Transcription Factors/physiology , Receptors, G-Protein-Coupled/physiology , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/physiology , Calcineurin/metabolism , Calcineurin Inhibitors , Cell Line, Tumor , Cyclosporine/pharmacology , Decidua/metabolism , Egtazic Acid/pharmacology , Endometrium/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flavonoids/pharmacology , Humans , Immunohistochemistry , Interleukin-11/genetics , NFATC Transcription Factors/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Pregnancy , Pregnancy Trimester, First , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/genetics , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/pharmacology
11.
BMC Cancer ; 10: 488, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20840749

ABSTRACT

BACKGROUND: An increase in cancer cell invasion and microvascular density is associated with a poorer prognosis for patients with endometrial cancer. In endometrial adenocarcinoma F-prostanoid (FP) receptor expression is elevated, along with its ligand prostaglandin (PG)F2α, where it regulates expression and secretion of a host of growth factors and chemokines involved in tumorigenesis. This study investigates the expression, regulation and role of a disintegrin and metalloproteinase with thrombospondin repeat 1 (ADAMTS1) in endometrial adenocarcinoma cells by PGF2α via the FP receptor. METHODS: Human endometrium and adenocarcinoma tissues were obtained in accordance with Lothian Research Ethics Committee guidance with informed patient consent. Expression of ADAMTS1 mRNA and protein in tissues was determined by quantitative RT-PCR analysis and immunohistochemistry. Signal transduction pathways regulating ADAMTS1 expression in Ishikawa cells stably expressing the FP receptor to levels seen in endometrial cancer (FPS cells) were determined by quantitative RT-PCR analysis. In vitro invasion and proliferation assays were performed with FPS cells and human umbilical vein endothelial cells (HUVECs) using conditioned medium (CM) from PGF2α-treated FPS cells from which ADAMTS1 was immunoneutralised and/or recombinant ADAMTS1. The role of endothelial ADAMTS1 in endothelial cell proliferation was confirmed with RNA interference. The data in this study were analysed by T-test or ANOVA. RESULTS: ADAMTS1 mRNA and protein expression is elevated in endometrial adenocarcinoma tissues compared with normal proliferative phase endometrium and is localised to the glandular and vascular cells. Using FPS cells, we show that PGF2α-FP signalling upregulates ADAMTS1 expression via a calmodulin-NFAT-dependent pathway and this promotes epithelial cell invasion through ECM and inhibits endothelial cell proliferation. Furthermore, we show that CM from FPS cells regulates endothelial cell ADAMTS1 expression in a rapid biphasic manner. Using RNA interference we show that endothelial cell ADAMTS1 also negatively regulates cellular proliferation. CONCLUSIONS: These data demonstrate elevated ADAMTS1 expression in endometrial adenocarcinoma. Furthermore we have highlighted a mechanism whereby FP receptor signalling regulates epithelial cell invasion and endothelial cell function via the PGF2α-FP receptor mediated induction of ADAMTS1.


Subject(s)
ADAM Proteins/metabolism , Adenocarcinoma/pathology , Cell Movement , Dinoprost/metabolism , Endometrial Neoplasms/pathology , Endometrium/pathology , Receptors, Prostaglandin/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAMTS1 Protein , Adenocarcinoma/metabolism , Adult , Aged , Apoptosis , Blotting, Western , Calmodulin/genetics , Calmodulin/metabolism , Cell Adhesion , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned/pharmacology , Dinoprost/genetics , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Middle Aged , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Receptors, Prostaglandin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Umbilical Veins/cytology , Umbilical Veins/metabolism , Young Adult
12.
Reproduction ; 138(6): 903-19, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19793840

ABSTRACT

Inflammation involves alterations to vascular and immune cell function. It is well recognised that many physiological reproductive events such as ovulation, menstruation, implantation and onset of labour display hallmark signs of inflammation. These are orchestrated by specific molecular pathways involving a host of growth factors, cytokines, chemokines and lipid mediators. Resumption of normal reproductive function involves prompt and proper resolution of these inflammatory pathways. Recent literature confirms that resolution of inflammatory pathways involves specific biochemical events that are activated to re-establish homeostasis in the affected tissue. Moreover, initiation and maintenance of inflammatory pathways are the key components of many pathologies of the reproductive tract and elsewhere in the body. The onset of reproductive disorders or disease may be the result of exacerbated activation and maintenance of inflammatory pathways or their dysregulated resolution. This review will address the role of inflammatory events in normal reproductive function and its pathologies.


Subject(s)
Disease/genetics , Inflammation Mediators/physiology , Inflammation/etiology , Reproduction/genetics , Animals , Female , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Models, Biological , Pregnancy , Pregnancy Complications/etiology , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Reproduction/physiology , Reproductive Health , Signal Transduction/genetics , Signal Transduction/physiology
13.
Mol Cell Endocrinol ; 285(1-2): 51-61, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18316157

ABSTRACT

In endometrial adenocarcinomas COX-2 and F-series prostanoid (FP) receptor expression and prostanoid biosynthesis (PGE(2) and PGF(2alpha)) are elevated. In the present study, we investigated the effect of PGE(2) and PGF(2alpha) on the expression of COX-2 via the FP receptor in endometrial adenocarcinoma cells stably expressing the FP receptor (FPS cells). Using chemical inhibitors of intracellular signaling pathways, reporter gene assays and quantitative RT-PCR analysis, we show that PGE(2) and PGF(2alpha) can mobilize inositol 1,4,5-trisphosphate, induce ERK1/2 phosphorylation via the phospholipase Cbeta-protein kinase A-epidermal growth factor receptor pathway and induce cyclooxygenase-2 (COX-2) expression via the FP receptor. In addition we show that the PGE(2) or PGF(2alpha)-regulation of COX-2 via the FP receptor is mediated via the cAMP response element (CRE) binding site on the COX-2 promoter. These data indicate that PGE(2) and PGF(2alpha) biosynthesized locally within endometrial adenocarcinomas can regulate tumor cell function in an autocrine/paracrine manner via the FP receptor.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprost/metabolism , Dinoprostone/metabolism , Gene Expression Regulation, Enzymologic , Response Elements , Signal Transduction/physiology , Adenocarcinoma/metabolism , Cell Line, Tumor , Cyclooxygenase 2/genetics , Dinoprost/analogs & derivatives , Endometrial Neoplasms/metabolism , Enzyme Activation , Enzyme Inhibitors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genes, Reporter , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Promoter Regions, Genetic , Prostaglandin Antagonists/metabolism , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E/metabolism , Xanthones/metabolism
14.
Endocrinology ; 148(8): 3635-44, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17478553

ABSTRACT

Prostaglandin (PG) F(2alpha) is a potent bioactive lipid in the female reproductive tract, and exerts its function after coupling with its heptahelical G-protein-coupled receptor [F-series-prostanoid (FP) receptor] to initiate cell signaling and target gene transcription. In the present study, we found elevated expression of fibroblast growth factor (FGF) 2, FGF receptor 1 (FGFR1), and FP receptor, colocalized within the neoplastic epithelial cells of endometrial adenocarcinomas. We investigated a role for PGF(2alpha)-FP receptor interaction in modulating FGF2 expression and signaling using an endometrial adenocarcinoma cell line stably expressing the FP receptor to the levels detected in endometrial adenocarcinomas (FPS cells) and endometrial adenocarcinoma tissue explants. PGF(2alpha)-FP receptor activation rapidly induced FGF2 mRNA expression, and elevated FGF2 protein expression and secretion into the culture medium in FPS cells and endometrial adenocarcinoma explants. The effect of PGF(2alpha) on the expression and secretion of FGF2 could be abolished by treatment of FPS cells and endometrial tissues with an FP receptor antagonist (AL8810) and inhibitor of ERK (PD98059). Furthermore, we have shown that FGF2 can promote the expression of FGF2 and cyclooxygenase-2, and enhance proliferation of endometrial adenocarcinoma cells via the FGFR1 and ERK pathways, thereby establishing a positive feedback loop to regulate neoplastic epithelial cell function in endometrial adenocarcinomas.


Subject(s)
Adenocarcinoma/metabolism , Endometrial Neoplasms/metabolism , Fibroblast Growth Factor 2/metabolism , MAP Kinase Signaling System/physiology , Receptors, Prostaglandin/metabolism , Adenocarcinoma/pathology , Adult , Aged , Autocrine Communication/physiology , Cell Line, Tumor , Cells, Cultured , Cyclooxygenase 2/metabolism , Dinoprost/metabolism , Dinoprost/pharmacology , Endometrial Neoplasms/pathology , Endometrium/cytology , Endometrium/physiology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological/physiology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Middle Aged , Paracrine Communication/physiology , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Prostaglandin/genetics
15.
Cancer Res ; 65(17): 7707-16, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140938

ABSTRACT

Prostaglandins have been implicated in several neovascular diseases. In the present study, we found elevated FP receptor and vascular endothelial growth factor (VEGF) expression colocalized in glandular epithelial and vascular cells lining the blood vessels in endometrial adenocarcinomas. We investigated the signaling pathways activated by the FP receptor and their role in modulating VEGF expression in endometrial adenocarcinoma (Ishikawa) cells. Ishikawa cells were stably transfected with FP receptor cDNA in the sense or antisense orientations. Treatment of Ishikawa cells with prostaglandin F2alpha (PGF2alpha) rapidly induced transphosphorylation of the epidermal growth factor receptor (EGFR) and phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 via the FP receptor. Activation of EGFR-Ras-mitogen-activated protein kinase/ERK kinase (MEK) signaling via the FP receptor resulted in an increase in VEGF promoter activity, expression of VEGF mRNA, and secretion of VEGF protein. These effects of PGF2alpha on the FP receptor could be abolished by treatment of cells with a specific FP receptor antagonist, chemical inhibitors of c-Src, matrix metalloproteinase, and EGFR kinase or by inactivation of signaling with dominant-negative mutant isoforms of EGFR, Ras, or MEK or with small inhibitory RNA oligonucleotides targeted against the EGFR. Finally, we confirmed that PGF2alpha could potentiate angiogenesis in endometrial adenocarcinoma explants by transactivation of the EGFR and induction of VEGF mRNA expression.


Subject(s)
Adenocarcinoma/blood supply , Adenocarcinoma/metabolism , Dinoprost/metabolism , Endometrial Neoplasms/blood supply , Endometrial Neoplasms/metabolism , MAP Kinase Signaling System/physiology , Receptors, Prostaglandin/metabolism , Aged , Dinoprost/pharmacology , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Middle Aged , Neovascularization, Pathologic/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Prostaglandin/biosynthesis , Transcriptional Activation , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Endocrinology ; 147(7): 3356-65, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16574793

ABSTRACT

E-series prostanoid (EP)4 receptor is up-regulated in numerous cancers, including cervical carcinomas, and has been implicated in mediating the effects of prostaglandin (PG)E(2) in tumorigenesis. In addition to regulation by endogenously biosynthesized PGE(2), neoplastic cervical epithelial cells in sexually active women may also be regulated by PGs present in seminal plasma. In this study, we investigated the signal transduction pathways mediating the role of seminal plasma and PGE(2) in the regulation of tumorigenic and angiogenic genes via the EP4 receptor in cervical adenocarcinoma (HeLa) cells. HeLa cells were stably transfected with EP4 receptor in the sense orientation. Seminal plasma and PGE(2) signaling via the EP4 receptor induced the activation of cyclooxygenase (COX)-2 and vascular endothelial growth factor (VEGF) promoters, expression of COX-2 and VEGF mRNA and protein, and secretion of VEGF protein into the culture medium. Treatment of HeLa cells with seminal plasma or PGE(2) also rapidly induced the phosphorylation of ERK1/2 via the EP4 receptor. Preincubation of cells with a specific EP4 receptor antagonist (ONO-AE2-227) or chemical inhibitors of epidermal growth factor receptor (EGFR) tyrosine kinase or MAPK kinase or cotransfection of cells with dominant-negative mutant cDNA targeted against the EGFR, serine/threonine kinase Raf, or MAPK kinase abolished the EP4-induced activation of COX-2, VEGF, and ERK1/2. Therefore, we have demonstrated that seminal plasma and PGE(2) can promote the expression of tumorigenic and angiogenic factors, in cervical adenocarcinoma cells via the EP4 receptor, EGFR, and ERK1/2 signaling pathways.


Subject(s)
Adenocarcinoma/metabolism , Neovascularization, Pathologic , Receptors, Prostaglandin E/metabolism , Semen/physiology , Uterine Cervical Neoplasms/metabolism , Cyclooxygenase 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , HeLa Cells , Humans , Male , Phosphorylation , Prostaglandins/metabolism , Receptors, Prostaglandin E, EP4 Subtype , Vascular Endothelial Growth Factor A/metabolism
17.
Endocrinology ; 147(4): 1697-705, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16373414

ABSTRACT

Prostacyclin (PGI) is a member of the prostanoid family of lipid mediators that mediates its effects through a seven-transmembrane G protein-coupled receptor (IP receptor). Recent studies have ascertained a role for prostanoid-receptor signaling in angiogenesis. In this study we examined the temporal-spatial expression of the IP receptor within normal human endometrium and additionally explored the signaling pathways mediating the role of IP receptor in activation of target angiogenic genes. Quantitative RT-PCR analysis demonstrated the highest endometrial expression of the IP receptor during the menstrual phase compared with all other stages of the menstrual cycle. Immunohistochemical analysis localized the site of IP receptor expression to the glandular epithelial compartment with stromal and perivascular cell immunoreactivity. Expression of the immunoreactive IP receptor protein was greatest during the proliferative and early secretory phases of the menstrual cycle. To explore the role of the IP receptor in glandular epithelial cells, we used the Ishikawa endometrial epithelial cell line. Stimulation of Ishikawa cells and human endometrial biopsy explants with 100 nm iloprost (a PGI analog) rapidly activated ERK1/2 signaling and induced the expression of proangiogenic genes, basic fibroblast growth factor, angiopoietin-1, and angiopoietin-2, in an epidermal growth factor receptor (EGFR)-dependent manner. Furthermore, EGFR colocalized with IP receptor in the glandular epithelial compartment. These data suggest that PGI-IP interaction within glandular epithelial cells can promote the expression of proangiogenic genes in human endometrium via cross talk with the EGFR.


Subject(s)
Endometrium/metabolism , ErbB Receptors/physiology , Gene Expression Regulation , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Receptors, Epoprostenol/physiology , Angiopoietin-1/genetics , Angiopoietin-2/genetics , Cell Line, Tumor , Female , Fibroblast Growth Factor 2/genetics , Humans , Iloprost/pharmacology , Phosphorylation , Receptors, Epoprostenol/genetics , Signal Transduction , Up-Regulation
18.
Endocrinology ; 147(2): 744-53, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16282352

ABSTRACT

The menstrual cycle is a complex interaction of sex steroids, prostanoids, and cytokines that lead to coordinated tissue degradation, regeneration and repair. The transcription factor hypoxia-inducible factor (HIF-1) plays critical roles in cellular responses to hypoxia, the generation of an inflammatory response and vasculogenesis through transcriptional activation of angiogenic genes. We hypothesize that HIF-1 is expressed in human endometrium and that locally synthesized prostaglandins (PGE2 and PGF(2alpha)) regulate HIF-1 activity. Here we demonstrate that PGE2 up-regulates HIF-1alpha mRNA and protein via the E-series prostanoid receptor 2 (EP2), and this up-regulation is dependent on epidermal growth factor receptor kinase activity. We show the tight temporal-spatial confinement of HIF-1alpha protein expression in endometrium across the cycle. HIF-1alpha is expressed exclusively during the secretory and menstrual phases. Protein expression is maximal at progesterone withdrawal during the late secretory and menstrual phase. HIF-1alpha protein colocalizes with prostaglandin EP2 receptor in glandular cells. In contrast, HIF-1beta/aryl receptor nuclear translocator 1 expression occurs throughout the cycle but is maximal in glandular cells during the proliferative phase. This provides evidence for a role for HIF-1 in the menstrual cycle and demonstrates that HIF-1 activation in human endometrium may occur via a PGE2-regulated pathway and provides a coordinated pathway from progesterone withdrawal through to angiogenic gene expression via HIF-1.


Subject(s)
Endometrium/metabolism , ErbB Receptors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Prostaglandins E/metabolism , Receptors, Prostaglandin E/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Menstrual Cycle/metabolism , RNA, Messenger/analysis , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Statistics, Nonparametric , Up-Regulation
19.
Mol Cell Endocrinol ; 252(1-2): 191-200, 2006 Jun 27.
Article in English | MEDLINE | ID: mdl-16701939

ABSTRACT

Prostaglandins are bioactive lipids produced from arachidonic acid by cyclooxygenase enzymes and specific terminal prostanoid synthase enzymes. Following biosynthesis, prostaglandins exert an autocrine/paracrine function by coupling to specific prostanoid G protein-coupled receptors to activate intracellular signaling and gene transcription. For many years prostaglandins have been recognised as key molecules in reproductive biology by regulating ovulation, endometrial physiology and proliferation of endometrial glands and menstruation. More recently a role for COX enzymes and prostaglandins has been ascertained in reproductive tract pathology, including dysmenorrhea, endometriosis, menorrhagia and cancer. Emerging evidence supports a role for COX enzymes, prostaglandins and prostaglandin receptor signaling pathways in a multitude of phenotypic changes in reproductive tissues including the promotion of angiogenesis and vascular function. Here we provide an overview of some of the findings from these studies with specific emphasis on the role of cyclooxygenase enzymes, prostaglandins and their receptors in benign and neoplastic pathologies of the human endometrium.


Subject(s)
Blood Vessels/physiology , Endometrium/enzymology , Endometrium/pathology , Neovascularization, Physiologic/physiology , Receptors, Prostaglandin/physiology , Animals , Dysmenorrhea/pathology , Electron Transport Complex IV/metabolism , Endometrial Neoplasms/pathology , Endometriosis/pathology , Female , Humans , Menorrhagia/pathology , Menstrual Cycle/physiology , Models, Biological , Prostaglandins/physiology
20.
Cancer Res ; 62(2): 424-32, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11809691

ABSTRACT

This study was designed to investigate the expression and molecular signaling of cyclooxygenase-1 (COX-1) in cervical carcinomas. Real-time quantitative reverse transcription-polymerase chain reaction and Western blot analysis confirmed enhanced expression of COX-1 RNA, and protein in squamous cell carcinomas and adenocarcinoma of the cervix. COX-1 expression in all carcinoma tissues was associated with enhanced expression of COX-2 RNA and protein. The site of COX-1 expression was localized by immunohistochemistry to the neoplastic epithelial cells in all squamous cell carcinomas and adenocarcinomas studied. Minimal COX-1 immunoreactivity was detected in normal cervix. To explore events associated with COX-1 up-regulation, we developed a doxycycline-regulated expression system in HeLa (cervical carcinoma) cells. Overexpression of COX-1 in HeLa cells resulted in induced expression of cyclooxygenase-2 (COX-2) and prostaglandin E synthase (PGES) concomitant with increased prostaglandin E(2) (PGE(2)) synthesis. Treatment of HeLa cells overexpressing COX-1 with the dual COX enzyme inhibitor indomethacin or selective COX-2 inhibitor NS-398 significantly reduced PGE(2) synthesis. Indomethacin, but not NS-398, treatment abolished the up-regulation of expression of COX-2 and PGES in HeLa cells, suggesting that the observed up-regulation of COX-2 and PGES was mediated by COX-1-enzyme products. To assess whether enhanced PGE(2) synthesis after COX-1 induction would act in an autocrine/paracrine manner, we investigated the effect of COX-1 on the expression of the different isoforms of PGE(2) receptors (EP1-EP4). We found that the cAMP-linked PGE(2) receptors were significantly up-regulated by COX-1 overexpression coincident with enhanced cAMP responsiveness of these cells to exogenous PGE(2) ligand. Finally, overexpression of COX-1 was associated with enhanced expression of the angiogenic factors basic fibroblast growth factor, vascular endothelial growth factor, angiopoietin-1, and angiopoietin-2. This up-regulation of angiogenic factor expression was abolished by indomethacin and partially reduced by NS-398. These data indicate that COX-1 up-regulation modulates the expression of factors that may act in an autocrine/paracrine manner to enhance and sustain tumorigenesis in neoplastic cervical epithelial cells. It is likely that similar mechanisms may act in vivo to modulate tumorigenesis of cervical carcinomas.


Subject(s)
Angiogenesis Inducing Agents/biosynthesis , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Receptors, Prostaglandin E/biosynthesis , Uterine Cervical Neoplasms/enzymology , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Cervix Uteri/enzymology , Cyclic AMP/biosynthesis , Cyclooxygenase 1 , Cyclooxygenase 2 , Dinoprostone/pharmacology , Endothelial Growth Factors/biosynthesis , Epithelial Cells/enzymology , Female , Fibroblast Growth Factor 2/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Intramolecular Oxidoreductases/biosynthesis , Isoenzymes/genetics , Isoenzymes/physiology , Lymphokines/biosynthesis , Membrane Proteins , Prostaglandin-E Synthases , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/physiology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Uterine Cervical Neoplasms/genetics , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL