Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
Hum Reprod ; 27(4): 1112-21, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22328559

ABSTRACT

BACKGROUND: The human endometrium efficiently repairs each month after menstruation. The mechanisms involved in this repair process remain undefined. Aberrations in endometrial repair may lead to the common disorder of heavy menstrual bleeding. We hypothesized that connective tissue growth factor (CTGF) is increased at the time of endometrial repair post-menses and that this increase is regulated by prostaglandins (PGs) and hypoxic conditions present during menstruation. METHODS AND RESULTS: Examination of 41 endometrial biopsies from 5 stages of the menstrual cycle revealed maximal CTGF mRNA expression (using quantitative RT-PCR) at menstruation and peak protein levels during the proliferative phase. CTGF was immunolocalized to epithelial and stromal cells, with intense staining of occasional stromal cells during the proliferative phase. Dual immunohistochemistry identified these cells as macrophages. Treatment of endometrial epithelial cells with 100 nM PGE(2), PGF(2α) or hypoxia (0.5% O(2)) revealed a significant increase in CTGF mRNA expression (P < 0.01 for all, versus vehicle control). Cells treated simultaneously with PGE(2) and hypoxia revealed a synergistic increase in CTGF expression (P < 0.05 versus PGE(2) or hypoxia alone) and maximal secreted CTGF protein levels (P < 0.05 versus control). CONCLUSIONS: CTGF is increased in the human endometrium at the time of endometrial repair post-menses. The increase in CTGF may be mediated by PG production and the transient hypoxic episode observed in the endometrium at menstruation.


Subject(s)
Connective Tissue Growth Factor/metabolism , Endometrium/metabolism , Cell Hypoxia , Connective Tissue Growth Factor/analysis , Connective Tissue Growth Factor/genetics , Female , Gene Expression Regulation/drug effects , Humans , Menstrual Cycle/metabolism , Menstruation/metabolism , Prostaglandins/pharmacology , RNA, Messenger/metabolism
2.
Placenta ; 32(3): 247-54, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21232790

ABSTRACT

BACKGROUND: Pre-gravid obesity is associated with increased morbidity and mortality for both mother and offspring. Recent studies have demonstrated a heightened inflammatory response both systemically and locally within the adipose and placental tissue in women with pre-gravid obesity, which may play a role in mediating the adverse pregnancy outcomes. The aim of this study was to characterise the maternal and placental inflammatory status and investigate associated changes in placental structure in obese women. METHODS: The pro-inflammatory status of a cohort of 47 non-obese (BMI 20-25 kg/m(2)) and 33 obese (≥30 kg/m(2)) women was characterised by measuring maternal circulating levels and placental gene expression of pro-inflammatory cytokines, and quantifying immune cell populations within the placenta. The effect of pre-gravid obesity on placental structure was investigated by examining placental maturity, vessel density, the formation of syncytial knots and sprouts, and the degree of fibrin deposition, chorangiosis and muscularisation of vessel walls. RESULTS: Maternal obesity was associated with significantly greater IL-1ß (p < 0.05), IL-8 (p < 0.05), MCP-1 (p < 0.001) and CXCR2 (p < 0.05) mRNA expression within the placenta and higher circulating maternal levels of IL-6 (3.30 ± 0.38 vs. 1.77 ± 0.15 pg/ml) (p < 0.001) compared with non-obese women. There were no differences in the number of CD14(+), CD68(+) cells or neutrophils within the placental villi of non-obese and obese women. However there were significantly higher numbers of neutrophils within the interstitial space (p < 0.05). Greater muscularity of placental vessel walls was associated with maternal obesity (p = 0.03), however no other associated structural changes were observed. CONCLUSIONS: Our findings show that although pre-gravid obesity was associated with greater expression of placental pro-inflammatory cytokines and higher circulating IL-6 in pregnancy, there were no major differences in immune cell populations within the placental villi and only a greater degree of muscularity in the vessel walls.


Subject(s)
Cytokines/immunology , Inflammation/immunology , Obesity/immunology , Placenta/immunology , Pregnancy Complications/immunology , Adult , Cell Count , Cohort Studies , Cytokines/genetics , Female , Histocytochemistry , Humans , Macrophages/immunology , Neutrophils/immunology , Placenta/cytology , Pregnancy , Pregnancy Complications/pathology , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
3.
Mol Cell Endocrinol ; 335(1): 52-9, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-20708653

ABSTRACT

Human parturition is an inflammatory event, modulated and influenced by a host of other environmental and physiological processes, including the endocrine hormones. Complex bidirectional communication occurs between the two systems to bring about some of the changes that are seen in labour, an event that is not yet fully understood. Preterm birth is a major problem in obstetrics and neonatology, with dysfunctional labour or prolonged pregnancy also making increasingly significant contributions to maternal morbidity. With better understanding of normal and abnormal parturition we may be able to develop novel ways of treating these complications of pregnancy and reduce maternal and neonatal morbidity and mortality. This review discusses the crucial role that endocrine-immune interaction plays in the process of labour and in the processes of abnormal and preterm labour. We propose that amongst these complex interactions it is the immune system that is the driving force behind human parturition.


Subject(s)
Inflammation/immunology , Parturition/immunology , Cervix Uteri/cytology , Cervix Uteri/immunology , Cervix Uteri/metabolism , Cytokines/metabolism , Extraembryonic Membranes/immunology , Extraembryonic Membranes/metabolism , Female , Hormones/metabolism , Humans , Myometrium/cytology , Myometrium/immunology , Myometrium/metabolism , Parturition/metabolism , Pregnancy , Premature Birth/immunology , Premature Birth/prevention & control
4.
Cell Signal ; 22(1): 71-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19782748

ABSTRACT

Prostaglandins exert their effects on target cells by coupling to specific G protein-coupled receptors (GPCRs) that are often co-expressed in the same cells and use alternate and in some cases opposing intracellular signaling pathways. This study investigated the cross-talk that influences intracellular signaling and gene expression profiling in response to co-activation of the EP2 and FP prostanoid receptors in Ishikawa cells stably expressing both receptors (FPEP2 cells). In this study we show that in FPEP2 cells, PGF alone does not alter adenosine 3',5'-cyclic monophosphate (cAMP) production, but in combination with Butaprost enhances EP2 receptor mediated cAMP release compared to treatment with Butaprost alone. PGF-mediated potentiation of cAMP release was abolished by antagonism of the FP receptor, inhibition of phospholipase C (PLC) and inositol phosphate receptor (IP3R) whereas inhibition of protein kinase C (PKC) had no effect. Moreover, inhibition of calcium effectors using calmodulin antagonist (W7) or Ca(2+)/calmodulin-dependent kinase II (CaMK-II) inhibitor (KN-93) abolished PGF potentiation of Butaprost-mediated cAMP release. Using siRNA molecules targeted against the adenylyl cyclase 3 (AC3) isoform, we show that AC3 is responsible for the cross-talk between the FP and EP2 receptors. Using gene array studies we have identified a candidate gene, Spermidine/N1-acetyltransferase (SAT1), which is regulated by this cAMP mediated cross-talk. In conclusion, this study demonstrates that co-activation of the FP and EP2 receptors results in enhanced release of cAMP via FP receptor-G alpha(q)-Ca(2+)-calmodulin pathway by activating calcium sensitive AC3 isoform.


Subject(s)
Calcium Signaling , Calmodulin/metabolism , Cyclic AMP/metabolism , Dinoprost/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line, Tumor , Enzyme Activation , Gene Expression Regulation , Humans , Oligonucleotide Array Sequence Analysis , Phosphoinositide Phospholipase C/metabolism , Receptors, Prostaglandin E, EP2 Subtype
6.
Oncogene ; 27(17): 2466-77, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-17968320

ABSTRACT

Cellular adhesion to extracellular matrix is a central phenomenon for the maintenance of tissue integrity and cellular movement. Collectively, these processes are regulated by a fine-tuned balance between the formation and loosening of adhesive contacts, a process involving integrins, and the elevation and diminution of cytoplasmic signalling molecules. We demonstrate that prostaglandin (PG) F(2alpha) stimulation rapidly increases the capacity of Ishikawa cells stably expressing the F-prostanoid receptor (FPS) to adhere to vitronectin. Coincident with this elevation in matrix adhesion, we demonstrate a profound PGF(2alpha)-induced alteration in cytoskeletal remodelling, characterized by polymerization of the actin cytoskeleton and recruitment of focal adhesion kinase at focal adhesions and enhanced cell migration. Moreover, we show that these PGF(2alpha)-induced alterations in adhesion and morphology on vitronectin and migration could be abolished by cultivating FPS cells in the presence of integrin alphavbeta3 antibody or alphavbeta3-directed tetrapeptide arg-gly-asp-ser or inhibition of FP receptor signalling with the FP receptor antagonist, chemical disruptors of the phospholipase C-beta, protein kinase A, c-Src and epidermal growth factor receptor kinase pathways or inhibition of the monomeric G proteins Rho, Rac and CDC42. These results reveal a mechanism by which prostanoids regulate cell movement, which may be relevant to pathologies of the endometrium.


Subject(s)
Cell Movement , Cell Shape , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Receptors, Prostaglandin/metabolism , Cell Adhesion , Cell Line, Tumor , Dinoprost/metabolism , Endometrial Neoplasms/genetics , Enzyme Activation , ErbB Receptors/metabolism , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin alphaVbeta3/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Prostaglandin/genetics , Vitronectin/metabolism
7.
Hum Reprod ; 22(5): 1450-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17264103

ABSTRACT

BACKGROUND: Although the mechanisms underlying the causes of heavy menstrual blood loss (MBL) remain to be elucidated, prostaglandins have been previously implicated. This study was initiated to elucidate a pattern of expression of the various components of the cyclooxygenase (COX)-prostaglandin signalling pathways present in the endometrium of women with normal and heavy MBLs. METHODS: Endometrial biopsies were collected at different stages of the menstrual cycle from women who underwent measurement of MBL. Tissue was divided for either examination of gene expression by quantitative RT-PCR analysis or in vitro culture experimentation. RESULTS: Analysis of gene expression demonstrated a significant elevation in expression of COX-1 and COX-2 mRNA in endometrium obtained from women with heavy MBL when compared with endometrium obtained from women with normal MBL. Tissue culture with PGE(2) stimulation caused a significantly elevated production of cyclic AMP (cAMP) by endometrium of women with heavy MBL when compared with normal MBL. Expression of phosphodiesterase 4B, an enzyme involved in cAMP breakdown, was reduced in these same endometrial samples obtained from women with heavy MBL. CONCLUSIONS: These data identify the E series prostaglandin receptors and their signalling pathways as potential therapeutic targets in the treatment of heavy menstruation.


Subject(s)
Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Menorrhagia/enzymology , Menorrhagia/physiopathology , Receptors, Prostaglandin E/physiology , Signal Transduction/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/physiology , Adult , Cyclic AMP/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 4 , Female , Follicular Phase/physiology , Humans , Luteal Phase/physiology , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
8.
Hum Reprod ; 22(1): 36-44, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16905765

ABSTRACT

BACKGROUND: Prostaglandin E(2) (PGE(2)) has been shown to modulate angiogenesis and tumour progression via the E-series prostanoid-2 (EP2) receptor. Endometrial adenocarcinomas may be exposed to endogenous PGE(2) and exogenous PGE(2), present at high concentration in seminal plasma. METHODS: This study investigated fibroblast growth factor 2 (FGF2) mRNA expression and cell signalling in response to seminal plasma or PGE(2), using an endometrial adenocarcinoma (Ishikawa) cell line stably expressing the EP2 receptor (EP2 sense cells) and endometrial adenocarcinoma explants. RESULTS: Seminal plasma and PGE(2) induced a significant up-regulation of FGF2 expression in EP2 sense but not parental untransfected Ishikawa (wild-type) cells (P < 0.05). These effects were inhibited by co-treatment with EP2 receptor antagonist or inhibitors of protein kinase A, c-Src, epidermal growth factor receptor (EGFR) kinase or extracellular signal-regulated kinase (ERK) signalling. The treatment of EP2 sense cells with seminal plasma induced cAMP accumulation and phosphorylation of c-Src, EGFR kinase and ERK via the EP2 receptor. Finally, seminal plasma and PGE(2) significantly increased FGF2 mRNA expression in endometrial adenocarcinoma tissue explants via the EP2 receptor (P < 0.05). CONCLUSIONS: Seminal plasma and PGE(2) can similarly activate FGF2 expression and EP2 receptor signalling in endometrial adenocarcinoma cells. These data highlight the potential for seminal plasma exposure to facilitate tumorigenesis-angiogenesis in endometrial adenocarcinomas in vivo.


Subject(s)
Adenocarcinoma/metabolism , Dinoprostone/physiology , Endometrial Neoplasms/metabolism , ErbB Receptors/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Fibroblast Growth Factor 2/biosynthesis , Receptors, Prostaglandin E/physiology , Semen/physiology , Cell Line, Tumor , Cyclic AMP/metabolism , Female , Gene Expression Regulation , Humans , Male , Microscopy, Confocal , Microscopy, Fluorescence , Phosphorylation/drug effects , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Signal Transduction , Transcriptional Activation/physiology , Up-Regulation
9.
Trends Endocrinol Metab ; 15(8): 398-404, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15380812

ABSTRACT

Prostaglandins are bioactive lipids that exert an autocrine or paracrine function by binding to specific G-protein-coupled receptors (GPCRs) to activate intracellular signalling and gene transcription. Prostaglandins are key regulators of reproductive processes, including ovulation, implantation and menstruation. Prostaglandins have been ascertained to have a role in various pathological changes of the reproductive tract including menorrhagia, dysmenorrhea, endometriosis and cancer. Although the mechanism by which prostaglandins modulate these changes remains unclear, much evidence suggests that prostaglandins and their receptors and downstream signalling pathways are involved in angiogenesis and in alterations in cell adhesion, morphology, motility, invasion and metastases. The potential role of prostaglandin receptors in pathological changes of the endometrium has significance for the future development of therapeutic interventions.


Subject(s)
Endometrium , Receptors, Prostaglandin/metabolism , Signal Transduction , Uterine Diseases/physiopathology , Animals , Female , Humans
10.
J Clin Endocrinol Metab ; 89(5): 2463-9, 2004 May.
Article in English | MEDLINE | ID: mdl-15126578

ABSTRACT

This study investigated the possible role of the newly discovered endocrine gland-derived vascular endothelial growth factors and their cognate receptors in the human endometrium during the menstrual cycle. Endocrine gland-derived vascular endothelial growth factors are also known as prokineticin (PK) 1 and PK2 and their receptors as PKR1 and PKR2. Expression of PK1 was elevated in the secretory compared with the proliferative phase of the menstrual cycle (P < 0.05). There was no temporal variation in expression of PK2, PKR1, or PKR2. PK1 and PK2 and their receptors were localized to multiple cellular compartments, including glandular epithelial, stromal, and endothelial cells in the endometrium and endothelial and smooth muscle cells in the myometrium. The elevation in PK1 expression in the secretory phase of the menstrual cycle indicated potential regulation of PK1 by progesterone. To investigate this, endometrial tissue was treated with 1 microM (micromol/liter(-1)) progesterone for 24 h, and PK1 expression was assessed by quantitative RT-PCR. Treatment with 1 microM ( micromol/liter(-1)) progesterone resulted in 2.91 +/- 0.75-fold elevation in PK1 expression, compared with controls (P < 0.05). These data identify a paracrine role for the PKs and their receptors in endometrial vascular function.


Subject(s)
Endometrium/metabolism , Gastrointestinal Hormones/metabolism , Menstrual Cycle/physiology , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/metabolism , Endometrium/cytology , Female , Follicular Phase/physiology , Humans , Luteal Phase/physiology , Myometrium/cytology , Myometrium/metabolism , Progesterone/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
11.
Reproduction ; 127(1): 79-86, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15056772

ABSTRACT

Prostacyclin (PGI(2)) synthesis and function in the human uterus has been implicated in the regulation of the process of normal and dysfunctional menstruation. PGI(2) synthesis is elevated during normal menstruation and is also associated with blood loss in women who suffer from heavy menses. This study was designed to outline further the role of PGI(2) in menstruation by investigating the temporal pattern and site of expression of prostaglandin I synthase (PGIS) and the prostacyclin receptor (IP receptor) in the non-pregnant human endometrium across the menstrual cycle. Quantitative RT-PCR demonstrated increased expression of PGIS and IP receptor during the menstrual phase of the cycle compared with all other phases (P < 0.05). Furthermore, PGIS and IP receptor were localised to the glandular epithelium, stromal and endothelial cells in the basal and functional layers of the endometrium. Functionality of the IP receptor in the human endometrium was assessed by measuring cAMP generation following treatment with 100 nmol l(-1) of the PGI(2) analogue, iloprost. cAMP generation was significantly higher in endometrial tissue collected during the proliferative compared with the secretory phase of the menstrual cycle (P < 0.05). In conclusion, this study has confirmed increased expression and signalling of PGIS and IP receptor during the menstrual phase and outlines a potential autocrine/paracrine role for PGI(2) on several cellular compartments in the endometrium including the endothelium. This may underscore a pivotal role for PGI(2) receptor signalling in normal and dysfunctional menstruation.


Subject(s)
Endometrium/metabolism , Menstrual Cycle/metabolism , Receptors, Epoprostenol/analysis , Signal Transduction/physiology , Cyclic AMP/metabolism , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Endometrium/chemistry , Epoprostenol/metabolism , Female , Humans , Iloprost/pharmacology , Immunohistochemistry/methods , In Situ Hybridization , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , RNA, Messenger/analysis , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Prostaglandins Other Lipid Mediat ; 71(3-4): 97-117, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14518555

ABSTRACT

Prostaglandins, thromboxanes (TX) and leukotrienes, collectively referred to as eicosanoids, are cyclooxygenase (COX) metabolites of arachidonic acid (AA). Prostaglandins, have been recognised for many years as key molecules in regulating reproductive tract physiology and pathology. Numerous recent studies in in vitro model systems and knockout mouse models have demonstrated specific functional roles for the respective cyclooxygenase enzymes, prostaglandins and prostanoid receptors. Here we review the findings obtained in several of these studies with emphasis on the roles played by cyclooxygenase enzymes and prostaglandins, specifically prostaglandin E2 (PGE2) and F2alpha in reproductive tract physiology and pathology.


Subject(s)
Genitalia, Female/physiology , Genitalia, Female/physiopathology , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , Animals , Dinoprost/metabolism , Dinoprostone/biosynthesis , Dinoprostone/metabolism , Female , Genital Neoplasms, Female/physiopathology , Humans , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/metabolism , Reproduction , Signal Transduction
13.
J Clin Endocrinol Metab ; 88(9): 4481-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12970327

ABSTRACT

In this study, we investigated the effect of prostaglandin E(2) (PGE(2)) on MAPK ERK1/2 protein phosphorylation and on proliferation of epithelial cells of the human endometrium. Treatment of proliferative phase endometrium with PGE(2) induced rapid phosphorylation of ERK1/2 proteins in glandular epithelial and endothelial cells. Treatment of human endometrial tissue with PGE(2) for 24 h resulted in increased incorporation of 5-bromo-2'-deoxyuridine (a marker of cellular proliferation) in glandular epithelial cells. To investigate further the effect of PGE(2) on proliferation of epithelial cells, we used an endometrial epithelial cell line (HES). HES cells express functional EP4 (with absence of expression of EP1, EP2, and EP3) receptors and stimulate cAMP release and rapid phosphorylation of ERK1/2 proteins in response to PGE(2) or forskolin. Treatment of HES cells with PGE(2) or forskolin alone resulted in a significant increase in HES cell proliferation compared with control untreated cells (P < 0.05). Cotreatment of the cells with PGE(2) or forskolin and PD98059 abolished the increase in cellular proliferation. These data demonstrate ERK1/2 phosphorylation in response to PGE(2) in the human endometrium and suggest that PGE(2) via EP4 receptor may induce glandular epithelial cell proliferation in ERK1/2- dependent manner during the proliferative phase of the menstrual cycle.


Subject(s)
Dinoprostone/pharmacology , Endometrium/cytology , Epithelial Cells/drug effects , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinases/physiology , Signal Transduction/drug effects , Adult , Antimetabolites/pharmacology , Blotting, Western , Bromodeoxyuridine/pharmacology , Cell Division/drug effects , Cyclic AMP/biosynthesis , Cyclic AMP/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Endometrium/drug effects , Female , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinase 3 , Phosphorylation , Receptors, Prostaglandin/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction
14.
Article in English | MEDLINE | ID: mdl-12445497

ABSTRACT

This study was designed to investigate the effect of IL-1alpha-induced up-regulation of cyclooxygenase-2 (COX-2) on prostaglandin E(2) (PGE(2)) secretion and the subsequent phenotypic effects of PGE(2) on epithelial cells. The effect of IL-1alpha on COX-2 expression was investigated in the T24 bladder epithelial cell line following treatment with 0, 0.05, 0.5, 1 or 10 ng/ml IL-1alpha for 1, 2, 4 or 6 h. Quantitative PCR confirmed up-regulation of expression of COX-2 with maximal expression observed following treatment with 0.5 ng/ml IL-1alpha for 1 h. Co-treatment of the cells with 0.5 ng/ml IL-1alpha in the presence or absence of 100 ng/ml IL-1 receptor antagonist (RA) abolished the up-regulation in COX-2 expression confirming that the effect of IL-1alpha is mediated via its membrane-bound receptors. Treatment with 0.5 ng/ml IL-1alpha resulted in a time-dependent increase in PGE(2) secretion with maximal secretion detected at 24 and 48 h after stimulation with IL-1alpha. Co-treatment of the cells with IL-1alpha and IL-1RA or the COX-2 enzyme inhibitor NS398 abolished the IL-1alpha mediated secretion of PGE(2). Treatment of T24 cells with 100 nM PGE(2) resulted in a significant elevation in cAMP generation confirming the expression of functional PGE(2) receptors. Finally, the effect of exogenous treatment with PGE(2) on apoptosis of T24 cells was assessed using cell death detection ELISA. T24 cells were treated with camptothecin to induce apoptosis in the presence or absence of 50 or 100 nM PGE(2) or 10 microM forskolin. Treatment of T24 cells with increasing doses of camptothecin alone resulted in a significant increase in the induction of apoptosis (P<0.01). However, co-treatment of the cells with 50 or 100 nM PGE(2) or 10 microM forskolin resulted in the inhibition of induction of the apoptotic pathway by camptothecin. These data demonstrate that PGE(2) inhibits apoptosis of epithelial cells possibly via cAMP-dependent pathway.


Subject(s)
Apoptosis/drug effects , Autocrine Communication , Dinoprostone/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Paracrine Communication , Camptothecin/pharmacology , Cell Line , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Interleukin-1/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Polymerase Chain Reaction , Prostaglandin-Endoperoxide Synthases/metabolism , Ribonucleases
15.
Mol Hum Reprod ; 8(7): 606-11, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12087074

ABSTRACT

Experimental studies in animals have established prolactin (PRL) as a progonadal hormone that promotes the function of the testis and reproductive accessory glands. The present study investigated the localization of PRL receptor (PRL-R) expression in the human testis and accessory tissues. Expression of PRL-R was identified in human testis and vas deferens by RT-PCR, and further localized by immunohistochemistry to the Leydig cells and differentiating germ cells of the testis (developmental stages extending from pachytene spermatocytes to elongating spermatids). Positive staining for PRL-R was also clearly evident in the epithelium of vas deferens, epididymis, prostate and seminal vesicles. Functional activation of PRL-R was demonstrated in fresh samples of vas deferens collected at vasectomy by examination of the JAK/STAT (Janus kinase/signal transducer and activator of transcription) and MAP (mitogen-activated protein) kinase ERK (extracellular signal-regulated kinase) signalling pathways. Within the vas deferens, PRL induced rapid tyrosine phosphorylation of JAK 2 and STAT 5 (after 10 and 20 min respectively), and tyrosine and threonine phosphorylation of ERK 1 and 2 (after 5 min). The demonstration of function and localization of PRL-R presented here suggests multiple roles for PRL in the human male reproductive tract.


Subject(s)
Receptors, Prolactin/biosynthesis , Testis/metabolism , Vas Deferens/metabolism , Humans , Immunohistochemistry , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinases/metabolism , RNA/metabolism , Receptors, Prolactin/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
J Clin Endocrinol Metab ; 87(5): 2329-35, 2002 May.
Article in English | MEDLINE | ID: mdl-11994384

ABSTRACT

Functional PRL receptors are expressed in the human endometrium during the secretory phase of the menstrual cycle in which PRL stimulates tyrosine phosphorylation of Janus kinase 2 and STAT (signal transducer and activator of transcription) 1 and 5. In this study, we investigated the effect of PRL on the MAPK/ERK pathway in the human endometrium. Human endometrial tissue was collected during the mid to late secretory phase of the menstrual cycle. Western blot analysis performed on proteins, extracted after up to 30 min culture with PRL, demonstrated rapid tyrosine and threonine phosphorylation of ERK 1 and 2 MAPKs. The phosphorylation of ERK, in response to PRL, was localized by immunohistochemistry to glandular epithelial cells and a subset of stromal cells. Using immunofluorescence histochemistry, PRL-induced phosphorylation of ERK in the stromal compartment was localized to the uterine-specific CD56(+) natural killer (NK) cells. We have demonstrated that the PRL receptor is expressed in uterine CD56(+) NK cells in situ by immunofluorescence and in purified decidual CD56(+) NK cells by RT-PCR and Western blotting analysis. We have further demonstrated phosphorylation of ERK 1 and 2 in cultures of purified uterine CD56(+) NK cells, in response to PRL. Our data demonstrate that PRL stimulates the ERK pathway in multiple cellular compartments of the human endometrium and identify uterine CD56(+) NK cells as novel PRL target cells.


Subject(s)
CD56 Antigen/metabolism , Endometrium/metabolism , Killer Cells, Natural/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prolactin/pharmacology , Cells, Cultured , Culture Techniques , Endometrium/cytology , Female , Humans , Phosphorylation/drug effects , Receptors, Prolactin/metabolism , Tissue Distribution , Uterus/cytology , Uterus/metabolism
17.
Br J Cancer ; 85(7): 1023-31, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11592775

ABSTRACT

This study was designed to investigate the possible role of cyclo-oxygenase-2 (COX-2) and prostaglandin E(2)(PGE(2)) in endometrial adenocarcinoma. COX-2 RNA expression was confirmed in various grades of adenocarcinoma by ribonuclease protection assay. COX-2 and microsomal glutathione-dependent prostaglandin E synthase (mPGES) expression and PGE(2)synthesis were localised to the neoplastic epithelial cells and endothelial cells. In order to establish whether PGE(2)has an autocrine/paracrine effect in adenocarcinomas, we investigated the expression of 2 subtypes of PGE(2)receptors, namely EP2 and EP4, by real time quantitative PCR. Expression of EP2 and EP4 receptors was detected in adenocarcinomas from all grades of differentiation and was significantly higher than that detected in normal secretory phase endometrium (P< 0.01). The fold induction of expression in adenocarcinoma compared with normal secretory phase endometrium was 28.0 +/- 7.4 and 52.5 +/- 10.1 for EP2 and EP4 receptors respectively. Immunohistochemistry localised the site of expression of EP4 receptor in neoplastic epithelial cells and in the endothelium of carcinomas of all grades of differentiation. Finally, the functionality of the EP2/EP4 receptors was assessed by investigating cAMP generation following in vitro culture of adenocarcinoma tissue in the presence or absence of 300 nM PGE(2). cAMP production in response to PGE(2)was significantly higher in carcinoma tissue than that detected in normal secretory phase endometrium (3.42 +/- 0.46 vs 1.15 +/- 0.05 respectively; P< 0.001). In conclusion, these data suggest that PGE(2)may regulate neoplastic cell function in an autocrine/paracrine manner via the EP2/EP4 receptors.


Subject(s)
Adenocarcinoma/physiopathology , Dinoprostone/biosynthesis , Endometrial Neoplasms/physiopathology , Gene Expression Regulation, Neoplastic , Isoenzymes/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Adenocarcinoma/genetics , Adult , Cell Differentiation , Cyclic AMP/analysis , Cyclic AMP/biosynthesis , Cyclooxygenase 2 , DNA, Neoplasm/analysis , Dinoprostone/analysis , Endometrial Neoplasms/genetics , Endothelium , Epithelial Cells , Female , Humans , Immunohistochemistry , Isoenzymes/analysis , Membrane Proteins , Neoplasm Staging , Polymerase Chain Reaction , Prostaglandin-Endoperoxide Synthases/analysis , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Tumor Cells, Cultured
18.
J Clin Endocrinol Metab ; 86(9): 4453-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549693

ABSTRACT

This study was designed to elucidate the sites of synthesis and action of PGE(2) in the nonpregnant human uterus across the menstrual cycle. The sites of expression of PGE synthase and synthesis of PGE(2) were investigated by immunohistochemistry using full thickness uterine biopsies. Expression of PGE synthase and synthesis of PGE(2) were localized to glandular epithelial and endothelial cells in both basalis and functionalis regions of the human endometrium. By contrast, stromal staining was predominantly localized in the functionalis layer. Some cyclical variation in expression of PGE synthase and PGE(2) synthesis was observed, with reduced expression/synthesis detected in the stromal compartment of the functionalis during the late secretory phase of the menstrual cycle. Subsequently, we assessed the site of action of PGE(2) by investigating the expression of two PGE(2) receptor isoforms, namely EP2 and EP4. Cyclical variation in endometrial EP2 and EP4 receptor mRNA expression was quantified by TaqMan quantitative RT-PCR using RNA isolated from endometrial tissue collected across the menstrual cycle. No differences in EP2 receptor mRNA expression were detected; however, EP4 receptor mRNA expression was significantly higher in late proliferative stage (P < 0.05) than in early, mid, and late secretory stage endometrium. Expression patterns of EP2 and EP4 receptors were localized by nonradioactive in situ hybridization using fluorescein isothiocyanate end- labeled oligonucleotide probes. Expression of both receptors was observed in endometrial glandular epithelial and vascular cells, with no notable spatial or temporal variation. Finally, signaling of EP2/EP4 receptors was assessed by investigating cAMP generation in vitro after stimulation with PGE(2). Endometrial cAMP generation in response to PGE(2) was significantly greater in proliferative tissue compared with early and midsecretory stage tissue (3.77 +/- 0.85 vs. 1.96 +/- 0.28 and 1.38 +/- 0.23, respectively; P < 0.05). In conclusion, this study demonstrates glandular and vascular coexpression of PGE synthase, PGE(2), EP2, and EP4 receptors and suggests an autocrine/paracrine role for PGE(2) in epithelial/endothelial cell function in the human endometrium.


Subject(s)
Dinoprostone/biosynthesis , Dinoprostone/physiology , Endometrium/metabolism , Menstrual Cycle/metabolism , Receptors, Prostaglandin E/biosynthesis , Receptors, Prostaglandin E/physiology , Adult , Autocrine Communication/physiology , Cyclic AMP/biosynthesis , Endometrium/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Humans , Immunohistochemistry , In Situ Hybridization , Neovascularization, Physiologic/physiology , Paracrine Communication/physiology , RNA, Messenger/biosynthesis , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Reverse Transcriptase Polymerase Chain Reaction , Uterus/cytology , Uterus/metabolism
19.
Biol Reprod ; 65(3): 936-43, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11514361

ABSTRACT

This study investigated whether prolactin (PRL) plays a priming role in the testis during the nonmating season and thereby facilitates gonadal reactivation. Sexually inactive Soay rams under long days were treated as follows: 1) group C (control) received vehicle, 2) group B received bromocriptine to suppress PRL secretion, 3) group B + PRL received bromocriptine + ovine PRL to reinstate physiological levels of PRL (n = 5/group). Treatments were for 10 wk. The photoperiod was then switched to short days to reactivate the reproductive axis. Testis diameter and sex skin coloration were recorded, and routine blood samples were collected to measure concentrations of FSH, inhibin A, and testosterone (T). At the end of the treatments, blood samples were collected every 10 min for 10 h to monitor LH pulses and the T-response to exogenous LH, and a testis biopsy was collected to assess spermatogenic activity (bromodeoxyuridine [BrDU] method) and expression of PRL receptor (reverse transcription-polymerase chain reaction and immunocytochemistry). There were no significant differences between groups in spermatogenesis (BrDU index) or steroidogenesis (T-response), and no difference in the time taken to achieve full testicular redevelopment under short days. Testis diameter and inhibin A were marginally increased in group B + PRL. Overall, this thorough experiment provides minimal support for the priming hypothesis.


Subject(s)
Models, Biological , Prolactin/physiology , Seasons , Testis/physiology , Animals , Bromocriptine/pharmacology , Follicle Stimulating Hormone/blood , Gene Expression , Hair/physiology , Hormone Antagonists/pharmacology , Inhibins/blood , Luteinizing Hormone/metabolism , Luteinizing Hormone/pharmacology , Male , Molting , Periodicity , Photoperiod , Prolactin/antagonists & inhibitors , Prolactin/blood , Receptors, Prolactin/analysis , Receptors, Prolactin/genetics , Sheep , Spermatogenesis , Testis/anatomy & histology , Testis/chemistry , Testosterone/blood
20.
J Clin Endocrinol Metab ; 86(5): 2243-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11344234

ABSTRACT

The prevalence of cervical cancer in South African women is reported as being the highest in the world, occurring, on the average, in 60 of every 100,000 women. Cervical cancer is thus considered an important clinical problem in sub-Saharan AFRICA: Recent studies have suggested that epithelial tumors may be regulated by cyclooxygenase (COX) enzyme products. The purpose of this study was to determine whether cyclooxygenase-2 (COX-2) expression and PGE(2) synthesis are up-regulated in cervical cancers. Real-time quantitative RT-PCR and Western blot analysis confirmed COX-2 ribonucleic acid and protein expression in all cases of squamous cell carcinoma (n = 8) and adenocarcinoma (n = 2) investigated. In contrast, minimal expression of COX-2 was detected in histologically normal cervix (n = 5). Immunohistochemical analyses localized COX-2 expression and PGE(2) synthesis to neoplastic epithelial cells of all squamous cell (n = 10) and adenocarcinomas (n = 10) studied. Immunoreactive COX-2 and PGE(2) were also colocalized to endothelial cells lining the microvasculature. Minimal COX-2 and PGE(2) immunoreactivity were detected in normal cervix (n = 5). To establish whether PGE(2) has an autocrine/paracrine effect in cervical carcinomas, we investigated the expression of two subtypes of PGE(2) receptors, namely EP2 and EP4, by real-time quantitative RT-PCR. Expression of EP2 and EP4 receptors was significantly higher in carcinoma tissue (n = 8) than in histologically normal cervix (n = 5; P < 0.01). Finally, the functionality of the EP2/EP4 receptors was assessed by investigating cAMP generation after in vitro culture of cervical cancer biopsies and normal cervix in the presence or absence of 300 nmol/L PGE(2). cAMP production was detected in all carcinoma tissue after treatment with exogenous PGE(2) and was significantly higher in carcinoma tissue (n = 7) than in normal cervix (n = 5; P < 0.05). The fold induction of cAMP in response to PGE(2) was 51.1 +/- 12.3 in cervical carcinoma tissue compared with 5.8 +/- 2.74 in normal cervix. These results confirm that COX-2, EP2, and EP4 expression and PGE(2) synthesis are up-regulated in cervical cancer tissue and suggest that PGE(2) may regulate neoplastic cell function in cervical carcinoma in an autocrine/paracrine manner via the EP2/EP4 receptors.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Squamous Cell/metabolism , Dinoprostone/biosynthesis , Gene Expression Regulation, Enzymologic , Isoenzymes/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Prostaglandin E/physiology , Uterine Cervical Neoplasms/metabolism , Cyclooxygenase 2 , Female , Humans , Membrane Proteins , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...