Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Immunol ; 192(5): 2315-25, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24501200

RESUMO

Parturition is associated with a leukocyte influx into the intrauterine tissues; however, the exact role these leukocytes play in the onset of labor remains unclear. Neutrophil infiltration of the uteroplacental tissues has been particularly associated with infection-associated preterm labor (PTL) in both women and mouse models. In this study, we investigated the role of neutrophils in a mouse model of infection-induced PTL. Intrauterine administration of LPS on day 17 of gestation resulted in a 7-fold increase in the number of decidual neutrophils compared with control mice receiving PBS (p < 0.01; n = 8-11). We hypothesized that neutrophil influx is necessary for PTL and that neutrophil depletion would abolish preterm birth. To test this hypothesis, mice were depleted of neutrophils by treatment with anti-Gr-1, anti-Ly-6G, or the appropriate IgG control Ab on day 16 of gestation prior to LPS on day 17 (n = 6-7). Successful neutrophil depletion was confirmed by flow cytometry and immunohistochemistry. Neutrophil depletion with Gr-1 resulted in reduced uterine and placental Il-1ß expression (p < 0.05). Neutrophil depletion with Ly-6G reduced uterine Il-1ß and Tnf-α expression (p < 0.05). However, neutrophil depletion with either Ab did not delay LPS-induced preterm birth. Collectively, these data show that decidual neutrophil infiltration is not essential for the induction of infection-induced PTL in the mouse, but that neutrophils contribute to the LPS-induced inflammatory response of the uteroplacental tissues.


Assuntos
Decídua/imunologia , Lipopolissacarídeos/toxicidade , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Trabalho de Parto Prematuro/induzido quimicamente , Animais , Decídua/patologia , Modelos Animais de Doenças , Feminino , Humanos , Infecções/imunologia , Infecções/patologia , Interleucina-1beta/imunologia , Camundongos , Neutrófilos/patologia , Trabalho de Parto Prematuro/imunologia , Trabalho de Parto Prematuro/patologia , Gravidez
2.
Reproduction ; 146(6): 581-91, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24051059

RESUMO

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.


Assuntos
Membranas Extraembrionárias/efeitos dos fármacos , Inflamação/induzido quimicamente , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/farmacologia , Animais , Membranas Extraembrionárias/imunologia , Membranas Extraembrionárias/metabolismo , Feminino , Inflamação/genética , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Injeções , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Gravidez , Nascimento Prematuro , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regulação para Cima/efeitos dos fármacos , Útero , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
3.
Am J Pathol ; 179(6): 2709-19, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21983634

RESUMO

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.


Assuntos
Endometrite/etiologia , Hormônios Gastrointestinais/fisiologia , Miométrio/metabolismo , Nascimento Prematuro/etiologia , Nascimento a Termo/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Endometrite/metabolismo , Feminino , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Expressão Gênica , Humanos , Lipopolissacarídeos/farmacologia , MicroRNAs/farmacologia , Gravidez , Nascimento Prematuro/metabolismo , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Regulação para Cima , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
4.
Mol Hum Reprod ; 17(1): 22-32, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20705717

RESUMO

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.


Assuntos
Gonadotropina Coriônica/farmacologia , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores CXCR4/metabolismo , Western Blotting , Linhagem Celular , Decídua/efeitos dos fármacos , Decídua/metabolismo , Implantação do Embrião/genética , Endométrio/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Ciclo Menstrual/metabolismo , Gravidez , Primeiro Trimestre da Gravidez/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos
5.
Mol Hum Reprod ; 17(3): 182-92, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21112968

RESUMO

Prostanoids are well-described primary mediators of inflammatory processes and are essential for the normal physiological function of the female reproductive system. The aim of this study was to determine the temporal expression of the prostanoid biosynthetic enzymes (PTGS1, PTGS2, PTGES, PTGES2, PTGES3, AKR1B1, AKR1C3, CBR1, HPGDS, PTGDS, PTGIS, TBXAS1 and HPGD) and the prostanoid receptors (PTGER1, PTGER2, PTGER3, PTGER4, PTGFR, PTGDR, GPR44, PTGIR and TBXA2R) in the human endometrium throughout the menstrual cycle. The analysis identified PTGFR to have a distinct expression profile compared with other components of the prostanoid system, as expression is maximal during the proliferative phase. Immunohistochemical analysis for PTGER1 suggests a dual function for this receptor depending on its temporal (proliferative versus secretory) and spatial (nuclear versus cell membrane) expression. The expression profiles of the PGF(2α) synthases identified AKR1B1 and CBR1 as the likely regulators of PGF(2α) production during the menstrual phase. Immunohistochemical analysis for AKR1B1, CBR1 and AKR1C3 suggest expression to be in the glandular epithelium and vasculature. This study represents the first comprehensive analysis of the components of prostanoid biosynthetic and signalling pathway in the human endometrium. The expression profiles described have the potential to identify specific prostanoid components that may be dysregulated in inflammatory-associated disorders of the endometrium.


Assuntos
Endométrio/metabolismo , Ciclo Menstrual/metabolismo , Prostaglandinas/metabolismo , Receptores de Prostaglandina/metabolismo , Adulto , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Aldeído Redutase , Aldo-Ceto Redutases , Endométrio/enzimologia , Feminino , Humanos , Prostaglandinas/análise , Prostaglandinas/genética , RNA Mensageiro/metabolismo , Receptores de Prostaglandina/análise , Receptores de Prostaglandina/genética
6.
Mol Hum Reprod ; 17(10): 626-36, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21546446

RESUMO

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.


Assuntos
Decídua/fisiologia , Hormônios Gastrointestinais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Adulto , Proliferação de Células , Células Cultivadas , AMP Cíclico/farmacologia , Decídua/efeitos dos fármacos , Implantação do Embrião , Células Epiteliais/fisiologia , Feminino , Hormônios Gastrointestinais/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Fase Luteal/metabolismo , Placentação/fisiologia , Gravidez , Progesterona/farmacologia , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Transdução de Sinais , Células Estromais/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
7.
Biochim Biophys Acta ; 1793(12): 1917-28, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19819266

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Calcineurina/metabolismo , Cálcio/metabolismo , Neoplasias do Endométrio/metabolismo , Regulação Neoplásica da Expressão Gênica , Interleucina-8/biossíntese , Fatores de Transcrição NFATC/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Prostaglandina/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Dinoprosta/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteína Quinase C/metabolismo , Elementos de Resposta , Transplante Heterólogo
8.
Mol Hum Reprod ; 16(5): 311-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20172976

RESUMO

The onset of labour is associated with inflammatory cascades in the uteroplacental unit, and these are prematurely activated in preterm labour. Infection can induce an inflammatory response, which results in premature onset of the pro-inflammatory parturient cascades and preterm delivery. We have become interested in two proteins called prokineticins and the role they may play in labour and its premature onset. Prokineticin 1 and 2 bind to two G-protein coupled receptors, called prokineticin receptor 1 and 2. Expression of the prokineticins and their receptors is elevated in the uteroplacental unit during labour and they can induce expression of a host of genes known to be important in initiating the inflammatory and contractile events of labour. Prokineticins have also been shown to directly induce contractility of smooth muscles. Analysing the promoter regions of the prokineticins and their receptors highlights their potential regulation by pathways activated by infectious agents. Hence, we propose that infection can result in premature activation of prokineticin expression and signalling in the uteroplacental unit and this initiates a premature inflammatory and contractile cascade leading to preterm birth. Antagonism of prokineticin action may provide a suitable therapy for preterm labour that targets both inflammation and contractile pathways.


Assuntos
Hormônios Gastrointestinais/metabolismo , Inflamação/metabolismo , Neuropeptídeos/metabolismo , Parto/metabolismo , Contração Uterina/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Feminino , Humanos , Trabalho de Parto Prematuro/metabolismo , Gravidez , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo
9.
FASEB J ; 23(7): 2165-75, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19255255

RESUMO

Implantation requires communication between a receptive endometrium and a healthy blastocyst. This maternal-embryonic crosstalk involves local mediators within the uterine microenvironment. We demonstrate that a secreted protein, prokineticin 1 (PROK1), is expressed in the receptive endometrium and during early pregnancy. PROK1 induces expression of leukemia inhibitory factor (LIF) in endometrial epithelial cells and first trimester decidua via a Gq-Ca(2+)-cSrc-mitogen-activated protein kinase kinase-mediated pathway. We show that human embryonic chorionic gonadotropin (hCG) induces sequential mRNA expression of PROK1 and LIF in an in vivo baboon model, in human endometrial epithelial cells, and in first-trimester decidua. We have used micro RNA constructs targeted to PROK1 to demonstrate that hCG-mediated LIF expression in the endometrium is dependent on prior induction of PROK1. Dual immunohistochemical analysis colocalized expression of the luteinizing hormone/chorionic gonadotropin receptor, PROK1, PROKR1, and LIF to the glandular epithelial cells of the first trimester decidual tissue. PROK1 enhances adhesion of trophoblast cells to fibronectin and laminin matrices, which are mediated predominantly via LIF induction. These data describe a novel signaling pathway mediating maternal-embryonic crosstalk, in which embryonic hCG via endometrial PROK1 may play a pivotal role in enhancing receptivity and maintaining early pregnancy.


Assuntos
Blastocisto/citologia , Implantação do Embrião , Endométrio/citologia , Fator Inibidor de Leucemia/genética , Comunicação Parácrina/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia , Animais , Blastocisto/fisiologia , Linhagem Celular , Gonadotropina Coriônica/análise , Endométrio/fisiologia , Células Epiteliais , Feminino , Hormônios Gastrointestinais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fator Inibidor de Leucemia/análise , Fator Inibidor de Leucemia/biossíntese , Camundongos , Papio , Gravidez , Receptores Acoplados a Proteínas G , Receptores do LH/análise , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/análise
10.
Reproduction ; 138(6): 903-19, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19793840

RESUMO

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.


Assuntos
Doença/genética , Mediadores da Inflamação/fisiologia , Inflamação/etiologia , Reprodução/genética , Animais , Feminino , Humanos , Inflamação/complicações , Inflamação/genética , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Modelos Biológicos , Gravidez , Complicações na Gravidez/etiologia , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Reprodução/fisiologia , Saúde Reprodutiva , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
11.
Endocrinology ; 149(6): 2877-87, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18339712

RESUMO

Prokineticin 1 (PROK1) is a recently described protein with a wide range of functions including tissue-specific angiogenesis, modulation of inflammatory responses, and regulation of hematopoiesis. The objective of this study was to investigate the role of PROK1 and prokineticin receptor 1 (PROKR1) in human endometrium during early pregnancy. PROK1 and PROKR1 expression is significantly elevated in first-trimester decidua, compared with nonpregnant endometrium. Expression of PROK1 and PROKR1 was localized in glandular epithelial and various cellular compartments within the stroma. To investigate the signaling pathways and target genes activated by PROK1, we generated an endometrial epithelial cell line stably expressing PROKR1 (Ishikawa PROKR1 cells). PROK1-PROKR1 interaction induced inositol phosphate mobilization and sequential phosphorylation of c-Src, epidermal growth factor receptor, and ERK 1/2. Gene microarray analysis on RNA extracted from Ishikawa PROKR1 cells treated with 40 nm PROK1 for 8 h revealed 49 genes to be differentially regulated. A number of these genes, including cyclooxygenase (COX)-2, leukemia inhibitory factor, IL-6, IL-8, and IL-11 are regulated in the endometrium during implantation and early pregnancy. We subsequently investigated the effect of PROK1 on expression of COX-2 in Ishikawa PROKR1 cells and first-trimester decidua. COX-2 mRNA and protein expression, and prostaglandin synthesis, were elevated in response to treatment with PROK1. Moreover, expression of COX-2 by PROK1 was dependent on activation of the Gq-phospholipase C-beta-cSrc-epidermal growth factor receptor-MAPK/ERK kinase pathway. These data demonstrate that PROK1 and PROKR1 expression is elevated in human decidua during early pregnancy and that PROK1-PROKR1 interaction regulates expression of a host of implantation-related genes.


Assuntos
Hormônios Gastrointestinais/fisiologia , Gravidez/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia , Células Cultivadas , Primers do DNA , Decídua/fisiologia , Implantação do Embrião/fisiologia , Endométrio/citologia , Endométrio/fisiologia , Feminino , Hormônios Gastrointestinais/genética , Humanos , Ciclo Menstrual/genética , Ciclo Menstrual/fisiologia , Gravidez/genética , Receptores Acoplados a Proteínas G/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
12.
Mol Endocrinol ; 17(3): 395-410, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12554761

RESUMO

We demonstrate that multiple promoters and alternate splicing regulate expression of the human CRH receptor type 2 (CRHR2) gene. We show that flanking regions to the first exons drive promoter activity in both endogenously and nonendogenously expressing cell lines. Putative promoter elements have been identified that are conserved between species, including the comparison of CRHR2gamma in nonhuman primates that was previously known only in humans, which may be responsible for subtype tissue specific regulation. We have identified novel transcripts produced by alternate splicing of the first exon of CRHR2beta (beta1a) with various combinations of the 5' exons including a novel exon (beta1c) spliced to the common exons. The 5' structure of the gene permits many other combinations of alternate splicing that may arise as part of a regulatory mechanism controlling functional receptor expression. The 5'-untranslated region of the first exons has been extended; and 3' acceptor sites identified within the 5' untranslated region of CRHR2gamma and CRHR2alpha are used during alternate splicing of CRHR2beta upstream exons. This has important implications because various reports on the expression of CRHR2gamma and CRHR2alpha have been unable to discriminate between the functional receptor and CRHR2beta alternate splice variants. Only the described sequences upstream of the 3' splice site are unique to CRHR2gamma and CRHR2alpha.


Assuntos
Processamento Alternativo/genética , Regulação da Expressão Gênica/genética , Receptores de Hormônio Liberador da Corticotropina/genética , Regiões 5' não Traduzidas/genética , Regiões 5' não Traduzidas/fisiologia , Processamento Alternativo/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Epiteliais , Éxons/genética , Éxons/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Dados de Sequência Molecular , Pan troglodytes , Papio , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia , Técnica de Amplificação ao Acaso de DNA Polimórfico , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Alinhamento de Sequência , Análise de Sequência de DNA , Transfecção
13.
Endocrinology ; 145(8): 3971-83, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15142984

RESUMO

We report the full genomic organization of the human gene for the corticotropin-releasing factor (CRF) receptor type 1 (CRFR1), with complete mapping of exons 1-14. The 5' flanking region (2.4 kb) of the gene encoding for human CRFR1 was isolated, sequenced, and characterized. Two major transcriptional start sites were determined at -265 and -238, relative to the ATG start site (+1). Transient expression of constructs containing sequentially deleted 5'-flanking sequences of CRFR1 fused to luciferase, revealed the minimal promoter sequence 370 bp in size, as shown by assays in neuroblastoma (SH-5YSY), teratocarcinoma (NT2), and adenocarcinoma (MCF 7) cell lines. CRF and UCN markedly increased promoter activity during transient CRFR1 expression studies. Similarly, CRF and UCN up-regulate the endogenous CRFR1 at the mRNA level in NT2 and MCF 7 cells. To dissect further the mechanisms involved, we have used primary myometrial cells transfected with the CRFR1 promoter. CRF and UCN increased the promoter activity, an effect blocked by protein kinase (PK)A and PKC inhibitors. Both CRF and UCN cause a positive feedback effect in primary cultures of human pregnant myometrial cells, by increasing mRNA expression of CRFR1. This effect appears to be dependent on activation of both PKA and PKC by CRF, whereas UCN's effect was mediated solely via PKC activation. Collectively, our data suggest that the CRFR1 gene is under the influence of both CRF and UCN, acting via distinct signaling pathways to create a positive feedback loop and regulate further the transcription of the receptor.


Assuntos
Hormônio Liberador da Corticotropina/fisiologia , Regiões Promotoras Genéticas , Receptores de Hormônio Liberador da Corticotropina/genética , Regiões 5' não Traduzidas/química , Sequência de Aminoácidos , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Transdução de Sinais , Transcrição Gênica , Urocortinas
14.
Mol Cell Endocrinol ; 331(1): 129-35, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20816914

RESUMO

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.


Assuntos
Adenocarcinoma/patologia , Quimiocina CCL20/genética , Neoplasias do Endométrio/patologia , Receptores de Prostaglandina/metabolismo , Transdução de Sinais , Adenocarcinoma/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quimiocina CCL20/metabolismo , Dinoprosta/farmacologia , Neoplasias do Endométrio/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Modelos Biológicos , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CCR6/genética , Receptores CCR6/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
PLoS One ; 6(5): e19209, 2011 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-21589857

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias do Endométrio/metabolismo , Hipóxia/fisiopatologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais , Adenocarcinoma/patologia , Linhagem Celular Tumoral , Neoplasias do Endométrio/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Microscopia Confocal , Receptores de Prostaglandina E Subtipo EP4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Reprod Sci ; 17(6): 590-611, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20360594

RESUMO

The changes in transcript profile induced by vascular endothelial growth factor (VEGF-A) and serum withdrawal in primary human endometrial endothelial cells (ECs) were investigated using microarrays, gene ontology and pathway analysis. Vascular endothelial growth factor A altered the levels of transcripts involved in angiogenesis, cell survival, and apoptosis, including up-and downregulation of AKT1, BAD, MIF, and IGFBP3 and ANGPT2, respectively. Serum deprivation induced downregulation of cell-cycle-related transcripts such as mitosis regulators CDC20 and SPC25. Of the transcripts regulated by both VEGF-A and partial serum deprivation, remarkably 88 of 89 showed reciprocal regulation (p < 1 x 10(-49)). These are predominantly cell-fate-associated transcripts and this novel observation suggests that endometrial ECs may be particularly dependant on the levels of these transcripts. Our results show that in addition to the known role of VEGF-A as an EC growth and survival promoter, it also regulates apoptosis-related messenger RNAs (mRNAs), many of which were reciprocally regulated following serum withdrawal.


Assuntos
Endométrio/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/fisiologia , Células Cultivadas , Meios de Cultura Livres de Soro , Endométrio/citologia , Feminino , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos
17.
Proc Natl Acad Sci U S A ; 102(24): 8585-90, 2005 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-15937114

RESUMO

Activation of the receptors for leukemia inhibitory factor (LIF) and IL-11 is essential for embryo attachment and decidualization in mice. Both receptors induce activation of the Stat family of signal transducers via the Jak/Stat pathway. Here, we aimed to establish whether activation of Stat3 in maternal endometrium is essential for successful implantation. Functional blockade of Stat3 before implantation, by injection into the uterine lumen of a cell-permeable Stat3 peptide inhibitor, reduced embryo implantation specifically by 70% (P < 0.001). Stat3 is phosphorylated in the luminal epithelium (LE) in response to LIF, and this phosphorylation was significantly reduced both in vitro and in vivo by the Stat3 inhibitor. The inhibitor also blocked induction by LIF of several LIF-regulated genes in the LE including Irg1, which has been shown previously to be essential for implantation. Successful implantation is therefore dependent on phosphorylation and activation of Stat3 in the endometrium before implantation. This finding provides a target for contraceptive development, based on selective blockade of signal transduction pathways essential for implantation. This study demonstrates that cell-permeable peptide inhibitors can be used effectively to target intracellular signaling pathways in the uterine LE.


Assuntos
Anticoncepção/métodos , Proteínas de Ligação a DNA/antagonistas & inibidores , Implantação do Embrião/fisiologia , Endométrio/metabolismo , Transdução de Sinais/fisiologia , Transativadores/antagonistas & inibidores , Animais , Proteínas de Ligação a DNA/metabolismo , Feminino , Immunoblotting , Interleucina-6 , Fator Inibidor de Leucemia , Camundongos , Camundongos Endogâmicos , Fosforilação , Proteínas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3 , Transativadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa