Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(6): 112561, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37243590

RESUMO

Glioblastoma (GBM) stem cells (GSCs) display phenotypic and molecular features reminiscent of normal neural stem cells and exhibit a spectrum of cell cycle states (dormant, quiescent, proliferative). However, mechanisms controlling the transition from quiescence to proliferation in both neural stem cells (NSCs) and GSCs are poorly understood. Elevated expression of the forebrain transcription factor FOXG1 is often observed in GBMs. Here, using small-molecule modulators and genetic perturbations, we identify a synergistic interaction between FOXG1 and Wnt/ß-catenin signaling. Increased FOXG1 enhances Wnt-driven transcriptional targets, enabling highly efficient cell cycle re-entry from quiescence; however, neither FOXG1 nor Wnt is essential in rapidly proliferating cells. We demonstrate that FOXG1 overexpression supports gliomagenesis in vivo and that additional ß-catenin induction drives accelerated tumor growth. These data indicate that elevated FOXG1 cooperates with Wnt signaling to support the transition from quiescence to proliferation in GSCs.


Assuntos
Fatores de Transcrição Forkhead , Glioblastoma , Via de Sinalização Wnt , Humanos , beta Catenina/metabolismo , Divisão Celular , Proliferação de Células , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Glioblastoma/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo
2.
Elife ; 112022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36476408

RESUMO

Extrachromosomal DNA (ecDNA) are frequently observed in human cancers and are responsible for high levels of oncogene expression. In glioblastoma (GBM), ecDNA copy number correlates with poor prognosis. It is hypothesized that their copy number, size, and chromatin accessibility facilitate clustering of ecDNA and colocalization with transcriptional hubs, and that this underpins their elevated transcriptional activity. Here, we use super-resolution imaging and quantitative image analysis to evaluate GBM stem cells harbouring distinct ecDNA species (EGFR, CDK4, PDGFRA). We find no evidence that ecDNA routinely cluster with one another or closely interact with transcriptional hubs. Cells with EGFR-containing ecDNA have increased EGFR transcriptional output, but transcription per gene copy is similar in ecDNA compared to the endogenous chromosomal locus. These data suggest that it is the increased copy number of oncogene-harbouring ecDNA that primarily drives high levels of oncogene transcription, rather than specific interactions of ecDNA with each other or with high concentrations of the transcriptional machinery.


Assuntos
Oncogenes , Células-Tronco , Humanos , Oncogenes/genética , DNA
3.
Nat Genet ; 53(8): 1221-1232, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34294917

RESUMO

Driver mutations in genes encoding histone H3 proteins resulting in p.Lys27Met substitutions (H3-K27M) are frequent in pediatric midline brain tumors. However, the precise mechanisms by which H3-K27M causes tumor initiation remain unclear. Here, we use human hindbrain neural stem cells to model the consequences of H3.3-K27M on the epigenomic landscape in a relevant developmental context. Genome-wide mapping of epitope-tagged histone H3.3 revealed that both the wild type and the K27M mutant incorporate abundantly at pre-existing active enhancers and promoters, and to a lesser extent at Polycomb repressive complex 2 (PRC2)-bound regions. At active enhancers, H3.3-K27M leads to focal H3K27ac loss, decreased chromatin accessibility and reduced transcriptional expression of nearby neurodevelopmental genes. In addition, H3.3-K27M deposition at a subset of PRC2 target genes leads to increased PRC2 and PRC1 binding and augmented transcriptional repression that can be partially reversed by PRC2 inhibitors. Our work suggests that, rather than imposing de novo transcriptional circuits, H3.3-K27M drives tumorigenesis by locking initiating cells in their pre-existing, immature epigenomic state, via disruption of PRC2 and enhancer functions.


Assuntos
Elementos Facilitadores Genéticos , Histonas/metabolismo , Células-Tronco Neurais/fisiologia , Complexo Repressor Polycomb 2/genética , Rombencéfalo/citologia , Animais , Neoplasias Encefálicas/genética , Diferenciação Celular/genética , Linhagem Celular , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigenoma , Regulação da Expressão Gênica no Desenvolvimento , Glioma/genética , Histonas/genética , Humanos , Lisina/metabolismo , Masculino , Camundongos Endogâmicos , Mutação , Células-Tronco Neurais/transplante , Oncogenes , Complexo Repressor Polycomb 2/antagonistas & inibidores , Complexo Repressor Polycomb 2/metabolismo , Regiões Promotoras Genéticas , Rombencéfalo/fisiologia
4.
Cell ; 184(9): 2454-2470.e26, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33857425

RESUMO

Glioblastoma multiforme (GBM) is an aggressive brain tumor for which current immunotherapy approaches have been unsuccessful. Here, we explore the mechanisms underlying immune evasion in GBM. By serially transplanting GBM stem cells (GSCs) into immunocompetent hosts, we uncover an acquired capability of GSCs to escape immune clearance by establishing an enhanced immunosuppressive tumor microenvironment. Mechanistically, this is not elicited via genetic selection of tumor subclones, but through an epigenetic immunoediting process wherein stable transcriptional and epigenetic changes in GSCs are enforced following immune attack. These changes launch a myeloid-affiliated transcriptional program, which leads to increased recruitment of tumor-associated macrophages. Furthermore, we identify similar epigenetic and transcriptional signatures in human mesenchymal subtype GSCs. We conclude that epigenetic immunoediting may drive an acquired immune evasion program in the most aggressive mesenchymal GBM subtype by reshaping the tumor immune microenvironment.


Assuntos
Neoplasias Encefálicas/imunologia , Epigênese Genética , Glioblastoma/imunologia , Evasão da Resposta Imune/imunologia , Células Mieloides/imunologia , Células-Tronco Neoplásicas/imunologia , Microambiente Tumoral/imunologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proliferação de Células , Metilação de DNA , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células Mieloides/metabolismo , Células Mieloides/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cell Stem Cell ; 28(5): 877-893.e9, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33631116

RESUMO

Point mutations within the histone H3.3 are frequent in aggressive childhood brain tumors known as pediatric high-grade gliomas (pHGGs). Intriguingly, distinct mutations arise in discrete anatomical regions: H3.3-G34R within the forebrain and H3.3-K27M preferentially within the hindbrain. The reasons for this contrasting etiology are unknown. By engineering human fetal neural stem cell cultures from distinct brain regions, we demonstrate here that cell-intrinsic regional identity provides differential responsiveness to each mutant that mirrors the origins of pHGGs. Focusing on H3.3-G34R, we find that the oncohistone supports proliferation of forebrain cells while inducing a cytostatic response in the hindbrain. Mechanistically, H3.3-G34R does not impose widespread transcriptional or epigenetic changes but instead impairs recruitment of ZMYND11, a transcriptional repressor of highly expressed genes. We therefore propose that H3.3-G34R promotes tumorigenesis by focally stabilizing the expression of key progenitor genes, thereby locking initiating forebrain cells into their pre-existing immature state.


Assuntos
Neoplasias Encefálicas , Glioma , Células-Tronco Neurais , Neoplasias Encefálicas/genética , Carcinogênese/genética , Glioma/genética , Histonas/genética , Humanos , Mutação/genética
6.
Genes Dev ; 31(8): 757-773, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28465359

RESUMO

Glioblastoma multiforme (GBM) is an aggressive brain tumor driven by cells with hallmarks of neural stem (NS) cells. GBM stem cells frequently express high levels of the transcription factors FOXG1 and SOX2. Here we show that increased expression of these factors restricts astrocyte differentiation and can trigger dedifferentiation to a proliferative NS cell state. Transcriptional targets include cell cycle and epigenetic regulators (e.g., Foxo3, Plk1, Mycn, Dnmt1, Dnmt3b, and Tet3). Foxo3 is a critical repressed downstream effector that is controlled via a conserved FOXG1/SOX2-bound cis-regulatory element. Foxo3 loss, combined with exposure to the DNA methylation inhibitor 5-azacytidine, enforces astrocyte dedifferentiation. DNA methylation profiling in differentiating astrocytes identifies changes at multiple polycomb targets, including the promoter of Foxo3 In patient-derived GBM stem cells, CRISPR/Cas9 deletion of FOXG1 does not impact proliferation in vitro; however, upon transplantation in vivo, FOXG1-null cells display increased astrocyte differentiation and up-regulate FOXO3. In contrast, SOX2 ablation attenuates proliferation, and mutant cells cannot be expanded in vitro. Thus, FOXG1 and SOX2 operate in complementary but distinct roles to fuel unconstrained self-renewal in GBM stem cells via transcriptional control of core cell cycle and epigenetic regulators.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Epigenômica , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/fisiopatologia , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/citologia , Fatores de Transcrição SOXB1/genética , Motivos de Aminoácidos , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Azacitidina/farmacologia , Neoplasias Encefálicas/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Cromatina/metabolismo , Metilação de DNA , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/genética , Humanos , Mutação , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Fatores de Transcrição SOXB1/metabolismo , Células Tumorais Cultivadas
7.
Development ; 144(4): 635-648, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28096221

RESUMO

Mammalian neural stem cell (NSC) lines provide a tractable model for discovery across stem cell and developmental biology, regenerative medicine and neuroscience. They can be derived from foetal or adult germinal tissues and continuously propagated in vitro as adherent monolayers. NSCs are clonally expandable, genetically stable, and easily transfectable - experimental attributes compatible with targeted genetic manipulations. However, gene targeting, which is crucial for functional studies of embryonic stem cells, has not been exploited to date in NSC lines. Here, we deploy CRISPR/Cas9 technology to demonstrate a variety of sophisticated genetic modifications via gene targeting in both mouse and human NSC lines, including: (1) efficient targeted transgene insertion at safe harbour loci (Rosa26 and AAVS1); (2) biallelic knockout of neurodevelopmental transcription factor genes; (3) simple knock-in of epitope tags and fluorescent reporters (e.g. Sox2-V5 and Sox2-mCherry); and (4) engineering of glioma mutations (TP53 deletion; H3F3A point mutations). These resources and optimised methods enable facile and scalable genome editing in mammalian NSCs, providing significant new opportunities for functional genetic analysis.


Assuntos
Neoplasias Encefálicas/genética , Sistemas CRISPR-Cas , Marcação de Genes/métodos , Glioma/genética , Células-Tronco Neurais/citologia , Alelos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neoplasias Encefálicas/metabolismo , Mapeamento de Epitopos , Epitopos , Glioma/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Recombinação Homóloga , Humanos , Camundongos , Camundongos Knockout , Mutação , Proteínas do Tecido Nervoso/genética , Fator de Transcrição 2 de Oligodendrócitos , Oligonucleotídeos/genética , Mutação Puntual , Recombinação Genética , Medicina Regenerativa , Transgenes
8.
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
9.
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
10.
Am J Pathol ; 176(1): 435-45, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20008143

RESUMO

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.


Assuntos
Calcineurina/metabolismo , Cálcio/metabolismo , Neoplasias do Endométrio/genética , Interleucina-11/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Musculares/metabolismo , Fatores de Transcrição NFATC/metabolismo , Receptores de Prostaglandina/metabolismo , Idoso , Diferenciação Celular , Proliferação de Células , Receptor gp130 de Citocina/genética , Receptor gp130 de Citocina/metabolismo , Proteínas de Ligação a DNA , Neoplasias do Endométrio/patologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-11/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Pessoa de Meia-Idade , Modelos Biológicos , Proteínas Musculares/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-11/genética , Receptores de Interleucina-11/metabolismo
11.
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
12.
Mol Cell Endocrinol ; 285(1-2): 51-61, 2008 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-18316157

RESUMO

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.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprosta/metabolismo , Dinoprostona/metabolismo , Regulação Enzimológica da Expressão Gênica , Elementos de Resposta , Transdução de Sinais/fisiologia , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Dinoprosta/análogos & derivados , Neoplasias do Endométrio/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Genes Reporter , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Regiões Promotoras Genéticas , Antagonistas de Prostaglandina/metabolismo , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Receptores de Prostaglandina E/metabolismo , Xantonas/metabolismo
13.
Biochem Biophys Res Commun ; 331(1): 187-93, 2005 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-15845377

RESUMO

This study demonstrates a synergistic action of prostaglandin E and GM-CSF which causes the release of pro-tolerant cytokines in two monocyte cell lines: U937 and ML-1. The prostaglandin effect is cyclic AMP dependent since stimulators of adenyl cyclase such as forskolin (fsk) can replace PGE. Fsk and GM-CSF combinations raised messenger RNA for IL-10, interleukin-1 receptor antagonist (IL-1ra), and CD14 as well as the released proteins. Effective levels of interleukin 12 are reduced. In these respects, the monocyte cells resemble the alternatively activated or tumour associated macrophages. A differential pattern in co-stimulatory molecule expression is seen; CD80 is unchanged but CD86 is markedly elevated and such a change is not seen in the alternatively activated macrophage but has been previously reported in monocytes resident in the non-inflamed gut. Control of leukocyte responses by two agents acting in synergy could be effective in critical situations such as discrimination between pathogens and commensal bacteria, etc. Monocytes modified in such a way could provide a pro-tolerant environment (high IL-10, low IL-12) for antigen presentation by dendritic cells and thus may contribute to a normally permissive milieu, e.g., for food absorption.


Assuntos
AMP Cíclico/metabolismo , Citocinas/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Monócitos/imunologia , Prostaglandinas E/farmacologia , Linhagem Celular , Colforsina/farmacologia , Sinergismo Farmacológico , Humanos , Tolerância Imunológica , Interleucina-12/biossíntese , Receptores de Lipopolissacarídeos/biossíntese , Monócitos/citologia , Monócitos/efeitos dos fármacos , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...