Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Gastroenterol ; 22(1): 234, 2022 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-35549670

RESUMEN

BACKGROUND: Aberrations in cyclooxygenase and lipoxygenase (LOX) pathways in non-neoplastic, normal appearing mucosa from patients with colorectal neoplasia (CRN), could hypothetically qualify as predisposing CRN-markers. METHODS: To test this hypothesis, biopsies were obtained during colonoscopy from macroscopically normal colonic mucosa from patients with and without CRN. Prostaglandin E2 (PGE2) receptors, EP1-4, were examined in Ussing-chambers by exposing biopsies to selective EP receptor agonists, antagonists and PGE2. Furthermore, mRNA expression of EP receptors, prostanoid synthases and LOX enzymes were evaluated with qPCR. RESULTS: Data suggest that PGE2 binds to both high and low affinity EP receptors. In particular, PGE2 demonstrated EP4 receptor potency in the low nanomolar range. Similar results were detected using EP2 and EP4 agonists. In CRN patients, mRNA-levels were higher for EP1 and EP2 receptors and for enzymes prostaglandin-I synthase, 5-LOX, 12-LOX and 15-LOX. CONCLUSIONS: In conclusion, normal appearing colonic mucosa from CRN patients demonstrates deviating expression in eicosanoid pathways, which might indicate a likely predisposition for early CRN development and furthermore that PGE2 potently activates high affinity EP4 receptor subtypes, supporting relevance of testing EP4 antagonists in colorectal neoplasia management.


Asunto(s)
Neoplasias Colorrectales , Subtipo EP4 de Receptores de Prostaglandina E , Neoplasias Colorrectales/patología , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Humanos , ARN Mensajero/análisis , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/genética
2.
Immunology ; 164(4): 777-791, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34529833

RESUMEN

Regulatory T (Treg) cells are essential for control of inflammatory processes by suppressing effector T-cell functions. The actions of PGE2 on the development and function of Treg cells, particularly under inflammatory conditions, are debated. In this study, we employed pharmacological and genetic approaches to examine whether PGE2  had a direct action on T cells to modulate de novo differentiation of Treg cells. We found that TGF-ß-induced Foxp3 expression and iTreg cell differentiation in vitro is markedly inhibited by PGE2 , which was mediated by the receptors EP2 and EP4. Mechanistically, PGE2 -EP2/EP4 signalling interrupts TGF-ß signalling during iTreg differentiation. Moreover, EP4 deficiency in T cells impaired iTreg cell differentiation in vivo. Thus, our results demonstrate that PGE2 negatively regulates iTreg cell differentiation through a direct action on T cells, highlighting the potential for selectively targeting the PGE2 -EP2/EP4 pathway to control T cell-mediated inflammation.


Asunto(s)
Dinoprostona/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Dinoprostona/farmacología , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Ratones , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/efectos de los fármacos
3.
Prostaglandins Other Lipid Mediat ; 152: 106498, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33035690

RESUMEN

The telencephalon of adult mammals shows constitutive neurogenesis, and disease or traumatic injuries alter the rate of neurogenesis in the adult brain. Understanding the molecular signals that control adult brain neurogenesis is of crucial importance for the development of therapies to promote regeneration in the injured or diseased brain. Here, I reviewed our current knowledge on the role of cyclooxygenases and prostaglandins in controlling adult brain neurogenesis. Current data indicate that cyclooxygenase-2 derived prostaglandin E2 acting through EP receptors promotes neurogenesis in adult neurogenic niches of the telencephalon and that manipulations of this signalling pathway could be used to promote neurogenesis under pathological conditions. In this review article, I also propose new research directions to increase our knowledge on the role of this signalling pathway in neurogenesis.


Asunto(s)
Encéfalo , Neurogénesis , Prostaglandina-Endoperóxido Sintasas , Prostaglandinas , Adulto , Proliferación Celular , Humanos
4.
J Allergy Clin Immunol ; 146(6): 1387-1396.e13, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32407837

RESUMEN

BACKGROUND: Reduced levels of prostaglandin E2 (PGE2) contribute to aspirin-induced hypersensitivity. COX inhibitors are also frequent cofactors in anaphylaxis. Whether alterations in the PGE2 system contribute to anaphylaxis independently of COX inhibitor intake is unclear. OBJECTIVE: Our aim was to test the hypothesis that relative PGE2 deficiency predisposes to anaphylaxis. METHODS: Sera from 48 patients with anaphylaxis and 27 healthy subjects were analyzed for PGE2 levels and correlated against severity; 9α,11ß-PGF2 and PGI2 metabolites were measured for control purposes. PGE2 stabilization by 15-hydroxyprostaglandin dehydrogenase inhibitor or EP2 or EP4 receptor agonists were used in a murine model of passive systemic anaphylaxis. FcεRI-triggered mediator release was determined in bone marrow-derived cultured mast cells (MCs) and human skin-derived MCs. Signaling was studied by Western blot analysis. RESULTS: Patients with anaphylaxis were characterized by markedly reduced PGE2 levels vis-à-vis healthy subjects, whereas prostacyclin metabolite levels were diminished only weakly, and 9α,11ß-PGF2 levels conversely increased. PGE2 was negatively correlated with severity. Lower PGE2 levels and higher susceptibility to anaphylaxis were also found in C57BL/6 mice vis-à-vis in Balb/c mice. Stabilization of PGE2 level by 15-hydroxyprostaglandin dehydrogenase inhibitor protected mice against anaphylaxis. Exogenous PGE2 attenuated bone marrow-derived cultured MC activation through EP2 and EP4 receptors. EP2 and EP4 agonism also curbed FcεRI-mediated degranulation of human MCs. Mechanistically, PGE2 interfered with the phosphorylation of phospholipase C gamma-1 and extracellular signal-regulated kinase. CONCLUSIONS: Homeostatic levels of PGE2 attenuate MC activation via EP2/EP4 and protect against anaphylaxis. Relative deficiency of PGE2 predisposes to anaphylaxis in humans and mice, whereas PGE2 stabilization protects against anaphylactic reactions.


Asunto(s)
Anafilaxia/inmunología , Dinoprostona/deficiencia , Mastocitos/inmunología , Anafilaxia/patología , Animales , Dinoprostona/inmunología , Susceptibilidad a Enfermedades/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Humanos , Mastocitos/patología , Ratones , Ratones Endogámicos BALB C , Fosfolipasa C gamma/inmunología , Subtipo EP2 de Receptores de Prostaglandina E/inmunología , Subtipo EP4 de Receptores de Prostaglandina E/inmunología , Índice de Severidad de la Enfermedad
5.
Handb Exp Pharmacol ; 262: 157-175, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31820176

RESUMEN

Prostaglandins (PGs) are highly bioactive fatty acids. PGs, especially prostaglandin E2 (PGE2), are abundantly produced by cells of both the bone-forming (osteoblast) lineage and the bone-resorbing (osteoclast) lineage. The inducible cyclooxygenase, COX-2, is largely responsible for most PGE2 production in bone, and once released, PGE2 is rapidly degraded in vivo. COX-2 is induced by multiple agonists - hormones, growth factors, and proinflammatory factors - and the resulting PGE2 may mediate, amplify, or, as we have recently shown for parathyroid hormone (PTH), inhibit responses to these agonists. In vitro, PGE2 can directly stimulate osteoblast differentiation and, indirectly via stimulation of RANKL in osteoblastic cells, stimulate the differentiation of osteoclasts. The net balance of these two effects of PGE2 in vivo on bone formation and bone resorption has been hard to predict and, as expected for such a widespread local factor, hard to study. Some of the complexity of PGE2 actions on bone can be explained by the fact that there are four receptors for PGE2 (EP1-4). Some of the major actions of PGE2 in vitro occur via EP2 and EP4, both of which can stimulate cAMP signaling, but there are other distinct signaling pathways, important in other tissues, which have not yet been fully elucidated in bone cells. Giving PGE2 or agonists of EP2 and EP4 to accelerate bone repair has been examined with positive results. Further studies to clarify the pathways of PGE2 action in bone may allow us to identify new and more effective ways to deliver the therapeutic benefits of PGE2 in skeletal disorders.


Asunto(s)
Resorción Ósea , Prostaglandinas , Humanos , Osteoclastos/química , Osteoclastos/fisiología , Subtipo EP4 de Receptores de Prostaglandina E/química
6.
Cancer Metastasis Rev ; 37(2-3): 369-384, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29858743

RESUMEN

Lymphangiogenesis (formation of new lymphatic vessels), unlike angiogenesis, has been a lesser-focused field in cancer biology, because of earlier controversy regarding whether lymphatic metastasis occurs via pre-existing or newly formed lymphatics. Recent evidence reveals that peri-tumoral or intra-tumoral lymphangiogenesis is a precursor for lymphatic metastasis in most carcinomas and melanomas. Two major lymphangiogenic factors, vascular endothelial growth factor (VEGF)-C and VEGF-D, are produced by cancer cells or immune cells such as macrophages in the tumor-stroma to promote sprouting of lymphatics from lymphatic endothelial cells (LEC) or LEC precursors (LECP) by binding to their primary (high affinity) receptor VEGF-R3 or secondary receptors VEGF-R2, neuropilin (NRP)2 and α9/ß1 integrin. Many other growth factors/receptors such as VEGF-A/VEGF-R2, fibroblast growth factor (FGF)2/FGF-R, platelet-derived growth factor (PDGF)/PDGF-R, hepatocyte growth factor (HGF)/C-Met, angiopoietins (Ang)1, 2/Tie2, and chemokines/ chemokine receptors (CCL21/CCR7, CCL12/CCR4) can also stimulate LEC sprouting directly or indirectly. This review deals with the roles of prostaglandins (PG), in particular PGE2, in cancer-associated lymphangiogenesis, with special emphasis on breast cancer. We show that cyclooxygenase (COX)-2 expression by breast cancer cells or tumor stroma leading to high PGE2 levels in the tumor milieu promotes lymphangiogenesis and lymphatic metastases, resulting from binding of PGE2 to PGE receptors (EP, in particular EP4) on multiple cell types: tumor cells, tumor-infiltrating immune cells, and LEC. EP4 activation on cancer cells and macrophages upregulated VEGF-C/D production to stimulate LEC sprouting. Furthermore, ligation of EP4 with PGE2 on cancer or host cells can initiate a new cascade of molecular events leading to cross-talk between cancer cells and LEC, facilitating lymphangiogenesis and lympho-vascular transport of cancer cells. We make a case for EP4 as a potential therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Linfangiogénesis , Neovascularización Patológica , Prostaglandinas/metabolismo , Microambiente Tumoral , Animales , Biomarcadores , Neoplasias de la Mama/complicaciones , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Eicosanoides/metabolismo , Células Endoteliales/metabolismo , Femenino , Humanos , Técnicas In Vitro , Metástasis Linfática , Linfedema/etiología , Redes y Vías Metabólicas , Receptores de Prostaglandina/metabolismo , Transducción de Señal
7.
Dig Dis Sci ; 64(10): 2806-2814, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30989466

RESUMEN

BACKGROUND: The prostaglandin D2 receptor DP2 has been implicated in eosinophil infiltration and the development of eosinophilic esophagitis (EoE). AIMS AND METHODS: In this study, we investigated an involvement of PGE2 (EP1-EP4) and PGD2 (DP1) receptors in EoE by measuring their expression in peripheral blood eosinophils and esophageal mucosal biopsies of EoE patients and by performing migration and adhesion assays with eosinophils from healthy donors. RESULTS: Expression of EP2 and EP4, but not EP1 and EP3, was decreased in blood eosinophils of patients with EoE vs. control subjects. Adhesion of eosinophils to esophageal epithelial cells was decreased by EP2 receptor agonist butaprost and EP4 agonist ONO-AE1-329, whereas DP1 agonist BW245C increased adhesion. In chemotaxis assays with supernatant from human esophageal epithelial cells, only ONO-AE1-329 but not butaprost or BW245C inhibited the migration of eosinophils. Expression of EP and DP receptors in epithelial cells and eosinophils was detected in sections of esophageal biopsies from EoE patients by immunohistochemistry. qPCR of biopsies from EoE patients revealed that gene expression of EP4 and DP1 was the highest among PGE2 and PGD2 receptors. Esophageal epithelial cells in culture showed high gene expression for EP2 and EP4. Activation of EP2 and EP4 receptors decreased barrier integrity of esophageal epithelial cells in impedance assays. CONCLUSIONS: Activation of EP2 and EP4 receptors may inhibit eosinophil recruitment to the esophageal mucosa. However, their activation could negatively affect esophageal barrier integrity suggesting that eosinophilic rather than epithelial EP2 and EP4 have a protective role in EoE.


Asunto(s)
Esofagitis Eosinofílica , Eosinófilos , Mucosa Esofágica , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E , Alprostadil/análogos & derivados , Alprostadil/farmacología , Adhesión Celular , Ensayos de Migración Celular/métodos , Células Cultivadas , Esofagitis Eosinofílica/sangre , Esofagitis Eosinofílica/metabolismo , Esofagitis Eosinofílica/patología , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Mucosa Esofágica/efectos de los fármacos , Mucosa Esofágica/metabolismo , Mucosa Esofágica/patología , Humanos , Inmunohistoquímica , Éteres Metílicos/farmacología , Proyectos Piloto , Prostaglandinas E Sintéticas/farmacología , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Subtipo EP2 de Receptores de Prostaglandina E/análisis , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/análisis
8.
Int J Mol Sci ; 19(4)2018 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-29596308

RESUMEN

G-protein-coupled receptors (GPCRs, also called seven-transmembrane or heptahelical receptors) are a superfamily of cell surface receptor proteins that bind to many extracellular ligands and transmit signals to an intracellular guanine nucleotide-binding protein (G-protein). When a ligand binds, the receptor activates the attached G-protein by causing the exchange of Guanosine-5'-triphosphate (GTP) for guanosine diphosphate (GDP). They play a major role in many physiological functions, as well as in the pathology of many diseases, including cancer progression and metastasis. Only a few GPCR members have been exploited as targets for developing drugs with therapeutic benefit in cancer. Present review briefly summarizes the signaling pathways utilized by the EP (prostaglandin E receptor) family of GPCR, their physiological and pathological roles in carcinogenesis, with special emphasis on the roles of EP4 in breast cancer progression. We make a case for EP4 as a promising newer therapeutic target for treating breast cancer. We show that an aberrant over-expression of cyclooxygenase (COX)-2, which is an inflammation-associated enzyme, occurring in 40-50% of breast cancer patients leads to tumor progression and metastasis due to multiple cellular events resulting from an increased prostaglandin (PG) E2 production in the tumor milieu. They include inactivation of host anti-tumor immune cells, such as Natural Killer (NK) and T cells, increased immuno-suppressor function of tumor-associated macrophages, promotion of tumor cell migration, invasiveness and tumor-associated angiogenesis, due to upregulation of multiple angiogenic factors including Vascular Endothelial Growth Factor (VEGF)-A, increased lymphangiogenesis (due to upregulation of VEGF-C/D), and a stimulation of stem-like cell (SLC) phenotype in cancer cells. All of these events were primarily mediated by activation of the Prostaglandin (PG) E receptor EP4 on tumor or host cells. We show that selective EP4 antagonists (EP4A) could mitigate all of these events tested with cells in vitro as well as in vivo in syngeneic COX-2 expressing mammary cancer bearing mice or immune-deficient mice bearing COX-2 over-expressing human breast cancer xenografts. We suggest that EP4A can avoid thrombo-embolic side effects of long term use of COX-2 inhibitors by sparing cardio-protective roles of PGI2 via IP receptor activation or PGE2 via EP3 receptor activation. Furthermore, we identified two COX-2/EP4 induced oncogenic and SLC-stimulating microRNAs-miR526b and miR655, one of which (miR655) appears to be a potential blood biomarker in breast cancer patients for monitoring SLC-ablative therapies, such as with EP4A. We suggest that EP4A will likely produce the highest benefit in aggressive breast cancers, such as COX-2 expressing triple-negative breast cancers, when combined with other newer agents, such as inhibitors of programmed cell death (PD)-1 or PD-L1.


Asunto(s)
Proteínas de Neoplasias , Subtipo EP4 de Receptores de Prostaglandina E , Neoplasias de la Mama Triple Negativas , Dinoprostona/genética , Dinoprostona/metabolismo , Femenino , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
9.
Int Ophthalmol ; 38(4): 1703-1708, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28695379

RESUMEN

PURPOSE: In the present study, we aimed to evaluate the expression of EP receptors in primary and recurrent human pterygium tissues. METHODS: Pterygium samples were collected from 65 patients with primary pterygium and 16 patients with recurrent pterygium. Normal conjunctival tissues were collected from nasal interpalpebral area from 17 patients without systemic and any other ocular pathology. Expression of EP receptors was evaluated by immunohistochemistry. The median value for each receptor staining score (RSS) was determined in normal conjunctival specimens. In this study, RSS of > median value was defined as positive staining or high expression and ≤ median value as negative staining or weak expression in specimens. Chi-square test was used for statistical analysis, and p value of less than 0.05 was considered significant. RESULTS: Stromal expression of EP1 was significantly higher in primary and recurrent pterygium specimens compared to normal conjunctival tissues (p = 0.007 and p = 0.002, respectively). Epithelial expressions of EP2 and EP3 were significantly lower in primary pterygium specimens compared to normal conjunctival tissues (p = 0.005 and p < 0.0001, respectively), and stromal expressions were insignificant. Stromal expression of EP4 was significantly higher in primary and recurrent pterygium specimens compared to normal conjunctival tissues (p = 0.002 and p = 0.012, respectively). CONCLUSIONS: Expression of EP receptors has been up- or downregulated in primary and recurrent pterygium tissues, and these receptors may play a role in formation and recurrence of pterygium.


Asunto(s)
Conjuntiva/metabolismo , Pterigion/metabolismo , Receptores de Prostaglandina E/metabolismo , Adulto , Estudios de Casos y Controles , Sustancia Propia/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad
10.
Brain Behav Immun ; 60: 27-31, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27375005

RESUMEN

From experiments in mice in which the prostaglandin E2 (PGE2) synthesizing enzyme mPGES-1 was genetically deleted, as well as from experiments in which PGE2 was injected directly into the brain, PGE2 has been implicated as a mediator of inflammatory induced anorexia. Here we aimed at examining which PGE2 receptor (EP1-4) that was critical for the anorexic response to peripherally injected interleukin-1ß (IL-1ß). However, deletion of neither EP receptor in mice, either globally (for EP1, EP2, and EP3) or selectively in the nervous system (EP4), had any effect on the IL-1ß induced anorexia. Because these mice were all on a C57BL/6 background, whereas previous observations demonstrating a role for induced PGE2 in IL-1ß evoked anorexia had been carried out on mice on a DBA/1 background, we examined the anorexic response to IL-1ß in mice with deletion of mPGES-1 on a C57BL/6 background and a DBA/1 background, respectively. We confirmed previous findings that mPGES-1 knock-out mice on a DBA/1 background displayed attenuated anorexia to IL-1ß; however, mice on a C57BL/6 background showed the same profound anorexia as wild type mice when carrying deletion of mPGES-1, while displaying almost normal food intake after pretreatment with a cyclooxygenase-2 inhibitor. We conclude that the involvement of induced PGE2 in IL-1ß evoked anorexia is strain dependent and we suggest that different routes that probably involve distinct prostanoids exist by which inflammatory stimuli may evoke an anorexic response and that these routes may be of different importance in different strains of mice.


Asunto(s)
Anorexia/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/metabolismo , Interleucina-1beta/metabolismo , Animales , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Oxidorreductasas Intramoleculares/efectos de los fármacos , Oxidorreductasas Intramoleculares/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Prostaglandina-E Sintasas/farmacología , Regulación hacia Arriba/efectos de los fármacos
11.
J Cell Mol Med ; 19(10): 2413-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26176998

RESUMEN

Previous work from both our lab and others have indicated that exposure to 50 Hz magnetic fields (ELF-MF) was able to modify ion channel functions. However, very few studies have investigated the effects of MF on γ-aminobutyric acid (GABA) type A receptors (GABA(A) Rs) channel functioning, which are fundamental to overall neuronal excitability. Here, our major goal is to reveal the potential effects of ELF-MF on GABA(A) Rs activity in rat cerebellar granule neurons (CGNs). Our results indicated that exposing CGNs to 1 mT ELF-MF for 60 min. significantly increased GABA(A) R currents without modifying sensitivity to GABA. However, activation of PKA by db-cAMP failed to do so, but led to a slight decrease instead. On the other hand, PKC activation or inhibition by PMA or Bis and Docosahexaenoic acid (DHA) mimicked or eliminated the field-induced-increase of GABA(A) R currents. Western blot analysis indicated that the intracellular levels of phosphorylated PKC (pPKC) were significantly elevated after 60 min. of ELF-MF exposure, which was subsequently blocked by application of DHA or EP1 receptor-specific (prostaglandin E receptor 1) antagonist (SC19220), but not by EP2-EP4 receptor-specific antagonists. SC19220 also significantly inhibited the ELF-MF-induced elevation on GABA(A) R currents. Together, these data obviously demonstrated for the first time that neuronal GABA(A) currents are significantly increased by ELF-MF exposure, and also suggest that these effects are mediated via an EP1 receptor-mediated PKC pathway. Future work will focus on a more comprehensive analysis of the physiological and/or pathological consequences of these effects.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Activación del Canal Iónico , Campos Magnéticos , Neuronas/metabolismo , Proteína Quinasa C/metabolismo , Receptores de GABA-A/metabolismo , Subtipo EP1 de Receptores de Prostaglandina E/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Modelos Biológicos , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Análisis de Regresión , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología
12.
Prostaglandins Other Lipid Mediat ; 121(Pt A): 4-16, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26463849

RESUMEN

The prostanoid E2 (PGE2) is known to modulate the aggregative response of platelets to their conventional agonists such as ADP, TXA2, thrombin or collagen. Through the activation of its receptor EP3, PGE2 sensitizes platelets to their agonists but also inhibits them through its two other receptors, EP2 and EP4. In mice, the net result of these opposed actions is the EP3-mediated potentiation of platelet aggregation and the in vivo aggravation of murine atherothrombosis. Since the pathway PGE2/EP3 is not involved in murine hemostasis, we propose a "platelet EP3 paradigm" to describe this apparently paradoxical association between the facilitating impact on atherothrombosis and the unaltered hemostasis. Consistent with this paradigm, a drug blocking EP3 dramatically decreased atherothrombosis without inducing bleeding in mice. In humans, several studies did not agree on the effect of PGE2 on platelets. Reinterpreting these data with the notion of "potentiation window" and taking the platelet initial cAMP level into account reconciled these inconsistent results. Thereby, the in vitro potentiating effect of PGE2 on human platelets becomes clear. In addition, the EP3 blocking drug DG-041 abrogated the potentiating effect of PGE2 in whole human blood but did not prolong bleeding times in volunteers. Thus, the murine "platelet EP3 paradigm" would apply to humans if the aggravating role of PGE2 on atherothrombosis is shown in patients. Therefore, testing an EP3 blocker in a phase III trial would be of high interest to fulfill the unmet medical need which is to control atherothrombosis without impacting hemostasis and thus to improve the prevention of myocardial infarction.


Asunto(s)
Aterosclerosis/complicaciones , Dinoprostona/metabolismo , Hemorragia/inducido químicamente , Terapia Molecular Dirigida/métodos , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo , Trombosis/complicaciones , Trombosis/prevención & control , Animales , Dinoprostona/biosíntesis , Humanos , Subtipo EP3 de Receptores de Prostaglandina E/antagonistas & inhibidores , Trombosis/metabolismo , Trombosis/fisiopatología
13.
Am J Physiol Renal Physiol ; 307(5): F539-50, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25007872

RESUMEN

The actions of prostaglandin E2 (PGE2) in the kidney are mediated by G protein-coupled E-prostanoid (EP) receptors, which affect renal growth and function. This report examines the role of EP receptors in mediating the effects of PGE2 on Madin-Darby canine kidney (MDCK) cell growth. The results indicate that activation of Gs-coupled EP2 and EP4 by PGE2 results in increased growth, while EP1 activation is growth inhibitory. Indeed, two EP1 antagonists (ONO-8711 and SC51089) stimulate, rather than inhibit, MDCK cell growth, an effect that is lost following an EP1 knockdown. Similar observations were made with M1 collecting duct and rabbit kidney proximal tubule cells. ONO-8711 even stimulates growth in the absence of exogenous PGE2, an effect that is prevented by ibuprofen (indicating a dependence upon endogenous PGE2). The involvement of Akt was indicated by the observation that 1) ONO-8711 and SC51089 increase Akt phosphorylation, and 2) MK2206, an Akt inhibitor, prevents the increased growth caused by ONO-8711. The involvement of the EGF receptor (EGFR) was indicated by 1) the increased phosphorylation of the EGFR caused by SC51089 and 2) the loss of the growth-stimulatory effect of ONO-8711 and SC51089 caused by the EGFR kinase inhibitor AG1478. The growth-stimulatory effect of ONO-8711 was lost following an EGFR knockdown, and transduction of MDCK cells with a dominant negative EGFR. These results support the hypothesis that 1) signaling via the EP1 receptor involves Akt as well as the EGFR, and 2), EP1 receptor pharmacology may be employed to prevent the aberrant growth associated with a number of renal diseases.


Asunto(s)
Proliferación Celular/fisiología , Receptores ErbB/fisiología , Riñón/patología , Proteínas Proto-Oncogénicas c-akt/fisiología , Subtipo EP1 de Receptores de Prostaglandina E/antagonistas & inhibidores , Transducción de Señal/fisiología , Animales , Compuestos Bicíclicos con Puentes/farmacología , Caproatos/farmacología , Proliferación Celular/efectos de los fármacos , Perros , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Hidrazinas/farmacología , Ibuprofeno/farmacología , Riñón/efectos de los fármacos , Riñón/fisiología , Células de Riñón Canino Madin Darby , Oxazepinas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Quinazolinas/farmacología , Subtipo EP1 de Receptores de Prostaglandina E/efectos de los fármacos , Subtipo EP1 de Receptores de Prostaglandina E/fisiología , Subtipo EP2 de Receptores de Prostaglandina E/fisiología , Subtipo EP4 de Receptores de Prostaglandina E/fisiología , Tirfostinos/farmacología
14.
Am J Physiol Renal Physiol ; 307(3): F243-50, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24966087

RESUMEN

Chronic kidney disease is a leading cause of morbidity and mortality in the world. A better understanding of disease mechanisms has been gained in recent years, but the current management strategies are ineffective at preventing disease progression. A widespread focus of research is placed on elucidating the specific processes implicated to find more effective therapeutic options. PGE2, acting on its four EP receptors, regulates many renal disease processes; thus EP receptors could prove to be important targets for kidney disease intervention strategies. This review summarizes the major pathogenic mechanisms contributing to initiation and progression of chronic kidney disease, emphasizing the role of hyperglycemia, hypertension, inflammation, and oxidative stress. We have long recognized the multifaceted role of PGs in both the initiation and progression of chronic kidney disease, yet studies are only now seriously contemplating specific EP receptors as targets for therapy. Given the plethora of renal complications attributed to PG involvement in the kidney, this review highlights these pathogenic events and emphasizes the PGE2 receptor targets as options available to complement current therapeutic strategies.


Asunto(s)
Progresión de la Enfermedad , Receptores de Prostaglandina E/fisiología , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/fisiopatología , Humanos , Hiperglucemia/fisiopatología , Hipertensión/fisiopatología , Inflamación/fisiopatología , Estrés Oxidativo/fisiología
15.
Prostate ; 74(10): 999-1011, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24802614

RESUMEN

BACKGROUND: Previous studies have shown that COX-2 inhibitors inhibit cancer cell proliferation. However, the molecular mechanism remains elusive. METHODS: Prostate cancer LNCaP, 22Rv1, and PC3 cells were cultured and treated with the COX-2 inhibitors celecoxib and CAY10404. Knockdown of COX-2 in LNCaP cells was carried out using lentiviral vector-loaded COX-2 shRNA. Cell cycle progression and cell proliferation were analyzed by flow cytometry, microscopy, cell counting, and the MTT assay. The antagonists of EP1, EP2, EP3, and EP4 were used to examine the effects of the PGE2 signaling. The effect of COX-2 inhibitors and COX-2 knockdown on expression of the kinetochore/centromere genes and proteins was determined by RT-PCR and immunoblotting. RESULTS: Treatment with the COX-2 inhibitors celecoxib and CAY10404 or knockdown of COX-2 significantly inhibited prostate cancer cell proliferation. Flow-cytometric analysis and immunofluorescent staining confirmed the cell cycle arrested at the G2/M phase. Biochemical analysis showed that inhibition of COX-2 or suppression of COX-2 expression induced a dramatic down-regulation of key proteins in the kinetochore/centromere assembly, such as ZWINT, Cdc20, Ndc80, CENP-A, Bub1, and Plk1. Furthermore, the EP1 receptor antagonist SC51322, but not the EP2, EP3, and EP4 receptor antagonists, produced similar effects to the COX-2 inhibitors on cell proliferation and down-regulation of kinetochore/centromere proteins, suggesting that the effect of the COX-2 inhibition is through inactivation of the EP1 receptor signaling. CONCLUSIONS: Our studies indicate that inhibition of COX-2 can arrest prostate cancer cell cycle progression through inactivation of the EP1 receptor signaling and down-regulation of kinetochore/centromere proteins.


Asunto(s)
Centrómero/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Cinetocoros/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Autoantígenos/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Centrómero/metabolismo , Proteína A Centromérica , Proteínas Cromosómicas no Histona/genética , Ciclooxigenasa 2/fisiología , Regulación hacia Abajo , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Cinetocoros/metabolismo , Masculino , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas/fisiología , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Subtipo EP1 de Receptores de Prostaglandina E/fisiología
16.
Prostaglandins Other Lipid Mediat ; 113-115: 62-8, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25263346

RESUMEN

Prostaglandin E2 (PGE2) is elevated during cardiac injury and we have previously shown that mice lacking the PGE2 EP4 receptor display dilated cardiomyopathy (DCM) with increased expression of the membrane type matrix metalloproteinase, MMP-14. We thus hypothesized that PGE2 regulates expression of MMP-14 and also affects fibroblast migration. Primary cultures of neonatal rat ventricular fibroblasts (NVFs) were used to test the effects of PGE2. Gene and protein expression was assessed by real time RT-PCR and Western blot, MMP activity was determined by zymography and migration of NVF was assessed by motility in a transwell system. PGE2 reduced expression of MMP-14 and these effects were antagonized by an EP4 antagonist. An EP4 agonist mimicked the effect of PGE2. PGE2 also increased mRNA and protein levels of plasminogen activator inhibitor-1 (PAI-1), an inhibitor of MMP activation. However, PGE2-stimulation of PAI-1 was mediated by the EP1/EP3 receptor and not EP4. Migration of NVF was assessed by motility in a transwell system. Treatment of NVFs with PGE2 reduced the number of cells migrating toward 10% FCS. Treatment with the EP2 agonist also reduced migration but did not affect MMP-14 expression or PAI-1. Our results suggest that PGE2 utilizes different receptors and mechanisms to ultimately decrease MMP expression and NVF migration.


Asunto(s)
Cardiomiopatías/metabolismo , Dinoprostona/metabolismo , Regulación de la Expresión Génica/fisiología , Metaloproteinasa 14 de la Matriz/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Animales Recién Nacidos , Cardiomiopatías/enzimología , Movimiento Celular/fisiología , Dinoprostona/análogos & derivados , Dinoprostona/farmacología , Fibroblastos/enzimología , Fibroblastos/metabolismo , Masculino , Metaloproteinasa 14 de la Matriz/genética , Éteres Metílicos/farmacología , Naftalenos/farmacología , Fenilbutiratos/farmacología , Inhibidor 1 de Activador Plasminogénico/genética , ARN Mensajero/química , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores
17.
Pharmacol Ther ; 241: 108313, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36427569

RESUMEN

Asthma is the most common airway chronic disease with treatments aimed mainly to control the symptoms. Adrenergic receptor agonists, corticosteroids and anti-leukotrienes have been used for decades, and the development of more targeted asthma treatments, known as biological therapies, were only recently established. However, due to the complexity of asthma and the limited efficacy as well as the side effects of available treatments, there is an urgent need for a new generation of asthma therapies. The anti-inflammatory and bronchodilatory effects of prostaglandin E2 in asthma are promising, yet complicated by undesirable side effects, such as cough and airway irritation. In this review, we summarize the most important literature on the role of all four E prostanoid (EP) receptors on the lung's immune and structural cells to further dissect the relevance of EP2/EP4 receptors as potential targets for future asthma therapy.


Asunto(s)
Asma , Dinoprostona , Humanos , Subtipo EP2 de Receptores de Prostaglandina E , Asma/tratamiento farmacológico , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Pulmón
18.
Life Sci ; 310: 121114, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36273629

RESUMEN

AIMS: Prostaglandins are important signaling lipids with prostaglandin E2 (PGE2) known to be the most abundant prostaglandin across tissues. In kidney, PGE2 plays an important role in the regulation of kidney homeostasis through its EP receptor signaling. Catabolism of PGE2 yields the metabolic products that are widely considered biologically inactive. Although recent in vitro evidence suggested the ability of 15-keto-PGE2 (a downstream metabolite of PGE2) to activate EP receptors, the question whether 15-keto-PGE2 exhibits physiological roles remains unresolved. MATERIALS AND METHODS: Pharmacological treatment was performed in transgenic zebrafish embryos using 500 µM 15-keto-PGE2 and 20 µM EP receptors antagonists' solutions during zebrafish embryonic development. After the exposure period, the embryos were fixed for confocal microscopy imaging and glomerular morphology analysis. KEY FINDINGS: Here, we show that 15-keto-PGE2 can bind and stabilize EP2 and EP4 receptors on the plasma membrane in the yeast model. Using lipidomic analysis, we demonstrate both PGE2 and 15-keto-PGE2 are present at considerable levels in zebrafish embryos. Our high-resolution image analysis reveals the exogenous treatment with 15-keto-PGE2 perturbs glomerular vascularization during zebrafish development. Specifically, we show that the increased levels of 15-keto-PGE2 cause intercalation defects between podocytes and endothelial cells of glomerular capillaries effectively reducing the surface area of glomerular filtration barrier. Importantly, 15-keto-PGE2-dependent defects can be fully reversed by combined blockade of the EP2 and EP4 receptors. SIGNIFICANCE: Altogether, our results reveal 15-keto-PGE2 to be a biologically active metabolite that modulates the EP receptor signaling in vivo, thus playing a potential role in kidney biology.


Asunto(s)
Subtipo EP2 de Receptores de Prostaglandina E , Pez Cebra , Animales , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Células Endoteliales/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E , Prostaglandinas
19.
Front Endocrinol (Lausanne) ; 13: 875425, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35813612

RESUMEN

Prostaglandin E2 (PGE2) is an important prostanoid expressing throughout the kidney and cardiovascular system. Despite the diverse effects on fluid metabolism and blood pressure, PGE2 is implicated in sustaining volume and hemodynamics homeostasis. PGE2 works through four distinct E-prostanoid (EP) receptors which are G protein-coupled receptors. To date, pharmacological specific antagonists and agonists of all four subtypes of EP receptors and genetic targeting knockout mice for each subtype have helped in uncoupling the diverse functions of PGE2 and discriminating the respective characteristics of each receptor. In this review, we summarized the functions of individual EP receptor subtypes in the renal and blood vessels and the molecular mechanism of PGE2-induced fluid metabolism and blood pressure homeostasis.


Asunto(s)
Dinoprostona , Receptores de Prostaglandina E , Animales , Presión Sanguínea , Dinoprostona/metabolismo , Dinoprostona/farmacología , Ratones , Ratones Noqueados , Receptores de Prostaglandina E/metabolismo , Equilibrio Hidroelectrolítico
20.
J Pers Med ; 11(8)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34442416

RESUMEN

Prostaglandin E2 (PGE2) is a major actor mediating renal injury. We aimed to determine genetic variability in the genes coding for its receptors (PTGER1-4) and study associations with nephrosclerosis risk and clinical outcomes. We identified 96 tag-SNPs capturing global variability in PTGER1-4 and screened 1209 nephrosclerosis patients and controls. The effect of these variants was evaluated by multivariate regression analyses. Two PTGER3 SNPs, rs11209730 and rs10399704, remained significant in a backward elimination regression model with other non-genetic variables (OR = 1.45 (1.07-1.95), p = 0.016 and OR = 0.71 (0.51-0.99), p = 0.041, respectively). In the nephrosclerosis patients, a proximal region of PTGER3 was tagged as relevant for eGFR (p values for identified SNPs ranged from 0.0003 to 0.038). Two consecutive PTGER3 SNPs, rs2284362 and rs2284363, significantly decreased systolic (p = 0.005 and p = 0.0005), diastolic (p = 0.039 and p = 0.005), and pulse pressure values (p = 0.038 and 0.014). Patients were followed for a median of 47 months (7-54) to evaluate cardiovascular (CV) risk. Cox regression analysis showed that carriers of the PTGER1rs2241360 T variant had better CV event-free survival than wild-type individuals (p = 0.029). In addition, PTGER3rs7533733 GG carriers had lower event-free survival than AA/AG patients (p = 0.011). Our results indicate that genetic variability in PGE2 receptors, particularly EP3, may be clinically relevant for nephrosclerosis and its associated CV risk.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA