Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 325
Filtrar
1.
Org Lett ; 22(1): 257-260, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31860319
2.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2504-2515, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31195118

RESUMEN

The therapeutic efficacy of the antineoplastic drug cisplatin is limited by its nephrotoxicity, which affects particularly to proximal tubular cells (PTC). Cisplatin-induced cytotoxicity appears to be multifactorial and involves inflammation, oxidative stress as well as apoptosis. We have recently shown that the cyclo-oxygenase-2 (COX-2)/intracellular prostaglandin E2 (iPGE2)/EP receptor pathway mediates the apoptotic effect of cisplatin on human proximal tubular HK-2 cells. Here, we studied the effects on HK-2 cells of apoptotic bodies (ABs) generated after treatment of HK-2 cells with cisplatin. We found that ABs inhibited cell growth, induced apoptosis and increased COX-2 expression and iPGE2 in ABs-recipient HK-2 cells. Inhibition of the COX-2/iPGE2/EP receptor pathway in these cells prevented the effects of ABs without interfering with their internalization. Interestingly, 2nd generation ABs (i.e. ABs released by cells undergoing apoptosis upon treatment with ABs) did not trigger apoptosis in naïve HK-2 cells, and stimulated cell proliferation through the COX-2/iPGE2/EP receptor pathway. These results suggest that ABs, through iPGE2-dependent mechanisms, might have a relevant role in the natural history of cisplatin-induced acute kidney failure because they contribute first to the propagation of the noxious effects of cisplatin to non-injured PTC and then to the promotion of the proliferative tubular response required for proximal tubule repair. Since iPGE2 also mediates both cisplatin-induced HK-2 cell apoptosis, intervention in the COX-2/iPGE2/EP receptor pathway might provide us with new therapeutic avenues in patients with cisplatin-induced acute kidney injury.


Asunto(s)
Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/toxicidad , Vesículas Extracelulares/metabolismo , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Túbulos Renales Proximales/citología , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
3.
BJU Int ; 124(5): 883-891, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31166645

RESUMEN

OBJECTIVES: To evaluate, using a rat model of non-bacterial prostatic inflammation, the prostaglandin production and expression profiles of E-series prostaglandin (EP) receptor subtypes, which are reportedly implicated in the development of overactive bladder, in the bladder mucosa, and to investigate the effect of EP receptor type 4 (EP4) blockade on bladder overactivity after prostatic inflammation. METHODS: Male Sprague-Dawley rats were used. Prostatic inflammation was induced by formalin injection (5%; 50 µL per lobe) into the bilateral ventral lobes of the prostate. At 10 days after induction of prostatic inflammation or vehicle injection, bladder tissues from the deeply anaesthetized rats were harvested and separated into mucosal and detrusor layers. Then, prostaglandin E2 (PGE2) concentrations and protein levels of PGE2 receptors (EP1-4) in the bladder mucosa and detrusor were measured by ELISA and Western blotting, respectively. In separate groups of control and formalin-treated rats, awake cystometry was performed to evaluate the changes in bladder activity after prostatic inflammation. In addition, the effect of intravesical administration of a selective EP4 antagonist (ONO-AE3-208; 30 µm) on bladder activity was evaluated in control rats and rats with prostatic inflammation. RESULTS: PGE2 concentration and protein levels of EP4, but not other EP receptor subtypes, in the bladder mucosa and detrusor layers were significantly increased in formalin-injected rats vs vehicle-injected control rats. In cystometry, rats with prostatic inflammation exhibited a significant decrease in intercontraction intervals (ICIs) compared with control rats. Intravesical application of ONO-AE3-208 (30 µm), but not vehicle application, significantly increased ICIs in rats with prostatic inflammation, whereas ONO-AE3-208 at this concentration did not significantly affect any cystometric values in control rats. CONCLUSIONS: Because intravesical administration of an EP4 antagonist effectively improved bladder overactivity after prostatic inflammation, EP4 activation, along with increased PGE2 production in the bladder mucosa, seems to be an important contributing factor to bladder overactivity induced by prostatic inflammation. Thus, blockade of EP4 in the bladder could be a therapeutic approach to male lower urinary tract symptoms attributable to benign prostatic hyperplasia with prostatic inflammation.


Asunto(s)
Inflamación , Prostaglandinas E/metabolismo , Prostatitis/metabolismo , Receptores de Prostaglandina E , Vejiga Urinaria Hiperactiva , Animales , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Membrana Mucosa/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Vejiga Urinaria/metabolismo , Vejiga Urinaria/fisiopatología , Vejiga Urinaria Hiperactiva/metabolismo , Vejiga Urinaria Hiperactiva/fisiopatología
4.
JCI Insight ; 4(9)2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31045572

RESUMEN

The physiological process of defecation is directly controlled by colorectal motility. The transient receptor potential ankyrin 1 (TRPA1) channel is expressed in small intestine enterochromaffin cells and is involved in gastrointestinal motility via serotonin release. In the colorectum, however, enterochromaffin cell localization is largely distinct from that in the small intestine. Here, we investigated the role of lower gastrointestinal tract TRPA1 in modulating colorectal motility. We found that in colonic tissue, TRPA1 is predominantly expressed in mesenchymal cells of the lamina propria, which are clearly distinct from those in the small intestine. These cells coexpressed COX1 and microsomal prostaglandin E synthase-1. Intracolonic administration of TRPA1 agonists induced colonic contraction, which was suppressed by a prostaglandin E2 (PGE2) receptor 1 antagonist. TRPA1 activation induced calcium influx and PGE2 release from cultured human fibroblastic cells. In dextran sulfate sodium-treated animals, both TRPA1 and its endogenous agonist were dramatically increased in the colonic lamina propria, accompanied by abnormal colorectal contractions. Abnormal colorectal contractions were significantly prevented by pharmacological and genetic inhibition of TRPA1. In conclusion, in the lower gastrointestinal tract, mesenchymal TRPA1 activation results in PGE2 release and consequently promotes colorectal contraction, representing what we believe is a novel physiological and inflammatory bowel disease-associated mechanism of gastrointestinal motility.


Asunto(s)
Colon/metabolismo , Motilidad Gastrointestinal/fisiología , Mesodermo/metabolismo , Membrana Mucosa/metabolismo , Canal Catiónico TRPA1/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Calcio/metabolismo , Colon/patología , Ciclooxigenasa 1/metabolismo , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Femenino , Fibroblastos , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Prostaglandina-E Sintasas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Canal Catiónico TRPA1/genética
5.
Immunity ; 49(1): 107-119.e4, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29958798

RESUMEN

Intestinal macrophages are critical for gastrointestinal (GI) homeostasis, but our understanding of their role in regulating intestinal motility is incomplete. Here, we report that CX3C chemokine receptor 1-expressing muscularis macrophages (MMs) were required to maintain normal GI motility. MMs expressed the transient receptor potential vanilloid 4 (TRPV4) channel, which senses thermal, mechanical, and chemical cues. Selective pharmacologic inhibition of TRPV4 or conditional deletion of TRPV4 from macrophages decreased intestinal motility and was sufficient to reverse the GI hypermotility that is associated with chemotherapy treatment. Mechanistically, stimulation of MMs via TRPV4 promoted the release of prostaglandin E2 and elicited colon contraction in a paracrine manner via prostaglandin E receptor signaling in intestinal smooth muscle cells without input from the enteric nervous system. Collectively, our data identify TRPV4-expressing MMs as an essential component required for maintaining normal GI motility and provide potential drug targets for GI motility disorders.


Asunto(s)
Colon/fisiología , Motilidad Gastrointestinal , Macrófagos/metabolismo , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Canales Catiónicos TRPV/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C/metabolismo , Colon/fisiopatología , Ciclooxigenasa 1/deficiencia , Ciclooxigenasa 1/metabolismo , Dinoprostona/análisis , Dinoprostona/metabolismo , Femenino , Mucosa Gástrica/citología , Expresión Génica , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Contracción Muscular , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética
6.
Pulm Pharmacol Ther ; 49: 75-87, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29408043

RESUMEN

Arachidonic acid is metabolized by cyclooxygenases (COX-1 and COX-2) into various prostanoids which exert different functions in mammalian physiology. One of these prostanoids, prostaglandin E2 (PGE2), interacts with four different G protein-coupled receptors, named EP1, EP2, EP3 and EP4, to initiate different downstream signalling pathways. Prostanoid receptors are diversely expressed throughout different tissues all over the body and PGE2 is responsible for a large variety of beneficial and disadvantageous effects. We have recently achieved a greater understanding of the biology of prostanoid E receptors and the potential for specific drug targeting with the advent of potent and selective EP receptor agonists and antagonists. This has important implications for lung health and disease as PGE2-mediated EP receptor activation impacts upon migration of airway smooth muscle cells, airway microvascular leak, tone regulation of pulmonary blood vessels, mast cell degranulation, bronchodilatation, cough, angiogenesis and airway inflammation, to name a few. In this review, we overview the EP receptor family and the related signalling pathways, summarize a variety of EP1-4 receptor agonists and antagonists, provide an overview of pharmacological tools used to implicate EP receptor function in the context of respiratory health and disease and finally highlight some of the more selective pharmacological reagents that have recently been developed. The availability of selective pharmacological agonists and antagonists for the distinct EP receptors, as well as the development of specific prostanoid receptor knock-out mice, offer hitherto unattainable opportunities for achieving an in depth understanding of the role and function of PGE2 in respiratory disease and the exciting potential of targeting EP receptors more broadly.


Asunto(s)
Desarrollo de Medicamentos/métodos , Enfermedades Pulmonares/tratamiento farmacológico , Receptores de Prostaglandina E/metabolismo , Animales , Ácido Araquidónico/metabolismo , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Enfermedades Pulmonares/fisiopatología , Ratones , Ratones Noqueados , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
7.
Curr Med Chem ; 25(22): 2566-2575, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28260499

RESUMEN

BACKGROUND: Pancreatic cancer is the fourth leading cause of cancer deaths with rising incidence and a high mortality rate. Smoking, psychological stress, diabetes, pancreatitis and alcohol abuse are known risk factors for pancreatic cancer. OBJECTIVE: Targeting G protein-coupled receptor signaling for the prevention and therapy of pancreatic cancer. METHOD: Review of published literature. RESULTS AND CONCLUSION: All known risk factors for pancreatic cancer cause hyperactive cyclic adenosine monophosphate (cAMP) signaling via cancer stimulating Gαs-coupled beta-adrenergic and prostaglandin E2 receptors and/or by suppressing signaling via inhibitory Gαi-coupled GABAB-receptors. Psychological stress in mice promotes the progression of pancreatic cancer xenografts via stress neurotransmitter-mediated increase in betaadrenergic signaling and suppression of GABA while stress reduction inhibits pancreatic cancer by reversing these effects. The activation of Gαi-coupled GABAB-receptor signaling by treatment with GABA, inhibition of beta-adrenergic signaling by a beta-blocker and/or suppression of Gαs-coupled PGE2 receptor signaling by a cyclooxygenase (COX) inhibitor prevent the development and progression of pancreatic cancer induced in hamsters by carcinogenic nitrosamines and in transgenic mice. The re-purposing of cardiovascular therapeutics (beta-blockers, COX-2 inhibitors, Ca2+-channel blockers) that inhibit betaadrenergic and PGE2 signaling for pancreatic cancer intervention is problematic due to undesirable side effects under chronic treatment protocols. To avoid such side effects while effectively reducing excessive cAMP signaling, nutritional GABA supplementation or positive allosteric modulators (PAMs) of Gαi-coupled receptors (GABAB-Rs) currently in clinical trials for the treatment of addiction should be explored for pancreatic cancer intervention.


Asunto(s)
Neoplasias Pancreáticas/patología , Receptores Acoplados a Proteínas G/metabolismo , Alcoholismo/complicaciones , Alcoholismo/patología , AMP Cíclico/metabolismo , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Complicaciones de la Diabetes , Progresión de la Enfermedad , Humanos , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Estrés Psicológico
8.
J Neuroinflammation ; 14(1): 262, 2017 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-29284502

RESUMEN

BACKGROUND: Astrocytic aromatization and consequent increases in estradiol are neuroprotective in the injured brain. In zebra finches, cyclooxygenase-activity is necessary for injury-induced aromatase expression, and increased central estradiol lowers neuroinflammation. The mechanisms underlying these influences are unknown. Here, we document injury-induced, cyclooxygenase-dependent increases in glial aromatase expression and replicate previous work in our lab showing increases in central prostaglandin E2 and estradiol following brain damage. Further, we describe injury-dependent changes in E-prostanoid and estrogen receptor expression and reveal the necessity of E-prostanoid and estrogen receptors in the injury-dependent, reciprocal interactions of neuroinflammatory and neurosteroidogenic pathways. METHODS: Adult male and female birds were shams or received bilateral injections of the appropriate drug or vehicle into contralateral telencephalic lobes. RESULTS: Injuries sustained in the presence of indomethacin (a cyclooxygenase inhibitor) had fewer aromatase-expressing reactive astrocytes relative to injuries injected with vehicle suggesting that cyclooxygenase activity is necessary for the induction of glial aromatase around the site of damage. Injured hemispheres had higher prostaglandin E2 and estradiol content relative to shams. Importantly, injured hemispheres injected with E-prostanoid- or estrogen receptor-antagonists showed elevated prostaglandin E2 and estradiol, respectively, but lower prostaglandin E2 or estradiol-dependent downstream activity (protein kinase A or phosphoinositide-3-kinase mRNA) suggesting that receptor antagonism did not affect injury-induced prostaglandin E2 or estradiol, but inhibited the effects of these ligands. Antagonism of E-prostanoid receptors 3 or 4 prevented injury-induced increases in neural estradiol in males and females, respectively, albeit this apparent sex-difference needs to be tested more stringently. Further, estrogen receptor-α, but not estrogen receptor-ß antagonism, exaggerated neural prostaglandin E2 levels relative to the contralateral lobe in both sexes. CONCLUSION: These data suggest injury-induced, sex-specific prostaglandin E2-dependent estradiol synthesis, and estrogen receptor-α dependent decreases in neuroinflammation in the vertebrate brain.


Asunto(s)
Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Dinoprostona/metabolismo , Estradiol/metabolismo , Transducción de Señal/fisiología , Animales , Aromatasa/metabolismo , Encéfalo/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Femenino , Pinzones , Masculino , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo
9.
Sci Rep ; 6: 27995, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27313009

RESUMEN

We explored the regulation of filaggrin, cyclooxygenase 2 (COX2) and prostaglandin E2 (PGE2) expression induced by urban particulate matter (PM) in human keratinocytes. In addition, we investigated the signaling pathways involved in PM-induced effects on COX2/PGE2 and filaggrin. PMs induced increases in COX2 expression and PGE2 production, and decreased filaggrin expression. These effects were attenuated by pretreatment with COX2 inhibitor and PGE2 receptor antagonist, or after transfection with siRNAs of the aryl hydrocarbon receptor (AhR), gp91phox and p47phox. Furthermore, PM-induced generation of reactive oxygen species (ROS) and NADPH oxidase activity was attenuated by pretreatment with an AhR antagonist (AhRI) or antioxidants. Moreover, Nox-dependent ROS generation led to phosphorylation of ERK1/2, p38, and JNK, which then activated the downstream molecules NF-κB and AP-1, respectively. In vivo studies in PMs-treated mice showed that AhRI and apocynin (a Nox2 inhibitor) had anti-inflammatory effects by decreasing COX2 and increasing filaggrin expression. Our results reveal for the first time that PMs-induced ROS generation is mediated through the AhR/p47 phox/NADPH oxidase pathway, which in turn activates ERK1/2, p38/NF-κB and JNK/AP-1, and which ultimately induces COX2 expression and filaggrin downregulation. Up-regulated expression of COX2 and production of PGE2 may lead to impairment of skin barrier function.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Proteínas de Filamentos Intermediarios/metabolismo , Material Particulado/toxicidad , Animales , Antioxidantes/farmacología , Línea Celular , Inhibidores de la Ciclooxigenasa 2/farmacología , Proteínas Filagrina , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , NADPH Oxidasa 2/antagonistas & inhibidores , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , Fosforilación/efectos de los fármacos , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Prostaglandina E/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos
10.
Biochim Biophys Acta ; 1863(2): 293-302, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26658718

RESUMEN

Nephrotoxicity, particularly in the proximal tubule, limits the therapeutic efficacy of the antineoplastic drug cisplatin. The signaling mechanisms appear to be multifactorial, involving inflammation, oxidative stress, and caspase. Here we studied the role of intracellular prostaglandin E2 (iPGE2) in cisplatin's cytotoxicity in human proximal tubular HK-2 cells. Cisplatin-induced apoptotic cell death was prevented by inhibitors of the prostaglandin transporter (PGT) or by PGT knock-down or by pharmacologic inhibition of PGE2 EP receptors or cyclo-oxygenase-2 (COX-2). iPGE2 also increased in cisplatin-treated cells, which was probably due to increased expression of COX-2, microsomal PGE2 synthase-1 and PGT, and was prevented by inhibitors of PGT or COX-2. Thus iPGE2, most likely acting through intracellular EP receptors, mediates cisplatin-induced HK-2 cell death. Importantly, the tumoricidal effect of cisplatin on human cervical adenocarcinoma HeLa cells was not affected by a pharmacologic inhibitor of PGT. In conclusion, iPGE2 may play a significant role in the pathogenesis of cisplatin's nephrotoxicity and treatment with PGT inhibitors might represent a novel strategy in its prevention.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/farmacología , Dinoprostona/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Antineoplásicos/farmacología , Western Blotting , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Expresión Génica , Humanos , Espacio Intracelular/metabolismo , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Microscopía Confocal , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Interferencia de ARN , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Clin Exp Metastasis ; 32(4): 335-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25749879

RESUMEN

Breast cancer can reoccur, often as bone metastasis, many years if not decades after the primary tumor has been treated. The factors that stimulate dormant metastases to grow are not known, but bone metastases are often associated with skeletal trauma. We used a dormancy model of MDA-MB-231BRMS1, a metastasis-suppressed human breast cancer cell line, co-cultured with MC3T3-E1 osteoblasts in a long term, three dimensional culture system to test the hypothesis that bone remodeling cytokines could stimulate dormant cells to grow. The cancer cells attached to the matrix produced by MC3T3-E1 osteoblasts but grew slowly or not at all until the addition of bone remodeling cytokines, TNFα and IL-ß. Stimulation of cell proliferation by these cytokines was suppressed with indomethacin, an inhibitor of cyclooxygenase and of prostaglandin production, or a prostaglandin E2 (PGE2) receptor antagonist. Addition of PGE2 directly to the cultures also stimulated cell proliferation. MCF-7, non-metastatic breast cancer cells, remained dormant when co-cultured with normal human osteoblast and fibroblast growth factor. Similar to the MDA-MB-231BRMS1 cells, MCF-7 proliferation increased in response to TNFα and IL-ß. These findings suggest that changes in the bone microenvironment due to inflammatory cytokines associated with bone repair or excess turnover may trigger the occurrence of latent bone metastasis.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/patología , Dinoprostona/farmacología , Interleucina-1beta/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Células 3T3 , Animales , Remodelación Ósea , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Indometacina/farmacología , Interleucina-6/farmacología , Células MCF-7 , Ratones , Osteoblastos , Receptores de Prostaglandina E/antagonistas & inhibidores , Microambiente Tumoral
12.
Mol Med Rep ; 11(6): 4454-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25625500

RESUMEN

Inflammation is a potent promoter of tumor metastasis. The aim of the present study was to explore the function of systemic inflammation in the formation of lung metastasis of breast cancer cells in a mouse model. BALB/c mice were injected intraperitoneally with lipopolysaccharide (LPS) in order to establish an inflammatory animal model and 4T1 murine breast cancer cells were injected through the tail vein to induce lung metastasis. The levels of proinflammatory cytokines were evaluated by ELISA. Metastases on the surface of the lungs were counted and histologically analyzed by hematoxylin and eosin staining. Angiogenesis in the lungs was examined by CD31 immunofluorescence. Mouse pulmonary endothelial cells (MPVECs) were isolated and used to assay endothelial tube formation and determine the protein expression levels of vascular endothelial growth factor (VEGF) in vitro. Serum levels of VEGF and prostaglandin E2 (PGE2), the number and size of metastatic lesions, and the expression levels of cyclooxygenase­2 were significantly greater in the lungs of LPS­treated mice, as compared with those in control mice threated with phosphate­buffered saline. Blood vessel density was also markedly increased in the LPS­treated mice. These increases were reversed by treatment with celecoxib. In vitro, the protein expression levels of VEGF produced by the PGE2­treated cells were significantly increased in a concentration­dependent manner. In addition, the production of VEGF was increased in response to treatment with the PGE2 receptor (EP2) agonist ONO­AE1­259­01; however, this increase was abrogated by treatment with AH6809, an EP2 receptor antagonist. Treatment with PGE2 or VEGF alone promoted the tube formation of MPVECs and this effect was reversed by treatment with celecoxib. These results demonstrated that PGE2 may regulate the release of VEGF by MPVECs through the EP2 receptor pathway and thereby promoted pulmonary angiogenesis and breast cancer metastasis in a mouse model.


Asunto(s)
Neoplasias de la Mama/patología , Dinoprostona/metabolismo , Lipopolisacáridos/toxicidad , Neoplasias Pulmonares/secundario , Pulmón/efectos de los fármacos , Receptores de Prostaglandina E/metabolismo , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/fisiología , Neoplasias de la Mama/metabolismo , Celecoxib/farmacología , Celecoxib/uso terapéutico , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Citocinas/análisis , Dinoprostona/sangre , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Inflamación/inducido químicamente , Pulmón/metabolismo , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Xantonas/farmacología
13.
Biochim Biophys Acta ; 1851(4): 414-21, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25038274

RESUMEN

Prostaglandin E2 (PGE2) is one of the most typical lipid mediators produced from arachidonic acid (AA) by cyclooxygenase (COX) as the rate-limiting enzyme, and acts on four kinds of receptor subtypes (EP1-EP4) to elicit its diverse actions including pyrexia, pain sensation, and inflammation. Recently, the molecular mechanisms underlying the PGE2 actions mediated by each EP subtype have been elucidated by studies using mice deficient in each EP subtype as well as several compounds highly selective to each EP subtype, and their findings now enable us to discuss how PGE2 initiates and exacerbates inflammation at the molecular level. Here, we review the recent advances in PGE2 receptor research by focusing on the activation of mast cells via the EP3 receptor and the control of helper T cells via the EP2/4 receptor, which are the molecular mechanisms involved in PGE2-induced inflammation that had been unknown for many years. We also discuss the roles of PGE2 in acute inflammation and inflammatory disorders, and the usefulness of anti-inflammatory therapies that target EP receptors. This article is part of a Special Issue entitled "Oxygenated metabolism of PUFA: analysis and biological relevance".


Asunto(s)
Dinoprostona/metabolismo , Inflamación/metabolismo , Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Animales , Antiinflamatorios/farmacología , Dinoprostona/química , Dinoprostona/inmunología , Diseño de Fármacos , Humanos , Inflamación/inmunología , Inflamación/prevención & control , Activación de Linfocitos , Mastocitos/inmunología , Mastocitos/metabolismo , Estructura Molecular , Terapia Molecular Dirigida , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/inmunología , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Relación Estructura-Actividad , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
14.
J Egypt Soc Parasitol ; 45(3): 511-20, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26939228

RESUMEN

Schistosomiasis is a chronic disease with considerable social impact. Despite the availability of affordable chemotherapy, drug treatment has not significantly reduced the overall number of disease cases. Among other mechanisms, the parasite produces PGE2 and PGD2 to evade host immune defenses. To investigate the role of PGE2 and PGD2 in schistosomiasis, we evaluated the effects of L-161,982, Ah6809 (PGE2 receptor antagonists alone of combined with each other) and MK-0524 (PGD2 receptor antagonist) during prepatent Schistosoma mansoni infection. Drugs were administered intraperitoneally an hour before and 24 hours after infection of C57BL/6 mice with 100 Schistosoma mansoni cercariae. L-161,982, Ah6809, their combination and MK-0524 caused partial protection against pre-patent S. mansoni infection which was mediated by biasing the immune response towards Th1 phenotype. These results showed that blockade of PGE2 and PGD2 receptors confers partial protection against pre-patent S. mansoni infection in mice and that they may be useful as adjunctive therapy to current anti-schistosomal drugs or vaccines.


Asunto(s)
Indoles/farmacología , Receptores Inmunológicos/antagonistas & inhibidores , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina/antagonistas & inhibidores , Esquistosomiasis mansoni/prevención & control , Tiofenos/farmacología , Triazoles/farmacología , Xantonas/farmacología , Animales , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Antagonistas de Prostaglandina/farmacología , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/fisiología
15.
J Pharmacol Exp Ther ; 351(1): 134-45, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25047516

RESUMEN

The excitotoxicity caused by excessive glutamate is a critical element in the neuropathology of acute and chronic brain disorders. Therefore, inhibition of glutamate release is a potentially valuable therapeutic strategy for treating these diseases. In this study, we investigated the effect of celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor that reduces the level of prostaglandin E2 (PGE2), on endogenous glutamate release in rat cerebral cortex nerve terminals (synaptosomes). Celecoxib substantially inhibited the release of glutamate induced by the K(+) channel blocker 4-aminopyridine (4-AP), and this phenomenon was prevented by chelating the extracellular Ca(2+) ions and by the vesicular transporter inhibitor bafilomycin A1. Celecoxib inhibited a 4-AP-induced increase in cytosolic-free Ca(2+) concentration, and the celecoxib-mediated inhibition of glutamate release was prevented by the Cav2.2 (N-type) and Cav2.1 (P/Q-type) channel blocker ω-conotoxin MVIIC. However, celecoxib did not alter 4-AP-mediated depolarization and Na(+) influx. In addition, this glutamate release-inhibiting effect of celecoxib was mediated through the PGE2 subtype 2 receptor (EP2) because it was not observed in the presence of butaprost (an EP2 agonist) or PF04418948 [1-(4-fluorobenzoyl)-3-[[6-methoxy-2-naphthalenyl)methyl]-3-azetidinecarboxylic acid; an EP2 antagonist]. The celecoxib effect on 4-AP-induced glutamate release was prevented by the inhibition or activation of protein kinase A (PKA), and celecoxib decreased the 4-AP-induced phosphorylation of PKA. We also determined that COX-2 and the EP2 receptor are present in presynaptic terminals because they are colocalized with synaptophysin, a presynaptic marker. These results collectively indicate that celecoxib inhibits glutamate release from nerve terminals by reducing voltage-dependent Ca(2+) entry through a signaling cascade involving EP2 and PKA.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Ácido Glutámico/metabolismo , Terminales Presinápticos/efectos de los fármacos , Prostaglandinas E/metabolismo , Pirazoles/farmacología , Receptores de Prostaglandina E/metabolismo , Sulfonamidas/farmacología , Animales , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/metabolismo , Celecoxib , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Exocitosis , Masculino , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Terminales Presinápticos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/antagonistas & inhibidores , Sodio/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo
16.
Hum Reprod ; 29(7): 1400-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24781425

RESUMEN

STUDY QUESTION: Can administration of a prostaglandin (PG) E2 receptor 2 (PTGER2) antagonist prevent pregnancy in adult female monkeys by blocking periovulatory events in the follicle without altering menstrual cyclicity or general health? SUMMARY ANSWER: This is the first study to demonstrate that a PTGER2 antagonist can serve as an effective non-hormonal contraceptive in primates. WHAT IS KNOWN ALREADY: The requirement for PGE2 in ovulation and the release of an oocyte surrounded by expanded cumulus cells (cumulus-oocyte expansion; C-OE) was established through the generation of PTGS2 and PTGER2 null-mutant mice. A critical role for PGE2 in primate ovulation is supported by evidence that intrafollicular injection of indomethacin in rhesus monkeys suppressed follicle rupture, whereas co-injection of PGE2 with indomethacin resulted in ovulation. STUDY DESIGN, SIZE, DURATION: First, controlled ovulation protocols were performed in adult, female rhesus monkeys to analyze the mRNA levels for genes encoding PGE2 synthesis and signaling components in the naturally selected pre-ovulatory follicle at different times after the ovulatory hCG stimulus (0, 12, 24, 36 h pre-ovulation; 36 h post-ovulation, n = 3-4/time point). Second, controlled ovarian stimulation cycles were utilized to obtain multiple cumulus-oocyte complexes (COCs) from rhesus monkeys to evaluate the role of PGE2 in C-OE in vitro (n = 3-4 animals/treatment; ≥3 COCs/animal/treatment). Third, adult cycling female cynomolgus macaques were randomly assigned (n = 10/group) to vehicle (control) or PTGER2 antagonist (BAY06) groups to perform a contraceptive trial. After the first treatment cycle, a male of proven fertility was introduced into each group and they remained housed together for the duration of the 5-month contraceptive trial that was followed by a post-treatment reversibility trial. PARTICIPANTS/MATERIALS, SETTING, METHODS: Quantitative real-time PCR, COC culture and expansion, immunofluorescence/confocal microscopy, enzyme immunoassay, contraceptive trial, ultrasonography, complete blood counts, serum biochemistry tests and blood lipid profiles. MAIN RESULTS AND THE ROLE OF CHANCE: Several mRNAs encoding proteins involved in PGE2 synthesis, metabolism and signaling increase (P < 0.05) in the periovulatory follicle after administration of an ovulatory hCG bolus. PGE2 signaling through PTGER2 induces cumulus cell expansion and production of hyaluronic acid, which are critical events for fertilization. Moreover, chronic administration of a selective PTGER2 antagonist resulted in a significant (P < 0.05 versus vehicle-treated controls) contraceptive effect without altering steroid hormone patterns or menstrual cyclicity during a 5-months contraceptive trial. Fertility recovered as early as 1 month after ending treatment. LIMITATIONS, REASONS FOR CAUTION: This is a proof-of-concept study in a non-human primate model. Further investigations are warranted to elucidate the mechanism(s) of PTGER2 antagonist action in the primate ovary. Although PTGER2 antagonist treatment did not produce any obvious undesirable effects, improvements in the mode of administration, as well as the efficacy of these compounds, are necessary to consider such a contraceptive for women. WIDER IMPLICATIONS OF THE FINDINGS: Monitoring as well as improving the efficacy and safety of female contraceptives is an important public health activity. Even though hormonal contraceptives are effective for women, concerns remain regarding their side-effects and long-term use because of the widespread actions of such steroidal products in many tissues. Moreover, some women cannot take hormones for medical reasons. Thus, development of non-hormonal contraceptives for women is warranted. STUDY FUNDING/COMPETING INTEREST(S): Supported by Bayer HealthCare Pharmaceuticals, The Eunice Kennedy Shriver NICHD Contraceptive Development and Research Center (U54 HD055744), NIH Office of the Director (Oregon National Primate Research Center P51 OD011092), and a Lalor Foundation Postdoctoral Basic Research Fellowship (MCP). The use of the Leica confocal was supported by grant number S10RR024585. Some of the authors (N.B., A.R., K.-H.F., U.F., B.B. and B.L.) are employees of Bayer Healthcare Pharma.


Asunto(s)
Anticoncepción/métodos , Anticonceptivos/uso terapéutico , Receptores de Prostaglandina E/antagonistas & inhibidores , Animales , Femenino , Regulación de la Expresión Génica , Indometacina/uso terapéutico , Macaca , Macaca fascicularis , Oocitos/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Ovario/efectos de los fármacos , Ovulación/efectos de los fármacos , Embarazo , Preñez , ARN Mensajero/metabolismo , Factores de Tiempo
17.
PLoS One ; 8(8): e73839, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24013220

RESUMEN

AIMS: Arachidonic acid (AA) and its metabolites, prostaglandins (PG) are known to be involved in regulation of vascular homeostasis including vascular tone and vessel wall tension, but their potential role in Hypoxic pulmonary vasoconstriction (HPV) remains unclear. In this study, we examined the effects of AA and PGE2 on the hypoxic response in isolated rat intrapulmonary arteries (IPAs). METHODS AND RESULTS: We carried out the investigation on IPAs by vessel tension measurement. Isotetrandrine (20 µM) significantly inhibited phase I, phase IIb and phase IIc of hypoxic vasoconstriction. Both indomethacin (100 µM) and NS398 attenuated KPSS-induced vessel contraction and phase I, phase IIb and phase IIc of HPV, implying that COX-2 plays a primary role in the hypoxic response of rat IPAs. PGE2 alone caused a significant vasoconstriction in isolated rat IPAs. This constriction is mediated by EP4. Blockage of EP4 by L-161982 (1 µM) significantly inhibited phase I, phase IIb and phase IIc of hypoxic vasoconstriction. However, AH6809 (3 µM), an antagonist of EP1, EP2, EP3 and DP1 receptors, exerted no effect on KPSS or hypoxia induced vessel contraction. Increase of cellular cAMP by forskolin could significantly reduce KPSS-induced vessel contraction and abolish phase I, phase II b and phase II c of HPV. CONCLUSION: Our results demonstrated a vasoconstrictive effect of PGE2 on rat IPAs and this effect is via activation of EP4. Furthermore, our results suggest that intracellular cAMP plays dual roles in regulation of vascular tone, depending on the spatial distribution of cAMP and its coupling with EP receptor and Ca(2+) channels.


Asunto(s)
Ácido Araquidónico/farmacología , Dinoprostona/farmacología , Hipoxia , Oxitócicos/farmacología , Arteria Pulmonar , Vasoconstricción/efectos de los fármacos , Animales , Calcio/metabolismo , AMP Cíclico/metabolismo , Ciclooxigenasa 2/metabolismo , Hipoxia/metabolismo , Hipoxia/fisiopatología , Masculino , Tono Muscular/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Ratas , Ratas Wistar , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Tiofenos/farmacología , Triazoles/farmacología , Xantonas/farmacología
18.
Am J Respir Cell Mol Biol ; 49(1): 105-13, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23526216

RESUMEN

Numerous epidemiological studies have linked exposure to particulate matter (PM) air pollution with acute respiratory infection and chronic respiratory and cardiovascular diseases. We have previously shown that soluble nickel (Ni), a common component of PM, alters the release of CXC chemokines from cultured human lung fibroblasts (HLF) in response to microbial stimuli via a pathway dependent on disrupted prostaglandin (PG)E2 signaling. The current study sought to identify the molecular events underlying Ni-induced alterations in PGE2 signaling and its effects on IL-8 production. PGE2 synergistically enhances Ni-induced IL-8 release from HLF in a concentration-dependent manner. The effects of PGE2 were mimicked by butaprost and PGE1-alcohol and inhibited with antagonists AH6809 and L-161,982, indicating PGE2 signals via PGE2 receptors 2 and 4. PGE2 and forskolin stimulated cAMP, but it was only in the presence of Ni-induced hypoxia-inducible factor 1, α subunit (HIF1A) that these agents stimulated IL-8 release. The Ni-induced HIF1A DNA binding was enhanced by PGE2 and mediated, in part, by activation of p38 MAPK. Negation of cAMP-response element binding protein 1 or HIF1A using short interfering RNA blocked the synergistic interactions between Ni and PGE2. The results of the current study provide novel information on the ability of atmospheric hypoxia-mimetic metals to disrupt the release of immune-modulating chemokines by HLF in response to PGE2. Moreover, in the presence of HIF1A, cAMP-mediated signaling pathways may be altered to exacerbate inflammatory-like processes in lung tissue, imparting a susceptibility of PM-exposed populations to adverse respiratory health effects.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dinoprostona/farmacología , Fibroblastos/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-8/metabolismo , Níquel/farmacología , Alprostadil/análogos & derivados , Alprostadil/farmacología , Biomimética , Células Cultivadas , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Sinergismo Farmacológico , Fibroblastos/metabolismo , Humanos , Inflamación/patología , Pulmón/citología , Pulmón/metabolismo , Níquel/metabolismo , Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/agonistas , Subtipo EP4 de Receptores de Prostaglandina E/agonistas , Transducción de Señal , Xantonas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Proc Natl Acad Sci U S A ; 110(9): 3603-8, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23401543

RESUMEN

It is well established that dorsal root ganglion (DRG) cells synthesize prostaglandin. However, the role that prostaglandin plays in the inflammatory hyperalgesia of peripheral tissue has not been established. Recently, we have successfully established a technique to inject drugs (3 µL) directly into the L5-DRG of rats, allowing in vivo identification of the role that DRG cell-derived COX-1 and COX-2 play in the development of inflammatory hyperalgesia of peripheral tissue. IL-1ß (0.5 pg) or carrageenan (100 ng) was administered in the L5-peripheral field of rat hindpaw and mechanical hyperalgesia was evaluated after 3 h. Administration of a nonselective COX inhibitor (indomethacin), selective COX-1 (valeryl salicylate), or selective COX-2 (SC-236) inhibitors into the L5-DRG prevented the hyperalgesia induced by IL-1ß. Similarly, oligodeoxynucleotide-antisense against COX-1 or COX-2, but not oligodeoxynucleotide-mismatch, decreased their respective expressions in the L5-DRG and prevented the hyperalgesia induced by IL-1ß in the hindpaw. Immunofluorescence analysis demonstrated that the amount of COX-1 and COX-2, constitutively expressed in TRPV-1(+) cells of the DRG, significantly increased after carrageenan or IL-1ß administration. In addition, indomethacin administered into the L5-DRG prevented the increase of PKCε expression in DRG membrane cells induced by carrageenan. Finally, the administration of EP1/EP2 (7.5 ng) or EP4 (10 µg) receptor antagonists into L5-DRG prevented the hyperalgesia induced by IL-1ß in the hindpaw. In conclusion, the results of this study suggest that the inflammatory hyperalgesia in peripheral tissue depends on activation of COX-1 and COX-2 in C-fibers, which contribute to the induction and maintenance of sensitization of primary sensory neurons.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Ganglios Espinales/enzimología , Hiperalgesia/enzimología , Hiperalgesia/patología , Inflamación/enzimología , Inflamación/patología , Proteínas de la Membrana/metabolismo , Animales , Carragenina/farmacología , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/farmacología , Activación Enzimática/efectos de los fármacos , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/patología , Técnicas de Silenciamiento del Gen , Hiperalgesia/complicaciones , Indometacina/administración & dosificación , Indometacina/farmacología , Inflamación/complicaciones , Interleucina-1beta/farmacología , Vértebras Lumbares/efectos de los fármacos , Vértebras Lumbares/patología , Masculino , Proteína Quinasa C-epsilon/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Prostaglandina E/antagonistas & inhibidores , Receptores de Prostaglandina E/metabolismo , Canales Catiónicos TRPV/metabolismo
20.
PLoS One ; 8(1): e54376, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23349866

RESUMEN

Although the modulation of Ca(2+) channel activity by extremely low-frequency electromagnetic fields (ELF-EMF) has been studied previously, few reports have addressed the effects of such fields on the activity of voltage-activated Na(+) channels (Na(v)). Here, we investigated the effects of ELF-EMF on Na(v) activity in rat cerebellar granule cells (GCs). Our results reveal that exposing cerebellar GCs to ELF-EMF for 10-60 min significantly increased Na(v) currents (I(Na)) by 30-125% in a time- and intensity-dependent manner. The Na(v) channel steady-state activation curve, but not the steady-state inactivation curve, was significantly shifted (by 5.2 mV) towards hyperpolarization by ELF-EMF stimulation. This phenomenon is similar to the effect of intracellular application of arachidonic acid (AA) and prostaglandin E(2) (PGE(2)) on I(Na) in cerebellar GCs. Increases in intracellular AA, PGE(2) and phosphorylated PKA levels in cerebellar GCs were observed following ELF-EMF exposure. Western blottings indicated that the Na(V) 1.2 protein on the cerebellar GCs membrane was increased, the total expression levels of Na(V) 1.2 protein were not affected after exposure to ELF-EMF. Cyclooxygenase inhibitors and PGE(2) receptor (EP) antagonists were able to eliminate this ELF-EMF-induced increase in phosphorylated PKA and I(Na). In addition, ELF-EMF exposure significantly enhanced the activity of PLA(2) in cerebellar GCs but did not affect COX-1 or COX-2 activity. Together, these data demonstrate for the first time that neuronal I(Na) is significantly increased by ELF-EMF exposure via a cPLA2 AA PGE(2) EP receptors PKA signaling pathway.


Asunto(s)
Dinoprostona/metabolismo , Campos Electromagnéticos , Receptores de AMP Cíclico/metabolismo , Receptores de Prostaglandina E/antagonistas & inhibidores , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Ácido Araquidónico/metabolismo , Encéfalo/metabolismo , Cerebelo/citología , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/efectos de la radiación , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Proteínas de la Membrana/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Ratas , Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Canales de Sodio Activados por Voltaje/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...