Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.335
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Cell Physiol ; 236(6): 4764-4777, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33275302

RESUMO

Primary cilia have been found to function as mechanosensors in low-magnitude high-frequency vibration (LMHFV)-induced osteogenesis. The PGE2 also regulates bone homeostasis and mechanical osteogenesis through its receptor EP4 signaling, but its involvement in LMHFV-induced or in primary cilia-induced osteogenesis has not been investigated. We hypothesized that LMHFV stimulates osteoblast (OB) differentiation by activating the COX2-PGE2-EP pathway in a manner dependent on primary cilia and that primary cilia are also affected by the PGE2 pathway. In this study, through western blot analysis, RNA interference, enzyme-linked immunosorbent assay, real-time quantitative polymerase chain reaction, and cytochemical staining, we observed that COX2, mPGES-1, and PGE2 levels were markedly elevated in cells treated with LMHFV and were greatly decreased in LMHFV-treated cells following IFT88 silencing. EP4 expression was significantly increased in OBs following LMHFV treatment, but IFT88 silencing significantly blocked this increase. EP4 localized to the bases of primary cilia. LMHFV reduced the length and abundance of primary cilia, but the cells could self-repair their primary cilia after mechanical damage. EP4 antagonism significantly blocked the LMHFV-induced increase in IFT88 expression and blocked the recovery of primary cilia length and the proportion of cells with primary cilia. In addition, COX2 or EP4 antagonism disrupted LMHFV-induced osteogenesis. These results demonstrate the integration of and crosstalk between primary cilia and the COX2-PGE2-EP4 signaling pathway under mechanical stimulation.


Assuntos
Diferenciação Celular , Cílios/enzimologia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Mecanotransdução Celular , Osteoblastos/enzimologia , Osteogênese , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células 3T3 , Animais , Diferenciação Celular/efeitos dos fármacos , Cílios/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/farmacologia , Camundongos , Osteoblastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Estimulação Física , Antagonistas de Prostaglandina/farmacologia , Prostaglandina-E Sintases/genética , Prostaglandina-E Sintases/metabolismo , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Vibração
2.
Am J Physiol Renal Physiol ; 320(4): F537-F547, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33491563

RESUMO

Local or systemic inflammation can severely impair urinary bladder functions and contribute to the development of voiding disorders in millions of people worldwide. Isoprostanes are inflammatory lipid mediators that are upregulated in the blood and urine by oxidative stress and may potentially induce detrusor overactivity. The aim of the present study was to investigate the effects and signal transduction of isoprostanes in human and murine urinary bladders in order to provide potential pharmacological targets in detrusor overactivity. Contraction force was measured with a myograph in murine and human urinary bladder smooth muscle (UBSM) ex vivo. Isoprostane 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the murine UBSM, which was abolished in mice deficient in the thromboxane prostanoid (TP) receptor. The responses remained unaltered after removal of the mucosa or incubation with tetrodotoxin. Smooth muscle-specific deletion of Gα12/13 protein or inhibition of Rho kinase by Y-27632 decreased the contractions. In Gαq/11-knockout mice, responses were reduced and in the presence of Y-27632 abolished completely. In human UBSM, the TP agonist U-46619 evoked dose-dependent contractions. Neither atropine nor the purinergic receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid decreased the effect, indicating that TP receptors directly mediate detrusor muscle contraction. 8-iso-PGE2 and 8-iso-PGF2α evoked dose-dependent contraction in the human UBSM, and these responses were abolished by the TP antagonist SQ-29548 and were decreased by Y-27632. Our results indicate that isoprostanes evoke contraction in murine and human urinary bladders, an effect mediated by the TP receptor. The G12/13-Rho-Rho kinase pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target in detrusor overactivity.NEW & NOTEWORTHY Voiding disorders affect millions of people worldwide. Inflammation can impair urinary bladder functions and contribute to the development of detrusor overactivity. The effects and signal transduction of inflammatory lipid mediator isoprostanes were studied in human and murine urinary bladders ex vivo. We found that isoprostanes evoke contraction, an effect mediated by thromboxane prostanoid receptors. The G12/13-Rho-Rho kinase signaling pathway plays a significant role in mediating the contraction and therefore may be a potential therapeutic target.


Assuntos
Isoprostanos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina/efeitos dos fármacos , Receptores de Tromboxanos/efeitos dos fármacos , Animais , Humanos , Prostaglandinas/farmacologia , Receptores de Tromboxanos/fisiologia
3.
Respir Res ; 22(1): 262, 2021 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-34620168

RESUMO

BACKGROUND: Prostaglandin D2 (PGD2) signaling via prostaglandin D2 receptor 2 (DP2) contributes to atopic and non-atopic asthma. Inhibiting DP2 has shown therapeutic benefit in certain subsets of asthma patients, improving eosinophilic airway inflammation. PGD2 metabolites prolong the inflammatory response in asthmatic patients via DP2 signaling. The role of PGD2 metabolites on eosinophil and ILC2 activity is not fully understood. METHODS: Eosinophils and ILC2s were isolated from peripheral blood of atopic asthmatic patients. Eosinophil shape change, ILC2 migration and IL-5/IL-13 cytokine secretion were measured after stimulation with seven PGD2 metabolites in presence or absence of the selective DP2 antagonist fevipiprant. RESULTS: Selected metabolites induced eosinophil shape change with similar nanomolar potencies except for 9α,11ß-PGF2. Maximal values in forward scatter of eosinophils were comparable between metabolites. ILC2s migrated dose-dependently in the presence of selected metabolites except for 9α,11ß-PGF2 with EC50 values ranging from 17.4 to 91.7 nM. Compared to PGD2, the absolute cell migration was enhanced in the presence of Δ12-PGD2, 15-deoxy-Δ12,14-PGD2, PGJ2, Δ12-PGJ2 and 15-deoxy-Δ12,14-PGJ2. ILC2 cytokine production was dose dependent as well but with an average sixfold reduced potency compared to cell migration (IL-5 range 108.1 to 526.9 nM, IL-13 range: 125.2 to 788.3 nM). Compared to PGD2, the absolute cytokine secretion was reduced in the presence of most metabolites. Fevipiprant dose-dependently inhibited eosinophil shape change, ILC2 migration and ILC2 cytokine secretion with (sub)-nanomolar potencies. CONCLUSION: Prostaglandin D2 metabolites initiate ILC2 migration and IL-5 and IL-13 cytokine secretion in a DP2 dependent manner. Our data indicate that metabolites may be important for in vivo eosinophil activation and ILC2 migration and to a lesser extent for ILC2 cytokine secretion.


Assuntos
Asma/tratamento farmacológico , Eosinófilos/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Prostaglandina D2/farmacologia , Receptores Imunológicos/agonistas , Receptores de Prostaglandina/agonistas , Adolescente , Adulto , Idoso , Asma/imunologia , Asma/metabolismo , Movimento Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Eosinófilos/imunologia , Eosinófilos/metabolismo , Feminino , Humanos , Ácidos Indolacéticos/farmacologia , Interleucina-13/metabolismo , Interleucina-5/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Antagonistas de Prostaglandina/farmacologia , Prostaglandina D2/análogos & derivados , Piridinas/farmacologia , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Transdução de Sinais , Adulto Jovem
4.
FASEB J ; 33(5): 6140-6153, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30735438

RESUMO

Nonsteroidal anti-inflammatory drugs interfere with the metabolism of arachidonic acid to proinflammatory prostaglandins and leukotrienes by targeting cyclooxygenases (COXs), 5-lipoxygenase (LOX), or the 5-LOX-activating protein (FLAP). These and related enzymes act in conjunction with marked crosstalk within a complex lipid mediator (LM) network where also specialized proresolving LMs (SPMs) are formed. Here, we present how prominent LM pathways can be differentially modulated in human proinflammatory M1 and proresolving M2 macrophage phenotypes that, upon exposure to Escherichia coli, produce either abundant prostaglandins and leukotrienes (M1) or SPMs (M2). Targeted liquid chromatography-tandem mass spectrometry-based metabololipidomics was applied to analyze and quantify the specific LM profiles. Besides expected on-target actions, we found that: 1) COX or 15-LOX-1 inhibitors elevate inflammatory leukotriene levels, 2) FLAP and 5-LOX inhibitors reduce leukotrienes in M1 but less so in M2 macrophages, 3) zileuton blocks resolution-initiating SPM biosynthesis, whereas FLAP inhibition increases SPM levels, and 4) that the 15-LOX-1 inhibitor 3887 suppresses SPM formation in M2 macrophages. Conclusively, interference with discrete LM biosynthetic enzymes in different macrophage phenotypes considerably affects the LM metabolomes with potential consequences for inflammation-resolution pharmacotherapy. Our data may allow better appraisal of the therapeutic potential of these drugs to intervene with inflammatory disorders.-Werner, M., Jordan, P. M., Romp, E., Czapka, A., Rao, Z., Kretzer, C., Koeberle, A., Garscha, U., Pace, S., Claesson, H.-E., Serhan, C. N., Werz, O., Gerstmeier, J. Targeting biosynthetic networks of the proinflammatory and proresolving lipid metabolome.


Assuntos
Leucotrienos/metabolismo , Macrófagos/metabolismo , Metaboloma , Prostaglandinas/metabolismo , Adulto , Anti-Inflamatórios não Esteroides/farmacologia , Células Cultivadas , Inibidores de Ciclo-Oxigenase/farmacologia , Humanos , Antagonistas de Leucotrienos/farmacologia , Lipoxigenase/metabolismo , Inibidores de Lipoxigenase/farmacologia , Macrófagos/efeitos dos fármacos , Antagonistas de Prostaglandina/farmacologia , Prostaglandina-Endoperóxido Sintases/metabolismo
5.
Adv Exp Med Biol ; 1274: 29-54, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32894506

RESUMO

Prostanoids (prostaglandins, prostacyclin and thromboxane) belong to the oxylipin family of biologically active lipids generated from arachidonic acid (AA). Protanoids control numerous physiological and pathological processes. Cyclooxygenase (COX) is a rate-limiting enzyme involved in the conversion of AA into prostanoids. There are two COX isozymes: the constitutive COX-1 and the inducible COX-2. COX-1 and COX-2 have similar structures, catalytic activities, and subcellular localizations but differ in patterns of expression and biological functions. Non-selective COX-1/2 or traditional, non-steroidal anti-inflammatory drugs (tNSAIDs) target both COX isoforms and are widely used to relieve pain, fever and inflammation. However, the use of NSAIDs is associated with various side effects, particularly in the gastrointestinal tract. NSAIDs selective for COX-2 inhibition (coxibs) were purposefully designed to spare gastrointestinal toxicity, but predisposed patients to increased cardiovascular risks. These health complications from NSAIDs prompted interest in the downstream effectors of the COX enzymes as novel drug targets. This chapter describes various safety issues with tNSAIDs and coxibs, and discusses the current development of novel classes of drugs targeting the prostanoid pathway, including nitrogen oxide- and hydrogen sulfide-releasing NSAIDs, inhibitors of prostanoid synthases, dual inhibitors, and prostanoid receptor agonists and antagonists.


Assuntos
Antagonistas de Prostaglandina/farmacologia , Antagonistas de Prostaglandina/uso terapêutico , Prostaglandinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Inibidores de Ciclo-Oxigenase 2/farmacologia , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Humanos , Prostaglandina-Endoperóxido Sintases/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(5): 1117-1122, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096371

RESUMO

In recent years, it has been established that programmed cell death protein ligand 1 (PD-L1)-mediated inhibition of activated PD-1+ T lymphocytes plays a major role in tumor escape from immune system during cancer progression. Lately, the anti-PD-L1 and -PD-1 immune therapies have become an important tool for treatment of advanced human cancers, including bladder cancer. However, the underlying mechanisms of PD-L1 expression in cancer are not fully understood. We found that coculture of murine bone marrow cells with bladder tumor cells promoted strong expression of PD-L1 in bone marrow-derived myeloid cells. Tumor-induced expression of PD-L1 was limited to F4/80+ macrophages and Ly-6C+ myeloid-derived suppressor cells. These PD-L1-expressing cells were immunosuppressive and were capable of eliminating CD8 T cells in vitro. Tumor-infiltrating PD-L1+ cells isolated from tumor-bearing mice also exerted morphology of tumor-associated macrophages and expressed high levels of prostaglandin E2 (PGE2)-forming enzymes microsomal PGE2 synthase 1 (mPGES1) and COX2. Inhibition of PGE2 formation, using pharmacologic mPGES1 and COX2 inhibitors or genetic overexpression of PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH), resulted in reduced PD-L1 expression. Together, our study demonstrates that the COX2/mPGES1/PGE2 pathway involved in the regulation of PD-L1 expression in tumor-infiltrating myeloid cells and, therefore, reprogramming of PGE2 metabolism in tumor microenvironment provides an opportunity to reduce immune suppression in tumor host.


Assuntos
Antígeno B7-H1/biossíntese , Células da Medula Óssea/metabolismo , Ciclo-Oxigenase 2/fisiologia , Dinoprostona/fisiologia , Macrófagos/metabolismo , Células Supressoras Mieloides/metabolismo , Prostaglandina-E Sintases/fisiologia , Animais , Antígeno B7-H1/genética , Comunicação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Hidroxiprostaglandina Desidrogenases/biossíntese , Hidroxiprostaglandina Desidrogenases/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos NOD , Camundongos SCID , Antagonistas de Prostaglandina/farmacologia , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/patologia
7.
Inflamm Res ; 67(4): 301-314, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29260240

RESUMO

OBJECTIVE: We investigated whether: (1) P2 × 7 receptor activation by its agonist (BzATP) induces articular hyperalgesia in the rat's knee joint via inflammatory mechanisms and (2) activation of P2 × 7 receptors by endogenous ATP contributes to the articular hyperalgesia induced by bradykinin, TNF-α, IL-1ß, CINC-1, PGE2, and dopamine. METHODS: The articular hyperalgesia was quantified using the rat knee joint incapacitation test. The knee joint inflammation, characterized by the concentration of pro-inflammatory cytokines and by neutrophil migration, was quantified in the synovial lavage fluid by ELISA and myeloperoxidase enzyme activity assay, respectively. RESULTS: BzATP induced a dose-dependent articular hyperalgesia in the rat's knee joint that was significantly reduced by the selective antagonists for P2 × 7, bradykinin B1 or B2 receptors, ß1 or ß2 adrenoceptors, and by pre-treatment with Indomethacin. BzATP induced a local increase of TNF-α, IL-1ß, IL-6, and CINC-1 concentration and neutrophil migration into the knee joint. The co-administration of the selective P2 × 7 receptor antagonist A-740003 significantly reduced the articular hyperalgesia induced by bradykinin and dopamine, but not by TNF-α, IL-1ß, CINC-1, and PGE2. CONCLUSIONS: P2 × 7 receptor activation induces articular hyperalgesia mediated by the previous inflammatory mediator release. P2 × 7 receptor-induced articular hyperalgesia is sustained by the involvement of this purinergic receptor in bradykinin and dopamine-induced hyperalgesia in the knee joint.


Assuntos
Hiperalgesia/etiologia , Articulação do Joelho/metabolismo , Receptores da Bradicinina/metabolismo , Receptores Dopaminérgicos/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Trifosfato de Adenosina/análogos & derivados , Antagonistas Adrenérgicos beta/farmacologia , Animais , Bradicinina , Antagonistas dos Receptores da Bradicinina/farmacologia , Citocinas/metabolismo , Dopamina , Hiperalgesia/induzido quimicamente , Hiperalgesia/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , Antagonistas de Prostaglandina/farmacologia , Antagonistas do Receptor Purinérgico P2/farmacologia , Agonistas do Receptor Purinérgico P2X/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Ratos Wistar
8.
Am J Physiol Renal Physiol ; 313(4): F1038-F1049, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28701311

RESUMO

During the early phase of ANG II-dependent hypertension, tubular PGE2 is increased. Renin synthesis and secretion in the collecting duct (CD) are upregulated by ANG II, contributing to further intratubular ANG II formation. However, what happens first and whether the triggering mechanism is independent of tubular ANG II remain unknown. PGE2 stimulates renin synthesis in juxtaglomerular cells via E-prostanoid (EP) receptors through the cAMP/cAMP-responsive element-binding (CREB) pathway. EP receptors are also expressed in the CD. Here, we tested the hypothesis that renin is upregulated by PGE2 in CD cells. The M-1 CD cell line expressed EP1, EP3, and EP4 but not EP2. Dose-response experiments, in the presence of ANG II type 1 receptor blockade with candesartan, demonstrated that 10-6 M PGE2 maximally increases renin mRNA (approximately 4-fold) and prorenin/renin protein levels (approximately 2-fold). This response was prevented by micromolar doses of SC-19220 (EP1 antagonist), attenuated by the EP4 antagonist, L-161982, and exacerbated by the highly selective EP3 antagonist, L-798106 (~10-fold increase). To evaluate further the signaling pathway involved, we used the PKC inhibitor calphostin C and transfections with PKCα dominant negative. Both strategies blunted the PGE2-induced increases in cAMP levels, CREB phosphorylation, and augmentation of renin. Knockdown of the EP1 receptor and CREB also prevented renin upregulation. These results indicate that PGE2 increases CD renin expression through the EP1 receptor via the PKC/cAMP/CREB pathway. Therefore, we conclude that during the early stages of ANG II-dependent hypertension, there is augmentation of PGE2 that stimulates renin in the CD, resulting in increased tubular ANG II formation and further stimulation of renin.


Assuntos
Proteína de Ligação a CREB/metabolismo , AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Proteína Quinase C/metabolismo , Receptores de Prostaglandina E Subtipo EP1/agonistas , Sistema Renina-Angiotensina/efeitos dos fármacos , Renina/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Proteína de Ligação a CREB/genética , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Túbulos Renais Coletores/enzimologia , Camundongos , Simulação de Acoplamento Molecular , Fosforilação , Antagonistas de Prostaglandina/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Receptores de Prostaglandina E Subtipo EP1/genética , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Renina/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Regulação para Cima
9.
Bioorg Med Chem Lett ; 27(5): 1186-1192, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28169162

RESUMO

Starting from acylsufonamide HTS hit 2, a novel series of para-N-acylaminomethylbenzoic acids was identified and developed as selective prostaglandin EP4 receptor antagonists. Structural modifications on lead compound 4a were explored with the aim of improving potency, physicochemical properties, and animal PK predictive of QD (once a day) dosing regimen in human. These efforts led to the discovery of the clinical candidate AAT-008 (4j), which exhibited significantly improved pharmacological profiles over grapiprant (1).


Assuntos
Benzoatos/farmacologia , Niacinamida/análogos & derivados , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Animais , Benzoatos/química , Benzoatos/farmacocinética , Descoberta de Drogas , Humanos , Niacinamida/química , Niacinamida/farmacocinética , Niacinamida/farmacologia , Antagonistas de Prostaglandina/química , Antagonistas de Prostaglandina/farmacocinética , Relação Estrutura-Atividade
10.
Eur J Appl Physiol ; 117(6): 1175-1180, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28432421

RESUMO

PURPOSE: Animal studies suggest that the inhibition of nitric oxide synthase (NOS) affects blood flow differently in different skeletal muscles according to their muscle fibre type composition (oxidative vs glycolytic). Quadriceps femoris (QF) muscle consists of four different muscle parts: vastus intermedius (VI), rectus femoris (RF), vastus medialis (VM), and vastus lateralis (VL) of which VI is located deep within the muscle group and is generally regarded to consist mostly of oxidative muscle fibres. METHODS: We studied the effect of NOS inhibition on blood flow in these four different muscles by positron emission tomography in eight young healthy men at rest and during one-leg dynamic exercise, with and without combined blockade with prostaglandins. RESULTS: At rest blood flow in the VI (2.6 ± 1.1 ml/100 g/min) was significantly higher than in VL (1.9 ± 0.6 ml/100 g/min, p = 0.015) and RF (1.7 ± 0.6 ml/100 g/min, p = 0.0015), but comparable to VM (2.4 ± 1.1 ml/100 g/min). NOS inhibition alone or with prostaglandins reduced blood flow by almost 50% (p < 0.001), but decrements were similar in all four muscles (drug × muscle interaction, p = 0.43). During exercise blood flow was also the highest in VI (45.4 ± 5.5 ml/100 g/min) and higher compared to VL (35.0 ± 5.5 ml/100 g/min), RF (38.4 ± 7.4 ml/100 g/min), and VM (36.2 ± 6.8 ml/100 g/min). NOS inhibition alone did not reduce exercise hyperemia (p = 0.51), but combined NOS and prostaglandin inhibition reduced blood flow during exercise (p = 0.002), similarly in all muscles (drug × muscle interaction, p = 0.99). CONCLUSION: NOS inhibition, with or without prostaglandins inhibition, affects blood flow similarly in different human QF muscles both at rest and during low-to-moderate intensity exercise.


Assuntos
Exercício Físico , Músculo Esquelético/irrigação sanguínea , Óxido Nítrico Sintase/antagonistas & inibidores , Antagonistas de Prostaglandina/farmacologia , Fluxo Sanguíneo Regional/efeitos dos fármacos , Adulto , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Músculo Esquelético/fisiologia , ômega-N-Metilarginina/farmacologia
11.
Brain Behav Immun ; 56: 209-220, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26944000

RESUMO

We recently reported that immune stimulation can be compromised if animals are simultaneously subjected to stressful conditions. To test the generalizability of these findings, and to elucidate neuroendocrine mediating mechanisms, we herein employed CpG-C, a novel TLR-9 immune-stimulating agent. Animals were subjected to ongoing stress (20-h of wet cage exposure) during CpG-C treatment, and antagonists to glucocorticoids, ß-adrenoceptor, COX2, or opioids were employed (RU486, nadolol, etodolac, naltrexone). In F344 rats, marginating-pulmonary NK cell numbers and cytotoxicity were studied, and the NK-sensitive MADB106 experimental metastasis model was used. In Balb/C mice, experimental hepatic metastases of the CT-26 colon tumor were studied; and in C57BL/6J mice, survival rates following excision of B16 melanoma was assessed - both mouse tumor models involved surgical stress. The findings indicated that simultaneous blockade of glucocorticoid and ß-adrenergic receptors improved CpG-C efficacy against MADB106 metastasis. In mice bearing B16 melanoma, long-term survival rate was improved by CpG-C only when employed simultaneously with blockers of glucocorticoids, catecholamines, and prostaglandins. Prolonged stress impaired CpG-C efficacy in potentiating NK activity, and in resisting MADB106 metastasis in both sexes, as also supported by in vitro studies. This latter effect was not blocked by any of the antagonists or by adrenalectomy. In the CT26 model, prolonged stress only partially reduced the efficacy of CpG-C. Overall, our findings indicate that ongoing behavioral stress and surgery can jeopardize immune-stimulatory interventions and abolish their beneficial metastasis-reducing impacts. These findings have implications for the clinical setting, which often involve psychological and physiological stress responses during immune-stimulation.


Assuntos
Catecolaminas/antagonistas & inibidores , Glucocorticoides/antagonistas & inibidores , Fatores Imunológicos/farmacologia , Células Matadoras Naturais , Metástase Neoplásica/prevenção & controle , Neoplasias/tratamento farmacológico , Oligodesoxirribonucleotídeos/farmacologia , Antagonistas de Prostaglandina/farmacologia , Estresse Psicológico/imunologia , Animais , Modelos Animais de Doenças , Feminino , Fatores Imunológicos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/administração & dosagem , Ratos , Ratos Endogâmicos F344
12.
Pharmacol Res ; 111: 208-216, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27317944

RESUMO

The chemoattractant receptor-homologous molecule expressed on T-helper type 2 cells (CRTh2) is a G protein-coupled receptor expressed on the leukocytes most closely associated with asthma and allergy like eosinophils, mast cells, Th2-lymphocytes and basophils. At present it is clear that CRTh2 mediates most prostaglandin D2 (PGD2) pro-inflammatory effects and as a result antagonists for this receptor have reached asthma clinical studies showing a trend of lung function improvement. The challenge remains to identify compounds with improved clinical efficacy when administered once a day. Herein we described the pharmacological profile of LAS191859, a novel, potent and selective CRTh2 antagonist. In vitro evidence in GTPγS binding studies indicate that LAS191859 is a CRTh2 antagonist with activity in the low nanomolar range. This potency is also maintained in cellular assays performed with human eosinophils and whole blood. The main differentiation of LAS191859 vs other CRTh2 antagonists is in its receptor binding kinetics. LAS191859 has a residence time half-life of 21h at CRTh2 that translates into a long-lasting in vivo efficacy that is independent of plasma levels. We believe that the strategy behind this compound will allow optimal efficacy and posology for chronic asthma treatment.


Assuntos
Antiasmáticos/farmacologia , Eosinófilos/efeitos dos fármacos , Antagonistas de Prostaglandina/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , Receptores Imunológicos/antagonistas & inibidores , Receptores de Prostaglandina/antagonistas & inibidores , Animais , Antiasmáticos/sangue , Antiasmáticos/química , Antiasmáticos/farmacocinética , Células CHO , Forma Celular/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , Cricetulus , Cães , Relação Dose-Resposta a Droga , Desenho de Fármacos , Eosinófilos/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Cobaias , Meia-Vida , Cinética , Masculino , Camundongos , Antagonistas de Prostaglandina/sangue , Antagonistas de Prostaglandina/química , Antagonistas de Prostaglandina/farmacocinética , Ligação Proteica , Piridinas/sangue , Piridinas/química , Piridinas/farmacocinética , Pirróis/sangue , Pirróis/química , Pirróis/farmacocinética , Ratos Wistar , Receptores Imunológicos/sangue , Receptores Imunológicos/genética , Receptores de Prostaglandina/sangue , Receptores de Prostaglandina/genética , Transfecção
13.
Bull Exp Biol Med ; 162(2): 187-190, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27909962

RESUMO

We performed a complex functional study of the effects of prostaglandin synthesis blockage with diclofenac on manifestation of the hydroosmotic effect of vasopressin V2-receptor agonist desmopressin in the kidneys of Wistar rats with normal synthesis of endogenous vasopressin and homozygous Brattleboro rats with hereditary impaired synthesis of neurohypophyseal hormone vasopressin. Blockage of prostaglandin synthesis led to more pronounced increase in urine osmolality in Brattleboro rats than in Wistar rats due to elevation of not only urine but also sodium gradient at the expense of elimination of the inhibitory effect of prostaglandins on sodium reabsorption and membrane permeability for urine. During combined treatment, the effects of the hormone predominated: the increase in urine osmolality in Wistar and Brattleboro rats did not differ from that after desmopressin administration.


Assuntos
Antidiuréticos/farmacologia , Diabetes Insípido Nefrogênico/tratamento farmacológico , Diclofenaco/farmacologia , Antagonistas de Prostaglandina/farmacologia , Prostaglandinas/biossíntese , Vasopressinas/farmacologia , Animais , Desamino Arginina Vasopressina/farmacologia , Diabetes Insípido Nefrogênico/patologia , Diabetes Insípido Nefrogênico/urina , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Córtex Renal/patologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Medula Renal/patologia , Concentração Osmolar , Ratos , Ratos Brattleboro , Ratos Wistar , Sódio/urina , Vasopressinas/deficiência
14.
Bull Exp Biol Med ; 161(6): 850-852, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27783283

RESUMO

Ultrastructural changes in cells of the renal inner medulla involved in the realization of the antidiuretic effect of vasopressin under conditions of prostaglandin synthesis blockade were studied in the kidneys of Wistar rats and endogenous vasopressin-deficient homozygous Brattleboro rats. The results indicated uniform trend to an increase in the number of clathrincoated vesicles under conditions of hormone treatment combined with prostaglandin synthesis blockade in animals with different neurohypophyseal status. These changes reflected translocation of aquaporins and an increase in the permeability of the collecting tubular epithelium for water. Brattleboro rats, but not Wistar rats, exhibited ultrastructural signs of synthesis activation in the epithelium and widening of the intercellular gaps, which could indicate more intense paracellular water transport.


Assuntos
Antidiuréticos/farmacologia , Diabetes Insípido Neurogênico/tratamento farmacológico , Medula Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Prostaglandinas/metabolismo , Vasopressinas/farmacologia , Animais , Aquaporinas/metabolismo , Transporte Biológico , Vesículas Revestidas por Clatrina/efeitos dos fármacos , Vesículas Revestidas por Clatrina/metabolismo , Diabetes Insípido Neurogênico/metabolismo , Diabetes Insípido Neurogênico/patologia , Diclofenaco/farmacologia , Medula Renal/metabolismo , Medula Renal/patologia , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/patologia , Microscopia Eletrônica , Concentração Osmolar , Antagonistas de Prostaglandina/farmacologia , Transporte Proteico , Ratos , Ratos Brattleboro , Ratos Wistar , Vasopressinas/deficiência , Água/metabolismo
15.
J Neurochem ; 135(5): 867-79, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25865705

RESUMO

The blood-brain barrier, formed by microvessel endothelial cells, is the restrictive barrier between the brain parenchyma and the circulating blood. Arachidonic acid (ARA; 5,8,11,14-cis-eicosatetraenoic acid) is a conditionally essential polyunsaturated fatty acid [20:4(n-6)] and is a major constituent of brain lipids. The current study examined the transport processes for ARA in confluent monolayers of human brain microvascular endothelial cells (HBMEC). Addition of radioactive ARA to the apical compartment of HBMEC cultured on Transwell(®) inserts resulted in rapid incorporation of radioactivity into the basolateral medium. Knock down of fatty acid transport proteins did not alter ARA passage into the basolateral medium as a result of the rapid generation of prostaglandin E2 (PGE2 ), an eicosanoid known to facilitate opening of the blood-brain barrier. Permeability following ARA or PGE2 exposure was confirmed by an increased movement of fluorescein-labeled dextran from apical to basolateral medium. ARA-mediated permeability was attenuated by specific cyclooxygenase-2 inhibitors. EP3 and EP4 receptor antagonists attenuated the ARA-mediated permeability of HBMEC. The results indicate that ARA increases permeability of HBMEC monolayers likely via increased production of PGE2 which acts upon EP3 and EP4 receptors to mediate permeability. These observations may explain the rapid influx of ARA into the brain previously observed upon plasma infusion with ARA. The blood-brain barrier, formed by microvessel endothelial cells, is a restrictive barrier between the brain parenchyma and the circulating blood. Radiolabeled arachidonic acid (ARA) movement across, and monolayer permeability in the presence of ARA, was examined in confluent monolayers of primary human brain microvessel endothelial cells (HBMECs) cultured on Transwell(®) plates. Incubation of HBMECs with ARA resulted in a rapid increase in HBMEC monolayer permeability. The mechanism was mediated, in part, through increased prostaglandin E2 production from ARA which acted upon EP3 and EP4 receptors to increase HBMEC monolayer permeability.


Assuntos
Ácido Araquidônico/farmacologia , Permeabilidade Capilar/efeitos dos fármacos , Dinoprostona/metabolismo , Células Endoteliais/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Encéfalo/anatomia & histologia , Antígenos CD36/metabolismo , Permeabilidade Capilar/fisiologia , Células Cultivadas , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dextranos/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Isótopos/farmacocinética , Microvasos/citologia , Ácido Oleico/farmacologia , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Antagonistas de Prostaglandina/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
16.
Cell Physiol Biochem ; 37(1): 1-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26277839

RESUMO

BACKGROUND: Recently, we gained evidence that impairment of rOat1 and rOat3 expression induced by ischemic acute kidney injury (AKI) is mediated by COX metabolites and this suppression might be critically involved in renal damage. METHODS: (i) Basolateral organic anion uptake into proximal tubular cells after model ischemia and reperfusion (I/R) was investigated by fluorescein uptake. The putative promoter sequences from hOAT1 (SLC22A6) and hOAT3 (SCL22A8) were cloned into a reporter plasmid, transfected into HEK cells and (ii) transcriptional activity was determined after model ischemia and reperfusion as a SEAP reporter gen assay. Inhibitors or antagonists were applied with the beginning of reperfusion. RESULTS: By using inhibitors of PKA (H89) and PLC (U73122), antagonists of E prostanoid receptor type 2 (AH6809) and type 4 (L161,982), we gained evidence that I/R induced down regulation of organic anion transport is mediated by COX1 metabolites via E prostanoid receptor type 4. The latter signaling was confirmed by application of butaprost (EP2 agonist) or TCS2510 (EP4 agonist) to control cells. In brief, the latter signaling was verified for the transcriptional activity in the reporter gen assay established. Therein, selective inhibitors for COX1 (SC58125) and COX2 (SC560) were also applied. CONCLUSION: Our data show (a) that COX1 metabolites are involved in the regulation of renal organic anion transport(ers) after I/R via the EP4 receptor and (b) that this is due to transcriptional regulation of the respective transporters. As the promoter sequences cloned were of human origin and expressed in a human renal epithelial cell line we (c) hypothesize that the regulatory mechanisms described after I/R is meaningful for humans as well.


Assuntos
Túbulos Renais Proximais/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Linhagem Celular , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células HEK293 , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Antagonistas de Prostaglandina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
17.
Mol Carcinog ; 54(3): 216-28, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24127286

RESUMO

Silymarin has been known as a chemopreventive agent, and possesses multiple anti-cancer activities including induction of apoptosis, inhibition of proliferation and growth, and blockade of migration and invasion. However, whether silymarin could inhibit prostaglandin (PG) E2 -induced renal cell carcinoma (RCC) migration and what are the underlying mechanisms are not well elucidated. Here, we found that silymarin markedly inhibited PGE2 -stimulated migration. PGE2 induced G protein-dependent CREB phosphorylation via protein kinase A (PKA) signaling, and PKA inhibitor (H89) inhibited PGE2 -mediated migration. Silymarin reduced PGE2 -induced CREB phosphorylation and CRE-promoter activity. PGE2 also activated G protien-independent signaling pathways (Src and STAT3) and silymarin reduced PGE2 -induced phosphorylation of Src and STAT3. Inhibitor of Src (Saracatinib) markedly reduced PGE2 -mediated migration. We found that EP2, a PGE2 receptor, is involved in PGE2 -mediated cell migration. Down regulation of EP2 by EP2 siRNA and EP2 antagonist (AH6809) reduced PGE2 -inudced migration. In contrast, EP2 agonist (Butaprost) increased cell migration and silymarin effectively reduced butaprost-mediated cell migration. Moreover, PGE2 increased EP2 expression through activation of positive feedback mechanism, and PGE2 -induced EP2 expression, as well as basal EP2 levels, were reduced in silymarin-treated cells. Taken together, our study demonstrates that silymarin inhibited PGE2 -induced cell migration through inhibition of EP2 signaling pathways (G protein dependent PKA-CREB and G protein-independent Src-STAT3).


Assuntos
Antioxidantes/farmacologia , Carcinoma de Células Renais/patologia , Movimento Celular/efeitos dos fármacos , Dinoprostona/antagonistas & inibidores , Neoplasias Renais/patologia , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Silimarina/farmacologia , Alprostadil/análogos & derivados , Alprostadil/farmacologia , Benzodioxóis/farmacologia , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Regulação para Baixo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Células HCT116 , Células HT29 , Humanos , Integrases/genética , Neoplasias Renais/metabolismo , Fosforilação , Antagonistas de Prostaglandina/farmacologia , Quinazolinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/biossíntese , Receptores de Prostaglandina E Subtipo EP2/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Cicatrização , Xantonas/farmacologia , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
18.
Respir Res ; 16: 149, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26684827

RESUMO

BACKGROUND: Cyclooxygenase-2-derived prostaglandin E2 (PGE2), a bioactive eicosanoid, has been implicated in many biological processes including reproduction, inflammation and tumor growth. We previously showed that PGE2 stimulated lung cancer cell growth and progression through PGE2 receptor EP2/EP4-mediated kinase signaling pathways. However, the role of PGE2 in controlling lung airway epithelial cell phenotype remains unknown. We evaluated the effects of c-Jun and 3-phosphoinositede dependent protein kinase-1 (PDK1) in mediating epithelial cell hyperplasia induced by PGE2. METHOD: The bronchial epithelial cell lines BEAS-2B and HBEc14-KT were cultured and then treated with PGE2. PDK1 small interfering RNA (siRNA) and a PDK1 inhibitor, an antagonist of the PGE2 receptor subtype EP4 and EP4 siRNA, c-Jun siRNA, and overexpressions of c-Jun and PDK1 have been used to evaluate the effects on cell proliferation. RESULTS: We demonstrated that PGE2 increased normal bronchial epithelial cell proliferation through induction of PDK1, an ankyrin repeat-containing Ser/Thr kinase implicated in the induction of apoptosis and the suppression of tumor growth. PDK1 siRNA and a PDK1 inhibitor blocked the effects of PGE2 on normal cell growth. The PGE2-induced PDK1 expression was blocked by an antagonist of the PGE2 receptor subtype EP4 and by EP4 siRNA. In addition, we showed that induction of PDK1 by PGE2 was associated with induction of the transcription factor, c-Jun protein. Silencing of c-Jun using siRNA and point mutations of c-Jun sites in the PDK1 gene promoter resulted in blockade of PDK1 expression and promoter activity induced by PGE2. In contrast, overexpression of c-Jun induced PDK1 gene promoter activity and expression followed increased cell proliferation. CONCLUSION: PGE2 increases normal bronchial epithelial cell proliferation through increased PDK1 gene expression that is dependent on EP4 and induction of c-Jun. Therewith, our data suggest a new role of c-Jun and PDK1 in mediating epithelial cell hyperplasia induced by PGE2.


Assuntos
Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Brônquios/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dinoprostona/farmacologia , Células Epiteliais/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/antagonistas & inibidores , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/genética , Sítios de Ligação , Brônquios/enzimologia , Brônquios/patologia , Linhagem Celular , Relação Dose-Resposta a Droga , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Humanos , Hiperplasia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Fosforilação , Regiões Promotoras Genéticas , Antagonistas de Prostaglandina/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Receptores de Prostaglandina E Subtipo EP4/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transfecção
19.
Arterioscler Thromb Vasc Biol ; 34(2): 285-93, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24285580

RESUMO

OBJECTIVE: Prompt post-hypoxia-ischemia (HI) revascularization has been suggested to improve outcome in adults and newborn subjects. Other than hypoxia-inducible factor, sensors of metabolic demand remain largely unknown. During HI, anaerobic respiration is arrested resulting in accumulation of carbohydrate metabolic intermediates. As such succinate readily increases, exerting its biological effects via a specific receptor, G-protein-coupled receptor (GPR) 91. We postulate that succinate/GPR91 enhances post-HI vascularization and reduces infarct size in a model of newborn HI brain injury. APPROACH AND RESULTS: The Rice-Vannucci model of neonatal HI was used. Succinate was measured by mass spectrometry, and microvascular density was evaluated by quantification of lectin-stained cryosection. Gene expression was evaluated by real-time polymerase chain reaction. Succinate levels rapidly increased in the penumbral region of brain infarcts. GPR91 was foremost localized not only in neurons but also in astrocytes. Microvascular density increased at 96 hours after injury in wild-type animals; it was diminished in GPR91-null mice leading to an increased infarct size. Stimulation with succinate led to an increase in growth factors implicated in angiogenesis only in wild-type mice. To explain the mode of action of succinate/GPR91, we investigated the role of prostaglandin E2-prostaglandin E receptor 4, previously proposed in neural angiogenesis. Succinate-induced vascular endothelial growth factor expression was abrogated by a cyclooxygenase inhibitor and a selective prostaglandin E receptor 4 antagonist. This antagonist also abolished succinate-induced neovascularization. CONCLUSIONS: We uncover a dominant metabolic sensor responsible for post-HI neurovascular adaptation, notably succinate/GPR91, acting via prostaglandin E2-prostaglandin E receptor 4 to govern expression of major angiogenic factors. We propose that pharmacological intervention targeting GPR91 could improve post-HI brain recovery.


Assuntos
Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/efeitos dos fármacos , Infarto Cerebral/tratamento farmacológico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Ácido Succínico/farmacologia , Proteínas Angiogênicas/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Linhagem Celular , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Infarto Cerebral/etiologia , Infarto Cerebral/genética , Infarto Cerebral/metabolismo , Infarto Cerebral/patologia , Infarto Cerebral/fisiopatologia , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Hipóxia-Isquemia Encefálica/etiologia , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Injeções Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/metabolismo , Antagonistas de Prostaglandina/farmacologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Prostaglandina E Subtipo EP4/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ácido Succínico/administração & dosagem , Ácido Succínico/metabolismo , Fatores de Tempo , Técnicas de Cultura de Tecidos
20.
Blood ; 119(10): 2358-67, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-22234697

RESUMO

Prostaglandin E(2) (PGE(2)) is a lipid mediator that acts by ligating 4 distinct G protein-coupled receptors, E prostanoid (EP) 1 to 4. Previous studies identified the importance of PGE(2) in regulating macrophage functions, but little is known about its effect on macrophage maturation. Macrophage maturation was studied in vitro in bone marrow cell cultures, and in vivo in a model of peritonitis. EP2 was the most abundant PGE(2) receptor expressed by bone marrow cells, and its expression further increased during macrophage maturation. EP2-deficient (EP2(-/-)) macrophages exhibited enhanced in vitro maturation compared with wild-type cells, as evidenced by higher F4/80 expression. An EP2 antagonist also increased maturation. In the peritonitis model, EP2(-/-) mice exhibited a higher percentage of F4/80(high)/CD11b(high) cells and greater expression of macrophage colony-stimulating factor receptor (M-CSFR) in both the blood and the peritoneal cavity. Subcutaneous injection of the PGE(2) analog misoprostol decreased M-CSFR expression in bone marrow cells and reduced the number of peritoneal macrophages in wild-type mice but not EP2(-/-) mice. The suppressive effect of EP2 ligation on in vitro macrophage maturation was mimicked by a selective protein kinase A agonist. Our findings reveal a novel role for PGE(2)/EP2/protein kinase A signaling in the suppression of macrophage maturation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Macrófagos/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Transdução de Sinais , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Citometria de Fluxo , Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Misoprostol/farmacologia , Peritonite/induzido quimicamente , Peritonite/genética , Peritonite/metabolismo , Antagonistas de Prostaglandina/farmacologia , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptores de Prostaglandina E Subtipo EP2/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tioglicolatos , Xantonas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA