44 resultados para Prostacyclin


Relevância:

20.00% 20.00%

Publicador:

Resumo:

BACKGROUND: Prostacyclin synthase (PGIS) metabolizes prostaglandin H(2), into prostacyclin. This study aimed to determine the expression profile of PGIS in nonsmall cell lung cancer (NSCLC) and examine potential mechanisms involved in PGIS regulation. METHODS: PGIS expression was examined in human NSCLC and matched controls by reverse transcriptase polymerase chain reaction (RT-PCR), Western analysis, and immunohistochemistry. A 204-patient NSCLC tissue microarray was stained for PGIS and cyclooxygenase 2 (COX2) expression. Staining intensity was correlated with clinical parameters. Epigenetic mechanisms underpinning PGIS promoter expression were examined using RT-PCR, methylation-specific PCR, and chromatin immunoprecipitation analysis. RESULTS: PGIS expression was reduced/absent in human NSCLC protein samples (P <.0001), but not mRNA relative to matched controls. PGIS tissue expression was higher in squamous cell carcinoma (P =.004) and in male patients (P <.05). No significant correlation of PGIS or COX2 expression with overall patient survival was observed, although COX2 was prognostic for short-term (2-year) survival (P <.001). PGIS mRNA expression was regulated by DNA CpG methylation and histone acetylation in NSCLC cell lines, with chromatin remodeling taking place directly at the PGIS gene. PGIS mRNA expression was increased by both demethylation agents and histone deacetylase inhibitors. Protein levels were unaffected by demethylation agents, whereas PGIS protein stability was negatively affected by histone deacetylase inhibitors. CONCLUSIONS: PGIS protein expression is reduced in NSCLC, and does not correlate with overall patient survival. PGIS expression is regulated through epigenetic mechanisms. Differences in expression patterns between mRNA and protein levels suggest that PGIS expression and protein stability are regulated post-translationally. PGIS protein stability may have an important therapeutic role in NSCLC. © 2011 American Cancer Society.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Prostacyclin synthase and thromboxane synthase signaling via arachidonic acid metabolism affects a number of tumor cell survival pathways such as cell proliferation, apoptosis, tumor cell invasion and metastasis, and angiogenesis. However, the effects of these respective synthases differ considerably with respect to the pathways described. While prostacyclin synthase is generally believed to be anti-tumor, a pro-carcinogenic role for thromboxane synthase has been demonstrated in a variety of cancers. The balance of oppositely-acting COX-derived prostanoids influences many processes throughout the body, such as blood pressure regulation, clotting, and inflammation. The PGI2/TXA2 ratio is of particular interest in-vivo, with the corresponding synthases shown to be differentially regulated in a variety of disease states. Pharmacological inhibition of thromboxane synthase has been shown to significantly inhibit tumor cell growth, invasion, metastasis and angiogenesis in a range of experimental models. In direct contrast, prostacyclin synthase overexpression has been shown to be chemopreventive in a murine model of the disease, suggesting that the expression and activity of this enzyme may protect against tumor development. In this review, we discuss the aberrant expression and known functions of both prostacyclin synthase and thromboxane synthase in cancer. We discuss the effects of these enzymes on a range of tumor cell survival pathways, such as tumor cell proliferation, induction of apoptosis, invasion and metastasis, and tumor cell angiogenesis. As downstream signaling pathways of these enzymes have also been implicated in cancer states, we examine the role of downstream effectors of PGIS and TXS activity in tumor growth and progression. Finally, we discuss current therapeutic strategies aimed at targeting these enzymes for the prevention/treatment of cancer. © 2010 Elsevier B.V. All rights reserved.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

The risk of diabetic retinopathy is associated with the presence of both oxidative stress and toxic eicosanoids. Whether oxidative stress actually causes diabetic retinopathy via the generation of toxic eicosanoids, however, remains unknown. The aim of the present study was to determine whether tyrosine nitration of prostacyclin synthase (PGIS) contributes to retinal cell death in vitro and in vivo. Exposure of human retinal pericytes to heavily oxidized and glycated LDL (HOG-LDL), but not native forms of LDL (N-LDL), for 24 hours significantly increased pericyte apoptosis, accompanied by increased tyrosine nitration of PGIS and decreased PGIS activity. Inhibition of the thromboxane receptor or cyclooxygenase-2 dramatically attenuated HOG-LDL-induced apoptosis without restoring PGIS activity. Administration of superoxide dismutase (to scavenge superoxide anions) or L-N(G)-nitroarginine methyl ester (L-NAME, a nonselective nitric oxide synthase inhibitor) restored PGIS activity and attenuated pericyte apoptosis. In Akita mouse retinas, diabetes increased intraretinal levels of oxidized LDL and glycated LDL, induced PGIS nitration, enhanced apoptotic cell death, and impaired blood-retinal barrier function. Chronic administration of tempol, a superoxide scavenger, reduced intraretinal oxidized LDL and glycated LDL levels, PGIS nitration, and retina cell apoptosis, thereby preserving the integrity of blood-retinal barriers. In conclusion, oxidized LDL-mediated PGIS nitration and associated thromboxane receptor stimulation might be important in the initiation and progression of diabetic retinopathy.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Background and purpose: The present study was designed to assess whether cyclooxygenase-2 (COX-2) activation is involved in the effects of chronic aldosterone treatment on endothelial function of mesenteric resistance arteries (MRA) from Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHR). Experimental approach: Relaxation to acetylcholine was measured in MRA from both untreated and aldosterone-treated strains. Vasomotor responses to prostacyclin and U46619 were also analysed. Release of 6-oxo-prostaglandin (PG)F(1 alpha) and thromboxane B(2) (TxB(2)) was determined by enzyme immunoassay. COX-2 protein expression was measured by western blot. Key results: Aldosterone reduced acetylcholine relaxation in MRA from both strains. In MRA from both aldosterone-treated strains the COX-1/2 or COX-2 inhibitor (indomethacin and NS-398, respectively), Tx2 synthesis inhibitor (furegrelate), prostacyclin synthesis inhibitor (tranylcypromine) or Tx2/PG2 receptor antagonist (SQ 29 548), but not COX-1 inhibitor SC-560, increased acetylcholine relaxation. In untreated rats this response was increased only in SHR. Prostacyclin elicited a biphasic vasomotor response: lower concentrations elicited relaxation, whereas higher concentrations elicited contraction that was reduced by SQ 29 548. Aldosterone increased the acetylcholine-stimulated production of 6-oxo-PGF(1 alpha) and TxB(2) in MRA from both strains. COX-2 expression was higher in both strains of rats treated with aldosterone. Conclusions and implications: Chronic treatment with aldosterone impaired endothelial function in MRA under normotensive and hypertensive conditions by increasing COX-2-derived prostacyclin and thromboxane A(2). As endothelial dysfunction participates in the pathogenesis of many cardiovascular disorders we hypothesize that anti-inflammatory drugs, specifically COX-2 inhibitors, could ameliorate vascular damage in patients with elevated aldosterone production.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

To better understand the mechanisms of how the human prostacyclin receptor (1P) mediates vasodilation and platelet anti-aggregation through Gs protein coupling, a strategy integrating multiple approaches including high resolution NMR experiments, synthetic peptide, fluorescence spectroscopy, molecular modeling, and recombinant protein was developed and used to characterize the structure/function relationship of important segments and residues of the IP receptor and the α-subunit of the Gs protein (Gαs). The first (iLP1) and third (iLP3) intracellular loops of the IP receptor, as well as the Gαs C-terminal domain, relevant to the Gs-mediated IP receptor signaling, were first identified by observation of the effects of the mini gene-expressed corresponding protein segments in HEK293 cells which co-expressed the receptor and Gαs. Evidence of the IP iLP1 domain interacted with the Gαs C-terminal domain was observed by fluorescence and NMR spectroscopic studies using a constrained synthetic peptide, which mimicked the IP iLP1 domain, and the synthetic peptide, which mimicked Gαs C-terminal domain. The solution structural models and the peptide-peptide interaction of the two synthetic protein segments were determined by high resolution NMR spectroscopy. The important residues in the corresponding domains of the IP receptor and the Gαs predicted by NMR chemical shift mapping were used to guide the identification of their protein-protein interaction in cells. A profile of the residues Arg42 - Ala48 of the IP iLP1 domain and the three residues Glu392 ∼ Leu394 of the Gαs C-terminal domain involved in the IP/Gs protein coupling were confirmed by recombinant proteins. The data revealed an intriguing speculation on the mechanisms of how the signal of the ligand-activated IP receptor is transmitted to the Gs protein in regulating vascular functions and homeostasis, and also provided substantial insights into other prostanoid receptor signaling. ^

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Although coronary artery disease (CAD) is appreciated to be accelerated in patients with chronic spinal cord injury (SCI), the underlying mechanism of CAD in SCI remains obscure. We have recently shown that platelets from subjects with SCI develop resistance to the inhibitory effect of prostacyclin (PGI2) on the platelet stimulation of thrombin generation. The loss of the inhibitory effect was due to the loss of high-affinity prostanoid receptors, which may contribute to atherogenesis in SCI. Incubation of normal, non-SCI platelets in SCI plasma (n = 12) also resulted in the loss of high-affinity binding of PGI2 (Kd1 = 9.1 ± 2.0 nM; n1 = 170 ± 32 sites per cell vs. Kd1 = 7.2 ± 1.1 nM; n1 = 23 ± 8 sites per cell), with no significant change in the low-affinity receptors (Kd2 = 1.9 ± 0.1 μM; n2 = 1,832 ± 232 sites per cell vs. Kd2 = 1.6 ± 0.1 μM; n2 = 1,740 ± 161 sites per cell) as determined by Scatchard analysis of the binding of [3H]PGE1. The loss of high-affinity PGI2 binding led to the failure of PGI2 to inhibit the platelet-stimulated thrombin generation. The increase of cellular cyclic AMP level, mediated through the binding of PGI2 to low-affinity receptors in platelets, was unaffected in SCI platelets. PAGE and immunoblot of SCI plasma showed the presence of an IgG band, which specifically blocked the binding of [3H]PGE1 to the high-affinity PGI2 receptors of normal platelets. PAGE of the reduced IgG band, the amino acid sequence of the novel band as a heavy chain of IgG that inhibits the binding of [3H]PGE1 to the high-affinity platelet PGI2 receptor, demonstrates that the specific recognition and inhibition of high-affinity PGI2 binding to platelets was due to an anti-prostacyclin receptor antibody present in SCI plasma.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

There is evidence from both genetic and pharmacologic studies to suggest that the cyclooxygenase-2 (COX-2) enzyme plays a causal role in the development of colorectal cancer. However, little is known about the identity or role of the eicosanoid receptor pathways activated by COX-derived prostaglandins (PG). We previously have reported that COX-2-derived prostacyclin promotes embryo implantation in the mouse uterus via activation of the nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR) δ. In light of the recent finding that PPARδ is a target of β-catenin transactivation, it is important to determine whether this signaling pathway is operative during the development of colorectal cancer. Analysis of PPARδ mRNA in matched normal and tumor samples revealed that expression of PPARδ, similar to COX-2, is up-regulated in colorectal carcinomas. In situ hybridization studies demonstrate that PPARδ is expressed in normal colon and localized to the epithelial cells at the very tips of the mucosal glands. In contrast, expression of PPARδ mRNA in colorectal tumors was more widespread with increased levels in transformed epithelial cells. Analysis of PPARδ and COX-2 mRNA in serial sections suggested they were colocalized to the same region within a tumor. Finally, transient transfection assays established that endogenously synthesized prostacyclin (PGI2) could serve as a ligand for PPARδ. In addition, the stable PGI2 analog, carbaprostacyclin, and a synthetic PPARδ agonist induced transactivation of endogenous PPARδ in human colon carcinoma cells. We conclude from these observations that PPARδ, similar to COX-2, is aberrantly expressed in colorectal tumors and that endogenous PPARδ is transcriptionally responsive to PGI2. However, the functional consequence of PPARδ activation in colon carcinogenesis still needs to be determined.

Relevância:

20.00% 20.00%

Publicador:

Resumo:

Coronary artery disease is a leading cause of death in individuals with chronic spinal cord injury (SCI). However, platelets of those with SCI (n = 30) showed neither increased aggregation nor resistance to the antiaggregatory effects of prostacyclin when compared with normal controls (n = 30). Prostanoid-induced cAMP synthesis was similar in both groups. In contrast, prostacyclin, which completely inhibited the platelet-stimulated thrombin generation in normal controls, failed to do so in those with SCI. Scatchard analysis of the binding of [3H]prostaglandin E1, used as a prostacyclin receptor probe, showed the presence of one high-affinity (Kd1 = 8.11 +/- 2.80 nM; n1 = 172 +/- 32 sites per cell) and one low-affinity (Kd2 = 1.01 +/- 0.3 microM; n2 = 1772 +/- 226 sites per cell) prostacyclin receptor in normal platelets. In contrast, the same analysis in subjects with SCI showed significant loss (P < 0.001) of high-affinity receptor sites (Kd1 = 6.34 +/- 1.91 nM; n1 = 43 +/- 10 sites per cell) with no significant change in the low affinity-receptors (Kd2 = 1.22 +/- 0.23; n2 = 1820 +/- 421). Treatment of these platelets with insulin, which has been demonstrated to restore both of the high- and low-affinity prostaglandin receptor numbers to within normal ranges in coronary artery disease, increased high-affinity receptor numbers and restored the prostacyclin effect on thrombin generation. These results demonstrate that the loss of the inhibitory effect of prostacyclin on the stimulation of thrombin generation was due to the loss of platelet high-affinity prostanoid receptors, which may contribute to atherogenesis in individuals with chronic SCI.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Arachidonic acid metabolism through cyclooxygenase (COX) pathways leads to the generation of biologically active eicosanoids. Eicosanoid expression levels vary during development and progression of gastrointestinal (GI) malignancies. COX-2 is the major COX-isoform responsible for G.I. cancer development/progression. COX-2 expression increases during progression from a normal to cancerous state. Evidence from observational studies has demonstrated that chronic NSAID use reduces the risk of cancer development, while both incidence and risk of death due to G.I. cancers were significantly reduced by daily aspirin intake. A number of randomized controlled trials (APC trial, Prevention of Sporadic Adenomatous Polyps trial, APPROVe trial) have also shown a significant protective effect in patients receiving selective COX-2 inhibitors. However, chronic use of selective COX-2 inhibitors at high doses was associated with increased cardiovascular risk, while NSAIDs have also been associated with increased risk. More recently, downstream effectors of COX-signaling have been investigated in cancer development/progression. PGE 2, which binds to both EP and PPAR receptors, is the major prostanoid implicated in the carcinogenesis of G.I. cancers. The role of TXA 2 in G.I. cancers has also been examined, although further studies are required to uncover its role in carcinogenesis. Other prostanoids investigated include PGD 2 and its metabolite 15d-PGJ2, PGF 1α and PGI 2. Targeting these prostanoids in G.I. cancers has the promise of avoiding cardiovascular toxicity associated with chronic selective COX-2 inhibition, while maintaining anti-tumor reactivity.A progressive sequence from normal to pre-malignant to a malignant state has been identified in G.I. cancers. In this review, we will discuss the role of the COX-derived prostanoids in G.I. cancer development and progression. Targeting these downstream prostanoids for chemoprevention and/or treatment of G.I. cancers will also be discussed. Finally, we will highlight the latest pre-clinical technologies as well as avenues for future investigation in this highly topical research field. © 2011 Elsevier B.V.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Exogenous prostacyclin is effective in reducing pulmonary vascular resistance in some forms of human pulmonary hypertension (PH). To explore whether endogenous prostaglandins played a similar role in pulmonary hypertension, we examined the effect of deleting cyclooxygenase (COX)-gene isoforms in a chronic hypoxia model of PH. Pulmonary hypertension, examined by direct measurement of right ventricular end systolic pressure (RVESP), right ventricular hypertrophy (n = 8), and hematocrit (n = 3), was induced by 3 weeks of hypobarichypoxia in wild-type and COX-knockout (KO) mice. RVESP was increased in wild-type hypoxic mice compared with normoxic controls (24.4 ± 1.4 versus 13.8 ± 1.9 mm Hg; n = 8; p < 0.05). COX-2 KO mice showed a greater increase in RVESP following hypoxia (36.8 ± 2.7 mm Hg; p < 0.05). Urinary thromboxane (TX)B2 excretion increased following hypoxia (44.6 ± 11.1 versus 14.7 ± 1.8 ng/ml; n = 6; p < 0.05), an effect that was exacerbated by COX-2 gene disruption (54.5 ± 10.8 ng/ml; n = 6). In contrast, the increase in 6-keto-prostacyclin1α excretion following hypoxia was reduced by COX-2 gene disruption (29 ± 3 versus 52 ± 4.6 ng/ml; p < 0.01). Tail cut bleed times were lower following hypoxia, and there was evidence of intravascular thrombosis in lung vessels that was exacerbated by disruption of COX-2 and reduced by deletion of COX-1. The TXA2/endoperoxide receptor antagonist ifetroban (50 mg/kg/day) offset the effect of deleting the COX-2 gene, attenuating the hypoxia-induced rise in RVESP and intravascular thrombosis. COX-2 gene deletion exacerbates pulmonary hypertension, enhances sensitivity to TXA2, and induces intravascular thrombosis in response to hypoxia. The data provide evidence that endogenous prostaglandins modulate the pulmonary response to hypoxia. Copyright © 2008 by The American Society for Pharmacology and Experimental Therapeutics.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

AIMS: Aldosterone plays a crucial role in cardiovascular disease. 'Systemic' inhibition of its mineralocorticoid receptor (MR) decreases atherosclerosis by reducing inflammation and oxidative stress. Obesity, an important cardiovascular risk factor, is an inflammatory disease associated with increased plasma aldosterone levels. We have investigated the role of the 'endothelial' MR in obesity-induced endothelial dysfunction, the earliest stage in atherogenesis. METHODS AND RESULTS: C57BL/6 mice were exposed to a normal chow diet (ND) or a high-fat diet (HFD) alone or in combination with the MR antagonist eplerenone (200 mg/kg/day) for 14 weeks. Diet-induced obesity impaired endothelium-dependent relaxation in response to acetylcholine, whereas eplerenone treatment of obese mice prevented this. Expression analyses in aortic endothelial cells isolated from these mice revealed that eplerenone attenuated expression of pro-oxidative NADPH oxidase (subunits p22phox, p40phox) and increased expression of antioxidative genes (glutathione peroxidase-1, superoxide dismutase-1 and -3) in obesity. Eplerenone did not affect obesity-induced upregulation of cyclooxygenase (COX)-1 or prostacyclin synthase. Endothelial-specific MR deletion prevented endothelial dysfunction in obese (exhibiting high 'endogenous' aldosterone) and in 'exogenous' aldosterone-infused lean mice. Pre-incubation of aortic rings from aldosterone-treated animals with the COX-inhibitor indomethacin restored endothelial function. Exogenous aldosterone administration induced endothelial expression of p22phox in the presence, but not in the absence of the endothelial MR. CONCLUSION: Obesity-induced endothelial dysfunction depends on the 'endothelial' MR and is mediated by an imbalance of oxidative stress-modulating mechanisms. Therefore, MR antagonists may represent an attractive therapeutic strategy in the increasing population of obese patients to decrease vascular dysfunction and subsequent atherosclerotic complications.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Les Cellules Endothéliales Progénitrices ("Endothelial Progenitor Cells", EPCs) sont des précurseurs endothéliaux qui jouent un rôle émergeant en biologie vasculaire. Les EPCs ont été localisées dans le cordon ombilical, la moelle osseuse, le sang périphérique et dans certains tissus régénérateurs. Les interactions des EPCs avec les cellules sanguines et vasculaires peuvent largement influencer leurs propriétés biologiques et dicter leur fonctionnement pendant la réparation endothéliale. Plus spécifiquement, les interactions des EPCs avec les plaquettes circulantes induisent leur migration, leur recrutement et leur différentiation en cellules endothéliales aux sites de lésions vasculaires. Cependant, l’impact d’une telle interaction sur la fonction plaquettaire n’a pas été recherché. Le but de mon projet était de :1) générer des EPCs à partir des cellules mononucléaires du sang humain périphérique ("Peripheral Blood Mononuclear Cells", PBMCs); 2) étudier les interactions adhésives entre les EPCs et les plaquettes; 3) déterminer leur impact sur la fonction plaquettaire et la formation du thrombus et 4) décrire le mécanisme d’action des EPCs sur les plaquettes et le thrombus. Mises en culture sur une surface de fibronectine dans un milieu conditionné, les PBMCs fraîchement isolées possédaient une morphologie ronde et une petite taille. Après cinq jours, les PBMCs adhérentes donnaient naissance à des colonies, puis formaient une monocouche de cellules aplaties caractéristiques des EPCs après dix jours de culture. Les EPCs différenciées étaient positives pour l’Ulex-lectine et l’Acétyle des lipoprotéines de faible densité ("Acetylated Low Density Lipoprotein", Ac-LDL), exprimaient les marqueurs progéniteurs (CD34, P-sélectine, VEGFR2, vWF et VE-Cadhérine) tandis que les marqueurs leucocytaires (CD14, PSGL-1 et L-sélectine) étaient absents. Ces EPCs interagissaient avec les plaquettes activées par un mécanisme dépendant de la P-sélectine plaquettaire, inhibaient l’activation et l’agrégation plaquettaire et réduisaient significativement l’adhésion plaquettaire, principalement par l’action de prostacycline (PGI2). En fait, ceci était associé avec une augmentation de l’expression de la cyclooxygénase-2 (COX-2) et du monoxyde d’azote (NO) synthéthase inductible (iNOS). Toutefois, les effets inhibiteurs des EPCs sur la fonction plaquettaire ont été renversés par une inhibition de la COX et non pas du NO. Bien que les EPCs fussent en mesure de lier les plaquettes via la P-sélectine, leurs effets prédominants étaient médiés essentiellement par une sécrétion paracrine, impliquant la PGI2. Néanmoins, un rapprochement étroit ou un bref contact entre les EPCs et les plaquettes était requis pour que cette fonction soit complètement réalisée. D’ailleurs, cet aspect a été investigué chez des souris déficientes en P-sélectine (P-sel-/-) et chez leurs congénères de phénotype sauvage (Wild Type, WT). Chez les souris WT, les EPCs inhibaient l’agrégation plaquettaire dans le sang complet de manière concentration-dépendante alors que dans les souris P-sel-/-, l’action des EPCs n’avait pas d’effet significatif. De plus, en utilisant un modèle murin de thrombose artérielle, nous avons démontré que l’infusion systémique des EPCs altéraient la formation du thrombus et réduisaient significativement sa masse chez les souris WT, mais non pas chez les souris P-sel-/-. En outre, le nombre des EPCs incorporées au niveau du thrombus et de la paroi vasculaire était visiblement réduit chez les P-sel-/- par rapport aux souris WT. Dans cette étude, nous sommes parvenus à différentier adéquatement des EPCs à partir des PBMCs, nous avons étudié les interactions adhésives entre les EPCs et les plaquettes, et nous avons décrit leur impact sur la fonction plaquettaire et la formation du thrombus. De plus, nous avons identifié la PGI2 comme étant le principal facteur soluble sécrété par les EPCs en culture et responsable de leurs effets inhibiteurs sur l’activation, l’adhésion et l’agrégation plaquettaire in vitro. De surcroît, nous avons élucidé le mécanisme d’action des EPCs sur l’agrégation plaquettaire et la formation du thrombus, in vivo, et nous avons souligné le rôle de la P-sélectine plaquettaire dans ce processus. Ces résultats ajoutent de nouvelles connaissances sur la biologie des EPCs et définissent leur rôle potentiel dans la régulation de la fonction plaquettaire et la thrombogenèse.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Notre laboratoire a démontré que la capacité proinflammatoire du vascular endothelial growth factor (VEGF-A165) implique la synthèse endothéliale du facteur d’activation plaquettaire (PAF) via l’activation du récepteur tyrosine kinase homodimérique VEGFR-2/R-2. La synthèse du PAF requiert l’activation de la p38 MAPK et p42/44 MAPK qui activent la phospholipase A2 secrétée de type V (sPLA2-V). Nous avons découvert que la synthèse aigue de prostacycline (PGI2) induite par le VEGF-A165 requiert l’activation des récepteurs hétérodimériques VEGFR-1/R-2. L’activation sélective des récepteurs du VEGF peut donc agir comme balance dans la synthèse de facteurs pro-(PAF) et anti-(PGI2) inflammatoire. Cependant, les tyrosines impliquées dans la transphosphorylation de VEGFR-2/R-2 menant à la synthèse du PAF sont inconnues. Par mutagenèse dirigée, nous avons effectué des transfections transitoires de cellules endothéliales avec des plasmides codant pour le VEGFR-2 dont les tyrosines ciblées ont été remplacées de façon séquentielle par une phénylalanine. Un vecteur vide pcDNA a été utilisé comme contrôle négatif. La stimulation des cellules endothéliales de l’aorte bovine (BAEC) transfectées avec le VEGF-A165 (1nM) pendant 15 minutes augmente la synthèse du PAF de 300%, laquelle était similaire dans les BAEC non transfectées. Dans les BAEC transfectées avec les vecteurs pcDNA codant pour les mutations Y801F, Y1059F, Y1175F et Y1214F, nous avons observé une réduction de 54, 73, 68, et 57% respectivement de la synthèse du PAF induite par le VEGF par rapport au pcDNA témoin. Nos résultats apportent un nouvel aperçu sur le mécanisme par lequel le VEGF induit la synthèse du PAF qui est connu pour sa contribution dans l’activité pro-inflammatoire du VEGF.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Thèse numérisée par la Division de la gestion de documents et des archives de l'Université de Montréal.

Relevância:

10.00% 10.00%

Publicador:

Resumo:

Au cours de la grossesse, une perfusion placentaire adéquate est indispensable au bon développement du fœtus. Dans certaines maladies comme la prééclampsie, celle-ci est altérée, compromettant ainsi la vie du fœtus, mais aussi celle de sa mère. Le retrait du placenta mène à la disparition des symptômes de la prééclampsie, suggérant un rôle central de ce dernier dans la maladie. Le placenta étant dépourvu d’innervation autonome, le tonus vasculaire placentaire doit être sous le contrôle de facteurs humoraux et tissulaires. Les vaisseaux placentaires sont très réactifs aux prostanoïdes. Le rapport thromboxane A2 (TXA2)/prostacycline (PGI2) est fortement augmenté dans les placentas de grossesses avec prééclampsie. De plus, le taux d’isoprostane, marqueur du stress oxydatif, est accru dans les placentas de femmes avec prééclampsie. Finalement, la prééclampsie s’accompagne d’un stress oxydatif placentaire marqué. Les espèces réactives de l’oxygène sont connues d’une part, pour oxyder l’acide arachidonique (AA), formant ainsi des isoprostanes et d’autre part, pour augmenter la production de TXA2 dans différents tissus, suite à l’activation des cyclooxygénases (COXs). Nous proposons que : 1. les prostanoïdes sont parmi les molécules endogènes qui contrôlent le tonus vasculaire placentaire. 2. la maladie modifie la réponse aux isoprostanes dans les vaisseaux placentaires. 3. l’induction d’un stress oxydatif placentaire entraîne une réponse vasoactive par activation de la voie du métabolisme de l’AA. Nous avons tout d’abord montré, dans des placentas obtenus de grossesses normotensives, que l’U-46619, un mimétique de la TXA2, de même que l’isoprostane, 8-iso-prostaglandine E2 (8-isoPGE2), ont augmenté fortement la pression de perfusion dans les cotylédons perfusés in vitro et la tension dans les anneaux d’artères chorioniques suspendus dans des bains à organe isolé. En revanche, dans les artères chorioniques de placentas obtenus de grossesses avec prééclampsie, ces réponses étaient modifiées puisque la réponse maximale à l’U-46619 était augmentée et celle à la 8-isoPGE2 diminuée. D’autre part, nous avons montré que les réponses maximales aux deux prostanoïdes étaient augmentées dans les vaisseaux placentaires de grossesse normale ou avec prééclampsie issus d’une délivrance prématurée par rapport à ceux d’une délivrance à terme. Ceci suggère une évolution de la réactivité des artères placentaires au cours du 3e trimestre de grossesse. En outre, les vaisseaux placentaires ont répondu aux prostanoïdes de façon semblable qu’ils aient été issus d’un accouchement vaginal ou d’une césarienne élective. Ceci indique que les prostanoïdes placentaires n’interviennent pas dans le processus de délivrance. D’un autre côté, l’utilisation de bloqueurs spécifiques des récepteurs TP à la TXA2, le SQ29,548 et l’ICI192,605, et des récepteurs EP à la prostaglandine E2, l’AH6809, nous ont permis de mettre en évidence le fait que l’U-46619 et la 8-isoPGE2 pouvaient agir de façon non-sélective sur l’un ou l’autre des récepteurs. Ces résultats supportent donc nos 2 premières hypothèses : les prostanoïdes font partie des molécules endogènes qui peuvent contrôler le tonus vasculaire placentaire et la prééclampsie modifie la réponse aux isoprostanes dans les artères chorioniques d’une manière compatible avec l’augmentation de la production de ces substances qui elle, est probablement le résultat du stress oxydatif. En revanche, en ce qui concerne les substances capables de jouer la contrepartie vasodilatatrice, l’utilisation d’un inhibiteur des synthases de monoxyde d’azote, le L-NAME, et celle d’inhibiteurs des COXs, l’ibuprofène, l’indométacine et le N-2PIA, ne nous a pas permis de mettre en évidence un quelconque rôle du monoxyde d’azote ou des prostanoïdes vasodilatatrices à ce niveau. Finalement, nous avons montré que l’induction d’un stress oxydatif dans les cotylédons perfusés in vitro et les artères chorioniques entraînait une vasoconstriction marquée. Celle-ci semble résulter de l’action des prostanoïdes puisqu’un blocage des récepteurs TP ou des COXs diminuait significativement la réponse maximale au peroxyde d’hydrogène. Les prostanoïdes impliquées dans la réponse au stress oxydatif proviendraient essentiellement d’une activation des COXs puisque l’étude ne nous permet pas de conclure à une quelconque implication des isoprostanes dans cette réponse. Ces observations confirment donc notre hypothèse que, dans le placenta, le stress oxydatif possède des propriétés vasoactives par activation du métabolisme de l’AA. En résumé, les résultats obtenus dans les placentas de grossesses normotensives et avec prééclampsie suggèrent que les prostanoïdes sont des molécules d’importance dans la régulation du tonus vasculaire placentaire. Le fait que la prééclampsie modifie la réponse aux prostanoïdes pourrait expliquer pourquoi la perfusion placentaire est altérée chez ces patientes. En outre, il apparaît évident qu’il existe un lien étroit entre le stress oxydatif et la voie de synthèse des prostanoïdes placentaires. Cependant d’autres études sont nécessaires pour mieux comprendre la nature de ce lien, qui pourrait, d’une certaine façon, jouer un rôle important dans le développement de la prééclampsie.