115 resultados para OXIDE SYNTHASE EXPRESSION
Resumo:
The cellular response to an inflammatory stressor requires a proinflammatory cellular activation followed by a controlled resolution of the response to restore homeostasis. We hypothesized that biliverdin reductase (BVR) by binding biliverdin (BV) quells the cellular response to endotoxin-induced inflammation through phosphorylation of endothelial nitric oxide synthase (eNOS). The generated NO, in turn, nitrosylates BVR, leading to nuclear translocation where BVR binds to the Toll-like receptor-4 (TLR4) promoter at the Ap-1 sites to block transcription. We show in macrophages that BV-induced eNOS phosphorylation (Ser-1177) and NO production are mediated in part by Ca(2+)/calmodulin-dependent kinase kinase. Furthermore, we show that BVR is S-nitrosylated on one of three cysteines and that this posttranslational modification is required for BVR-mediated signaling. BV-induced nuclear translocation of BVR and inhibition of TLR4 expression is lost in macrophages derived from Enos(-/-) mice. In vivo in mice, BV provides protection from acute liver damage and is dependent on the availability of NO. Collectively, we elucidate a mechanism for BVR in regulating the inflammatory response to endotoxin that requires eNOS-derived NO and TLR4 signaling in macrophages.
Resumo:
Transcorneoscleral iontophoresis was used to enhance ocular penetration of a 21-bp NH(2) protected anti-NOSII oligonucleotides (ODNs) (fluorescein or infrared-41 labeled) in Lewis rats. Both histochemical localization and acrylamide sequencing gels were used. To evaluate the potential to down-regulate NOSII expression in the rat model of endotoxin-induced uveitis (EIU), anti-sense NOSII ODN, scrambled ODN or saline were iontophorezed into these animals' eyes. Iontophoresis facilitated the penetration of intact ODNs into the intraocular tissues of the rat eye and only the eyes receiving ODNs and electrical current demonstrated intact ODNs within the ocular tissues of both segments of the eye. Iontophoresis of anti-NOSII ODN significantly down-regulated the expression of NOSII expression in iris/ciliary body compared to the saline or scrambled ODN treated eyes. Nitrite production was also significantly reduced in the anti-NOSII applied eyes compared to those treated with saline. Using this system, intraocular delivery of ODNs can be significantly enhanced increasing the potential for successful gene therapy for human eye diseases.
Resumo:
Exercise is known to reduce cardiovascular risk. However, its role on atherosclerotic plaque stabilization is unknown. Apolipoprotein E(-/-) mice with vulnerable (2-kidney, 1-clip: angiotensin [Ang] II-dependent hypertension model) or stable atherosclerotic plaques (1-kidney, 1-clip: Ang II-independent hypertension model and normotensive shams) were used for experiments. Mice swam regularly for 5 weeks and were compared with sedentary controls. Exercised 2-kidney, 1-clip mice developed significantly more stable plaques (thinner fibrous cap, decreased media degeneration, layering, macrophage content, and increased smooth muscle cells) than sedentary controls. Exercise did not affect blood pressure. Conversely, swimming significantly reduced aortic Ang II type 1 receptor mRNA levels, whereas Ang II type 2 receptor expression remained unaffected. Sympathetic tone also significantly diminished in exercised 2-kidney, 1-clip mice compared with sedentary ones; renin and aldosterone levels tended to increase. Ang II type 1 downregulation was not accompanied by improved endothelial function, and no difference in balance among T-helper 1, T-helper 2, and T regulatory cells was observed between sedentary and exercised mice. These results show for the first time, in a mouse model of Ang II-mediated vulnerable plaques, that swimming prevents atherosclerosis progression and plaque vulnerability. This benefit is likely mediated by downregulating aortic Ang II type 1 receptor expression independent from any hemodynamic change. Ang II type 1 downregulation may protect the vessel wall from the Ang II proatherogenic effects. Moreover, data presented herein further emphasize the pivotal and blood pressure-independent role of Ang II in atherogenesis.
Resumo:
Adrenal chromaffin cells synthesize and secrete catecholamines and neuropeptides that may regulate hormonal and paracrine signaling in stress and also during inflammation. The aim of our work was to study the role of the cytokine interleukin-1beta (IL-1beta) on catecholamine release and synthesis from primary cell cultures of human adrenal chromaffin cells. The effect of IL-1beta on neuropeptide Y (NPY) release and the intracellular pathways involved in catecholamine release evoked by IL-1beta and NPY were also investigated. We observed that IL-1beta increases the release of NPY, norepinephrine (NE), and epinephrine (EP) from human chromaffin cells. Moreover, the immunoneutralization of released NPY inhibits catecholamine release evoked by IL-1beta. Moreover, IL-1beta regulates catecholamine synthesis as the inhibition of tyrosine hydroxylase decreases IL-1beta-evoked catecholamine release and the cytokine induces tyrosine hydroxylase Ser40 phosphorylation. Moreover, IL-1beta induces catecholamine release by a mitogen-activated protein kinase (MAPK)-dependent mechanism, and by nitric oxide synthase activation. Furthermore, MAPK, protein kinase C (PKC), protein kinase A (PKA), and nitric oxide (NO) production are involved in catecholamine release evoked by NPY. Using human chromaffin cells, our data suggest that IL-1beta, NPY, and nitric oxide (NO) may contribute to a regulatory loop between the immune and the adrenal systems, and this is relevant in pathological conditions such as infection, trauma, stress, or in hypertension.
Resumo:
Résumé: Les environnements hémodynamiques, favorisant ou protégeant contre la formation de la plaque, induisent tout deux une augmentation de la production d'anion superoxide dans les cellules endothéliales (ECs). Par ailleurs, une régulation différente de l'expression des gènes a été décrite dans les cellules exposées à ces différentes conditions. Dans le but d'investiguer le rôle de l'augmentation du stress oxydatif dans l'expression des gènes régulée par le flux, nous avons d'abord exposé les EC à un flux unidirectionnel, non pulsé. Dans ces conditions, l'état oxydatif des cellules endothéliales est augmenté de façon transitoire. L'expression du gène de l'endothéline 1 (ET-1) est aussi induite de façon transitoire par un tel flux, alors que l'expression du gène de la nitiric oxyde synthase endothéliale (NOS III) est stimulé de façon durable. Au contraire, un flux unidirectionnel pulsé, qui induit une augmentation durable de la production d'anion superoxide, augmente aussi de façon durable l'expression des gènes de ET-1 comme de NOS III. Un flux oscillatoire (favorisant la plaque), qui lui aussi ,a des effets à long terme sur la production d'anion superoxide, a uniquement augmenté l'expression de ET-1. De plus, l'utilisation d'un antioxydant, a seulement partiellement inhibé la stimulation de l'expression du gène NOS III par le flux unidirectionnel pulsé, alors qu'il a complètement abrogé la stimulation de l'expression du gène ET-1 par le flux unidirectionnel pulsé et oscillatoire. Ceci suggère que les forces mécaniques régulent l'expression des gènes dans les EC par un double mécanisme dépendant et indépendant du stress oxidatif des cellules. Par ailleurs, ces résultats supportent ultérieurement l'hypothèse que la balance entre la réponse oxidative et anti-oxidante dans les cellules endothéliales exposées à un environnement hémodynamique est une des clés de la prédisposition à un dysfonctionnement endothélial observé dans des régions exposées à des flux perturbés. Abstract: Both plaque-free and plaque-prone hemodynamic environments induce an increase in the oxidative state of endothelial cells (ECs), whereas differential gene expression regulation was described in cells exposed to these conditions. In order to investigate the role of the increased oxidative state in flow-regulation of gene expression, we first exposed EC to non-pulsed unidirectional shear stress. These conditions only slightly increases ECs oxidative state and endothelin-1 (ET-1) mRNA expression, whereas endothelial nitric oxide synthase (NOS III) mRNA level were significantly up-regulated. On the contrary, both ET-1 and NOS III gene expression were significantly induced in EC exposed to pulsed-unidirectional flow (plaque-free). Only ET-1 gene expression was up-regulated by oscillatory flow (plaque-prone). Moreover, use of an antioxidant only partially inhibited NOS III gene up-regulation by unidirectional flow, whereas it completely abrogated ET-1 gene up-regulation by unidirectional and oscillatory flows. Thus suggesting that mechanical forces regulate gene expression in ECs both via oxidative stress-dependent and -independent mechanisms.
Resumo:
RESUME GENERAL Au cours de ces dernières années, le monoxyde d'azote (NO) produit par une famille d'enzymes, les NO synthases (NOS), est apparu comme un effecteur central dans la régulation du système cardiovasculaire et du métabolisme énergétique. Chez l'homme, un défaut de production du NO est associé à des maladies cardiovasculaires et métaboliques comme la résistance à l'insuline ou le diabète de type 2. Ces pathologies se retrouvent chez les souris invalidées pour la NO synthase endothéliale (eN0S-/-) qui présentent non seulement une hypertension mais également une résistance à l'insuline et une dyslipidémie (augmentation des triglycérides et des acides gras libres). Ces anomalies sont étroitement associées et impliquées dans le développement du diabète de type 2. Dans cette étude, nous avons essayé de déterminer à partir du modèle de souris eN0S-/-, l'influence de la eNOS et de son produit, le NO, sur la régulation du métabolisme lipidique intracellulaire. Ainsi, nous avons montré que cette enzyme et le NO régulent directement l'activité β-oxydative des mitochondries isolées du muscle squelettique, du muscle cardiaque et du tissu adipeux blanc. Par ailleurs, dans le muscle de ces souris, le contenu des mitochondries et l'expression des gènes impliqués dans leur biogénèse sont diminués, ce qui suggère que la eNOS et/ou le NO contrôlent également la synthèse de ces organelles. Les mitochondries, via la β-oxydation, sont impliquées dans la production d'énergie à partir des acides gras libres. Dans notre modèle animal, la diminution de la β-oxydation dans le muscle, s'accompagne d'une accumulation des triglycérides intramyocellulaires. Cette accumulation prédispose fortement au développement de la résistance à l'insuline. Les anomalies du métabolisme β-oxydatif favorisent donc probablement l'apparition de la dyslipidémie et le développement de la résistance à l'insuline observées chez les souris eN0S-/-. Cette hypothèse est soutenue par différentes études effectuées chez l'homme et l'animal qui suggèrent qu'une dysfonction mitochondriale peut être à l'origine de la résistance à l'insuline. Ces données récentes et les résultats de ce travail apportent un regard nouveau sur le rôle du NO dans le développement des maladies métaboliques que sont la résistance à l'insuline, le diabète de type 2 et l'obésité. Elles placent aux centres de ces mécanismes une organelle, la mitochondrie, située au carrefour des métabolismes glucidiques et lipidiques. SUMMARY Over the last years, nitric oxide (NO), synthesized by a family of enzymes, the NO synthases, has become a central regulator of the cardiovascular system and energy metabolism. In humans, defective NO production is found in cardiovascular and metabolic diseases such as insulin resistance or type 2 diabetes mellitus. These alterations are also found in knockout mice for the endothelial nitric oxide synthase (eN0S-/-), which are not only hypertensive but also display insulin resistance and dyslipidemia (with increased triglyceride and free fatty acid levels). These pathologic features are tightly linked and involved in the pathogenesis of type 2 DM. In this study, using eN0S-/- mice, we determined the role played by this enzyme and its product, NO, on intracellular lipid metabolism. We show that eNOS and NO directly regulate β-oxidation in mitochondria isolated from skeletal and cardiac muscle as well as white adipose tissue. Furthermore, in the skeletal muscle of these mice, the mitochondrial content and the expression of genes involved in mitochondrial biogenesis are decreased, suggesting that eNOS and/or NO also regulate the synthesis of this intracellular organelle. Mitochondria, through β-oxidation, play a role in energy production from free fatty acids. In our animal model, decreased β-oxidation in skeletal muscle is associated with accumulation of intramyocellular lipids. This increased lipid content plays an important role in the pathogenesis of insulin resistance. Defective β-oxidation, therefore, probably favours the development of insulin resistance and dyslipidemia as seen in these animals. This hypothesis is strengthened by studies in humans and animals indicating that mitochondrial dysfunction is associated with insulin resistance. These recent data and the results of this work provide evidence for a role of NO in the development of metabolic diseases such as insulin resistance or type diabetes mellitus. They put as a central player, an organelle, the mitochondria, which lies at the crossway of carbohydrate and lipid metabolism. RESUME DIDACTIQUE Le maintien des fonctions vitales et l'accomplissement d'une activité physique nécessitent, chez l'homme, un apport quotidien d'énergie. Cette énergie est présente, dans l'alimentation, principalement sous forme de graisses (lipides) ou de sucres. La production d'énergie s'effectue en majorité dans le muscle au niveau d'une organelle particulière, la mitochondrie. La régulation du métabolisme énergétique fait intervenir de nombreux facteurs de régulation dont l'un des plus connu est l'insuline. De nombreuses maladies comme le diabète de type 2, l'obésité ou le syndrome métabolique découlent de la dérégulation du métabolisme énergétique. Un mécanisme particulier, la résistance à l'insuline, qui se caractérise par un défaut d'action de l'insuline au niveau de ses tissus cibles (foie, muscle...) est souvent impliqué dans le développement de ces pathologies. L'étude de ces anomalies métaboliques nécessite l'utilisation de modèles, notamment animaux, qui ont la particularité de reproduire partiellement un état pathologique caractéristique de certaines maladies humaines. Dans ce travail, nous avons utilisé un modèle de souris dont la particularité est de ne pas exprimer une enzyme, la monoxyde d'azote (NO) synthase endothéliale (eNOS), responsable de la synthèse d'un gaz, le NO. Ces souris présentent une hypertension artérielle, des anomalies du métabolisme des lipides et une résistance à l'insuline. Or, de récents travaux effectués chez l'homme montrent que des individus insulino-résistants ou diabétiques de type 2 ont une diminution de la production de NO. Lors de nos investigations, nous avons démontré que la quantité et la capacité des mitochondries à utiliser les lipides comme substrat énergétique est diminuée dans les muscles des souris eN0S-/-. Par ailleurs, ces deux anomalies sont associées dans ce tissu à une accumulation des lipides. De façon très intéressante, ce phénomène est décrit dans de nombreuses études effectuées chez l'homme et l'animal comme favorisant le développement de la résistance à l'insuline. Les résultats de ce travail suggèrent donc que la eNOS et/ou le NO joue un rôle important dans l'activité et la synthèse des mitochondries. Le NO pourrait donc constituer une cible thérapeutique dans le traitement des maladies métaboliques.
Resumo:
Doxorubicin (DOX) is a potent available antitumor agent; however, its clinical use is limited because of its cardiotoxicity. Cell death is a key component in DOX-induced cardiotoxicity, but its mechanisms are elusive. Here, we explore the role of superoxide, nitric oxide (NO), and peroxynitrite in DOX-induced cell death using both in vivo and in vitro models of cardiotoxicity. Western blot analysis, real-time PCR, immunohistochemistry, flow cytometry, fluorescent microscopy, and biochemical assays were used to determine the markers of apoptosis/necrosis and sources of NO and superoxide and their production. Left ventricular function was measured by a pressure-volume system. We demonstrated increases in myocardial apoptosis (caspase-3 cleavage/activity, cytochrome c release, and TUNEL), inducible NO synthase (iNOS) expression, mitochondrial superoxide generation, 3-nitrotyrosine (NT) formation, matrix metalloproteinase (MMP)-2/MMP-9 gene expression, poly(ADP-ribose) polymerase activation [without major changes in NAD(P)H oxidase isoform 1, NAD(P)H oxidase isoform 2, p22(phox), p40(phox), p47(phox), p67(phox), xanthine oxidase, endothelial NOS, and neuronal NOS expression] and decreases in myocardial contractility, catalase, and glutathione peroxidase activities 5 days after DOX treatment to mice. All these effects of DOX were markedly attenuated by peroxynitrite scavengers. Doxorubicin dose dependently increased mitochondrial superoxide and NT generation and apoptosis/necrosis in cardiac-derived H9c2 cells. DOX- or peroxynitrite-induced apoptosis/necrosis positively correlated with intracellular NT formation and could be abolished by peroxynitrite scavengers. DOX-induced cell death and NT formation were also attenuated by selective iNOS inhibitors or in iNOS knockout mice. Various NO donors when coadministered with DOX but not alone dramatically enhanced DOX-induced cell death with concomitant increased NT formation. DOX-induced cell death was also attenuated by cell-permeable SOD but not by cell-permeable catalase, the xanthine oxidase inhibitor allopurinol, or the NADPH oxidase inhibitors apocynine or diphenylene iodonium. Thus, peroxynitrite is a major trigger of DOX-induced cell death both in vivo and in vivo, and the modulation of the pathways leading to its generation or its effective neutralization can be of significant therapeutic benefit.
Resumo:
Nitric oxide synthase (NOS) is strongly and transiently expressed in the developing heart but its function is not well documented. This work examined the role, either protective or detrimental, that endogenous and exogenous NO could play in the functioning of the embryonic heart submitted to hypoxia and reoxygenation. Spontaneously beating hearts isolated from 4-day-old chick embryos were either homogenized to determine basal inducible NOS (iNOS) expression and activity or submitted to 30 min anoxia followed by 100 min reoxygenation. The chrono-, dromo- and inotropic responses to anoxia/reoxygenation were determined in the presence of NOS substrate (L-arginine 10 mM), NOS inhibitor L-NIO (1-5 mM), or NO donor (DETA NONOate 10-100 microM). Myocardial iNOS was detectable by immunoblotting and its activity was specifically decreased by 53% in the presence of 5 mM L-NIO. L-Arginine, L-NIO and DETA NONOate at 10 microM had no significant effect on the investigated functional parameters during anoxia/reoxygenation. However, irrespective of anoxia/reoxygenation, DETA NONOate at 100 microM decreased ventricular shortening velocity by about 70%, and reduced atrio-ventricular propagation by 23%. None of the used drugs affected atrial activity and hearts of all experimental groups fully recovered at the end of reoxygenation. These findings indicate that (1) by contrast with adult heart, endogenously released NO plays a minor role in the early response of the embryonic heart to reoxygenation, (2) exogenous NO has to be provided at high concentration to delay postanoxic functional recovery, and (3) sinoatrial pacemaker cells are the less responsive to NO.
Resumo:
Adaptive immune responses are initiated when T cells encounter antigen on dendritic cells (DC) in T zones of secondary lymphoid organs. T zones contain a 3-dimensional scaffold of fibroblastic reticular cells (FRC) but currently it is unclear how FRC influence T cell activation. Here we report that FRC lines and ex vivo FRC inhibit T cell proliferation but not differentiation. FRC share this feature with fibroblasts from non-lymphoid tissues as well as mesenchymal stromal cells. We identified FRC as strong source of nitric oxide (NO) thereby directly dampening T cell expansion as well as reducing the T cell priming capacity of DC. The expression of inducible nitric oxide synthase (iNOS) was up-regulated in a subset of FRC by both DC-signals as well as interferon-γ produced by primed CD8+ T cells. Importantly, iNOS expression was induced during viral infection in vivo in both LN FRC and DC. As a consequence, the primary T cell response was found to be exaggerated in Inos(-/-) mice. Our findings highlight that in addition to their established positive roles in T cell responses FRC and DC cooperate in a negative feedback loop to attenuate T cell expansion during acute inflammation.
Resumo:
Connexins (Cxs) and endothelial nitric oxide synthase (eNOS) contribute to the adaptation of endothelial and smooth muscle cells to hemodynamic changes. To decipher the in vivo interplay between these proteins, we studied Cx40-null mice, a model of renin-dependent hypertension which displays an altered endothelium-dependent relaxation of the aorta because of reduced eNOS levels. These mice, which were either untreated or subjected to the 1-kidney, 1-clip (1K1C) procedure, a model of volume-dependent hypertension, were compared with control mice submitted to either the 1K1C or the 2-kidney, 1-clip (2K1C) procedure, a model of renin-dependent hypertension. All operated mice became hypertensive and featured hypertrophy and altered Cx expression of the aorta. The combination of volume- and renin-dependent hypertension in Cx40-/- 1K1C mice raised blood pressure and cardiac weight index. Under these conditions, all aortas showed increased levels of Cx40 in endothelial cells and of both Cx37 and Cx45 in smooth muscle cells. In the wild-type 1K1C mice, the interactions between Cx40 and Cx37 with eNOS were enhanced, resulting in increased NO release. The Cx40-eNOS interaction could not be observed in mice lacking Cx40, which also featured decreased levels of eNOS. In these animals, the volume overload caused by the 1K1C procedure resulted in increased phosphorylation of eNOS and in a higher NO release. The findings provide evidence that Cx40 and Cx37 play an in vivo role in the regulation of eNOS.
Resumo:
To investigate the influence of glutathione (GSH) on cellular effects of nitric oxide (NO) formation, human colon adenocarcinoma cells were transfected with a vector allowing controlled expression of inducible nitric oxide synthase (iNOS). Protein levels of oxidative stress-sensitive heme oxygenase-1 (HO-1) were analyzed in the presence or absence of GSH depletion using L-buthionine-[S,R]-sulfoximine and iNOS induction. While no effect was observed in the presence of iNOS activity alone, a synergistic effect on HO-1 expression was observed in the presence of iNOS expression and GSH depletion. This effect was prevented by addition of N-methyl-L-arginine. Therefore, targeting of endogenous NO may be modulated by intracellular GSH.
Resumo:
Rationale: Peroxisome proliferator activated receptor (PPAR)-beta/delta is a transcription factor that belongs to the PPAR nuclear hormone receptor family, but the role of PPAR-beta/delta in sepsis is unknown. Objectives: We investigated the role of PPAR-beta/delta in murine models of LPS-induced organ injury and dysfunction and cecal ligation and puncture (CLP)-induced polymicrobial sepsis. Methods: Wild-type (WT) and PPAR-beta/delta knockout (1(0) mice and C57BL/6 mice were subjected to LPS for 16 hours. C57BL/6 mice received the PPAR-beta/delta agonist GW0742 (0.03 mg/kg intravenously, 1 h after LPS) or GW0742 plus the PPAR-beta/delta antagonist GSK0660 (0.1 mg/kg intravenously, 30 min before LPS). CD-1 mice subjected to CLP received GW0742 or GW0742 plus GSK0660. Measurements and Main Results: In PPAR-beta/delta KO mice, endotoxemia exacerbated organ injury and dysfunction (cardiac, renal, and hepatic) and inflammation (lung) compared with WT mice. In C57BL/6 mice subjected to endotoxemia, GW0742 significantly (1) attenuated organ (cardiac and renal) dysfunction and inflammation (lung); (2) increased the phosphorylation of Akt and glycogen synthase kinase (GSK)-3 beta; (3) attenuated the increase in extracellular signal-regulated kinase (ERK)1/2 and signal transducer and activator of transcription (STAT)-3 phosphorylation; and (4) attenuated the activation of nuclear factor (NF)-kappa B and the expression of inducible nitric oxide synthase (iNOS). In CD-1 mice subjected to CLP, GW0742 improved 10-day survival. All the observed beneficial effects of GW0742 were attenuated by the PPAR-beta/delta antagonist GSK0660. Conclusions: PPAR-beta/delta protects against multiple organ injury and dysfunction, and inflammation caused by endotoxic shock and improves survival in polymicrobial sepsis by a mechanism that may involve activation of Akt and inhibition of GSK-3 beta and NF-kappa B.
Resumo:
Insulin-dependent diabetes mellitus is an autoimmune disease in which pancreatic islet beta cells are destroyed by a combination of immunological and inflammatory mechanisms. In particular, cytokine-induced production of nitric oxide has been shown to correlate with beta cell apoptosis and/or inhibition of insulin secretion. In the present study, we investigated whether the interleukin (IL)-1beta intracellular signal transduction pathway could be blocked by overexpression of dominant negative forms of the IL-1 receptor interacting protein MyD88. We show that overexpression of the Toll domain or the lpr mutant of MyD88 in betaTc-Tet cells decreased nuclear factor kappaB (NF-kappaB) activation upon IL-1beta and IL-1beta/interferon (IFN)-gamma stimulation. Inducible nitric oxide synthase mRNA accumulation and nitrite production, which required the simultaneous presence of IL-1beta and IFN-gamma, were also suppressed by approximately 70%, and these cells were more resistant to cytokine-induced apoptosis as compared with parental cells. The decrease in glucose-stimulated insulin secretion induced by IL-1beta and IFN-gamma was however not prevented. This was because these dysfunctions were induced by IFN-gamma alone, which decreased cellular insulin content and stimulated insulin exocytosis. These results demonstrate that IL-1beta is involved in inducible nitric oxide synthase gene expression and induction of apoptosis in mouse beta cells but does not contribute to impaired glucose-stimulated insulin secretion. Furthermore, our data show that IL-1beta cellular actions can be blocked by expression of MyD88 dominant negative proteins and, finally, that cytokine-induced beta cell secretory dysfunctions are due to the action of IFN-gamma.
Resumo:
Upon agonist stimulation, endothelial cells trigger smooth muscle relaxation through the release of relaxing factors such as nitric oxide (NO). Endothelial cells of mouse aorta are interconnected by gap junctions made of connexin40 (Cx40) and connexin37 (Cx37), allowing the exchange of signaling molecules to coordinate their activity. Wild-type (Cx40(+/+)) and hypertensive Cx40-deficient mice (Cx40(-/-)), which also exhibit a marked decrease of Cx37 in the endothelium, were used to investigate the link between the expression of endothelial connexins (Cx40 and Cx37) and endothelial nitric oxide synthase (eNOS) expression and function in the mouse aorta. With the use of isometric tension measurements in aortic rings precontracted with U-46619, a stable thromboxane A(2) mimetic, we first demonstrate that ACh- and ATP-induced endothelium-dependent relaxations solely depend on NO release in both Cx40(+/+) and Cx40(-/-) mice, but are markedly weaker in Cx40(-/-) mice. Consistently, both basal and ACh- or ATP-induced NO production were decreased in the aorta of Cx40(-/-) mice. Altered relaxations and NO release from aorta of Cx40(-/-) mice were associated with lower expression levels of eNOS in the aortic endothelium of Cx40(-/-) mice. Using immunoprecipitation and in situ ligation assay, we further demonstrate that eNOS, Cx40, and Cx37 tightly interact with each other at intercellular junctions in the aortic endothelium of Cx40(+/+) mice, suggesting that the absence of Cx40 in association with altered Cx37 levels in endothelial cells from Cx40(-/-) mice participate to the decreased levels of eNOS. Altogether, our data suggest that the endothelial connexins may participate in the control of eNOS expression levels and function.
Resumo:
Adverse events in utero are associated with the occurrence of chronic diseases in adulthood. We previously demonstrated in mice that perinatal hypoxia resulted in altered pulmonary circulation in adulthood, with a decreased endothelium-dependent relaxation of pulmonary arteries, associated with long-term alterations in the nitric oxide (NO)/cyclic GMP pathway. The present study investigated whether inhaled NO (iNO) administered simultaneously to perinatal hypoxia could have potential beneficial effects on the adult pulmonary circulation. Indeed, iNO is the therapy of choice in humans presenting neonatal pulmonary hypertension. Long-term effects of neonatal iNO therapy on adult pulmonary circulation have not yet been investigated. Pregnant mice were placed in hypoxia (13% O2) with simultaneous administration of iNO 5 days before delivery until 5 days after birth. Pups were then raised in normoxia until adulthood. Perinatal iNO administration completely restored acetylcholine-induced relaxation, as well as endothelial nitric oxide synthase protein content, in isolated pulmonary arteries of adult mice born in hypoxia. Right ventricular hypertrophy observed in old mice born in hypoxia compared to controls was also prevented by perinatal iNO treatment. Therefore, simultaneous administration of iNO during perinatal hypoxic exposure seems able to prevent adverse effects of perinatal hypoxia on the adult pulmonary circulation.