898 resultados para B ACTIVATION
Resumo:
Viral infection triggers an early host response through activation of pattern recognition receptors, including Toll-like receptors (TLR). TLR signaling cascades induce production of type I interferons and proinflammatory cytokines involved in establishing an anti-viral state as well as in orchestrating ensuing adaptive immunity. To allow infection, replication, and persistence, (herpes)viruses employ ingenious strategies to evade host immunity. The human gamma-herpesvirus Epstein-Barr virus (EBV) is a large, enveloped DNA virus persistently carried by more than 90% of adults worldwide. It is the causative agent of infectious mononucleosis and is associated with several malignant tumors. EBV activates TLRs, including TLR2, TLR3, and TLR9. Interestingly, both the expression of and signaling by TLRs is attenuated during productive EBV infection. Ubiquitination plays an important role in regulating TLR signaling and is controlled by ubiquitin ligases and deubiquitinases (DUBs). The EBV genome encodes three proteins reported to exert in vitro deubiquitinase activity. Using active site-directed probes, we show that one of these putative DUBs, the conserved herpesvirus large tegument protein BPLF1, acts as a functional DUB in EBV-producing B cells. The BPLF1 enzyme is expressed during the late phase of lytic EBV infection and is incorporated into viral particles. The N-terminal part of the large BPLF1 protein contains the catalytic site for DUB activity and suppresses TLR-mediated activation of NF-κB at, or downstream of, the TRAF6 signaling intermediate. A catalytically inactive mutant of this EBV protein did not reduce NF-κB activation, indicating that DUB activity is essential for attenuating TLR signal transduction. Our combined results show that EBV employs deubiquitination of signaling intermediates in the TLR cascade as a mechanism to counteract innate anti-viral immunity of infected hosts.
Resumo:
Despite advancement in breast cancer treatment, 30% of patients with early breast cancers experience relapse with distant metastasis. It is a challenge to identify patients at risk for relapse; therefore, the identification of markers and therapeutic targets for metastatic breast cancers is imperative. Here, we identified DP103 as a biomarker and metastasis-driving oncogene in human breast cancers and determined that DP103 elevates matrix metallopeptidase 9 (MMP9) levels, which are associated with metastasis and invasion through activation of NF-κB. In turn, NF-κB signaling positively activated DP103 expression. Furthermore, DP103 enhanced TGF-β-activated kinase-1 (TAK1) phosphorylation of NF-κB-activating IκB kinase 2 (IKK2), leading to increased NF-κB activity. Reduction of DP103 expression in invasive breast cancer cells reduced phosphorylation of IKK2, abrogated NF-κB-mediated MMP9 expression, and impeded metastasis in a murine xenograft model. In breast cancer patient tissues, elevated levels of DP103 correlated with enhanced MMP9, reduced overall survival, and reduced survival after relapse. Together, these data indicate that a positive DP103/NF-κB feedback loop promotes constitutive NF-κB activation in invasive breast cancers and activation of this pathway is linked to cancer progression and the acquisition of chemotherapy resistance. Furthermore, our results suggest that DP103 has potential as a therapeutic target for breast cancer treatment.
Resumo:
Background: Cigarette smoke induces a pro-inflammatory response in airway epithelial cells but it is not clear which of the various chemicals contained within cigarette smoke (CS) should be regarded as predominantly responsible for these effects. We hypothesised that acrolein, nicotine and acetylaldehyde, important chemicals contained within volatile cigarette smoke in terms of inducing inflammation and causing addiction, have immunomodulatory effects in primary nasal epithelial cell cultures (PNECs).
Methods: PNECs from 19 healthy subjects were grown in submerged cultures and were incubated with acrolein, nicotine or acetylaldehyde prior to stimulation with Pseudomonas aeruginosa lipopolysaccharide (PA LPS). Experiments were repeated using cigarette smoke extract (CSE) for comparison. IL-8 was measured by ELISA, activation of NF-κB by ELISA and Western blotting, and caspase-3 activity by Western blotting. Apoptosis was evaluated using Annexin-V staining and the terminal transferase-mediated dUTP nick end-labeling (TUNEL) method.
Results: CSE was pro-inflammatory after a 24 h exposure and 42% of cells were apoptotic or necrotic after this exposure time. Acrolein was pro-inflammatory for the PNEC cultures (30 μM exposure for 4 h inducing a 2.0 fold increase in IL-8 release) and also increased IL-8 release after stimulation with PA LPS. In contrast, nicotine had anti-inflammatory properties (0.6 fold IL-8 release after 50 μM exposure to nicotine for 24 h), and acetylaldehyde was without effect. Acrolein and nicotine had cellular stimulatory and anti-inflammatory effects respectively, as determined by NF-κB activation. Both chemicals increased levels of cleaved caspase 3 and induced cell death.
Conclusions: Acrolein is pro-inflammatory and nicotine anti-inflammatory in PNEC cultures. CSE induces cell death predominantly by apoptotic mechanisms.
Resumo:
Tese de doutoramento, Farmácia (Biologia Celular e Molecular), Universidade de Lisboa, Faculdade de Farmácia, 2016
Resumo:
Les cellules gliales sont essentielles au fonctionnement du système nerveux. Dans la rétine, les cellules gliales de Müller assurent à la fois l’homéostasie du tissu et la protection des neurones, notamment celle des cellules ganglionnaires de la rétine (CGRs). L’hypothèse principale de la thèse est que les cellules de Müller joueraient un rôle primordial dans la survie neuronale tant au plan de la signalisation des neurotrophines/proneurotrophines par suite d’une blessure que lors des mécanismes d’excitotoxicité. Contrairement au brain-derived neurotrophic factor (BDNF), le nerve growth factor (NGF) n’est pas en mesure d’induire la survie des CGRs après une section du nerf optique. Le premier objectif de la thèse a donc été de localiser les récepteurs p75NTR et TrkA du NGF dans la rétine adulte et d’établir leur fonction respective en utilisant des ligands peptidomimétiques agonistes ou antagonistes spécifiques pour chacun des récepteurs. Nos résultats ont démontré que TrkA est surexprimé par les CGRs après l’axotomie, tandis que p75NTR est spécifiquement exprimé par les cellules de Müller. Alors que NGF n’est pas en mesure d’induire la survie des CGRs, l’activation spécifique de TrkA par des ligands peptidomimétique est nettement neuroprotectrice. De façon surprenante, le blocage sélectif de p75NTR ou l’absence de celui-ci protège les CGRs de la mort induite par l’axotomie. De plus, la combinaison de NGF avec l’antagoniste de p75NTR agit de façon synergique sur la survie des CGRS. Ces résultats révèlent un nouveau mécanisme par lequel le récepteur p75NTR exprimé par les cellules gliales de Müller peut grandement influencer la survie neuronale. Ensuite, nous avons voulu déterminer l’effet des proneurotrophines dans la rétine adulte. Nous avons démontré que l’injection de proNGF induit la mort des CGRs chez le rat et la souris par un mécanisme dépendant de p75NTR. L’expression de p75NTR étant exclusive aux cellules de Müller, nous avons testé l’hypothèse que le proNGF active une signalisation cellulaire non-autonome qui aboutit à la mort des CGRs. En suivant cette idée, nous avons montré que le proNGF induit une forte expression du tumor necrosis factor α (TNFα) dans les cellules de Müller et que l’inhibition du TNF bloque la mort neuronale induite par le proNGF. L’utilisation de souris knock-out pour la protéine p75NTR, son co-récepteur sortiline, ou la protéine adaptatrice NRAGE a permis de montrer que la production de TNF par les cellules gliales était dépendante de ces protéines. Le proNGF semble activer une signalisation cellulaire non-autonome qui cause la mort des neurones de façon dépendante du TNF in vivo. L’hypothèse centrale de l’excitotoxicité est que la stimulation excessive des récepteurs du glutamate sensibles au N-Methyl-D-Aspartate (NMDA) est dommageable pour les neurones et contribue à plusieurs maladies neurodégénératives. Les cellules gliales sont soupçonnées de contribuer à la mort neuronale par excitotoxicité, mais leur rôle précis est encore méconnu. Le dernier objectif de ma thèse était d’établir le rôle des cellules de Müller dans cette mort neuronale. Nos résultats ont démontré que l’injection de NMDA induit une activation du nuclear factor κB (NF-κB) dans les cellules de Müller, mais pas dans les CGRs, et que l’utilisation d’inhibiteurs du NF-κB empêche la mort des CGRs. De plus, nous avons montré que les cellules de Müller en réaction à l’activation du NF-κB produisent la protéine TNFα laquelle semble être directement impliquée dans la mort des CGRs par excitotoxicité. Cette mort cellulaire se produit principalement par l’augmentation à la surface des neurones des récepteurs AMPA perméables au Ca2+, un phénomène dépendant du TNFα. Ces donnés révèlent un nouveau mécanisme cellululaire non-autonome par lequel les cellules gliales peuvent exacerber la mort neuronale lors de la mise en jeu de mécanismes excitotoxiques.
Resumo:
L’ostéoarthrose (OA) est une maladie articulaire dont l’incidence augmente avec le vieillissement de la population. Elle se caractérise par une détérioration progressive du cartilage articulaire accompagnée du remodelage de l’os sous-chondral et du changement des tissus mous de l’articulation. La douleur et le dysfonctionnement de l’articulation affectée sont généralement attribués à l’inflammation et l’épanchement de la synovie. Plusieurs évidences indiquent que l’inflammation de la membrane synoviale contribue grandement à la pathogenèse de l’OA. En effet, la synthèse et l’expression des enzymes protéolytiques qui dégradent la matrice cartilagineuse sont régulées par de nombreuses cytokines retrouvées au sein de ce foyer inflammatoire. Deux d’entre elles, l’interleukine-1 beta (IL-1β) et le «tumor necrosis factor » alpha (TNF-α), jouent un rôle majeur dans le déclenchement de l’inflammation associée à l’OA. Ces cytokines pro-inflammatoires agissent notamment sur les synoviocytes et les chondrocytes en activant NF-κB qui, à son tour, active les gènes de cytokines. Cette boucle de régulation positive amplifie et perpétue la réponse inflammatoire. Récemment, il a été rapporté que l’activation de NF-κB par TNF-α peut être potentialisée par EXTL3, un récepteur transmembranaire ; mais le mécanisme sous-jacent de cet effet demeure inconnu. Toutefois, les niveaux important d’EXTL3 et de son ligand Reg1B chez les patients arthrosiques, laissent croire que ces protéines jouent un rôle dans le développement de l’OA. Notre objectif était d’étudier le mécanisme par lequel EXTL3 amplifie l’activation de NF-κB par TNF-α et d’examiner si ce phénomène se produit aussi avec l’IL-1β. Nous avons utilisé les cellules C28/I2, une lignée cellulaire de chondrocytes, comme modèle d’étude. Les transfections transitoires avec un vecteur d’expression, les techniques d’immunofluorescence (IF), d’immunoprécipitation (IP) et d’immunobuvardage de type Western (IB); ont été utilisées dans le cadre de diverses approches expérimentales. Les résultats obtenus par transfection ont révélé que la protéine EXTL3 potentialisait l’activation de NF-κB aussi bien par IL-1β que par TNF-α. Ce résultat signifie que la potentialisation de l’activité NF-κB par EXTL3 n’est pas spécifique à TNF-α. D’autre part, l’IP avec TNFRI et TRAF2 a révélé la présence d’EXTL3 dans le complexe TNF-α/TNFRI/TRAF2 qui se forme au niveau de la membrane plasmique. De plus, ceci a été confirmé in vivo par microscopie confocale montrant la co-localisation de TNFRI-TRAF2-EXTL3 dans la membrane nucléaire, suggérant ainsi la formation d’un complexe identique au niveau des membranes plasmique et nucléaires. Toutefois, la présence du ligand Reg1B et/ou de la glucosamine inhibait la formation de ce complexe au niveau de la membrane plasmique, tout comme ils abolissaient la potentialisation de l’activité NF-κB par EXTL3. Ces résultats suggèrent non seulement que le recrutement d’EXTL3 libre dans le complexe TNF-α/TNFR1 est requis pour amplifier l’activation de NF-κB par TNF-α, mais aussi la capacité du ligand Reg1B et de la glucosamine à moduler cette activation à travers la baisse ou l’inhibition de l’interaction EXTL3-TNFR1. Les données de cette étude constituent une avancée majeure dans la compréhension des événements moléculaires qui contrôlent l’activation de NF-κB par les cytokines pro-inflammatoires. Ces résultats pourraient conduire au développement de nouvelles approches thérapeutiques pour le traitement de l’inflammation associée à l’OA et impliquant une activation incessante de NF-κB.
Resumo:
El marcaje de proteínas con ubiquitina, conocido como ubiquitinación, cumple diferentes funciones que incluyen la regulación de varios procesos celulares, tales como: la degradación de proteínas por medio del proteosoma, la reparación del ADN, la señalización mediada por receptores de membrana, y la endocitosis, entre otras (1). Las moléculas de ubiquitina pueden ser removidas de sus sustratos gracias a la acción de un gran grupo de proteasas, llamadas enzimas deubiquitinizantes (DUBs) (2). Las DUBs son esenciales para la manutención de la homeostasis de la ubiquitina y para la regulación del estado de ubiquitinación de diferentes sustratos. El gran número y la diversidad de DUBs descritas refleja tanto su especificidad como su utilización para regular un amplio espectro de sustratos y vías celulares. Aunque muchas DUBs han sido estudiadas a profundidad, actualmente se desconocen los sustratos y las funciones biológicas de la mayoría de ellas. En este trabajo se investigaron las funciones de las DUBs: USP19, USP4 y UCH-L1. Utilizando varias técnicas de biología molecular y celular se encontró que: i) USP19 es regulada por las ubiquitin ligasas SIAH1 y SIAH2 ii) USP19 es importante para regular HIF-1α, un factor de transcripción clave en la respuesta celular a hipoxia, iii) USP4 interactúa con el proteosoma, iv) La quimera mCherry-UCH-L1 reproduce parcialmente los fenotipos que nuestro grupo ha descrito previamente al usar otros constructos de la misma enzima, y v) UCH-L1 promueve la internalización de la bacteria Yersinia pseudotuberculosis.
Resumo:
Background— T NADPH oxidase, by generating reactive oxygen species, is involved in the pathophysiology of many cardiovascular diseases and represents a therapeutic target for the development of novel drugs. A single-nucleotide polymorphism (SNP) C242T of the p22phox subunit of NADPH oxidase has been reported to be negatively associated with coronary heart disease (CHD) and may predict disease prevalence. However, the underlying mechanisms remain unknown. Methods and Results— Using computer molecular modelling we discovered that C242T SNP causes significant structural changes in the extracellular loop of p22phox and reduces its interaction stability with Nox2 subunit. Gene transfection of human pulmonary microvascular endothelial cells showed that C242T p22phox reduced significantly Nox2 expression but had no significant effect on basal endothelial O2.- production or the expression of Nox1 and Nox4. When cells were stimulated with TNFα (or high glucose), C242T p22phox inhibited significantly TNFα-induced Nox2 maturation, O2.- production, MAPK and NFκB activation and inflammation (all p<0.05). These C242T effects were further confirmed using p22phox shRNA engineered HeLa cells and Nox2-/- coronary microvascular endothelial cells. Clinical significance was investigated using saphenous vein segments from non CHD subjects after phlebectomies. TT (C242T) allele was common (prevalence of ~22%) and compared to CC, veins bearing TT allele had significantly lower levels of Nox2 expression and O2.- generation in response to high glucose challenge. Conclusions— C242T SNP causes p22phox structural changes that inhibit endothelial Nox2 activation and oxidative response to TNFα or high glucose stimulation. C242T SNP may represent a natural protective mechanism against inflammatory cardiovascular diseases.
Resumo:
The temporal organization of mammals presents a daily adjustment to the environmental light/dark cycle. The environmental light detected by the retina adjusts the central clock in the suprachiasmatic nuclei, which innervate the pineal gland through a polysynaptic pathway. During the night, this gland produces and releases the nocturnal hormone melatonin, which circulates throughout the whole body and adjusts several bodily functions according to the existence and duration of darkness. We have previously shown that during the time frame of an inflammatory response, pro-inflammatory cytokines, such as tumor necrosis factor-a, inhibit while anti-inflammatory mediators, such as glucocorticoids, enhance the synthesis of melatonin, interfering in the daily adjustment of the light/dark cycle. Therefore, injury disconnects the organism from environmental cycling, while recovery restores the light/dark information to the whole organism. Here, we extend these observations by evaluating the effect of a mild restraint stress, which did not induce macroscopic gastric lesions. After 2 h of restraint, there was an increase in circulating corticosterone, indicating activation of the hypothalamus-pituitary-adrenal (HPA) axis. In parallel, an increase in melatonin production was observed. Taking into account the data obtained with models of inflammation and stress, we reinforce the hypothesis that the activity of the pineal gland is modulated by the state of the immune system and the HPA axis, implicating the darkness hormone melatonin as a modulator of defense responses.
Resumo:
Interleukin-10 (IL-10) is an endogenous factor that restrains hepatic insulin resistance in diet-induced steatosis Reducing IL-10 expression increases proinflammatory activity in the steatotic liver and worsens insulin resistance As the transcriptional coactivator proliferator-activated receptor gamma coactivator-1 alpha (PGC-1 alpha) plays a central role in dysfunctional hepatocytic activity in diet-induced steatosis, we hypothesized that at least part of the action of PGC-1 alpha could be mediated by reducing the transcription of the IL-10 gene Here, we used immunoblotting, real-time polymerase chain reaction, immunocytochemistry, and chromatin immunoprecipitation assay to investigate the role of PGC-1 alpha in the control of IL-10 expression in hepatic cells First, we show that, in the intact steatotic liver, the expressions of IL-10 and PGC-1 alpha are increased Inhibiting PGC-1 alpha expression by antisense oligonucleotide increases IL-10 expression and reduces the steatotic phenotype. In cultured hepatocytes, the treatment with saturated and unsaturated fatty acids increased IL-10 expression. This was accompanied by increased association of PGC-1 alpha with c-Maf and p50-nuclear factor (NF) kappa B, 2 transcription factors known to modulate IL-10 expression In addition, after fatty acid treatment. PGC-1 alpha, c-Maf, and p50-NF kappa B migrate from the cytosol to the nuclei of hepatocytes and bind to the IL-10 promoter region Inhibiting NF kappa B activation with salicylate reduces IL-10 expression and the association of PGC-1 alpha with p50-NF kappa B Thus, PGC-1 alpha emerges as a potential transcriptional regulator of the inflammatory phenomenon taking place in the steatotic liver (C) 2010 Elsevier Inc All rights reserved
Resumo:
Short chain fatty acids (SCFAs) are fermentation products of anaerobic bacteria. More than just being an important energy source for intestinal epithelial cells, these compounds are modulators of leukocyte function and potential targets for the development of new drugs. The aim of this study was to evaluate the effects of SCFAs (acetate, propionate and butyrate) on production of nitric oxide (NO) and proinflammatory cytokines [tumor necrosis factor alpha (TNF-alpha) and cytokine-induced neutrophil chemoattractant-2 (CINC-2 alpha beta)] by rat neutrophils. The involvement of nuclear factor kappa B (NF-kappa B) and histone deacetylase (HDAC) was examined. The effect of butyrate was also investigated in vivo after oral administration of tributyrin (a pro-drug of butyrate). Propionate and butyrate diminished TNF-alpha, CINC-2 alpha beta and NO production by LPS-stimulated neutrophils. We also observed that these fatty acids inhibit HDAC activity and NF-kappa B activation, which might be involved in the attenuation of the LPS response. Products of cyclooxygenase and 5-lipoxygenase are not involved in the effects of SCFAs as indicated by the results obtained with the inhibitors of these enzymes. The recruitment of neutrophils to the peritonium after intraperitoneal administration of a glycogen solution (1%) and the ex vivo production of cytokines and NO by neutrophils were attenuated in rats that previously received tributyrin. These results argue that this triglyceride may be effective in the treatment of inflammatory conditions. Crown Copyright (C) 2011 Published by Elsevier Inc. All rights reserved.
Resumo:
Background and purpose: The inflammation-resolving lipid mediator resolvin E1 (RvE1) effectively stops inflammation-induced bone loss in vivo in experimental periodontitis. It was of interest to determine whether RvE1 has direct actions on osteoclast (OC) development and bone resorption. Experimental approach: Primary OC cultures derived from mouse bone marrow were treated with RvE1 and analysed for OC differentiation, cell survival and bone substrate resorption. Receptor binding was measured using radiolabelled RvE1. Nuclear factor (NF)-kappa B activation and Akt phosphorylation were determined with western blotting. Lipid mediator production was assessed with liquid chromatography tandem mass spectrometry. Key results: OC growth and resorption pit formation were markedly decreased in the presence of RvE1. OC differentiation was inhibited by RvE1 as demonstrated by decreased number of multinuclear OC, a delay in the time course of OC development and attenuation of receptor activator of NF-kappa B ligand-induced nuclear translocation of the p50 subunit of NF-kappa B. OC survival and apoptosis were not altered by RvE1. Messenger RNA for both receptors of RvE1, ChemR23 and BLT(1) is expressed in OC cultures. Leukotriene B(4) (LTB(4)) competed with [(3)H] RvE1 binding on OC cell membrane preparations, and the LTB(4) antagonist U75302 prevented RvE1 inhibition of OC growth, indicating that BLT(1) mediates RvE1 actions on OC. Primary OC synthesized the RvE1 precursor 18R-hydroxy-eicosapentaenoic acid and LTB(4). Co-incubation of OC with peripheral blood neutrophils resulted in transcellular RvE1 biosynthesis. Conclusions and implications: These results indicate that RvE1 inhibits OC growth and bone resorption by interfering with OC differentiation. The bone-sparing actions of RvE1 are in addition to inflammation resolution, a direct action in bone remodelling.
Resumo:
Background: Allergic lung inflammation is impaired in diabetic rats and is restored by insulin treatment. In the present study we investigated the effect of insulin on the signaling pathways triggered by allergic inflammation in the lung and the release of selected mediators. Methods: Diabetic male Wistar rats (alloxan, 42 mg/kg, i.v., 10 days) and matching controls were sensitized by s.c. injections of ovalbumin (OA) in aluminium hydroxide, 14 days before OA (1 mg/0.4 ml) or saline intratracheal challenge. A group of diabetic rats were treated with neutral protamine Hagedorn insulin (NPH, 4 IU, s.c.), 2 h before the OA challenge. Six hours after the challenge, bronchoalveolar lavage (BAL) was performed for mediator release and lung tissue was homogenized for Western blotting analysis of signaling pathways. Results: Relative to non-diabetic rats, the diabetic rats exhibited a significant reduction in OA-induced phosphorylation of the extracellular signal-regulated kinase (ERK, 59%), p38 (53%), protein kinase B (Akt, 46%), protein kinase C (PKC)-alpha (63%) and PKC-delta (38%) in lung homogenates following the antigen challenge. Activation of the NF-kappa B p65 subunit and phosphorylation of I kappa B alpha were almost suppressed in diabetic rats. Reduced expression of inducible nitric oxide synthase (iNOS, 32%) and cyclooxygenase-2 (COX-2, 46%) in the lung homogenates was also observed. The BAL concentration of prostaglandin (PG)-E(2), nitric oxide (NO) and interleukin (IL)-6 was reduced in diabetic rats (74%, 44% and 65%, respectively), whereas the cytokine-induced neutrophil chemoattractant (CINC)-2 concentration was not different from the control animals. Treatment of diabetic rats with insulin completely or partially restored all of these parameters. This protocol of insulin treatment only partially reduced the blood glucose levels. Conclusion: The data presented show that insulin regulates MAPK, PI3K, PKC and NF-kappa B pathways, the expression of the inducible enzymes iNOS and COX-2, and the levels of NO, PGE(2) and IL-6 in the early phase of allergic lung inflammation in diabetic rats. It is suggested that insulin is required for optimal transduction of the intracellular signals that follow allergic stimulation. Copyright (C) 2010 S. Karger AG, Basel
Resumo:
Diabetic individuals are more susceptible to infections and this seems to be related to impaired phagocyte function. Alveolar macrophages (AMs) are the first barrier to prevent respiratory infections Leukotrienes (LTs) increase AM phagocytic activity via Fc gamma R. In this study, we compared AMs from diabetic and nondiabetic rats for phagocytosis via Fc gamma R and the roles of LTs and insulin Diabetes was induced in male Wistar rats by alloxan (42 mg/kg, i.v); macrophages were obtained by bronchoalveolar lavage and IgG-opsonised sheep red blood cells (IgG-SRBC) were used as targets. LTs were added to the AMs 5 min before the addition of IgG-SRBC. AMs were treated with a LT synthesis inhibitor (zileuton, 10 mu M), or antagonists of the LTB(4) receptor (CP105 696, 10 mu M) cys-LT receptor (MK571, 10 mu M), 30 or 20 min before the addition of IgG-SRBC, respectively. We found that the phagocytosis of IgG-SRBC by AMs from diabetic rats is impaired compared with non-diabetic rats. Treatment with the LT inhibitor/antagonists significantly reduced AM phagocytosis in non-diabetic but not diabetic rats. During the phagocytosis of IgG-SRBC LTB(4) and LTC(4) were produced by AMs from both groups. The addition of exogenous LTB(4) or LTD(4) potentiated phagocytosis similarly in both groups Phagocytosis was followed by the phosphorylation of PKC-delta. ERK and Akt This was reduced by zileuton treatment in AMs from non-diabetic but not diabetic rats The addition of insulin to AMs further increased the phagocytosis by increasing PKC-delta phosphorylation These results suggest that the impaired phagocytosis found in AMs from diabetic rats is related to a deficient coupling of LTs to the Fc gamma R signaling cascade and that insulin has a key role in this coupling An essential role for insulin in Innate immunity is suggested (C) 2010 Elsevier Ltd. All rights reserved.
Resumo:
Considering that inflammation contributes to obesity-induced insulin resistance and that statins have been reported to have other effects beyond cholesterol lowering, the present study aimed to it whether atorvastatin treatment has anti-inflammatory action in white adipose tissue of obese mice, consequently improving insulin sensitivity. Insulin sensitivity in vivo (by insulin tolerance test); metabolic-hormonal profile; plasma tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, and adiponectin; adipose tissue immunohistochemistry; glucose transporter (GLUT) 4; adiponectin; INF-alpha; IL-1 beta; and IL-6 gene expression; and I kappa B kinase (IKK)-alpha/beta activity were assessed in 23-week-old monosodium glutamate induced obese mice untreated or treated with atorvastatin for 4 weeks. Insulin-resistant obese mice had increased plasma triglyceride, insulin, TNF-alpha, and IL-6 plasma levels. Adipose tissue of obese animals showed increased macrophage infiltration, IKK-alpha (42%, P < .05) and IKK-beta (73%, P < .05) phosphorylation, and INF-alpha and IL-6 messenger RNA (mRNA) (similar to 15%, P < .05) levels, and decreased GLUT4 mRNA and protein (30%, P < .05) levels. Atorvastatin treatment lowered cholesterol, triglyceride, insulin, INF-alpha, and IL-6 plasma levels, and restored whole-body insulin sensitivity. In adipose tissue, atorvastatin decreased macrophage in and normalized IKK-alpha/beta phosphorylation; INF-alpha, IL-6, and GLUT4 mRNA; and GLUT4 protein to control levels. The present findings demonstrate that atorvastatin has anti-inflammatory effects on adipose tissue of obese mice, which may be important to its local and whole-body insulin-sensitization effects. (C) 2010 Published by Elsevier Inc.