239 resultados para Activation C-H bond
Resumo:
The class B scavenger receptor CD36 is a component of the pattern recognition receptors on monocytes that recognizes a variety of molecules. CD36 expression in monocytes depends on exposure to soluble mediators. We demonstrate here that CD36 expression is induced in human monocytes following exposure to IL-13, a Th2 cytokine, via the peroxisome proliferator-activated receptor (PPAR)gamma pathway. Induction of CD36 protein was paralleled by an increase in CD36 mRNA. The PPARgamma pathway was demonstrated using transfection of a PPARgamma expression plasmid into the murine macrophage cell line RAW264.7, expressing very low levels of PPARgamma, and in peritoneal macrophages from PPARgamma-conditional null mice. We also show that CD36 induction by IL-13 via PPARgamma is dependent on phospholipase A2 activation and that IL-13 induces the production of endogenous 15-deoxy-Delta12,14-prostaglandin J2, an endogenous PPARgamma ligand, and its nuclear localization in human monocytes. Finally, we demonstrate that CD36 and PPARgamma are involved in IL-13-mediated phagocytosis of Plasmodium falciparum-parasitized erythrocytes. These results reveal a novel role for PPARgamma in the alternative activation of monocytes by IL-13, suggesting that endogenous PPARgamma ligands, produced by phospholipase A2 activation, could contribute to the biochemical and cellular functions of CD36.
Resumo:
Ag-experienced or memory T cells have increased reactivity to recall Ag, and can be distinguished from naive T cells by altered expression of surface markers such as CD44. Memory T cells have a high turnover rate, and CD8(+) memory T cells proliferate upon viral infection, in the presence of IFN-alphabeta and/or IL-15. In this study, we extend these findings by showing that activated NKT cells and superantigen-activated T cells induce extensive bystander proliferation of both CD8(+) and CD4(+) memory T cells. Moreover, proliferation of memory T cells can be induced by an IFN-alphabeta-independent, but IFN-gamma- or IL-12-dependent pathway. In these conditions of bystander activation, proliferating memory (CD44(high)) T cells do not derive from activation of naive (CD44(low)) T cells, but rather from bona fide memory CD44(high) T cells. Together, these data demonstrate that distinct pathways can induce bystander proliferation of memory T cells.
Resumo:
Upon infection with the protozoan parasite Leishmania major, susceptible BALB/c mice develop unhealing lesions associated with the maturation of CD4(+)Th2 cells secreting IL-4. In contrast, resistant C57BL/6 mice heal their lesions, because of expansion and secretion of IFN-gamma of CD4(+) Th1 cells. The Fas-FasL pathway, although not involved in Th cell differentiation, was reported to be necessary for complete resolution of lesions. We investigate here the role of IFN-gamma and IL-4 on Fas-FasL nonapoptotic signaling events leading to the modulation of macrophage activation. We show that addition of FasL and IFN-gamma to BMMø led to their increased activation, as reflected by enhanced secretion of TNF, IL-6, NO, and the induction of their microbicidal activity, resulting in the killing of intracellular L. major. In contrast, the presence of IL-4 decreased the synergy of IFN-gamma/FasL significantly on macrophage activation and the killing of intracellular L. major. These results show that FasL synergizes with IFN-gamma to activate macrophages and that the tight regulation by IFN-gamma and/or IL-4 of the nonapoptotic signaling events triggered by the Fas-FasL pathway affects significantly the activation of macrophages to a microbicidal state and may thus contribute to the pathogenesis of L. major infection.
Resumo:
RESUMESuite à un accident vasculaire cérébral (AVC) ischémique, les cellules gliales ducerveau deviennent activées, de nombreuses cellules inflammatoires pénètrent dans letissu lésé et sécrètent une grande variété de cytokines et chémokines. Aujourd'hui, ilexiste des interrogations sur les effets bénéfiques ou délétères de cette inflammation surla taille de la lésion et le pronostic neurologique.Ce projet vise à évaluer l'effet d'un peptide neuroprotecteur, D-JNKI1, inhibiteur de lavoie pro-apoptotique de signalisation intracellulaire c-Jun N-terminal kinase (JNK), surl'inflammation post-ischémique.Nous montrons d'abord que la microglie est largement activée dans toute la région lésée48 h après l'induction d'une ischémie chez la souris. Cependant, malgré l'inhibition dela mort neuronale par D-JNKI1 évaluée à 48 h, nous n'observons de modification ni del'activation de la microglie, ni de son nombre. Ensuite, nous montrons que le cerveaupeut être protégé même s'il y a une augmentation massive de la sécrétion de médiateursinflammatoires dans la circulation systémique très tôt après induction d'un AVCischémique. De plus, nous notons que la sécrétion de molécules inflammatoires dans lecerveau n'est pas différente entre les animaux traités par D-JNKI1 ou une solutionsaline, bien que nous ayons obtenu une neuroprotection significative chez les animauxtraités.En conclusion, nous montrons que l'inhibition de la voie de JNK par D-JNKI1n'influence pas directement l'inflammation post-ischémique. Ceci suggère quel'inhibition de l'inflammation n'est pas forcément nécessaire pour obtenir en hautdegré de neuroprotection du parenchyme lésé après ischémie cérébrale, et que lesmécanismes inflammatoires déclenchés lors d'une ischémie cérébrale ne sont pasforcément délétères pour la récupération du tissu endommagé.SUMMARYAfter cerebral ischemia, glial cells become activated and numerous inflammatory cellsinfiltrate the site of the lesion, secreting a large variety of cytokines and chemokines. Itis controversial whether this brain inflammation is detrimental or beneficial and how itinfluences lesion size and neurological outcome.This project was aimed at critically evaluating whether the neuroprotective peptide DJNKI,an inhibitor of the pro-apopotic c-Jun N-terminal kinase (JNK) pathway,modulates post-ischemic inflammation in animal models of stroke. Specifically, it wasasked whether JNK inhibition prevents microglial activation and the release ofinflammatory mediators.In the first part of this study, we showed that microglia was activated throughout thelesion 48 h after experimental stroke. However, the activation and accumulation ofmicroglia was not reduced by D-JNKI1, despite a significant reduction of the lesionsize. In the second part of this project, we demonstrated that neuroprotection measuredat 48 h occurs even though inflammatory mediators are released in the plasma veryearly after the onset of cerebral ischemia. Furthermore, we found that secretion ofinflammatory mediators in the brain was not different in groups treated with D-JNKI1or not, despite a significant reduction of the lesion size in the treated group.Altogether, we show that inhibition of the JNK pathway using D-JNKI1 does notinfluence directly post-stroke inflammation. Inhibition of inflammation is therefore notnecessarily required for neuroprotection after cerebral ischemia. Thus, post-strokeinflammation might not be detrimental for the tissue recovery.
Resumo:
NK cell function is regulated by a dual receptor system, which integrates signals from triggering receptors and MHC class I-specific inhibitory receptors. We show here that the src family kinase Fyn is required for efficient, NK cell-mediated lysis of target cells, which lack both self-MHC class I molecules and ligands for NKG2D, an activating NK cell receptor. In contrast, NK cell inhibition by the MHC class I-specific receptor Ly49A was independent of Fyn, suggesting that Fyn is specifically required for NK cell activation via non-MHC receptor(s). Compared to wild type, significantly fewer Fyn-deficient NK cells expressed the inhibitory Ly49A receptor. The presence of a transgenic Ly49A receptor together with its H-2(d) ligand strongly reduced the usage of endogenous Ly49 receptors in Fyn-deficient mice. These data suggest a model in which the repertoire of inhibitory Ly49 receptors is formed under the influenced of Fyn-dependent NK cell activation as well as the respective MHC class I environment. NK cells may acquire Ly49 receptors until they generate sufficient inhibitory signals to balance their activation levels. Such a process would ensure the induction of NK cell self-tolerance.
Resumo:
A BALB/c cloned T cell line directed against beef apo cytochrome c was shown to exhibit the Lyt-1+2- cell surface phenotype. The fine specificity of antigen recognition exhibited by the T cell clone was assessed by using a variety of peptide preparations obtained from cytochrome c of different sources. The peptide segment recognized by this T cell clone, in conjunction with I-A region gene products, appeared similar to that bound by a monoclonal antibody specific for beef apo cytochrome c derived from the same strain of mice.
Resumo:
Chromatin insulators are defined as transcriptionally neutral elements that prevent negative or positive influence from extending across chromatin to a promoter. Here we show that yeast subtelomeric anti-silencing regions behave as boundaries to telomere-driven silencing and also allow discontinuous propagation of silent chromatin. These two facets of insulator activity, boundary and silencing discontinuity, can be recapitulated by tethering various transcription activation domains to tandem sites on DNA. Importantly, we show that these insulator activities do not involve direct transcriptional activation of the reporter promoter. These findings predict that certain promoters behave as insulators and partition genomes in functionally independent domains.
Resumo:
OBJECTIVE: Blood-sucking arthropods' salivary glands contain a remarkable diversity of antihemostatics. The aim of the present study was to identify the unique salivary anticoagulant of the sand fly Lutzomyia longipalpis, which remained elusive for decades. METHODS AND RESULTS: Several L. longipalpis salivary proteins were expressed in human embryonic kidney 293 cells and screened for inhibition of blood coagulation. A novel 32.4-kDa molecule, named Lufaxin, was identified as a slow, tight, noncompetitive, and reversible inhibitor of factor Xa (FXa). Notably, Lufaxin's primary sequence does not share similarity to any physiological or salivary inhibitors of coagulation reported to date. Lufaxin is specific for FXa and does not interact with FX, Dansyl-Glu-Gly-Arg-FXa, or 15 other enzymes. In addition, Lufaxin blocks prothrombinase and increases both prothrombin time and activated partial thromboplastin time. Surface plasmon resonance experiments revealed that FXa binds Lufaxin with an equilibrium constant ≈3 nM, and isothermal titration calorimetry determined a stoichiometry of 1:1. Lufaxin also prevents protease-activated receptor 2 activation by FXa in the MDA-MB-231 cell line and abrogates edema formation triggered by injection of FXa in the paw of mice. Moreover, Lufaxin prevents FeCl(3)-induced carotid artery thrombus formation and prolongs activated partial thromboplastin time ex vivo, implying that it works as an anticoagulant in vivo. Finally, salivary gland of sand flies was found to inhibit FXa and to interact with the enzyme. CONCLUSIONS: Lufaxin belongs to a novel family of slow-tight FXa inhibitors, which display antithrombotic and anti-inflammatory activities. It is a useful tool to understand FXa structural features and its role in prohemostatic and proinflammatory events.
Resumo:
Summary : Platelet Derived Growth Factor (PDGF) and Transforming Growth Factor-ß (TGF-ß) are two crucial growth factors in tissue repair and regeneration. They control migration and proliferation of macrophages and fibroblasts, as well as myofibroblast differentiation and synthesis of the new connective tissue. The transcription factor Nuclear Factor I-C (NFI-C) has been implicated in the TGF-ß pathway and regulation of extracellular matrix proteins in vitro. This suggests a possible implication of NFI-C in tissue repair. In this study, our purpose was to identify the NFI-C target genes in TGF-ß1 pathway activation and define the relationship between these two factors in cutaneous wound healing process. High-throughput genomic analysis in wild-type and NFI-C knock-out embryonic fibroblasts indicated that NFI-C acts as a repressor of the expression of genes which transcriptional activity is enhanced by TGF-ß. Interestingly, we found an over representation of genes involved in connective tissue inflammation and repair. In accordance with the genomic analysis, NFI-C-/- mice showed an improvement of skin healing during the inflammatory stage. Analysis of this new phenotype indicated that the expression of PDGFA and PDGF-Ra genes were increased in the wounds of NFI-C-/- mice resulting in early recruitment of macrophages and fibroblasts in the granulation tissue. In correlation with the stimulation effect of TGF-ß on myofibroblast differentiation we found an increased differentiation of these cells in null mice, providing a rationale for rapid wound closure. Thus, in the absence of NFI-C, both TGF-ß and PDGF pathways may be activated, leading to enhanced healing process. Therefore, the inhibition of NFI-C expression could constitute a suitable therapy for healing improvement. In addition, we identified a delay of hair follicle cycle initiation in NFI-C-/- mice. This prompted us to investigate the role of NFI-C in skin appendage. The transition from a quiescent to a proliferative phase requires a perfect timing of signalling modulation, leading to stem cell activation. As a consequence of cycle initiation delay in null mice, the activation of signalling involved in cell proliferation was also retarded. Interestingly, at the crucial moment of cell fate determination, we identified a decrease of CD34 gene in mutant mice. Since CD34 protein is involved in migration of multipotent cells, we suggest that NFI-C may be involved in stem cell mobilisation required for hair follicle renewal. Further investigations of the role of NFI-C in progenitor cell activation will lead to a better understanding of tissue regeneration and raise the possibility of treating alopecia with NFI-C-targeting treatment. In summary, this study demonstrates new regenerative functions of NFI-C in adult mice, which regulates skin repair and hair follicle renewal. Résumé : PDGF et TGF-ß sont des facteurs important du mécanisme de défense immunitaire. Ils influencent la prolifération et migration des macrophages et des fibroblastes, ainsi que la différenciation des myofibroblastes et la formation du nouveau tissu conjonctif. Le facteur de transcription NFI-C a été impliqué dans la voie de signalisation de TGF-ß et dans 1a régulation de l'expression des protéines de la matrice extracellulaire in vitro. Ces études antérieures laissent supposer que NFI-C serait un facteur important du remodelage tissulaire. Cependant le rôle de NFI-C dans un tissu comme la peau n'a pas encore été étudié. Dans ce travail, le but a été de d'identifier la relation qu'il existe entre I~1FI-C et TGF-ßl à un niveau transcriptionnel et dans le processus de cicatrisation cutanée in vivo. Ainsi, une analyse génétique à grande échelle, a permis d'indiquer que NFI-C agit comme un répresseur sur l'expression des gènes dont l'activité transcriptionnelle est activée par TGF-ß. De plus nous avons identifié un groupe de gènes qui controlent le développement et l'inflammation du tissue conjonctif. En relation avec ce résultat, l'absence de NFI-C dans la peau induit une cicatrisation plus rapide pendant la phase inflammatoire. Durant cette période, nous avons montré que les expressions de PDGFA et PDGFRa seraient plus élevées en absence de NFI-C. En conséquence, l'activation de la voie de PDGF induit une infiltration plus importante des macrophages et fibroblastes dans le tissue granuleux des souris mutantes. De plus, en corrélation avec le rôle de TGF-ßl dans la différenciation des myofibroblasts, nous avons observé une différenciation plus importante de ces cellules chez les animaux knock-out, ce qui peut expliquer une contraction plus rapide de la plaie. De plus, nous avons découvert que NFI-C est impliqué dans l'initiation du cycle folliculaire. La caractérisation de ce nouveau phénotype a montré un ralentissement de la transition telogène-anagène des souris NFI-C-/-. Or, un événement clé de cette transition est la modulation de plusieurs signaux moléculaires aboutissant à' l'activation des cellules souches. En corrélation avec le decalage du cycle, l'activation de ces signaux est également décalée dans les souris NFI-C-/-. Ainsi, au commencement de l'anagène, la prolifération des keratinocytes,NFI-C-/- est retardée et corrèle avec une diminution de l'expression de CD34, une protéine responsable de la détermination du migration des cellules multipotentes. Ainsi, NFI-C semble être impliqué dans la mobilisation des cellules souches qui sont nécessaires au renouvellement folliculaire. En résumé, NFI-C est impliqué dans la régulation des signaux moléculaires nécessaires à la réparation tissulaire et son inhibition pourrait constituer un traitement de la cicatrisation. L'analyse de son rôle dans l'activation des cellules souches permettrait de mieux comprendre le renouvellement tissulaire et, à long terme, d'améliorer les techniques de greffe des cellules souches épithéliales ou consituter une cible pour le traitement de l'alopecie.
Resumo:
OBJECTIVE: To weight the rod-, cone-, and melanopsin-mediated activation of the retinal ganglion cells, which drive the pupil light reflex by varying the light stimulus wavelength, intensity, and duration. DESIGN: Experimental study. PARTICIPANTS: Forty-three subjects with normal eyes and 3 patients with neuroretinal visual loss. METHODS: A novel stimulus paradigm was developed using either a long wavelength (red) or short wavelength (blue) light given as a continuous Ganzfeld stimulus with stepwise increases over a 2 log-unit range. The pupillary movement before, during, and after the light stimulus was recorded in real time with an infrared illuminated video camera. MAIN OUTCOME MEASURES: The percent pupil contraction of the transient and sustained pupil response to a low- (1 cd/m(2)), medium- (10 cd/m(2)), and high-intensity (100 cd/m(2)) red- and blue-light stimulus was calculated for 1 eye of each subject. From the 43 normal eyes, median and 25th, 75th, 5th, and 95th percentile values were obtained for each stimulus condition. RESULTS: In normal eyes at lower intensities, blue light evoked much greater pupil responses compared with red light when matched for photopic luminance. The transient pupil contraction was generally greater than the sustained contraction, and this disparity was greatest at the lowest light intensity and least apparent with bright (100 cd/m(2)) blue light. A patient with primarily rod dysfunction (nonrecordable scotopic electroretinogram) showed significantly reduced pupil responses to blue light at lower intensities. A patient with achromatopsia and an almost normal visual field showed selective reduction of the pupil response to red-light stimulation. A patient with ganglion cell dysfunction owing to anterior ischemic optic neuropathy demonstrated global loss of pupil responses to red and blue light in the affected eye. CONCLUSIONS: Pupil responses that differ as a function of light intensity and wavelength support the hypothesis that selected stimulus conditions can produce pupil responses that reflect phototransduction primarily mediated by rods, cones, or melanopsin. Use of chromatic pupil responses may be a novel way to diagnose and monitor diseases affecting either the outer or inner retina.
Resumo:
MCT2 is the predominant neuronal monocarboxylate transporter allowing lactate use as an alternative energy substrate. It is suggested that MCT2 is upregulated to meet enhanced energy demands after modifications in synaptic transmission. Brain-derived neurotrophic factor (BDNF), a promoter of synaptic plasticity, significantly increased MCT2 protein expression in cultured cortical neurons (as shown by immunocytochemistry and western blot) through a translational regulation at the synaptic level. Brain-derived neurotrophic factor can cause translational activation through different signaling pathways. Western blot analyses showed that p44/p42 mitogen-activated protein kinase (MAPK), Akt, and S6 were strongly phosphorylated on BDNF treatment. To determine by which signal transduction pathway(s) BDNF mediates its upregulation of MCT2 protein expression, the effect of specific inhibitors for p38 MAPK, phosphoinositide 3-kinase (PI3K), mammalian target of rapamycin (mTOR), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK), p44/p42 MAPK (ERK), and Janus kinase 2 (JAK2) was evaluated. It could be observed that the BDNF-induced increase in MCT2 protein expression was almost completely blocked by all inhibitors, except for JAK2. These data indicate that BDNF induces an increase in neuronal MCT2 protein expression by a mechanism involving a concomitant stimulation of PI3K/Akt/mTOR/S6, p38 MAPK, and p44/p42 MAPK. Moreover, our observations suggest that changes in MCT2 expression could participate in the process of synaptic plasticity induced by BDNF.
Resumo:
Aeromonas hydrophila is a Gram-negative pathogen that causes serious infectious disease in humans. A. hydrophila induces apoptosis in infected macrophages, but the host proinflammatory responses triggered by macrophage death are largely unknown. Here, we demonstrate that the infection of mouse macrophages with A. hydrophila triggers the activation of caspase-1 and release of IL-1β. Caspase-1 activation was abrogated in macrophages deficient in Nod-like receptor family, pyrin domain containing 3 (NLRP3) and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), but not NLR family, CARD domain containing 4 (NLRC4). The activation of the NLRP3 inflammasome was mediated by three cytotoxins (aerolysin, hemolysin and multifunctional repeat-in-toxin) produced by A. hydrophila. Our results indicated that the NLRP3 inflammasome senses A. hydrophila infection through the action of bacterial cytotoxins.
Resumo:
The purpose of this study was to test melanoma vaccines consisting of peptides and immunological adjuvants for optimal immunogenicity and to evaluate laboratory immune monitoring for in vivo relevance. Forty-nine HLA-A2 positive patients with Melan-A positive melanoma were repeatedly vaccinated with Melan-A peptide, with or without immune adjuvant AS02B (QS21 and MPL) or IFA. Peptide-specific CD8 T cells in PBLs were analyzed ex vivo using fluorescent HLA-A2/Melan-A multimers and IFN-gamma ELISPOT assays. The vaccines were well tolerated. In vivo expansion of Melan-A-specific CD8 T cells was observed in 13 patients (1/12 after vaccination with peptide in AS02B and 12/17 after vaccination with peptide in IFA). The T cells produced IFN-gamma and downregulated CD45RA and CD28. T-cell responses correlated with inflammatory skin reactions at vaccine injection sites (P < 0.001) and with DTH reaction to Melan-A peptide (P < 0.01). Twenty-six of 32 evaluable patients showed progressive disease, whereas 4 patients had stable disease. The two patients with the strongest Melan-A-specific T-cell responses experienced regression of metastases in skin, lymph nodes, and lung. We conclude that repeated vaccination with Melan-A peptide in IFA frequently leads to sustained responses of specific CD8 T cells that are detectable ex vivo and correlate with inflammatory skin reactions.
Resumo:
CARMA1 is a lymphocyte-specific member of the membrane-associated guanylate kinase (MAGUK) family of scaffolding proteins, which coordinate signaling pathways emanating from the plasma membrane. CARMA1 interacts with Bcl10 via its caspase-recruitment domain (CARD). Here we investigated the role of CARMA1 in T cell activation and found that T cell receptor (TCR) stimulation induced a physical association of CARMA1 with the TCR and Bcl10. We found that CARMA1 was constitutively associated with lipid rafts, whereas cytoplasmic Bcl10 translocated into lipid rafts upon TCR engagement. A CARMA1 mutant, defective for Bcl10 binding, had a dominant-negative (DN) effect on TCR-induced NF-kappa B activation and IL-2 production and on the c-Jun NH(2)-terminal kinase (Jnk) pathway when the TCR was coengaged with CD28. Together, our data show that CARMA1 is a critical lipid raft-associated regulator of TCR-induced NF-kappa B activation and CD28 costimulation-dependent Jnk activation.
Resumo:
We have examined the effects of two agents depleting the intracellular pool of glutathione (GSH) on macrophage activation induced by IFN-gamma + LPS, as measured by nitrite production and leishmanicidal activity. Diethylmaleate (DEM), which depletes intracellular GSH by conjugation via a reaction catalyzed by the GSH-S-transferase, strongly inhibited nitrite secretion and leishmanicidal activity when added before or at the time of addition of IFN-gamma + LPS; this inhibition was progressively lost when addition of DEM was delayed up to 10 hr. A close correlation was observed between levels of intracellular soluble GSH during activation and nitrite secretion. Inhibition was partially reversed by the addition of glutathione ethyl ester (GSH-Et). Buthionine sulfoximine (BSO), a specific inhibitor of gamma-glutamylcysteine synthetase, also inhibited macrophage activation, although to a lesser extent than DEM despite a more pronounced soluble GSH depletion. This inhibition was completely reversed by the addition of GSH-Et. DEM and BSO did not alter cell viability or PMA-triggered O2- production by activated macrophages, suggesting that the inhibitory effects observed on nitrite secretion and leishmanicidal activity were not related to a general impairment of macrophage function. DEM and BSO treatment reduced iNOS specific activity and iNOS protein in cytosolic extracts. DEM also decreased iNOS mRNA expression while BSO had no effect. Although commonly used as a GSH-depleting agent, DEM may have additional effects because it can also act as a sulhydryl reagent; BSO, on the other hand, which depletes GSH by enzymatic inhibition, has no effect on protein-bound GSH. Our results suggest that both soluble and protein-bound GSH may be important for the induction of NO synthase in IFN-gamma + LPS-activated macrophages.