12

12

30 Abstract / Cytokine 70 (2014) 28–79 IL-2. In addition, IL-15 decreased the frequency of IL-2-producing CD4+ T cells, while increasing their abili...

57KB Sizes 2 Downloads 123 Views

30

Abstract / Cytokine 70 (2014) 28–79

IL-2. In addition, IL-15 decreased the frequency of IL-2-producing CD4+ T cells, while increasing their ability to secrete IFNc. These data suggest that hetIL-15 treatment limits Treg fitness through a concerted action of both CD8+ and CD4+ T cells, that results in an IL-2 deprived environment. Preclinical cancer studies support the use of hetIL-15 in tumor immunotherapy approaches to promote the development of anti-tumor responses by favoring effector over regulatory cells. http://dx.doi.org/10.1016/j.cyto.2014.07.015

9 RAGE is a cell surface nucleic acid sensor Damien Bertheloot 1, Cherilyn M. Sirois 2, Allison L. Miller 3, Tengchuan Jin 4, Natalio Garbi 5, T. Sam Xiao 4, Eicke Latz 1,6,7, 1 Institute of Innate Immunity, University of Bonn, Bonn, NRW, Germany, 2 Center for Translational Research, College of Health Sciences, Universidad de las Américas, Quito, Ecuador, 3 Division of Respiratory Inflammation & Autoimmunity, MedImmune LLC, Gaithersburg, MD, USA, 4 Structural Immunobiology Unit, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA, 5 Institutes of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, NRW, Germany, 6 Department of Medicine, Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA, 7 German Center for Neurodegenerative Diseases, University of Bonn, Bonn, NRW, Germany RAGE is a multi-ligand cell surface receptor of the Immunoglobulin super-family. It is composed of three extracellular immunoglobulin-like domains (V, C1 and C2), a single trans-membrane domain and a short cytoplasmic domain. RAGE is predominately expressed in the lung under normal conditions. However, during inflammation, RAGE expression increases dramatically in tissues like the vascular system, CNS and some hematopoietic cells. RAGE ligands are structurally heterogeneous and bind to RAGE through sequence nonspecific electrostatic interactions. They include advanced glycation end products, the DNA binding protein HMGB1, several proteins of the S100 family and certain types of b-sheet fibrils such as b-amyloid. Hence, RAGE is involved in many chronic inflammatory diseases such as diabetes, Alzheimer’s disease, asthma and systemic lupus erythematosus. Using EMSA and AlphaScreen, we showed that DNA and RNA bind to RAGE in a sequence nonspecific manner through electrostatic interaction of their phosphate backbone with the V and C1 domains. Human embryonic kidney (HEK) cells expressing RAGE bound more fluorescently labelled RNA and DNA than non-expressing cells as shown by microscopy and flow cytometry. In order to investigate the effect of RAGE expression on nucleic acid (NA) sensing by endosomal TLRs, we used HEK cells expressing TLR7, TLR8, TLR13 (ssRNA) or TLR9 (DNA) together with RAGE and found that RAGE increased NA induced activation of the transcription factor NFjB. Finally, in a mouse model of acute lung inflammation, DNA inhalation induced a strong inflammatory response in WT mice, while RAGE knockout mice displayed dramatically reduced inflammation. We show that RAGE binds NAs at the cell surface, increases their uptake and thereby amplifies the activation of endosomal TLRs. This work helps to further elucidate the function of RAGE in the development and perpetuation of chronic inflammation and presents RAGE as a promising target in the development of antiinflammatory treatments. http://dx.doi.org/10.1016/j.cyto.2014.07.016

10 Cell-restricted deletion of MyD88 affects tau pathology in hTau mouse model of tauopathy Kiran Bhaskar 1, Nicole Maphis 1, Zhizen Kang 2, Xiaoxia Li 2, Bruce T. Lamb 3, University of New Mexico, Albuquerque, NM, USA, 2 Immunology, Cleveland Clinic, Cleveland, OH, USA, 3 Neuroscience, Cleveland Clinic, Cleveland, OH, USA 1

Background: Tangle pathology (hyperphosphorylated and aggregation of tau) is a major neuropathological hallmark of Alzheimer’s disease (AD) and related tauopathies. Increasing evidence suggests that inflammatory processes in the brain precede tau pathology. A previous study from our group has demonstrated that enhancing neuroinflammation either via a Toll-Like Receptor-4 (TLR-4) ligand lipopolysaccharide (LPS) or genetic ablation of the Cx3cr1 (fractalkine receptor) led to increased tau phosphorylation, aggregation and working memory impairment in a manner dependent upon the activation of interleukin-1 receptor (IL-1R)-p38 mitogen activated protein kinase (p38 MAPK) pathway. Aims and methods: To block IL-1R and TLR-4 receptor signaling, we targeted and performed cell-specific deletion of MyD88 – a common adapter protein of IL-1 and TLR-4 receptors using Cre-LoxP technology.

Results and conclusions: First, in contrast to what was expected, microglia-restricted deletion of MyD88 (in CD11bCreMyD88f/fmice) led to enhanced AT8- and AT180-site tau phosphorylation. Notably, this effect was exacerbated upon LPS administration, suggesting the MyD88 may be required for homeostasis of tau. Second, infiltration of CD45 + peripheral monocytes was increased within the brains of CD11bCreMyD88f/f mice compared to MyD88f/f mice. Third, gene expression analysis comparing the CD11bCreMyD88f/f to MyD88f/f suggested alterations in key inflammatory cytokines. Finally, neuronal-restricted deletion of MyD88 in hTau mouse model of tauopathy (hTau-Mapt//CamKIIaCreMyD88f/f) showed drastic reduction in the levels of truncated neurotoxic tau in the cortex of hTauMapt//CamKIIaCreMyD88f/f mice compared to hTauMapt/ mice, suggesting neuron-restricted deletion of MyD88 is neuroprotective. Further studies on the characterization of truncated tau species and the effect of hTau on microglia-restricted (CD11b) deletion of MyD88 on tangle pathology and cognitive function are ongoing. http://dx.doi.org/10.1016/j.cyto.2014.07.017

11 Perinatal exposure to galactooligosaccharides/inulin prebiotics prevent food allergy by protecting intestine and promoting tolerance Gregory Bouchaud 1, Laure Castan 1, July Chabauty 1, Philippe Aubert 2, Michel Neunlist 2, Marie Bodinier 1, 1 BIA UR1268, INRA, Nantes, France, 2 INSERM UMR_S 913, Institut des Maladies de l’Appareil Digestif (IMAD), Nantes, France Aims: Food allergies are increasing, and no treatment exists, thus a strategy to prevent or reduce allergies would consist of modulating the host microbiota using either allochthonous bacteria (probiotics) or nondigestible food ingredients that can regulate the autochthonous microbiota (composition and metabolism), such as prebiotics. This study aimed to analyze the preventive effects of prebiotic feeding during perinatal and postweaning periods in a mouse model of food allergy. Method: In our model, mice were exposed to prebiotics (GOS/Inulin) either via their mother during pregnancy and breast feeding period only or also via food during postweaning period. Then, they were intraperitoneally sensitized to wheat proteins (modified gliadins) to induce a systemic response and then exposed orally to the same allergen. Results: Regardless of the exposure period to prebiotics, mice were protected against food allergy and display lower clinical score, body temperature variation and specific IgE and histamine levels compared to mice with normal diet. Moreover prebiotics intake either via mother alone or combine with post-weaning period reinforces intestinal permeability without affecting motility. Immunity was also modified by prebiotics supplementation in diet as shown by the increase of TGF-b secretion by splenocytes associated with the increase of regulatory T cells frequency in mesenteric lymph nodes and Peyer patches. A local decrease in IL-5 and IL-10 production together with an increase of IFN-c production is also observed in mice reflecting the immune modulatory effect of prebiotics during perinatal periods. Conclusion: Prebiotic supplemented diet during perinatal period protect against food allergy by decreasing levels of marker of allergy, clinical symptoms and intestinal permeability. Moreover, prebiotics also modulate immune reaction during allergy toward tolerance. http://dx.doi.org/10.1016/j.cyto.2014.07.018

12 Local IFN-I responses orchestrate cytokine responses and confer protection upon vaccinia virus infection Katharina Borst 1, Theresa Frenz 1, Stefan Lienenklaus 2, Mario Köster 2, Sukumar Namineni 3, Mathias Heikenwälder 3, Gerd Sutter 4, Ulrich Kalinke 1, 1 Twincore GmbH, Hanover, Germany, 2 Helmholtz Centre for Infection Research, Brunswick, Germany, 3 Technical University Munich, Munich, Germany, 4 Ludwig-Maximilians University, Munich, Germany Viruses express a wide range of immune evasion proteins to modulate host immunity. The DNA-encoded poxvirus vaccinia virus (VACV) carries IFN-I inhibitors of which the viral IFN-I receptor B18 binds to the surface of host cells to compete with the endogenous host IFN-I receptor. Upon VACV infection WT mice survive infection with mild signs of disease, while IFN-I is not detectable in the serum. Surprisingly, IFN-I receptor deficient (IFNAR/) mice succumb to VACV infection within 5 days. Thus, we hypothesized that upon VACV infection local IFN-I responses were induced which significantly affected the disease course. To monitor local IFN-I responses, we infected IFN-b-luciferase reporter mice with VACV. By in vivo imaging we found that upon VACV infection local IFN-b responses were induced within secondary lymphoid

Abstract / Cytokine 70 (2014) 28–79 organs. Studies with Mx2-luciferase reporter mice, which express luciferase upon IFNAR triggering, revealed significant IFNAR triggering within the liver and intermediate triggering in secondary lymphoid organs. Of note, viral titers in the liver of infected IFNAR/ mice were increased when compared to WT mice. Additionally, VACV infected IFNAR/ mice showed dramatically enhanced cytokine responses as well as elevated liver enzyme levels in the serum. Histological analysis of liver of VACV infected WT mice revealed that such mice developed mild hepatitis around day 5 post infection. In contrast, livers of infected IFNAR/ mice showed signs of severe hepatitis, massive influx of lymphocytes and inflammatory islets including acute necrotic areas. VACV infection of macrophage-specific IFNAR/ (LysMCre[wt/+]IFNARfl/fl) mice revealed that IFNAR signaling of macrophages was needed to protect mice from severe liver damage, whereas IFNAR signaling of hepatocytes (Alb-Cre[wt/+]IFNAR[fl/fl ]mice) was dispensable. Collectively our results indicated that locally induced IFN-I played a crucial role in balancing cytokine responses and were necessary to trigger macrophage-mediated liver protection.

31

phagoides farinae extract) a respiratory allergen without adjuvant to assess an impact on lung mucosa. Results: After food and respiratory allergen exposures, mice displayed stronger amount of blood markers: IgE specific and histamine. Moreover, splenocyte secretion of IL-4 and IL-17 were increased whereas weaker levels of IFNc were observed In parallel Peyer patches lymphocytes secreted higher amount of IL-4 with a decreased in IFNc, IL-10 and TGF-b productions. These mice exhibited intestine damages, higher paracellular flux and modification of transcellular permeability. In contrast, airway hyper-responsiveness, inflammatory cells and cytokines in lung remained unchanged compared to the respiratory allergy model. Conclusion: We show that dual exposure induces a raise in specific IgE and local Th2 and Th17 cytokines secretion before triggering phase. During the latter, gut morphology and functions were affected but not lung in dual exposed mice compared to single one underlying the organospecific impact. Altogether, our data make a step further in the elucidation of the mechanisms linking allergy history to immunological and clinical status potentially linked to atopic March development.

http://dx.doi.org/10.1016/j.cyto.2014.07.019 http://dx.doi.org/10.1016/j.cyto.2014.07.021

13 Crosstalk between interferon-beta, foam cell formation and inflammatory responses Marieke C.S. Boshuizen, Marten A. Hoeksema, Annette E. Neele, Saskia van der Velden, Sophie M.G. Pijnaker, Jan Van den Bossche, Menno P.J. de Winher, Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands Macrophage-derived foam cells are critical components of atherosclerotic lesions and the ways in which the inflammatory response of foam cells influences atherogenesis is of great interest. Previously we demonstrated that interferon-beta (IFN-bÞ promotes atherogenesis. But how IFN-b influences foam cell formation and inflammation is not understood yet. Hence, we assessed the functional involvement of IFN-b in these processes in a normal versus high cholesterol environment. First, we performed a microarray study on IFN-b-stimulated bone marrow-derived macrophages (BMDMS) under normocholesterolemic conditions, which showed upregulation of immune response pathways and downregulation of cholesterol biosynthesis. Secondly, we loaded BMDMs with acLDL followed by 6 h IFN-b treatment, which surprisingly impaired the induction of IFN-b target genes, like CCL5 and CXCL10. To validate these findings in vivo, LDLR/ mice were put on normal chow (NC) or a high cholesterol diet (HCD) for 10 weeks. Peritoneal macrophages (PEMs) were collected 4 days after intraperitoneal thioglycolate administration, combined with IFN-b (5000 U/ml) or PBS administration 24 and 8 h before sacrifice. Lipid loading increased following IFN-b treatment, accompanied by increased scavenger receptor-A (SR-A) gene expression. This lipid loading also resulted in PEM IFN-b-hyporesponsiveness, since several IFN-b target genes were again less expressed compared to NC PEMs. In addition, ex vivo culturing of PEMs from IFN-b-treated animals on HCD versus NC showed an overall decreased inflammatory activity, as gene expression of inflammatory markers was reduced and secretion of IL-6, TNF and NO was decreased. Currently we assess how HCD interferes with IFN-b-induced effects by investigating the IFN-b-induced activation of transcription factors like STAT1, IRF3 and IRF9 in control and acLDL loaded BMDMs. Altogether, IFN-b promotes foam cell formation, possibly by increased lipid influx via SR-A. Interestingly, increased lipid loading results in hyporesponsiveness to IFN-b. More research is needed to reveal the biological relevance for this hypercholesterolemia-induced hyporesponsive state. http://dx.doi.org/10.1016/j.cyto.2014.07.020

14 Previous food allergy aggravates allergic markers and intestinal damages in a mouse model of asthma Gregory Bouchaud 1, Pascal Gourbeyre 1, Tiphaine Bihouee 2, Philippe Aubert 3, David Lair 2, Maire-Aude Cheminant 2, Sandra Denery 1, Michel Neunlist 3, Antoine Magnan 2, Marie Bodinier 1, 1 BIA UR1268, INRA, Nantes, France, 2 INSERM UMR1087, CNRS UMR 6291, L’institut du thorax, Nantes, France, 3 INSERM UMR_S 913, Institut des Maladies de l’Appareil Digestif (IMAD), Nantes, France Aims: Increasing clinical data suggest a link between food allergy and the later development of respiratory allergy. This progression may be triggered by exposures to different allergens but the mechanism implicated remains unknown. This study aimed to identify the impact of a first exposure to food allergen on the development of a new form of allergy caused by exposure to a novel allergen using a mouse model. Method: In our model, mice were intraperitoneally sensitized to wheat proteins (modified gliadins) to induce a systemic response, then they were exposed orally to the same allergen and finally they were intranasally exposed to HDM (Dermato-

15 The role of cytokines in the cerebrospinal fluids of patients with Chronic Fatigue Syndrome/Myalgic Encephalomyelitis (CFS/ME) Sonya Marshall-Gradisnik 1, Gunnar Gottschalk 2, Sandra Ramos 1, Ekua Brenu 1, Don Staines 1, Dan Peterson 2, 1 Griffith University, Parklands, QLD, Australia, 2 Simmaron Research, Incline Village, NV, USA Objectives: Previous research has provided evidence for a dysregulation in cytokine levels in the periphery of patients with Chronic Fatigue Syndrome/Myalgic Encephalomyelitis (CFS/ME). To date few studies have examined cytokines in the cerebrospinal fluid. The purpose of this research is to examine the role of cytokines in the symptom presentation of CFS/ME patients. Methods: Cerebrospinal fluid (CSF) was collected from 18 CFS/ME patients and 5 healthy controls. The CSF samples were examined for the expression of 27 cytokines [interleukin (IL)-1b, IL-1ra, IL-2, IL-4, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12p70, IL-13, IL-15, IL-17, basic FGF, eotaxin, G-CSF, GM-CSF, IFN-c, IP-10, MCP-1 (MCAF), MIP-1a, MIP1b, PDGF-BB, RANTES, TNF-a and VEGF] using the bio-plex human cytokine 27-plex assay. Results: Of the cytokines examined, only four were significantly reduced in the CFS/ ME patients in comparison to the controls. Conclusions: The results show a decrease in pro-inflammatory cytokines in the CSF of CFS/ME patients and this may contribute to the clinical disease progression. http://dx.doi.org/10.1016/j.cyto.2014.07.022

16 Deregulated gp130/Stat3 signalling in lung cancer development Gavin D. Brooks, Saleela M Ruwanpura, Brendan J. Jenkins, Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research, Clayton, VIC, Australia Lung cancer (LC) is the most common and lethal cancer worldwide and, in Australia, accounts for 7500 deaths annually. A causal correlation between LC and cigarette smoking is well established, however only 10–15% of smokers develop LC, suggesting there are other ill-defined genetic, epigenetic and/or environmental factors which predispose individuals to LC. Regarding the former, interleukin (IL)-6 signals via the gp130 signal-transducing receptor subunit to primarily activate the latent transcription factor STAT3, as such, the pro-inflammatory and oncogenic properties of the IL-6/gp130/ STAT3 signalling axis can be established. Since IL-6 expression and STAT3 activity are up-regulated in human LC, we explored the downstream consequences of increased IL6/STAT3 activity in the well documented Kras[G12D] mouse LC model. For this purpose, we utilised gp130[F/F] (FF) mice that carry a knock-in mutation in gp130 leading to deregulated IL-6/STAT3 signalling, and crossed these mice with the LoxP-Stop-LoxP Kras[G12D] mice which harbour a conditionally-activated oncogenic Kras allele. Inhalation of FF:Kras[G12D] and WT:Kras[G12D] mice with a Cre recombinase adenovirus activated the Kras allele at 6 week of age, and mice were observed over 6 weeks. There was a severe and diffuse development of invasive adenocarcinoma in situ (AIS) in the lungs of FF:Kras[G12D] mice only. The percentage density of lesions in the lungs of FF:Kras[G12D] mice was increased compared to WT:Kras[G12D] littermate controls, and associated with an increase in the number of proliferative cells and inflammatory infiltrates (determined by immunohistochemistry). In contrast, partial suppression of deregulated IL-6/STAT3 signalling by crossing FF:Kras[G12D] mice onto an IL-6/ or Stat3[/+] background led to a recovery of normal lung tissue and a decrease of both proliferative and inflammatory cells.