• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 36
  • 33
  • 11
  • 5
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 99
  • 23
  • 22
  • 20
  • 20
  • 19
  • 18
  • 16
  • 15
  • 12
  • 12
  • 12
  • 11
  • 11
  • 11
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
71

Initiation of Autoimmunity in Experimental Autoimmune Encephalomyelitis

Isaksson, Magnus January 2012 (has links)
The events that trigger an autoimmune disease remain largely unknown. To study these events animal models are necessary because symptoms of autoimmune diseases are preceded by a long asymptomatic period in humans. Experimental autoimmune encephalomyelitis (EAE) is the best characterized model for cell mediated autoimmunity and an animal model for the human disease multiple sclerosis. EAE is induced in rodents by immunization with myelin antigens (Ags) together with adjuvants. After immunization, T cells are primed in the periphery by Ag presenting cells and subsequently invade the central nervous system where they mediate parenchymal inflammation, resulting in demyelination and clinical symptoms of an ascending paralysis. It is now generally recognised that the main cell type mediating EAE is the T helper type 17 (Th17) cell. Tolerance to EAE can be attained by DNA vaccination, but how the immune response against the myelin Ags is abrogated after DNA vaccination is not known. By employing short interfering RNA technology, induction of the innate immune signalling molecule interferon (IFN) -β was found to be necessary for the protective effect of DNA vaccination in EAE. In addition, DNA vaccination inhibited subsequent autoimmune Th17 cell responses. The Toll-like receptors (TLRs) of the innate immune system have evolved to recognise conserved molecular structures on microbes and signalling through them almost exclusively converge on the molecule MyD88. Signalling via MyD88 was found to be required for induction of EAE since mice deficient in this molecule did not develop disease. Upstream signalling via TLR4 and TLR9 had tolerogenic properties. In studies of Ag presentation in EAE, two major subtypes of dendritic cells (DCs) were examined. Plasmacytoid DCs were found to have a promoting role in the induction of EAE, partly via type 1 IFNs. Myeloid DCs had a redundant role in the induction phase of EAE, neither disease severity nor encephalitogenic Th17 responses were affected by their absence during priming. These studies further demonstrate that the cells and molecules of the innate immune system exhibit a crucial role in controlling the adaptive immune system which mediates tissue damage in autoimmune diseases.
72

The Role of Natural Killer Cells and Interferon in Virus Infections: A Thesis

Bukowski, Jack F. 01 August 1984 (has links)
Definitive evidence that natural killer (NK) cells mediate an antiviral effect in vivo was obtained using murine cytomegalovirus (MCMV) as a model system. Adoptive transfer studies using a variety of physical and immunochemical techniques to enrich and deplete NK cell activity showed that the cell population capable of mediating resistance (as assayed by enhanced survival and reduction in spleen virus titers) had the phenotype of an NK cell: a nylon wool nonadherent, asialo GM1+, NK 1.2+, ly 5+, Thy-1-, Ia-, low-density lymphocyte. Adoptive transfer of IL-2-dependent cloned NK cells (but not T cells) also provided resistance. NK cells did not provide resistance to lymphocytic choriomeningitis virus (LCMV). Selective depletion of NK cell activity by injection of mice with antibody to anti-asialo GM1 lowered resistance to MCMV, mouse hepatitis virus, and vaccinia virus but not to LCMV. NK cell depletion resulted in up to 1000-fold increases in spleen and liver virus titers, correlating with more severe pathology in these organs. NK cells were found to have antiviral effects early (0-3 days) but not late (6-9 days) postinfection. NK cell depletion resulted in markedly increased MCMV-induced suppression of T cell function, which is probably responsible for the delayed clearance of virus seen in these mice. NK cell depletion resulted in increased virus synthesis during persistent MCMV infection, but had no effect on the course of persistent LCMV infection, despite elevated NK cell and interferon (IFN) levels found in these LCMV-infected mice. The reason why NK cells play a role against MCMV but not LCMV infection was not due to differences in NK cells induced by these 2 viruses, but more likely due to target cell susceptibility. IFN pretreatment of MCMV-infected cells failed to protect them against NK cell-mediated lysis, whereas uninfected and LCMV-infected cells were almost totally protected. These IFN-pretreated, LCMV-infected cells were not resistant to cell-mediated lysis in general, as this treatment increased their sensitivity to virus-specific T cell-mediated lysis by 2- to 3-fold. This enhanced sensitivity to lysis correlated with increased surface expression of H-2 antigens, but not viral antigens. In summary, these studies provide compelling evidence that NK cells can mediate antiviral effects in vivo, and provide some insights into their mode of action and consequences of their disfunction.
73

The Role of Type I Interferon in Vitiligo Pathogenesis and Melanoma Immunotherapy

Riding, Rebecca L. 05 March 2020 (has links)
Vitiligo is an autoimmune skin disease in which the pigment producing cells of the epidermis, melanocytes, are targeted for destruction by CD8+ T cells specific for melanocyte/melanoma-shared antigens. Previous work has identified IFNg as the central cytokine driving disease pathogenesis in both human patients and in our mouse model of vitiligo. IFNg signaling induces production of the chemokines CXCL9 and CXCL10, which trigger autoreactive T cell migration into the epidermis where effector T cells can target and destroy melanocytes. However, both IFNg and type I IFN signaling through activation of STAT1 proteins can induce transcription of the chemokines CXCL9 and CXCL10. Therefore, it seems reasonable that type I IFN signaling may also contribute to disease pathogenesis. The role of type I IFN in vitiligo is still unclear. Genome wide association studies identified multiple genes within the type I IFN pathway including TICAM1 and IFIH1 as susceptibility loci in vitiligo. One additional study reported increased epidermal staining of CD123, a marker expressed by pDCs, and the type I IFN induced gene MX1 in vitiligo patient skin. However, this study did not show any functional data to support the role of type I IFN signaling in vitiligo pathogenesis. Since the role of type I IFN in vitiligo is ill-defined, we used two different mouse models of vitiligo to functionally determine the role of type I IFN in disease by inducing vitiligo in hosts which lack the type I IFN receptor (IFNaR). In the first model, we induced vitiligo by adoptive transfer of melanocyte-specific CD8 T cells, which are activated in vivo by infection with recombinant vaccinia virus (VACV) expressing their cognate antigen. Vitiligo induction in IFNaR-deficient mice led to the development of severe disease compared to wild type mice. Acceleration and severity of disease was characterized by increased early recruitment of melanocyte-specific CD8 T cells to the skin, increased production of effector cytokines TNFa and IFNg, and reduced PD-1 expression. Increased production of IFNg by CD8 T cells in the skin of IFNaR-deficient mice led to increased expression of the chemokines CXCL9 and CXCL10 driving disease progression. IFNaR-deficient mice also displayed significantly increased VACV titters compared to wild type hosts. This data reveals a role of type I IFN in the clearance of recombinant VACV. This data also suggests that persistent VACV infection and prolonged antigen exposure in IFNaR deficient hosts is likely driving enhanced activation of melanocyte specific CD8 T cells and the subsequent development of severe vitiligo. Since melanocytes and melanoma cells express shared antigens that can be recognized by CD8 T cells, and because the development of vitiligo after melanoma immunotherapy is a positive prognostic factor for patients, we asked whether VACV vaccine therapy in IFNaR deficient mice would enhance the anti-tumor response to melanoma. B16-F10 inoculated wild type and IFNaR-deficient mice received adoptive transfer of melanocyte-specific CD8 T cells in combination with vaccinia virus expressing their cognate antigen to activate the cells in vivo. Treatment of adoptive T cell transfer and infection with VACV in IFNaR-deficient mice revealed significantly reduced tumor burden compared to wild type mice. Improved tumor regression in IFNaR-deficient hosts was characterized by increased infiltrating cytotoxic T lymphocytes and reduced PD-1 expression. These results further demonstrate that in the absence of type I IFN, hosts mount a robust cytotoxic CD8 T cell response against melanocyte/melanoma antigens and this is likely a result of persistent VACV that leads to prolonged CD8 T cell priming. As a result, IFNaR deficient hosts kill tumor cells more efficiently. To determine whether type I IFN regulates disease pathogenesis in the absence of virus infection, we generated a model of vitiligo in which bone marrow derived dendritic cells (BMDCs) pulsed with the cognate antigen were used to prime melanocyte-specific T cells in place of the viral vector. Induction of vitiligo in IFNaR-deficient hosts using BMDCs revealed no significant differences in disease score compared to wild type hosts. This data clearly demonstrates that type I IFN, in contrast to IFNg, is not required during the effector stage of vitiligo pathogenesis in mice. However, since we intentionally activate transferred melanocyte-specific CD8 T cells with VACV or BMDCs expressing their cognate antigen, our mouse models may circumvent the role of type I IFNs in initiating activation of autoreactive cells and driving autoimmunity. Type I IFN is critical for providing innate immune signals that drive the priming of autoreactive T cells through maturation of DCs by inducing antigen presentation, co-stimulatory molecule expression, and migration to the lymph nodes to encounter naïve T cells. Our mouse models of vitiligo may not capture this process. We have addressed this question by using a TLR ligand to activate BMDCs before transfer into hosts. In fact, activation of BMDCs before transfer leads to significantly enhanced vitiligo in mice and this is partially a result of type I IFN signaling on host cells. Thus, we provide evidence that type I IFNs can enhance the activation of melanocyte-specific CD8 T cells and drive autoimmunity. Collectively, our results show that type I IFN signaling has disparate effects on autoreactive T cell priming in a context dependent manner. We reveal that although type I IFN is not required for the effector phase of vitiligo in mice, maturation of DCs and subsequent type I IFN production can enhance the priming of autoreactive T cells and enhance vitiligo severity. Our studies also reveal that type I IFN is required to clear recombinant attenuated VACV infection and vaccine administration in IFNaR deficient hosts led to a robust autoreactive and anti-tumor response. These insights describing the role of type I IFN in autoimmunity and tumor immunology could have important implications for T cell dependent tumor immunotherapy.
74

Low dose of lipopolysaccharide protects mice from lethal paramyxovirus infection and post-viral airway disease

Resiliac, Jenny January 2022 (has links)
No description available.
75

Zellspezifische Funktionen des Typ 1 Interferonrezeptors bei der experimentellen autoimmunen Enzephalomyelitis / Cell specific functions of the type 1 interferon receptor during experimental autoimmune encephalomyelitis

Schmidt, Hauke 17 October 2007 (has links)
No description available.
76

Le rôle et la régulation du pyroglutamylated RF-amide peptide dans le tissu adipeux lors de l’obésité

Jossart, Christian 08 1900 (has links)
L’obésité est définie comme un surplus de masse adipeuse. Cette condition représente un problème de santé publique devenu pandémique dans les pays industrialisés. Elle prédispose à des maladies potentiellement mortelles comme le diabète de type 2, les maladies cardiovasculaires et la stéatose hépatique non-alcoolique. L’accumulation du tissu adipeux intra-abdominal, formé d’adipocytes, est corrélée avec la résistance à l’insuline. L’augmentation de la masse adipeuse se fait par l’hyperplasie des préadipocytes, la différenciation des préadipocytes en adipocytes et l’hypertrophie des adipocytes. La différenciation des préadipocytes se fait selon l’adipogenèse qui est régulée par une multitude de facteurs, mais qui est inhibée pas les stimuli inflammatoires qui sont aussi responsables de la résistance à l’insuline et de l’apparition des problèmes de santé liés à l’obésité. Nous avons identifié un nouveau système de régulation autocrine/paracrine de l’adipogenèse dans les cellules du tissu adipeux. Le pyroglutamylated RF-amide peptide (QRFP), qui était connu pour son rôle dans la régulation de l’appétit, est un activateur de l’adipogenèse par l’activation de son récepteur, le G protein-coupled receptor 103 (GPR103). Le QRFP est exprimé dans les macrophages et les adipocytes alors que le GPR103 de sous-type b est exprimé dans les adipocytes seulement. Un traitement des adipocytes avec le QRFP augmente le captage des acides gras, l’accumulation de lipides ainsi que l’expression et l’activité de l’enzyme LPL. Le QRFP augmente aussi l’expression des gènes des transporteurs d’acides gras CD36 et FATP1, de l’enzyme activatrice d’acides gras ACSL1 et des facteurs de transcription PPAR-γ et C/EBP-α, qui sont tous impliqués dans l’adipogenèse. En plus de ses effets sur l’adipogenèse, le QRFP possède aussi un effet inhibiteur sur l’activité lipolytique induite par les catécholamines. Nous avons montré que l’expression du QRFP est diminuée dans le tissu adipeux des souris obèses. Selon nos résultats, cette diminution pourrait être expliquée par une augmentation des endotoxines circulantes chez les obèses, appelée endotoxémie métabolique, qui agirait, entre autres, par l’induction des interférons dans les macrophages. Les voies de signalisation de ces effets ont aussi été identifiées. Nous avons montré un autre exemple de stimulus inflammatoire qui régule les signaux adipogènes à la baisse. / Obesity is defined as an excess of fat tissue mass. Obesity is a public health problem which became pandemic in developed countries. The condition of obesity predisposes to potentially fatal diseases like type 2 diabetes, cardiovascular diseases and non-alcoholic steatohepatitis. The increase in intra-abdominal adipose tissue mass is intimately associated with the development of insulin resistance. An increase in fat tissue mass occurs by preadipocytes hyperplasia, preadipocytes differentiation into adipocytes and adipocyte hypertrophy. The differentiation of preadipocytes occurs during adipogenesis and is regulated by multiple factors but inhibited by inflammatory stimuli that are responsible for insulin resistance and the emergence of obesity-related dysfunctions. We identified a new autocrine/paracrine system of regulation of adipogenesis in adipose tissue cells. The pyroglutamylated RF-amide peptide (QRFP), previously known for its role in the regulation of appetite, is an activator of adipogenesis by activating its receptor, G protein-coupled receptor 103 (GPR103). QRFP is expressed in adipocytes and macrophages whereas the GPR103 subtype b is expressed in adipocytes only. Treatment of adipocytes with QRFP increases fatty acids uptake, lipid accumulation, LPL enzyme expression and activity. QRFP upregulates gene expressions of fatty acids transporters CD36 and FATP1, of the fatty acid activating enzyme ACSL1 and of transcription factors PPAR-γ and C/EBP-α, which are all involved in adipogenesis. In addition to its effects on adipogenesis, QRFP shows an inhibitory effect on lipolytic activity induced by catecholamines. We have shown that QRFP expression is decreased in adipose tissues of obese mice. According to our results, this decrease could be explained by an increase of circulating endotoxins in obesity, called metabolic endotoxemia, which mediate its effect, in part, by the induction of interferons in macrophages. Signaling pathways of these effects have been identified. We demonstrated another example of inflammatory stimulus downregulating adipogenic signals.
77

Role of type I interferons in Streptococcus pneumoniae pneumonia

Koppe, Uwe Moritz Eberhard 25 June 2012 (has links)
Streptococcus pneumoniae ist die häufigste Ursache für ambulant erworbene Pneumonien weltweit. Daher müssen die Wirts-Pathogen-Interaktionen erforscht werden, um neue Therapiestrategien zu entwickeln. In dieser Studie habe ich 1. den Typ I Interferon (IFN)-stimulierenden Signalweg des angeborenen Immunsystems in Pneumokokken-infizierten Wirtszellen sowie 2. dessen Bedeutung in der Pneumokokkenpneumonie untersucht. Humane und murine Makrophagen, aber nicht alveolare Epithelzellen, produzierten Typ I IFNs nach Infektion mit S. pneumoniae. Dieses war abhängig vom Virulenzfaktor Pneumolysin und erforderte sowohl die Phagozytose der Bakterien als auch die Ansäuerung der Endosomen. Die Induktion der Typ I IFNs wird durch einen zytosolischen Signalweg vermittelt, welcher wahrscheinlich DNA erkennt und sowohl das Adapterprotein STING als auch den Transkriptionsfaktor IRF3 aktiviert. Typ I IFNs, welche von infizierten Makrophagen gebildet wurden, regulierten die Expression von IFN-stimulierten Genen (ISGs) und Chemokinen in Makrophagen und co-kultivierten alveolaren Epithelzellen in vitro und in Mauslungen in vivo. In einem murinen Pneumoniemodell hatten die Typ I IFNs jedoch einen negativen Effekt für den Wirt. Mäuse mit einem Defekt im Typ I IFN-Rezeptor oder mit einem Knockout im Typ I und Typ II IFN-Rezeptor hatten eine signifikant geringere Bakterienlast in der Lunge und eine verminderte Reduktion der Körpertemperatur und des Körpergewichtes als wild-typ Mäuse. Diese Effekte waren nicht durch Änderungen in der Zellrekrutierung oder durch Änderungen der Zytokin-/Chemokinexpression erklärbar. Zusammenfassend lässt sich feststellen, dass Typ I IFNs durch Pneumokokken induziert werden, aber dass sie trotz einiger positiver Effekte auf die Expression von ISGs einen negativen Gesamteffekt in einem murinen Pneumoniemodell aufweisen. Ein detailliertes Verständnis der Typ I IFN-Antwort während der Pneumokokkeninfektion kann die Entwicklung neuer Therapiestrategien unterstützen. / Streptococcus pneumoniae is the leading cause of community-acquired pneumonia world-wide. A detailed understanding of the host-pathogen interactions is required in order to foster the development of new therapeutic strategies. Here, I (I) characterized an innate immune recognition pathway that senses pneumococcal infection and triggers the production of type I interferons (IFNs), and (II) examined the role of type I IFNs in pneumococcal pneumonia in mice. Human and murine macrophages, but not alveolar epithelial cells, produced type I IFNs after infection with S. pneumoniae. This induction was dependent on the virulence factor pneumolysin, the phagocytosis of the bacteria, and the acidification of the endosome. Moreover, it appeared to be mediated by a cytosolic DNA-sensing pathway involving the adaptor molecule STING and the transcription factor IRF3. Type I IFNs produced by S. pneumoniae-infected macrophages positively regulated the expression of IFN-stimulated genes (ISGs) and chemokines in macrophages and co-cultured alveolar epithelial cells in vitro and in mouse lungs in vivo. However, in a murine model of pneumococcal pneumonia, type I IFN signaling was detrimental to the host defense. Mice deficient in the type I IFN signaling or double deficient in type I and type II IFN signaling had a significantly reduced bacterial load in the lung and a diminished reduction of body temperature and body weight compared to wild-type mice. The decreased susceptibility of the knockout mice was unlikely to be attributable to alterations in cell recruitment or cytokine/chemokine production. In conclusion, type I IFNs are induced during pneumococcal infection. However, despite their positive effects on the expression of some ISGs and chemokines, they negatively affect the outcome of pneumococcal pneumonia in an in vivo mouse model. Targeting the type I IFN system could potentially be an effective way in enhancing the immune response in patients with S. pneumoniae pneumonia.
78

Studies of interferon-inducible transmembrane proteins and interferons on DNA synthesis and proliferation in H9C2 cardiomyoblasts.

January 2006 (has links)
Lau Lai Yee. / Thesis (M.Phil.)--Chinese University of Hong Kong, 2006. / Includes bibliographical references (leaves 125-141). / Abstracts in English and Chinese. / Abstract --- p.i / 論文摘要 --- p.iii / Acknowledgement --- p.v / Table of Contents --- p.vii / List of Figures --- p.xii / List of Tables --- p.xiv / Abbreviations --- p.xvii / Chapter CHAPTER 1 --- INTRODUCTION / Chapter 1.1 --- Research initiative and significance --- p.1 / Chapter 1.2 --- Terminal differentiation --- p.4 / Chapter 1.3 --- Controversial terminal differentiation in cardiomyocytes --- p.5 / Chapter 1.4 --- Molecular switch from hyperplasia to hypertrophy in neonatal myocardial development --- p.7 / Chapter 1.5 --- Interferons --- p.8 / Chapter 1.6 --- Functions induced by interferons --- p.9 / Chapter 1.7 --- Interferons in cardiomyocytes --- p.12 / Chapter 1.8 --- Interferon-inducible transmembrane gene family --- p.13 / Chapter 1.9 --- Our hypothesis and objective --- p.16 / Chapter CHAPTER 2 --- MATERIALS AND METHODS / Chapter 2.1 --- Sequence analysis --- p.18 / Chapter 2.2 --- Cell culture --- p.18 / Chapter 2.3 --- Induction of differentiation of H9C2 cells --- p.19 / Chapter 2.4 --- In vitro induction of IFITMs by interferon treatments --- p.19 / Chapter 2.5 --- RNA isolation --- p.20 / Chapter 2.5.1 --- Experimental animals and sampling --- p.20 / Chapter 2.5.2 --- Total RNA Isolation --- p.20 / Chapter 2.5.3 --- RNA Quantification and Quality Check --- p.21 / Chapter 2.5.4 --- Purification by Qiagen-RNeasy Column and DNase I Digestion --- p.21 / Chapter 2.6 --- First-strand cDNA synthesis --- p.22 / Chapter 2.7 --- Quantitative real-time polymerase chain reaction --- p.22 / Chapter 2.8 --- Cloning protocol --- p.25 / Chapter 2.8.1 --- "Construction of pEGFP-IFITMl, pEGFP-IFITM2 and pEGFP-IFITM3 fusion proteins" --- p.25 / Chapter 2.8.1.1 --- Amplification of DNA fragments --- p.25 / Chapter 2.8.1.2 --- Purification of PCR product --- p.26 / Chapter 2.8.1.3 --- Restriction endonuclease digestion --- p.26 / Chapter 2.8.1.4 --- Insert/vector ligation --- p.27 / Chapter 2.8.1.5 --- Preparation of chemically competent bacterial cells --- p.27 / Chapter 2.8.1.6 --- Transformation of ligation product into chemically competent bacterial cells DH5a --- p.28 / Chapter 2.8.1.7 --- Recombinant clone screening by PCR --- p.29 / Chapter 2.8.1.8 --- Small-scale preparation of recombinant plasmid DNA --- p.29 / Chapter 2.8.1.9 --- Dideoxy DNA sequencing --- p.30 / Chapter 2.8.1.10 --- Large-scale preparation of recombinant plasmid DNA --- p.30 / Chapter 2.8.2 --- "Construction of IFITMl-pcDNA4, IFITM2-pcDNA4 and IFITM3- pcDNA4 constructs" --- p.33 / Chapter 2.8.2.1 --- Amplification of DNA fragments --- p.33 / Chapter 2.8.2.2 --- Insert/vector ligation --- p.33 / Chapter 2.8.2.3 --- Transformation of ligation product into one shot® TOP1 OF´ة chemically competent E. coli cells --- p.34 / Chapter 2.9 --- Transient transfection --- p.36 / Chapter 2.10 --- Subcellular fractionation --- p.37 / Chapter 2.11 --- Isolation of total protein cell lysate --- p.38 / Chapter 2.12 --- Protein concentration determination --- p.38 / Chapter 2.13 --- Protein gel electrophoresis and western blotting --- p.39 / Chapter 2.13.1 --- Preparation of SDS-polyacrylamide gel --- p.39 / Chapter 2.13.2 --- Preparation of protein samples --- p.39 / Chapter 2.13.3 --- SDS-polyacrylamide gel electrophoresis --- p.40 / Chapter 2.13.4 --- Protein transfer to nylon membrane --- p.40 / Chapter 2.13.5 --- Antibodies and detection --- p.40 / Chapter 2.13.6 --- Stripping membrane --- p.41 / Chapter 2.14 --- Bromodeoxyuridine proliferation assay --- p.42 / Chapter 2.14.1 --- Bromodeoxyuridine labeling and detection --- p.42 / Chapter 2.14.2 --- Cell number determination --- p.42 / Chapter 2.15 --- Fluorescence microscopy --- p.43 / Chapter 2.16 --- Confocal microscopy --- p.43 / Chapter 2.17 --- Statistical analysis --- p.44 / Chapter CHAPTER 3 --- RESULTS / Chapter 3.1 --- Sequence analysis --- p.45 / Chapter 3.1.1 --- Primary structure analysis --- p.45 / Chapter 3.1.2 --- Transmembrane he lice prediction --- p.46 / Chapter 3.1.3 --- Conserved domain prediction --- p.51 / Chapter 3.1.4 --- Sequence alignments across different species --- p.52 / Chapter 3.2 --- Differential expression during rat myocardial development --- p.53 / Chapter 3.3 --- Altered mRNA levels during differentiation of H9C2 cells --- p.55 / Chapter 3.4 --- "Cloning of IFITMl, IFITM2 and IFITM3" --- p.60 / Chapter 3.5 --- Subcellular localization --- p.61 / Chapter 3.5.1 --- Fluorescence microscopy --- p.61 / Chapter 3.5.2 --- Subcellular fractionation --- p.70 / Chapter 3.6 --- "In vitro induction by interferons-α, β and γ" --- p.72 / Chapter 3.7 --- "DNA synthesis after in vitro induction of interferons-α, β and γ" --- p.79 / Chapter 3.8 --- "Proliferating cell nuclear antigen expression after in vitro induction of interferons-α, β and γ" --- p.87 / Chapter 3.9 --- "DNA synthesis after overexpression of IFITM1, IFITM2 and IFITM3" --- p.93 / Chapter 3.10 --- "Proliferating cell nuclear antigen expression after overexpression of IFITM1, IFITM2 and IFITM3" --- p.95 / Chapter 3.11 --- "β-catenin and cyclin D1 expression after in vitro induction of interferons-α, β and γ" --- p.97 / Chapter 3.12 --- "β-catenin and cyclin D1 expression after overexpression of IFITMl, IFITM2 and IFITM3" --- p.101 / Chapter CHAPTER 4 --- DISCUSSION / Chapter 4.1 --- "Upregulation of IlFITMl, IFITM2 and IFITM3 during myocardial development" --- p.103 / Chapter 4.2 --- "Subcellular localization of IFITMl, IFITM2 and IFITM3" --- p.105 / Chapter 4.3 --- "Induction by interferons-α, β and γ" --- p.107 / Chapter 4.4 --- Inhibition of DNA synthesis by interferons-α and β and IFITM1 --- p.109 / Chapter 4.5 --- Involvement of IFITM family in canonical Wnt pathway --- p.112 / Chapter 4.6 --- Other possible pathways involved --- p.117 / Chapter CHAPTER 5 --- FUTURE PROSPECTS / Chapter 5.1 --- Production of antibodies --- p.118 / Chapter 5.2 --- Silencing or knockout approach --- p.118 / Chapter 5.3 --- Target genes of Wnt/β-catenin signaling --- p.119 / Chapter 5.4 --- Other signaling pathways involved --- p.119 / Chapter 5.5 --- Use of primary cardiomyocytes --- p.120 / APPENDIX --- p.121 / REFERENCES --- p.124
79

Étude des mécanismes de la réponse interféron de type I précoce et du contrôle à long terme de la virémie dans le modèle d’infection du macaque cynomolgus par le SIV : implications dans la physiopathologie du VIH / Deciphering early type I interferon response and long-term control of viremia in cynomolgus macaque model of AIDS : contribution to HIV pathogenesis

Bruel, Timothée 29 May 2013 (has links)
L’infection par le VIH induit une activation immunitaire chronique qui est suspectée d’être un des moteurs de la pathogénèse du SIDA. L’identification des mécanismes de contrôle de cette activation immunitaire, ainsi qu’une meilleure compréhension du contrôle spontané de l’infection chez certains patients, sont des étapes essentielles vers la conception de thérapies innovantes. Nous avons utilisé le modèle d’infection du macaque cynomolgus (Macaca fascicularis) par le virus de l’immunodéficience simienne (SIV) pour étudier ces deux points fondamentaux de la physiopathologie de l’infection.Il est proposé que l’induction précoce de l’activation immunitaire chronique soit une conséquence de la surexpression des gènes induits par les interférons (ISG) en réponse aux interférons de type I (IFN-I). Ces IFN-I ( et ) sont notamment produits par les cellules dendritiques plasmacytoïdes (pDC) en réponse aux virus. Notre premier objectif a été d’étudier la dynamique de production des IFN-I et celle des pDC durant l’infection, en parallèle de celle de l’activation immunitaire chronique et de la virémie. Nos résultats montrent que les pDC sont activées dans les tissus et qu’elles sont responsables de la production d’IFN-I observée transitoirement au cours de la primo-infection. La dynamique des pDC (apoptose, activation, renouvellement) entraîne un épuisement de la capacité des pDC à produire de l’IFN-I, qui pourrait rendre compte à la fois de l’altération fonctionnelle des pDC et de l’arrêt de la production d’IFN-I. De manière surprenante, ce contrôle de la production d’IFN-I n’est pas suivi d’un contrôle de la surexpression des ISG, ce qui suggère l’existence d’autres mécanismes inducteurs des ISG et souligne l’origine multifactorielle de l’activation immunitaire chronique.Dans une seconde étude, nous avons analysé l’impact d’une déplétion in vivo des lymphocytes exprimant le CD8 chez des animaux contrôlant spontanément la réplication virale sur le long terme. Quatre des cinq animaux de l’étude n’expriment pas de complexe majeur d’histocompatibilité (CMH) précédemment associé au contrôle, et aucun d’entre eux ne présente de forte réponse LT CD8. La déplétion transitoire des cellules CD8 entraîne chez quatre des contrôleurs une augmentation transitoire de la virémie qui se stabilise ensuite à des valeurs similaires aux niveaux pré-déplétion lors du retour des cellules CD8+. Un de ces animaux contrôle sa virémie avant la restauration des LT CD8. Chez le cinquième animal, la déplétion des CD8 n’a pas été accompagnée d’une élévation de virémie. Globalement, le contrôle de la virémie après l’élévation transitoire n’a pas été accompagné d’une augmentation de leur fonction antivirale. En revanche, une expansion et une activation des LT CD4 consécutive à la déplétion des CD8 ont été remarquées et corrélées positivement avec la virémie plasmatique. Ces résultats suggèrent que les réponses LT CD8 ne sont pas les principales responsables du contrôle à long terme de la virémie chez ces animaux. Dans notre modèle, d’autres mécanismes, tels qu’un réservoir de virus limité ou un meilleur contrôle de l’activation immunitaire, semblent participer à ce phénotype de contrôle.En conclusion, ces résultats éclairent la contribution des pDC, des IFN-I et des LT CD8 dans la physiopathologie du VIH, et permettent de proposer un nouveau modèle d’étude des mécanismes immunologiques précoces mis en place chez les patients contrôleurs de la virémie à long terme. / HIV infection induces a chronic immune activation, which is suspected to be a driving force in the pathogenesis of AIDS. Identifying control mechanisms of this immune activation, and a better understanding of the spontaneous control observed in some patients, are essential steps towards the development of innovative therapies. We used the model of cynomolgus macaques (Macaca fascicularis) infected by the simian immunodeficiency virus (SIV) to study these two fundamental fields of HIV pathogenesis.It is proposed that early induction of chronic immune activation is a consequence of an interferon-induced genes (ISG) overexpression in response to type I interferons (IFN-I). These I IFN (α and β) are preferentially produced by plasmacytoid dendritic cells (pDC) in response to the virus. Our first objective was to study the dynamics of pDC and IFN-I production during infection, together with chronic immune activation and viremia analysis. Our results indicate that pDCs are activated in tissues and are responsible for the transient IFN-I production observed during primary infection. The dynamics of pDCs (apoptosis, activation, renewal) induces an impaired IFN-I production by pDC, which could account both the functional defect of pDCs and the arrest of IFN-I production. Surprisingly, control of IFN-I production is not followed by down-regulation of ISG, which suggests the existence of other mechanisms that induce ISG and emphasizes the multifactorial origin of chronic immune activation.In a second study, we analyzed the impact of a in vivo CD8 T cells depletion in animals which spontaneously control viral replication in the long term. Importantly, four of the five animals in the study do not express major histocompatibility complex (MHC) previously associated with control, and none of them display strong CD8 response. The transient depletion of CD8 cells results in four controllers in a transient increase in viremia, which then stabilizes at values similar to pre-depletion levels when CD8+ cells come back. One of these animals controls their viremia before the restoration of CD8. In the fifth animal, CD8 depletion was not followed by a rise in viremia. Overall, the control of viremia after the transient increase was not associated to an increase of the antiviral function of CD8 T cells. In contrast, CD4 T cells expansion and activation were noticed and positively correlated to plasma viremia. These results suggest that CD8 responses are not the main cause of long-term control of viremia in these animals. In our model, other mechanisms, such as smaller reservoir or better control of immune activation, seem to be involved in this controller phenotype.In conclusion, these results shed light on the contribution of pDCs, IFN-I and CD8 T cells in the pathogenesis of HIV, and allow us to propose a new model for studying early immunological mechanisms in HIV controllers.
80

A Study On The Roles Of The Ras Activation Pathway During Interferonγ Mediated Functional Responses And Acetaminophen-induced Liver Injury In Mice

Saha, Banishree 05 1900 (has links)
Interferons (IFNs) perform a wide range of biological activities: anti-microbial, anti-proliferative, immunomodulatory etc. The IFN family includes three main classes: Type I, Type II and the recently identified Type III. The two main members of Type I class are IFNα and IFNβ, which are well known for their anti-viral roles. IFNλ, a member of the Type III class of IFNs, also exhibits antiviral activity. IFNγ, also known as immune IFN, is a Type II IFN which is secreted, primarily, by activated T cells, NK cells and macrophages. IFNγ is a potent immunomodulator which plays important roles in host defense. The diverse functions of this cytokine are demonstrated in Ifnγ-/- mice which display increased sensitivity to several pathogens, high incidences of tumors, reduced inflammatory response etc. IFNγ binds to its cognate receptors, which consist of two subunits, IFNγ receptor (IFNGR) 1 and IFNGR2. IFNγ mediates its multifarious biological actions by activating the Janus activated kinase (Jak)-Signal transducer and activator of transcription (Stat) 1 signaling pathway. Jaks belong to a family of non-receptor protein tyrosine kinases and phosphorylate the IFNγ receptor and the transcriptional co-activator, Stat. IFNGR1, the larger subunit, is required for ligand binding and its carboxyl terminus is involved in binding to Jak1, which in turn phosphorylates Stat1. The smaller subunit, IFNGR2, is required for signaling and contains the Jak2 binding site. After binding of IFNγ to its receptor, a series of phosphorylation events occur, resulting in Stat1 phosphorylation and homodimerization of Stat1 to form the gamma activating factor (GAF). These activated molecules translocate to the nucleus and bind to gamma activating sequence (GAS) present in the promoters of several IFNγ-modulated genes. Thus, the cellular responses mediated by IFNγ are, primarily, due to modulation of gene expression. Therefore, the identification and study of IFNγ stimulated genes, signaling mediators and their cross talk with other cellular pathways is an active area of research. The system of our study was a hepatoma cell line, H6, which is derived from a spontaneous tumor from B10.A mice and selected for in vitro cell culture. It is an IFNγ inducible system and has been used to study IFNγ-induced gene expression and functional responses. Treatment of H6 cells with IFNγ greatly enhanced MHC class I levels but also reduced cell growth. High amounts of reactive oxygen species (ROS) and reactive nitrogen intermediates (RNI) play crucial roles in the growth suppressive effect of IFNγ. To better understand the signaling pathways involved in the generation of ROS and RNI, the involvement of Ras was investigated. Ras-GTP levels were determined by pull down assays using GST-Raf1-Ras binding domain fusion protein bound to glutathione agarose. Ras activation (conversion of Ras-GDP to Ras-GTP) was observed in H6 cells upon IFNγ treatment by ~12 hr. To assess the functional role of Ras activation, studies with Manumycin A, a farnesyl transferase inhibitor (FTI), were performed. The formation of functional Ras requires farnesylation, a post-translational modification, which is inhibited by FTIs. Treatment with Manumycin A blocked Ras activation but did not significantly modulate the IFNγ-induced MHC class I. However, the inhibitor reduced ROS amounts leading to increased cell growth in the presence of IFNγ. Together, these results delineated the role of Ras and ROS in modulating some functions of IFNγ. To further understand the mechanisms by which Ras mediates its functions during IFNγ mediated growth suppression, the activation and function of Ras effectors was evaluated. In particular, the role of Ras-like (Ral) guanyl nucleotide-binding proteins, RalA and RalB, was investigated. IFNγ induced transcripts of RalA but not RalB. Also, the induction of RalA and IFNγ induced growth suppression were Stat1-dependent. Studies involving chemical inhibitors and genetic studies revealed that Ras played a role in the induction of RalA during IFNγ treatment. The role of c-Jun N-terminal kinase (JNK), a stress induced kinase, was also elucidated in this system. Together, IFNγ induced activation of Ras and its effectors RalA and JNK, leading to high amounts of ROS that suppressed cell growth. To evaluate the physiological significance of Ras activation during inflammatory responses, the mouse model of acetaminophen (APAP) induced liver injury was established. Hepatotoxicity due to overdose of the analgesic and antipyretic, APAP, is a major cause of liver failure in adults. APAP is metabolized into a reactive metabolite which binds to glutathione. Consequently, the depletion of intracellular glutathione stores leads to oxidative stress and liver injury. Notably, Ifnγ-/- mice are resistant to APAP-induced liver damage demonstrating a crucial role for this cytokine. The role of Ras activation was evaluated after oral dosing of BALB/c mice with APAP. Ras-GTP was induced early and decreased amounts were observed upon treatment with L-methionine, which replenished glutathione amounts. Injection with L-methionine or Manumycin A rescued liver injury as assessed by lowered serum alanine aminotransferase amounts and histological analysis. Kinetic studies were also performed, under different treatment conditions, to estimate different biochemical parameters: glutathione amounts, JNK activation, protein carbonylation, ROS amounts, serum amounts of cytokines, TNFα and IFNγ etc. This study reveals a role of Ras activation in stimulating proinflammatory responses and demonstrates the therapeutic efficacy of FTIs during APAP-induced liver injury. In addition the role of RalA during APAP-induced liver injury was also studied. In summary, this study, involving in vitro cell culture and in vivo liver injury model systems, sheds light on the significant contributions of Ras and its effector, RalA, during IFNγ mediated growth suppression and APAP-induced liver injury.

Page generated in 0.0477 seconds