• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 8
  • Tagged with
  • 12
  • 7
  • 6
  • 4
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 2
  • 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.
1

"Toll-Free" Pathways for Production of Type I Interferons

Wang, Ling, Ning, Shunbin 06 November 2017 (has links) (PDF)
Pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) are recognized by different cellular pathogen recognition receptors (PRRs), which are expressed on cell membrane or in the cytoplasm of cells of the innate immune system. Nucleic acids derived from pathogens or from certain cellular conditions represent a large category of PAMPs/DAMPs that trigger production of type I interferons (IFN-I) in addition to pro-inflammatory cytokines, by specifically binding to intracellular Toll-like receptors or cytosolic receptors. These cytosolic receptors, which are not related to TLRs and we call them "Toll-free" receptors, include the RNA-sensing RIG-I like receptors (RLRs), the DNA-sensing HIN200 family, and cGAS, amongst others. Viruses have evolved myriad strategies to evoke both host cellular and viral factors to evade IFN-I-mediated innate immune responses, to facilitate their infection, replication, and establishment of latency. This review outlines these "Toll-free" innate immune pathways and recent updates on their regulation, with focus on cellular and viral factors with enzyme activities.
2

Multimodal study of the interactions between the hepatitis B virus and the cyclic GMP-AMP synthase cGAS / Etude multimodale des interactions entre le virus de l’hépatite B et la cyclic AMP-GMP synthase, cGAS

Yim, Seung-Ae 12 September 2017 (has links)
Le virus de l’hépatite B (HBV) est l’agent étiologique de l’hépatite B. Ce virus est responsable d’hépatite chronique B, de cirrhose et de cancer du foie au niveau mondial. L’absence d’activation de la voie Interféron (IFN) suite à l’infection par HBV est encore mal comprise. Récemment, le senseur cellulaire cytosolic GMP-AMP synthase (cGAS) a été décrit comme un senseur efficace de DNA double brin possédant également une activité antivirale envers des virus à ADN et à ARN. Le but de mes travaux de thèse a été de contribuer à la compréhension des relations existants entre le HBV et cGAS, à des stades précoces et tardifs de l’infection HBV en utilisant des expériences de perte- et gain- de function ainsi que du profilage génomique des génes apparentés à cGAS dans un modéle cellulaire permissif au HBV. Mes travaux ont démontré (1) que cGAS exerce une forte activité antivirale envers le HBV incluant une réduction de la forme nucléaire du génome, le cccDNA; (2) alors que le rcDNA génomique nu est reconnu par la voie cGAS/STING et induit une réponse IFN efficace, la nucléocapside virale protège le DNA génomique viral et l’empêche d’être détecté par la réponse immunitaire innée; et (3) que l’infection par HBV diminue l’expression des acteurs de la voie cGAS-STING et des gènes impliqués dans la réponse immunitaire innée in vitro et in vivo. Ce dernier point met en lumière le rôle de cGAS dans un nouveau mécanisme d’échappement du HBV au système immunitaire inné dans les cellules hépatocytaires et dans ce mécanisme. / Chronic hepatitis B virus (HBV) infection is a major cause of liver disease and cancer worldwide. The mechanisms of viral genome sensing and the evasion of innate immune responses by HBV infection are still poorly understood. Recently, the cyclic GMP-AMP synthase (cGAS) was identified as a DNA sensor. In this PhD work, we aimed to investigate the functional role of cGAS in sensing of HBV infection and elucidate the mechanisms of viral evasion. We performed functional studies including loss- and gain-of-function experiments combined with cGAS effector gene expression profiling in an HBV infection-susceptible cell culture model. Collectively, our data show that (1) the cGAS-STING pathway exhibits robust antiviral activity against HBV infection including reduction of viral cccDNA levels; (2) naked HBV genomic rcDNA is sensed in a cGAS-dependent manner whereas packaging of the viral genome during infection abolishes host cell recognition of viral nucleic acids; (3) HBV infection down-regulates the cGAS/STING pathway actors as well as innate immune effector gene expression in vitro and vivo. Overall, this work led to describing new aspects of the complex interaction between HBV and the DNA sensor cGAS in hepatocytes.
3

Identification of viral and host mechanisms that determine innate immune activation by the DNA sensor cGAS / Identification des mécanismes viraux et hôtes qui déterminent l'activation de l’immunité innée par le senseur d'ADN cGAS

Gentili, Matteo 25 September 2017 (has links)
Les acides nucléiques sont des activateurs puissants des réponses immunitaires innées. Pour lutter contre les infections, les organismes multicellulaires ont développé de multiples façons de détecter les agents pathogènes. L'une d'entre elles est la reconnaissance de l'ADN dans le cytoplasme. La présence d'ADN dans le cytoplasme est généralement liée à des infections par des bactéries ou des virus comme le VIH. La GMP-AMP synthase cyclique (cGAS) est un capteur cytosolique essentiel de l'ADN chez les mammifères. Lors de la reconnaissance de l'ADN, cGAS synthétise le petit second messager cyclique GMP-AMP (cGAMP), qui active les voies de signalisation de l’immunité innée. De plus, cGAS joue un rôle central en réponse aux microbes et au développement de maladies auto-immunes comme les interféronopathies. Cette thèse examine le rôle de cGAS en réponse aux virus et à l’ADN du soi. En explorant la réponse médiée par cGAS en présence du VIH, nous avons constaté que dans les cellules produisant du virus, le cGAMP synthétisé par cGAS est contenu dans des particules virales et des vésicules extracellulaires. Les particules virales délivrent efficacement cGAMP aux cellules cibles et activent une réponse antivirale puissante. Le transfert de cGAMP nécessite une activité catalytique de cGAS et une fusion des particules virales avec les cellules cibles. Nous avons également montré que dans le contexte d'une infection, les virus à ADN tels que MVA et mCMV peuvent conditionner cGAMP. Par conséquent, le transfert de cGAMP médié par une infection virale est un mécanisme de défense généralisé du système immunitaire inné. Nous avons également étudié l'activation du cGAS par l'ADN en se concentrant sur l'ADN de la chromatine nucléaire. Dans les cellules eucaryotes, l'ADN est compartimenté dans deux organelles: le noyau et les mitochondries. La compartimentation nucléaire de l'ADN peut être transitoirement perdue lors de la rupture de l'enveloppe nucléaire pendant la division cellulaire et lors des ruptures d'enveloppes nucléaires dans les cellules dendritiques migrantes (DC). Nous avons constaté que la rupture de l'enveloppe nucléaire ou la perte de l'intégrité de l'enveloppe nucléaire entraînent un recrutement de cGAS sur l'ADN nucléaire. Nous avons également montré que les DC présentent un pool nucléaire de cGAS à l'état basal. La rupture de l'enveloppe nucléaire pendant le cycle cellulaire conduit au recrutement de cGAS à l'ADN nucléaire. En jouant sur la localisation cGAS, nous avons montré que le cGAS est peu actif lorsqu'il se trouve dans le noyau et la transfection d'ADN exogène permet son activité enzymatique complète. L'expression du cGAS localisé dans le noyau des DC a conduit à la maturation des DC et à la production d'interféron de type I (IFN). La région N-terminale de cGAS régule la distribution nucléaire / cytoplasmique de cGAS dans les cellules en interphases et nous avons établi que les lamines A / C sont requises pour l'accès de cGAS à l'ADN nucléaire dans les DC migrantes. De façon inattendue, nous montrons que cGAS n'est pas activé lors de la rupture de l'enveloppe nucléaire. Enfin, nous identifions l'hétérochromatine pericentromérique comme étant l'ADN de liaison préférentielle pour le cGAS nucléaire exprimé dans les DC. Au total, notre étude montre que le noyau agit comme un site intracellulaire immunitaire privilégié pour l'activation de cGAS. Collectivement, nous avons montré une pertinence de l'activation de cGAS lors d'une infection virale et découvert un mécanisme « cheval de Troie » de l'incorporation de cGAMP dans la progénie virale. Nous avons également décrit des mécanismes, encore à définir, qui limitent les réponses à l'ADN de la chromatine dans le noyau. À mesure que les preuves augmentent sur la prise en charge de cGAS en réponse à des agents pathogènes, dans des maladies auto-immunes, en réponse aux dommages causés par l'ADN et dans le développement de la sénescence cellulaire, (...) / Nucleic acids are potent activators of innate immune responses. To control infections, multicellular organisms have developed multiple ways to detect pathogens. One of these is the recognition of DNA in the cytoplasm. Presence of DNA in the cytoplasm is usually linked to infections by bacteria or viruses, such as HIV. Cyclic GMP-AMP synthase (cGAS) is an essential cytosolic sensor for DNA in mammals. Upon DNA recognition, cGAS synthetizes the small second messenger cyclic GMP-AMP (cGAMP), which activates innate immune signaling pathways. cGAS plays a pivotal role in response to microbes and in the development of autoimmune diseases such as interferonopathies. This thesis investigate the role of cGAS in response to viruses and to self DNA. By exploring the cGAS-mediated response to HIV, we found that in cells producing virus, cGAS-synthesized cGAMP is packaged in viral particles and extracellular vesicles. Viral particles efficiently deliver cGAMP to target cells and activate a potent antiviral response. The transfer of cGAMP requires cGAS catalytic activity and fusion of the viral particles with the target cells. We also showed that in the context of an infection, DNA viruses such as MVA and mCMV can package cGAMP. Therefore viral mediated cGAMP transfer is a generalized defense mechanism of the innate immune system. We further investigated cGAS activation by DNA focusing our attention on nuclear chromatin DNA. In eukaryotic cells, DNA is compartmentalized in two organelles: the nucleus and the mitochondrion. Nuclear compartmentalization of DNA can be transiently lost upon nuclear envelope breakdown during cell division and upon nuclear envelope ruptures in migrating dendritic cells (DCs). We found that nuclear envelope breakdown or loss of nuclear envelope integrity leads to cGAS recruitment on the nuclear DNA. We also showed that DCs present with a nuclear pool of cGAS at steady state. Nuclear envelope breakdown during cell cycle leads to cGAS recruitment to the nuclear DNA. By manipulating cGAS localization, we showed that cGAS is poorly active when in the nucleus, and that providing exogenous DNA unmasked its full enzymatic activity. Expression of nuclear localized-cGAS in DCs leads to DCs maturation and Type I interferon (IFN) production. The N-terminal region of cGAS regulates cGAS nuclear/cytoplasmic distribution in interphase cells and we established Lamin A/C as required for cGAS accessibility to nuclear DNA in migrating DCs. Unexpectedly, we show that cGAS is not activated upon nuclear envelope rupture. Finally, we identify the pericentromeric heterochromatin as the preferential binding DNA for expressed nuclear cGAS in DCs. Altogether, our study shows the nucleus to act as an intracellular immune-privileged site for the activation of cGAS. Collectively, we have shown relevance of cGAS activation upon viral infection and uncovered a Trojan horse mechanism of cGAMP incorporation in the viral progeny. We have also described yet to be defined regulatory mechanisms that limit responses towards chromatin DNA in the nucleus. As evidence grows for cGAS involvement in response to pathogens, in autoimmune diseases, in response to DNA damage and in the development of cell senescence, fully understanding the mechanisms of distinction between self and pathogen related DNA by the sensor will be important to develop effective means to regulate the pathway.
4

Exploiting DNA Repair Vulnerabilities to Modulate Anti-Cancer Immunity : a Study of the Immunological Potential of PARP inhibitors / Exploiter les défauts de réparation de l’ADN pour moduler l’immunité anti-cancéreuse : une étude du potentiel immunologique des inhibiteurs de PARP

Chabanon, Roman 31 January 2019 (has links)
Les inhibiteurs de poly(ADP-ribose) polymérase (PARPi) ciblent sélectivement les cellules porteuses de défauts des voies de réparation de l’ADN tels que les mutations de BRCA1/2 et les défauts d’ERCC1. Sur le plan clinique, plusieurs PARPi ont été approuvés pour le traitement des cancers BRCA-mutés ou platine-sensibles du sein et de l’ovaire, et des essais cliniques sont en cours pour évaluer l’efficacité des PARPi dans le cancer bronchique non-à-petites cellules (CBNPC) platine-sensible. Alors que les PARPi ont un fort potentiel thérapeutique dans les cancers comportant des défauts de réparation de l’ADN, de plus en plus d’essais cliniques évaluent également l’efficacité de ces médicaments en combinaison avec les « inhibiteurs d’immune checkpoints » (ICI) dans diverses populations de patients. Dans ce contexte, il est essentiel de mieux comprendre comment les PARPi modulent la réponse immunitaire anti-tumorale, et d’étudier le potentiel immunologique inhérent de ces médicaments.Dans cette étude, nous avons établi que les cellules de CBNPC déficientes en ERCC1 expriment fortement la signature interféron (IFN) de type I, et que les tumeurs de CBNPC ayant une faible expression d’ERCC1 ont un infiltrat lymphocytaire renforcé. En utilisant des lignées cellulaires isogéniques et des xénogreffes dérivées de patients, nous avons montré que plusieurs PARPi, notamment l’olaparib et le rucaparib, ont des propriétés immunomodulatrices dans les modèles de CBNPC ERCC1-déficients et de cancers du sein triple-négatifs (CSTN) BRCA1-mutés. D’un point de vue mécanistique, les PARPi génèrent des fragments d’ADN cytoplasmiques ayant les caractéristiques de micronoyaux ; ceux-ci activent la voie cGAS/STING et déclenchent une réponse IFN de type I, associée à la sécrétion de la cytokine CCL5. De manière importante, ces effets sont largement diminués dans les cellules de CSTN BRCA1-révertantes et les cellules de CBNPC ré-exprimant ERCC1, ce qui suggère que les défauts de réparation de l’ADN amplifient les phénotypes immunitaires associés au traitement par PARPi. En outre, ces effets sont totalement abrogés dans les cellules de CSTN PARP1-neutralisées, ce qui confirme que les phénotypes observés dépendent d’un effet spécifique des PARPi sur leur cible.Au-delà de leur potentiel d’activation d’une immunité spécifique des cellules cancéreuses via cGAS/STING et la signalisation IFN de type I, nous avons également constaté que les PARPi potentialisent les effets inducteurs de l‘IFN de type II sur l’expression de PD-L1 dans des lignées cellulaires et cellules tumorales fraîches de patients CBNPC, surtout en présence de défauts d’ERCC1. De plus, nous avons montré que certains PARPi, utilisés à des concentrations létales, activent de manière indépendante les éléments moléculaires clés de la mort cellulaire immunogénique, dont l’exposition de la calréticuline à la surface des cellules cancéreuses, la sécrétion d’ATP et le relargage d’HMGB1 en grandes quantités dans le milieu extracellulaire.Dans l’ensemble, ces données précliniques suggèrent que les PARPi ont des propriétés immunomodulatrices intrinsèques qui participent à l’activation de réponses immunitaires anti-tumorales ; ce potentiel pourrait être exploité cliniquement en combinaison avec les ICI dans des populations adéquatement sélectionnées au plan moléculaire. / Poly(ADP-ribose) polymerase inhibitors (PARPi) selectively target cancer cells with DNA repair deficiencies such as BRCA1/2 mutations or ERCC1 defects. Clinically, several PARPi are currently approved for the treatment of BRCA-mutant or platinum-sensitive advanced ovarian and breast cancers, and ongoing clinical trials are investigating the efficacy of PARPi in platinum-sensitive Non-Small Cell Lung Cancer (NSCLC). While PARPi constitute potent targeted therapies for the treatment of DNA repair-deficient malignancies, an increasing number of clinical trials are also evaluating their efficacy in combination with immune checkpoint inhibitor (ICI) in various populations. In this context, it is of critical importance to better understand how PARPi might modulate immune responses against cancer, and to investigate the inherent immunological potential of these agents.In this study, we show that ERCC1-defective NSCLC cells exhibit an enhanced type I interferon (IFN) transcriptomic signature and that low ERCC1 expression correlates with increased lymphocytic infiltration in human NSCLC tumours. Using isogenic cell lines and patient-derived xenografts, we further demonstrate that several clinical PARPi, including olaparib and rucaparib, display cell-autonomous immunomodulatory properties in ERCC1-defective NSCLC and BRCA1-mutant triple-negative breast cancer (TNBC) models. Mechanistically, PARPi generate cytoplasmic chromatin fragments with micronuclei characteristics; this activates the cGAS/STING pathway and elicits downstream type I IFN signalling and CCL5 secretion. Importantly, these effects are suppressed in BRCA1-reverted TNBC cells and ERCC1-rescued NSCLC cells, suggesting that DNA repair defects exacerbate the innate immunity-related phenotypes triggered by PARPi. Similarly, these effects are totally abrogated in PARP1-null TNBC cells, supporting the on-target effect of PARPi in mediating such phenotypes. Besides this potential to activate tumour cell-autonomous immunity through cGAS/STING and type I IFN signalling, we also observed that PARPi synergize with type II IFN to induce PD-L1 expression in NSCLC cell lines and fresh patient tumour cells, especially in the ERCC1-deficient setting. Moreover, we show that lethal concentrations of some PARPi independently activate the key damage-associated molecular patterns dictating the immunogenicity of cancer cell death, including calreticulin exposure at the tumour cell surface, ATP secretion and HMGB1 release in the extracellular compartment.Together, these preclinical data suggest that PARPi have intrinsic immunomodulatory properties that activate anti-cancer immune responses; this could be exploited clinically in combination with ICI in appropriately molecularly-selected populations.
5

Recognition of Neutrophil Extracellular Traps by the Cytosolic DNA Sensor cGAS

Apel, Falko 11 February 2019 (has links)
Neutrophile Granulozyten produzieren „Neutrophil Extracellular Traps“ (NETs), ein mit antimikrobiellen Molekülen bestücktes Netzwerk aus Chromatinfasern, das während eines Zelltodprogramms namens „NETosis“ von den sterbenden Neutrophilen ausgestoßen wird. Ihre netzartige Struktur erlaubt es ihnen, eine weitere Verbreitung des Infektionserregers zu verhindern; zudem erzeugen sie eine hohe lokale Konzentration an toxischen Molekülen, die Mikroorganismen töten können. Unter normalen Bedingungen werden NETs von Nukleasen zerkleinert und anschließend von Makrophagen entfernt. Wenn dieser Aufräummechanismus gestört ist, aktivieren NETs das Immunsystem und führen zur Produktion von Autoantikörpern oder entzündungsfördernden Zytokinen. NETs werden mit einer wachsenden Liste von inflammatorischen und Autoimmunerkrankungen in Verbindung gebracht. Wie genau dabei NETs durch das Immunsystem erkannt werden, ist noch nicht bekannt. In der vorliegenden Arbeit zeige ich, dass NETs durch den zytosolischen DNA Sensor „cyclic GMP-AMP synthase“ (cGAS) detektiert werden können und dass dadurch die Expression von Typ I Interferonen (TIIFN) induziert wird. Zu Beginn demonstriere ich, dass NETs durch rekombinantes cGAS erkannt werden und dass mit isolierten NETs stimulierte Immunzellen cGAS-abhängig TIIFN produzieren. Des Weiteren zeige ich, dass Neutrophile, die NETosis begehen, in Nachbarzellen ebenfalls cGAS-anhängig TIIFN induzieren können. Abschließend konnte ich diese Ergebnisse in einem in vivo Mausmodel für systemische NET-Produktion bestätigen. Die vorliegende Arbeit zeigt einen Mechanismus, wie NETs durch das Immunsystem erkannt werden und dadurch sowohl zur Entstehung als auch zur Progression von Krankheiten beitragen kann. Sie ermöglicht dementsprechend die Entwicklung neuer Interventionsstrategien, welche zur Heilung oder Linderung einer Vielzahl von Erkrankungen beitragen können. / The first line of cellular defense of the immune system are neutrophils. They are the most abundant white blood cell, which exert an array of antimicrobial effector functions. Neutrophils release neutrophil extracellular traps (NETs), a composite of chromatin and antimicrobial molecules, into the extracellular space during a form of regulated cell death called NETosis. Their net-like structure prevent further dissemination of the invader and establishes a high local concentration of toxic molecules that mediate pathogen killing. NETs provide a platform for undesired immune activation and contribute to the production of autoantibodies and pro-inflammatory cytokines. NETs are implicated in a growing list of inflammatory and autoimmune diseases, but the exact mechanism how NETs are recognized by the immune system is not fully understood. In this study, I demonstrate that the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) senses NETs and induces the production of type I interferons (TIIFN). I first showed that NETs are recognized by recombinant cGAS and that cells treated with isolated NETs produce TIIFN in a cGAS dependent mechanism. Secondly, I demonstrate that neutrophils undergoing NETosis are taken up by neighboring immune cells and induce cGAS-dependent TIIFN expression. Lastly, I confirmed our in vitro results in a mouse model of systemic NET induction. Wildtype mice injected with Concanavalin A significantly upregulate the expression of interferon stimulated genes, while cGAS-/- mice and Cybb-/- mice, which are incapable of producing NETs, fail to induce this response.
6

Pathways Involved in Recognition and Induction of Trained Innate Immunity by Plasmodium falciparum

Schrum, Jacob E. 07 August 2017 (has links)
Malarial infection in naïve individuals induces a robust innate immune response, but our understanding of the mechanisms by which the innate immune system recognizes malaria and regulates its response remain incomplete. Our group previously showed that stimulation of macrophages with Plasmodium falciparum genomic DNA (gDNA) and AT-rich oligodeoxynucleotides (ODNs) derived from this gDNA induces the production of type I interferons (IFN-I) through a STING/TBK1/IRF3-dependent pathway; however, the identity of the upstream cytosolic DNA receptor remained elusive. Here, we demonstrate that this IFN-I response is dependent on cyclic GMP-AMP synthase (cGAS). cGAS produced the cyclic dinucleotide 2’3’-cGAMP in response to P. falciparum gDNA and AT-rich ODNs, inducing IRF3 phosphorylation and IFNB transcription. In the recently described model of innate immune memory, an initial stimulus primes the innate immune system to either hyperrespond (termed “training”) or hyporespond (“tolerance”) to subsequent immune challenge. Previous work in mice and humans demonstrated that infection with malaria can both serve as a priming stimulus and promote tolerance to subsequent infection. In this study, we demonstrate that initial stimulation with P. falciparum-infected red blood cells (iRBCs) or the malaria crystal hemozoin (Hz) induced human adherent peripheral blood mononuclear cells (PBMCs) to hyperrespond to subsequent Toll-like receptor (TLR) challenge. This hyperresponsiveness correlated with increased H3K4me3 at important immunometabolic promoters, and these epigenetic modifications were also seen in monocytes from Kenyan children naturally infected with malaria. However, the use of epigenetic and metabolic inhibitors indicated that malaria-induced trained immunity may occur via previously unrecognized mechanism(s).
7

Cytosolic DNA sensing in autoimmune and autoinflammatory diseases

Motwani, Mona 20 March 2020 (has links)
Cytosolic DNA sensing plays a key role in autoimmune and autoinflammatory diseases. STING is a cytosolic adaptor protein which upon activation leads to induction of type I interferons and inflammatory cytokines. Recently, gain-of-function mutations in STING have been identified in patients with an autoinflammatory disease called STING-associated vasculopathy with onset in infancy (SAVI). We compared two independent SAVI mutant mouse models and revealed a hierarchy of immune abnormalities which were dependent on SAVI mutation in lymphocytes. We also showed that bone marrow from the V154M mutant mice transfers disease to the wild-type host, whereas the N153S does not, indicating mutation-specific disease outcomes. Collectively, these mutant mice recapitulate disease features seen in SAVI patients and highlight mutation-specific functions of STING. Other autoimmune mouse models such as DNAseII and DNAseIII-deficient mice, that fail to degrade DNA result in activation of the cGAS STING pathway. Deficiency of this pathway in these mouse models ameliorates lethality. By contrast, we previously reported that STING potently suppresses inflammation in a pristane-induced model of autoimmunity. In this model, we show that both cGAS- and STING-deficient mice exhibit exacerbated disease phenotypes compared to controls. We report that STING constrained TLR activation, and thereby limited autoimmune manifestations. Consistent with this premise, cGAS or STING deficient mice that lack a common TLR chaperone UNC93b develop less severe systemic autoimmunity than cGAS or STING deficient mice that are UNC93b sufficient. Overall, this study demonstrates that STING activation constrains systemic autoimmune disease and has important implications for cGAS STING-directed therapies.
8

Myší polyomavirus:Způsob translokace do buněčného jádra a rozpoznání virových genomů sensory vrozené imunity / Mouse polyomavirus:The way of virus translocation to the cell nucleus and sensing of viral genomes by sensors of innate immunity

Soldatova, Irina January 2021 (has links)
To understand molecular mechanisms of individual steps of virus infection is a prerequisite for successful design of specific and effective antiviral drugs. Polyomaviruses, replicating in the cell nucleus, travel from plasma membrane to the endoplasmic reticulum (ER) in endosomes. However, it is not clear how they deliver their DNA genomes from ER to the nucleus. In this thesis, we found that partially disassembled virions of the Murine polyomavirus (MPyV) interact with importin β1 at around 6 hours post infection. Mutational disruption of the nuclear localization signal (NLS) of the major capsid protein, VP1, and/or common NLS sequence of the minor capsid proteins VP2 and VP3 did not affect the structure and composition of virions, but it resulted in decreased viral infectivity (up to 80%). Virions are thus released from ER to cytosol and translocate to the nucleus via nucleopores. Mutation analyses of NLSs of individual capsid proteins showed that MPyV virions can utilize VP1 and VP2/VP3 NLSs in concert. However, one functional NLS, either that of VP1 or VP2/3 seems to be sufficient for the delivery of VP1-VP2/3 complexes into the nucleus, although none of these proteins is delivered into the nucleus separately. Thus, the conformation of NLS regions given by the presence of all three capsid...
9

A POTENT PYRAZOLE-CONTAINING STING ANTAGONIST SYNTHESIZED VIA DOEBNER-POVAROV MULTICOMPONENT REACTION

Wei Shiuan Wilson Ong (12442317) 21 April 2022 (has links)
<p>  </p> <p>The cGAS-STING axis represents a key pathway towards the activation of innate immunity against pathogens. However, persistent activation can lead to the development of autoimmune diseases, driving the need for the development of antagonists of cGAS-STING pathway. Herein, we describe the discovery of a small molecule STING binder HSD1077 through a STING based fluorescence polarization (FP) displacement assay. Initial SAR studies utilizing the FP displacement assay suggests the presence of pyrazole moieties critical for HSD1077 towards STING Binding. Additionally, we show that HSD1077 serves as an antagonist of the cGAS-STING pathway and effectively suppresses type-1 interferon expression upon 2’-3’cGAMP induction in both murine RAW macrophages and human THP-1 monocytes. HSD1077 in conclusion shows potential as a lead compound towards the further development of anti-inflammatory drugs. </p>
10

Studium rozpoznáni polyomavirové infekce sensory vrozené imunity / Sensing of MPyV infection by innate immunity sensors

Rjabčenko, Boris January 2021 (has links)
Host sensors that recognize pathogen associated molecular patterns and the mechanisms of innate immune response to mouse polyomavirus (MPyV) infection were the main topics of current work. We found that MPyV did not induce interferon (IFN) production during early events of infection, but induced interleukin-6 (IL-6) and other cytokine production without inhibiting virus multiplication. Cytokine microenvironment changed the phenotype of adjacent non infected fibroblasts toward the cancer-associated fibroblast (CAF)-like phenotype. We identified Toll-like receptor 4, a sensor of the innate immunity system, to be responsible for infection dependent IL-6 production. In an effort to determine whether and where virions are released from endosomal compartments into the cytosol, we found that the hydrophobic domains of minor capsid proteins, exposed on the surface of virions after their partial disassembly in the ER, play an important role in effective escape of virions from the lumen part of endoplasmic reticulum into the cytosol, Although naked, partially disassembled virions appear before translocation to the nucleus in the cytosol, viral DNA is not recognized by cytosolic sensors at this phase of infection Sensing of MPyV resulting in IFN production occurs first during viral replication. Mutant virus,...

Page generated in 0.0317 seconds