• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 6
  • 2
  • 2
  • 1
  • Tagged with
  • 11
  • 11
  • 9
  • 6
  • 5
  • 5
  • 5
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 3
  • 3
  • 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

Destabilization of IL-8 mRNA by Anthrax Lethal Toxin: Demonstration of the Requirement for TTP and Examination of its Cellular Interactions

Chow, Man Chi Edith 06 December 2012 (has links)
Control of mRNA stability is an important aspect in the regulation of gene expression. A well studied signal for rapid transcript decay in mammalian cells is the AU-rich element (ARE), which is found in the 3’ untranslated region (UTR) of many labile transcripts. These sequence elements confer destabilization of transcripts by binding to AU-binding proteins (AUBPs) that can recruit cellular decay enzymes. The stability of ARE-containing mRNAs can be regulated by extracellular stimuli, which allows for cells to adapt to the changing environment. AREs are found in many transcripts that encode for inflammatory genes, including TNF, GM-CSF, and IL-8. Pathogens evolve and devise mechanisms to subvert the immune response of the host to aid in its infection. Bacillus anthracis is one such infectious agent that can disable numerous arms of the host immune response. Its secreted toxin, anthrax lethal toxin (LeTx), causes the accelerated decay of the IL-8 mRNA. IL-8 is a dual function cytokine and chemokine that can recruit and activate neutrophils at the site of infection. Through the inactivation of MAPK pathways, LeTx activity causes the destabilization of IL-8 transcripts through its ARE. In this thesis, I show that an AUBP, TTP, is dephosphorylated by LeTx and MAPK inhibitors, and knock-down of its expression stabilized IL-8 transcripts. LeTx activity also increased the colocalization of TTP to P-bodies, cytoplasmic sites concentrated with RNA decay enzymes. This suggests that the post-translational modification of TTP induced by LeTx led to its enhanced destabilization function. Identified TTP-associated proteins, non-muscle myosin heavy chain 9 (myosin-9) and HSC-70, were examined for their role in IL-8 transcript decay. Knock-down of each protein led to a slower rate of IL-8 mRNA destabilization. However, treatment of LeTx continued to mediate accelerated destabilization of IL-8 in these siRNA-transfected cells. This suggests that LeTx, myosin-9, and HSC-70 modulate the destabilization function of TTP independently.
2

Destabilization of IL-8 mRNA by Anthrax Lethal Toxin: Demonstration of the Requirement for TTP and Examination of its Cellular Interactions

Chow, Man Chi Edith 06 December 2012 (has links)
Control of mRNA stability is an important aspect in the regulation of gene expression. A well studied signal for rapid transcript decay in mammalian cells is the AU-rich element (ARE), which is found in the 3’ untranslated region (UTR) of many labile transcripts. These sequence elements confer destabilization of transcripts by binding to AU-binding proteins (AUBPs) that can recruit cellular decay enzymes. The stability of ARE-containing mRNAs can be regulated by extracellular stimuli, which allows for cells to adapt to the changing environment. AREs are found in many transcripts that encode for inflammatory genes, including TNF, GM-CSF, and IL-8. Pathogens evolve and devise mechanisms to subvert the immune response of the host to aid in its infection. Bacillus anthracis is one such infectious agent that can disable numerous arms of the host immune response. Its secreted toxin, anthrax lethal toxin (LeTx), causes the accelerated decay of the IL-8 mRNA. IL-8 is a dual function cytokine and chemokine that can recruit and activate neutrophils at the site of infection. Through the inactivation of MAPK pathways, LeTx activity causes the destabilization of IL-8 transcripts through its ARE. In this thesis, I show that an AUBP, TTP, is dephosphorylated by LeTx and MAPK inhibitors, and knock-down of its expression stabilized IL-8 transcripts. LeTx activity also increased the colocalization of TTP to P-bodies, cytoplasmic sites concentrated with RNA decay enzymes. This suggests that the post-translational modification of TTP induced by LeTx led to its enhanced destabilization function. Identified TTP-associated proteins, non-muscle myosin heavy chain 9 (myosin-9) and HSC-70, were examined for their role in IL-8 transcript decay. Knock-down of each protein led to a slower rate of IL-8 mRNA destabilization. However, treatment of LeTx continued to mediate accelerated destabilization of IL-8 in these siRNA-transfected cells. This suggests that LeTx, myosin-9, and HSC-70 modulate the destabilization function of TTP independently.
3

The FIIND Domain of Nlrp1b Promotes Oligomerization and Pro-caspase-1 Activation in Response to Lethal Toxin of Bacillus anthracis

Joag, Vineet 29 November 2012 (has links)
Lethal toxin (LeTx) of Bacillus anthracis kills murine macrophages in a caspase-1 and Nod-like-receptor-protein 1b (Nlrp1b)-dependent manner. Nlrp1b detects intoxication, and self-associates to form a macromolecular complex called the inflammasome, which activates the pro-caspase-1 zymogen. I heterologously reconstituted the Nlrp1b inflammasome in human fibroblasts to characterize the role of the FIIND domain of Nlrp1b in pro-caspase-1 activation. Amino-terminal truncation analysis of Nlrp1b revealed that Nlrp1b1100-1233, containing the CARD domain and amino-terminal 42 amino acids within the FIIND domain was the minimal region that self-associated and activated pro-caspase-1. Residues 1100EIKLQIK1106 within the FIIND domain were critical for self-association and pro-caspase-1 activation potential of Nlrp1b1100-1233, but not for binding to pro-caspase-1. Furthermore, residues 1100EIKLQIK1106 were critical for cell death and pro-caspase-1 activation potential of full-length Nlrp1b upon intoxication. These data suggest that after Nlrp1b senses intoxication, the FIIND domain promotes self-association of Nlrp1b, which activates pro-caspase-1 zymogen due to induced pro-caspase-1 proximity.
4

The FIIND Domain of Nlrp1b Promotes Oligomerization and Pro-caspase-1 Activation in Response to Lethal Toxin of Bacillus anthracis

Joag, Vineet 29 November 2012 (has links)
Lethal toxin (LeTx) of Bacillus anthracis kills murine macrophages in a caspase-1 and Nod-like-receptor-protein 1b (Nlrp1b)-dependent manner. Nlrp1b detects intoxication, and self-associates to form a macromolecular complex called the inflammasome, which activates the pro-caspase-1 zymogen. I heterologously reconstituted the Nlrp1b inflammasome in human fibroblasts to characterize the role of the FIIND domain of Nlrp1b in pro-caspase-1 activation. Amino-terminal truncation analysis of Nlrp1b revealed that Nlrp1b1100-1233, containing the CARD domain and amino-terminal 42 amino acids within the FIIND domain was the minimal region that self-associated and activated pro-caspase-1. Residues 1100EIKLQIK1106 within the FIIND domain were critical for self-association and pro-caspase-1 activation potential of Nlrp1b1100-1233, but not for binding to pro-caspase-1. Furthermore, residues 1100EIKLQIK1106 were critical for cell death and pro-caspase-1 activation potential of full-length Nlrp1b upon intoxication. These data suggest that after Nlrp1b senses intoxication, the FIIND domain promotes self-association of Nlrp1b, which activates pro-caspase-1 zymogen due to induced pro-caspase-1 proximity.
5

Identification of Host Factors Required for Anthrax Lethal Toxin Intoxication Using Chemical Genetic and RNAi Approaches

Slater, Louise January 2011 (has links)
Bacterial toxins have co-opted host cell machinery in order to enter cells and exert their deleterious effects. Anthrax toxin is composed of the receptor binding protein protective antigen (PA), and the enzymatic subunits lethal factor (LF) and edema factor (EF), which form the binary toxin complexes lethal toxin, LeTx (PA + LF), and edema toxin, EdTx (PA + EF). PA binds to receptors on the surface of host cells and shuttles LF and EF into cells through the endocytic pathway. Upon endosome acidification, PA oligomers insert into the endosomal membrane and form functional pores that deliver LF and EF into the cytoplasm. Translocation of the N-terminal domain of LF, \(LF_N\), through PA pores formed in lipid bilayers in vitro does not require host machinery. However, translocation of the related fusion protein \(LF_N\)-DTA across the membrane of toxin-loaded endosomes in vitro requires the addition of cytosolic translocation factors that include the COPI coatamer complex. We performed high-throughput small molecule and RNAi screens to identify host factors required for LF translocation, using LeTx-induced cell death as a phenotype. We describe the characterization of small molecule inhibitors of LeTx-induced cell death that inhibit toxin entry. Further, we describe the role of the endosomal chaperone GRP78 and the cytoplasmic CCT chaperonin in toxin translocation. RNAi knockdown of GRP78 and CCT subunits inhibited LeTx and EdTx delivered through the endocytic pathway. CCT knockdown additionally inhibited translocation of LF through PA pores formed directly in the plasma membrane, while GRP78 had no effect. Furthermore, we show that the role of GRP78 in toxin translocation is specific to translocation from the early endosome. Together with biochemical data, we propose that GRP78 facilitates translocation by unfolding LF and EF at near-neutral pH. In addition, we show that in CCT-knockdown cells, lethal levels of toxin reach the endosome, suggesting that CCT has a role in translocation and/or refolding of LF and EF. These studies highlight previously unidentified strategies used by anthrax toxin to hijack host cellular machinery in order to gain access to the cytosol.
6

CHARACTERIZATION OF NEUTRALIZING RESPONSES TO ANTHRAX TOXINS AND ISOLATION AND CHARACTERIZATION OF THE SHIGA-TOXIN ENCODING PHAGE OF ESCHERICHIA COLI 0157:H7

HANSON, JAMES F. 05 October 2004 (has links)
No description available.
7

Investigation and characterization of the enhanced humoral response following immunization with the lethal and edema toxins of bacillus anthracis

Brenneman, Karen Elaine 27 March 2007 (has links)
No description available.
8

Effets des toxines de Bacillus anthracis sur le cytosquelette des cellules immunitaires : implication sur la phagocytose et les fonctions immunitaires / Effects of bacillus anthracis toxins on the cytoskeleton of immune cells : involvement in phagocytosis and immune fonctions

Trescos, Yannick 09 December 2015 (has links)
Bacillus anthracis, agent de la maladie du charbon, est aussi un agent majeur de la menace biologique. Sa virulence est liée à deux principaux facteurs : une capsule et deux toxines, la toxine oedémateuse (ET = EF + PA) et la toxine létale (LT = LF + PA). EF est une adénylate cyclase, calcium et calmoduline dépendante, produisant une élévation de la concentration en AMPc intracellulaire tandis que LF est une métalloprotéase à zinc clivant la majorité des Mitogen Activated Protein Kinase Kinases. Les toxines jouent un rôle central dans la pathogénie de la maladie du charbon et dans la dérégulation des fonctions des cellules du système immunitaire. Le cytosquelette d'actine participe activement aux fonctions de phagocytose et de migration des macrophages et des cellules dendritiques.Cependant, peu d'études analysent l'implication du cytosquelette d'actine des cellules immunitaires dans la physiopathologie des toxines. ET induit une rétraction temps-dépendant des cellules dendritiques et des macrophages normalisés sur des micropatterns de fibronectine, s'accompagnant d'une dépolymérisation de l'actine et d'une perte des points d'ancrage des cellules dendritiques. Précocement, ET active la cofiline par l'activation de la voie de signalisation AMPc – PKA – Protéines phosphatases. Malgré ces altérations du cytosquelette d'actine, ET n'induit pas de modification des capacités phagocytaires des cellules dendritiques, à l'exception d'une dérégulation de la maturation des phagosomes. ET conduit également à une augmentation de la migration des cellules dendritiques in vitro par activation et expression de CCR7 et CXCR4 à la surface des cellules dendritiques.A l'inverse, LT conduit à un étalement temps-dépendant des cellules dendritiques normalisées, accompagné d'une dérégulation de la dynamique de l'actine provoquant des regroupements anormaux d'actine filament. LT active les myosines phosphatases via la voie RhoA-ROCK pour déphosphoryler les myosines II. A la différence de ET, LT inhibe les capacités phagocytaires des cellules dendritiques mais ne conduit pas à une modification de la migration des cellules dendritiques in vitro. / Bacillus anthracis, the agent of anthrax, is also a major agent of biological warfare threat. Its virulence is caused by two main factors : the capsule and two toxins, edema toxin (ET = PA + EF) and lethal toxin (LT = LF + PA). EF is a calcium and calmodulin-dependent adenylate cyclase, producing a rise in intracellular cAMP concentration, while LF is a zinc metalloprotease cleaving the majority of Mitogen Activated Protein Kinase Kinases. The toxins play a central role in the pathogenesis of the disease and the deregulation of the functions of immune cells. The actin cytoskeleton is actively participating in the phagocytosis and the migration of macrophages and dendritic cells.However, few studies analyze the involvement of the actin cytoskeleton of immune cells in the pathogenesis of toxins. ET induces a time-dependent retraction of dendritic cells and macrophages on fibronectin micropatterns, accompanied by actin depolymerization and a loss of the anchor points of dendritic cells. ET early activates cofilin by activating the cAMP - PKA - Protein phosphatases signaling pathway. Despite these alterations of the actin cytoskeleton, ET does not induce any change in the phagocytic capacity of dendritic cells, except for a deregulation of the phagosomes maturation. ET also leads to an increase in the migration of dendritic cells in vitro by activation and expression of CCR7 and CXCR4 on the surface of dendritic cells.In contrast, LT results in a time-dependent spreading of micropatterned dendritic cells, accompanied by a dysregulation of actin dynamics causing abnormal combinations of actin filament. LT activates myosin phosphatase via the RhoA-ROCK pathway to dephosphorylate myosin II. Unlike ET, LT inhibits the dendritic cells phagocytosis but does not lead to a change in dendritic cells migration in vitro.
9

Inflammasomes : des mécanismes d’activation de la caspase-1 à la progression tumorale / Inflammasomes : from caspase-1 activation mechanisms to tumor progression

Guey, Baptiste 01 July 2016 (has links)
L'inflammasome est une plateforme moléculaire composée d'un récepteur de l'immunité innée tels que NLRP3 ou NLRP1b, de la protéine adaptatrice ASC et de la caspase-1. Il joue un rôle essentiel dans le déclenchement de la réponse inflammatoire via l'activation de la caspase-1 qui mène à la sécrétion de cytokines pro-inflammatoires telle que l'IL-1ß. Dans un premier axe de recherche, nous avons mis en évidence que les macrophages de souris déficientes pour ASC traités avec l'activateur de l'inflammasome NLRP1b, la toxine létale de bacillus anthracis, étaient capables de sécréter la forme mature de l'IL-1ß en absence de clivage de la caspase-1 pourtant décrit comme un événement indispensable à son activité. En reconstituant des macrophages caspase-1 KO avec une forme mutante non-clivable de la caspase-1, nous avons démontré que la forme entière de la caspase-1 est capable d'induire la sécrétion d'IL-1ß en réponse à la toxine létale alors qu'elle n'est pas fonctionnelle au sein de l'inflammasome NLRP3.Dans un second axe de recherche, mon travail de thèse s'est intéressé à comprendre le rôle de l'inflammasome au cours de la progression tumorale. En effet, l'IL-1ß est une cytokine fréquemment retrouvée dans le microenvironnement tumorale mammaire suggérant donc l'activation de l'inflammasome au sein des tumeurs. Au moyen d'un modèle de tumeurs in vivo, nous avons montré que l'absence de la caspase-1 et de ASC dans les cellules immunitaires chez la souris conduit à une réduction de la croissance tumorale. De plus, l'absence de ces deux protéines provoque également un plus fort recrutement et une meilleure activité des cellules NK au sein des tumeurs.En plus d'identifier un nouveau mécanisme d'activation de la caspase-1, mon travail de thèse a permis de mettre en évidence le rôle de l'inflammasome dans l'altération des cellules NK au cours de la progression tumorale mammaire. Ces données permettent d'envisager l'inflammasome comme une cible thérapeutique dans le cancer / The inflammasome is a molecular platform composed of an innate immune receptor such as NLRP3 or NLRP1b, of the adaptor protein ASC and of the caspase-1. It plays an essential role in triggering inflammatory response through the activation of caspase-1 that leads to the secretion of pro-inflammatory cytokines such as IL-1ß.In a first research axis, we showed that ASC deficient mice macrophages treated with an NLRP1b inflammasome activator, the lethal toxin of Bacillus anthracis, were able to secrete the mature form of IL-1ß in absence of any cleavage of caspase-1 previously described as an essential event for its function. By reconstituting caspase-1 KO macrophages with an uncleavable mutant form of caspase-1, we showed that the entire form of the protein is able to induce IL-1ß secretion upon lethal toxin treatment but is nonfunctional upon NLRP3 inflammasome activation.In a second research axis, my PhD work focused on underlying the inflammasome role during tumor progression. Indeed, IL-1ß is frequently found within breast tumor microenvironment suggesting that inflammasome is activated in tumors. Using in vivo tumor model, we showed that the absence of caspase-1 of ASC in immune cells lead to a delay tumor growth. In addition, the absence of these two proteins also causes a stronger recruitment and an enchanced activity of NK cells within mammary tumors
10

Proteolytic Processing of Nlrp1b in the FIIND Domain is Required for Inflammasome Activity

Frew, Bradley 21 March 2012 (has links)
Nlrp1b is a NOD-like receptor of the innate immune system that upon sensing of anthrax lethal toxin oliogmerizes and forms a protein scaffold that binds to and activates pro-caspase-1; this complex is called an inflammasome. Nlrp1b is highly polymorphic and different alleles display an all or none ability to sense lethal toxin. Here I show that Nlrp1b is cleaved in the FIIND domain, and that the cleaved fragments remain associated even after activation by lethal toxin. The inflammasome activity of an inactive allele was restored by three mutations, one of which also restored cleavage. A heterologous cleavage site was inserted into an uncleaved mutant of Nlrp1b; induced proteolysis of the cleavage site rescued inflammasome activity. An uncleaved mutant of Nlrp1b showed no deficiency in FIIND self-association, but did have reduced recruitment of pro-caspase-1. These data provide evidence that cleavage of Nlrp1b is required for proper recruitment and activation of caspase-1.

Page generated in 0.0426 seconds