• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 10
  • 8
  • 3
  • 2
  • 1
  • Tagged with
  • 29
  • 19
  • 17
  • 13
  • 12
  • 10
  • 10
  • 9
  • 8
  • 8
  • 8
  • 8
  • 8
  • 8
  • 8
  • 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.
21

Adesina Aae de Aggregatibacter actinomycetemcomitans: envolvimento na adesão a proteínas da matriz extracelular, polimorfismo genético e resposta imune humoral. / Aae adhesin of A. actinomycetemcomitans: Implication in binding to extracellular matrix proteins, genetic polymorphisms and humoral immune response.

Almeida, Lucas Ribeiro de Sousa 13 July 2017 (has links)
Aggregatibacter actinomycetemcomitans está associado à etiologia da periodontite agressiva localizada. A colonização de tecidos do hospedeiro é necessária para patogênese e a adesão é fundamental. A proteína autotransportada Aae faz a adesão da bactéria a células epiteliais gengivais. Proteínas autotransportadas de diferentes espécies apresentam múltiplas funções e podem ser antígenos vacinais na prevenção de infecções. Para entender o papel de Aae na ligação ao hospedeiro e efeito de anticorpos contra Aae, o polimorfismo de aae na região que codifica o domínio de ligação a células epiteliais foi determinado e relacionado à adesão a células epiteliais KB . Aae recombinante foi obtida, e a capacidade de ligação a proteínas da matriz extracelular e soro foi determinada em ensaios com a recombinante e com uma amostra deficiente na expressão de Aae obtida (ensaios comparativos com amostra selvagem). Títulos de IgG contra Aae em pacientes com periodontite agressiva e saudáveis foram determinados e relacionados à resposta humoral contra sorotipos de A. actinomycetemcomitans. Por fim, o efeito de anticorpos contra Aae e/ou seu domínio efetor, produzidos em modelo animal, foi determinado na inibição da adesão mediada por Aae e opsonização por fagócitos. / Aggregatibacter actinomycetemcomitans is related with etiology of localized aggressive periodontitis. Colonization of host tissues is necessary to pathogenesis and adhesion is essential. The autotransporter protein Aae mediates the adhesion of bacteria to gingival epithelial cells. Autotransporter proteins from different species have multiple functions and could be vaccine antigens to prevent infections. To understand the role of Aae in host interaction and the effect of antibodies against Aae, polymorphism of aae in codifying effector domain region of ligation to epithelial cells was determined and related with adhesion to these cells. Recombinant Aae was obtained and the ability of interaction with Extracellular matrix and serum proteins was determined through assays using the recombinant and an obtained defective sample in Aae expression (comparative assays with wild type). IgG titters against Aae were determined in patients with aggressive periodontitis and healthy and related to humoral response against A. actinomycetemcomitans serotypes. At last, the effect of antibodies against Aae and/or its effector domain, obtained in animal model, was determined in inhibition of adhesion to epithelial cells and macrophages oopsonization.
22

Sat (Secreted autotransporter toxin): ação citotóxica da toxina bacteriana em diferentes linhagens celulares e na infecção in vitro por uma cepa de Escherichia coli enteroagregativa (EAEC) sorotipo O125ab:H21. / Sat (Secreted autotransporter toxin): cytotoxic action of the bacterial toxin in different cellular lineages and in an in vitro infection with an enteroaggregative Escherichia coli (EAEC) serotype O125ab:H21.

Vieira, Paulo Cesar Gomes 07 August 2018 (has links)
As serino-proteases autotransportadoras de Enterobacteriaceae (SPATE) constituem uma família de proteases secretadas pelo sistema de secreção do tipo V, cujos genes foram estudados em Escherichia coli intestinal e extra-intestinal. Sat é uma SPATE citotóxica de 107 kDa, cujo gene foi identificado pela primeira vez em UPEC isolada da urina de um paciente com pielonefrite. A maioria dos estudos envolvendo Sat foram realizados em células renais e da bexiga, embora seu gene seja encontrado em DAEC, EAEC e, mais recentemente, em amostras bacterianas isoladas de septicemia neonatal e meningite. Os objetivos deste trabalho foram: i) purificar Sat; ii) determinar a ação da Sat purificada em diferentes tipos celulares e iii) caracterizar o papel de Sat na infecção in vitro por EAEC. Desta foram, a presença de Sat nos sobrenadantes do cultivo das cepas EAEC CV323 e DEC/Sat, isoladas de diarreia, foi confirmada por LC-MS/MS. Sat foi purificada da cultura de DEC/Sat+ e utilizada para a obtenção de anticorpos anti-Sat em coelho. O efeito citotóxico de Sat purificada foi investigado em células derivadas do endotélio (HUVEC) e do sistema urinário (Y1, LLC-PK1 e HEK-293) e gastrointestinal (Caco-2). Os parâmetros citotóxicos analisados foram: o descolamento celular e alterações na morfologia, permeabilidade e metabolismo mitocondrial das células. Para investigar o papel de Sat na infecção por EAEC, células Y-1 foram infectadas com EAEC CV323 e DEC/Sat+ na presença ou ausência de PMSF (inibidor de serino-protease) e anticorpos anti-Sat. Os parâmetros de citotoxicidade analisados nas culturas infectadas foram descolamento celular e alteração na morfologia. Os resultados demonstraram que: i) Sat é secretada por EAEC CV323 e DEC/Sat+ e, em ambas as cepas, há duas mutações em resíduos de aminoácidos que não interferiram na atividade enzimática; ii) as células do endotélio são mais sensíveis à Sat do que as células derivadas do trato urinário, sendo a linhagem gastrointestinal a mais resistente; iii) Sat secretada por EAEC CV323 durante a infecção induziu intenso dano celular, o qual, em presença de anticorpos anti-Sat e PMSF foi reduzido em cerca de 80 a 90%, respectivamente. Este é o primeiro trabalho que demonstra a expressão de Sat pela EAEC e a ação da toxina em células endoteliais sugerindo que o papel de Sat possa ser mais amplo na patogenia do que o proposto até o momento. / The serine protease autotransporters of Enterobacteriaceae (SPATEs) constitute a family of proteases secreted by the type V secretion system whose genes have been studied in intestinal and extra intestinal Escherichia coli. Sat is a 107 kDa cytotoxic SPATE and its gene was first identified in UPEC isolated from the urine of a patient with pyelonephritis. Most studies involving Sat were performed in renal and bladder cells, although the gene encoding Sat is encountered in other strains of E. coli such as DAEC, EAEC and more recently, in bacterial samples isolated from neonatal septicemia and meningitis. The objectives of this work were: i) purify Sat; ii) to determine the action of Sat in different types of cells and iii) to characterize in vitro the role of Sat in EAEC infection. Accordingly, the presence of Sat in the culture supernatant of EAEC CV323 and DEC/Sat+ derived from diarrhea was confirmed by LCMS/MS. Sat was purified from the culture of DEC/Sat+ and utilized to produce rabbit antibodies anti-Sat. The cytotoxic effect of Sat was investigated in cells derived from the endothelium (HUVEC) and the urinary (Y1, LLC-PK1, HEK-293) and gastrointestinal (Caco-2) systems. The cytotoxic parameters analyzed were cellular detachment and alterations in the morphology, permeability and mitochondrial metabolism of the cells. To investigate the role of Sat in EAEC infection, Y-1 cells were incubated with EAEC CV323 and DEC/Sat+ in the presence or absence of PMSF (a serine protease inhibitor) and rabbit antibodies anti-Sat. The parameters analyzed were cellular detachment and alteration in the morphology of the cells. The results demonstrated that: i) Sat is secreted by EAEC CV323 and DEC/Sat + and in both strains there are two mutations in amino acid residues that did not interfere with enzymatic activity; ii) endothelium cells are more sensitive to the Sat effect than the cells derived from urinary tract system, being the gastrointestinal cell lineage the most resistant one; iii) Sat secreted by EAEC CV323 during infection induced intense cellular damage which in the presence of anti-Sat antibodies and PMSF was reduced in about 80 to 90%, respectively. This is the first work demonstrating the expression of Sat by EAEC and the action of the toxin on endothelial cells suggesting that the role of Sat may be broader in pathogenesis than has been proposed so far.
23

Impact of glucose uptake rate on recombinant protein production in Escherichia coli

Bäcklund, Emma January 2011 (has links)
Escherichia coli (E. coli) is an attractive host for production of recombinant proteins, since it generally provides a rapid and economical means to achieve high product quantities. In this thesis, the impact of the glucose uptake rate on the production of recombinant proteins was studied, aiming at improving and optimising production of recombinant proteins in E. coli. E. coli can be cultivated to high cell densities in bioreactors by applying the fed-batch technique, which offers a means to control the glucose uptake rate. One objective of this study was to find a method for control of the glucose uptake rate in small-scale cultivation, such as microtitre plates and shake flasks. Strains with mutations in the phosphotransferase system (PTS) where used for this purpose. The mutants had lower uptake rates of glucose, resulting in lower growth rates and lower accumulation of acetic acid in comparison to the wild type. By using the mutants in batch cultivations, the formation of acetic acid to levels detrimental to cell growth could be avoided, and ten times higher cell density was reached. Thus, the use of the mutant strains represent a novel, simple alternative to fed-batch cultures.   The PTS mutants were applied for production of integral membrane proteins in order to investigate if the reduced glucose uptake rate of the mutants was beneficial for their production. The mutants were able to produce three out of five integral membrane proteins that were not possible to produce by the wild-type strain. The expression level of one selected membrane protein was increased when using the mutants and the expression level appeared to be a function of strain, glucose uptake rate and acetic acid accumulation. For production purposes, it is not uncommon that the recombinant proteins are secreted to the E. coli periplasm. However, one drawback with secretion is the undesired leakage of periplasmic products to the medium. The leakage of the product to the medium was studied as a function of the feed rate of glucose in fed-batch cultivations and they were found to correlate. It was also shown that the amount of outer membrane proteins was affected by the feed rate of glucose and by secretion of a recombinant product to the periplasm. The cell surface is another compartment where recombinant proteins can be expressed. Surface display of proteins is a potentially attractive production strategy since it offers a simple purification scheme and possibilities for on-cell protein characterisation, and may in some cases also be the only viable option. The AIDA-autotransporter was applied for surface display of the Z domain of staphylococcal protein A under control of the aidA promoter. Z was expressed in an active form and was accessible to the medium. Expression was favoured by growth in minimal medium and it seemed likely that expression was higher at higher feed rates of glucose during fed-batch cultivation. A repetitive batch process was developed, where relatively high cell densities were achieved whilst maintaining a high expression level of Z. / QC 20110608
24

Étude et inhibition de l'adhésine impliquée dans l'adhérence diffuse (AIDA-I) d'escherichia coli

Girard, Victoria January 2008 (has links)
Thèse numérisée par la Division de la gestion de documents et des archives de l'Université de Montréal
25

Étude de la biogenèse de l'autotransporteur AIDA-I d'Escherichia coli

Charbonneau, Marie-Ève 04 1900 (has links)
Les autotransporteurs monomériques, appartenant au système de sécrétion de type V, correspondent à une famille importante de facteurs de virulence bactériens. Plusieurs fonctions, souvent essentielles pour le développement d’une infection ou pour le maintien et la survie des bactéries dans l’organisme hôte, ont été décrites pour cette famille de protéines. Malgré l’importance de ces protéines, notre connaissance de leur biogenèse et de leur mécanisme d’action demeure relativement limitée. L’autotransporteur AIDA-I, retrouvé chez diverses souches d’Escherichia coli, est un autotransporter multifonctionnel typique impliqué dans l’adhésion et l’invasion cellulaire ainsi que dans la formation de biofilm et d’agrégats bactériens. Les domaines extracellulaires d’autotransporteurs monomériques sont responsables de la fonctionnalité et possèdent pratiquement tous une structure caractéristique d’hélice β. Nous avons mené une étude de mutagenèse aléatoire avec AIDA-I afin de comprendre la base de la multifonctionnalité de cette protéine. Par cette approche, nous avons démontré que les domaines passagers de certains autotransporteurs possèdent une organisation modulaire, ce qui signifie qu’ils sont construits sous la forme de modules fonctionnels. Les domaines passagers d’autotransporteurs peuvent être clivés et relâchés dans le milieu extracellulaire. Toutefois, malgré la diversité des mécanismes de clivage existants, plusieurs protéines, telles qu’AIDA-I, sont clivées par un mécanisme qui demeure inconnu. En effectuant une renaturation in vitro d’AIDA-I, couplée avec une approche de mutagenèse dirigée, nous avons démontré que cette protéine se clive par un mécanisme autocatalytique qui implique deux acides aminés possédant un groupement carboxyle. Ces résultats ont permis la description d’un nouveau mécanisme de clivage pour la famille des autotransporteurs monomériques. Une des particularités d’AIDA-I est sa glycosylation par une heptosyltransférase spécifique nommée Aah. La glycosylation est un concept plutôt récent chez les bactéries et pour l’instant, très peu de protéines ont été décrites comme glycosylées chez E. coli. Nous avons démontré que Aah est le prototype pour une nouvelle famille de glycosyltransférases bactériennes retrouvées chez diverses espèces de protéobactéries. La glycosylation d’AIDA-I est une modification cytoplasmique et post-traductionnelle. De plus, Aah ne reconnaît pas une séquence primaire, mais plutôt un motif structural. Ces observations sont uniques chez les bactéries et permettent d’élargir nos connaissances sur la glycosylation chez les procaryotes. La glycosylation par Aah est essentielle pour la conformation d’AIDA-I et par conséquent pour sa capacité de permettre l’adhésion. Puisque plusieurs homologues d’Aah sont retrouvés à proximité d’autotransporteurs monomériques putatifs, cette famille de glycosyltranférases pourrait être importante, sinon essentielle, pour la biogenèse et/ou la fonction de nombreux autotransporteurs. En conclusion, les résultats présentés dans cette thèse apportent de nouvelles informations et permettent une meilleure compréhension de la biogenèse d’une des plus importantes familles de protéines sécrétées chez les bactéries Gram négatif. / Monomeric autotransporters, a family of proteins that use the type V secretion pathway, are important mediators of virulence for many bacterial pathogens. Many functions important for host colonization and survival have been described for these proteins. Despite the recognized importance of this family of proteins, the mechanisms that are required for the biogenesis and functionality of monomeric autotransporters still remain poorly understood. The Escherichia coli adhesin involved in diffuse adherence (AIDA-I) is a classical multifunctional autotransporter protein that mediates bacterial aggregation and biofilm formation, as well as adhesion and invasion of cultured epithelial cells. Extracellular domains of autotransporters are responsible for the protein function and fold into a characteristic β-helical structure. We performed a random mutagenesis of the AIDA-I passenger domain in order to identify regions involved in the various phenotypes associated with the expression of this protein. Our study suggests that the passenger domain of AIDA-I possesses a modular organization, which means that AIDA-I is built with individual functional modules. Autotransporter passenger domains can be cleaved from the β-domain and released into the extracellular milieu. However, despite the fact that diverse cleavage mechanisms have been previously described, many autotransporters, like AIDA-I, are cleaved by an unknown mechanism. By monitoring the in vitro refolding and cleavage following by site-directed mutagenesis, we showed that AIDA-I processing is an autocatalytic event that involves two acidic residues. Our results unveil a new mechanism of auto-processing in the autotransporter family. AIDA-I is one of the few glycosylated proteins found in Escherichia coli. Glycosylation is mediated by a specific heptosyltransferase encoded by the aah gene, but little is known about the role of this modification and the mechanism involved. Our findings suggest that Aah represents the prototype of a new large family of bacterial protein O-glycosyltransferases that modify various substrates recognized through a structural motif. Furthermore, we showed that glycosylation occurs in the cytoplasm by a cotranslational mechanism. These observations are unique in bacteria and represent a significant advance in our comprehension of prokaryotic glycosylation. We also showed that glycosylation is required to ensure a normal conformation of AIDA-I and, as a consequence, is necessary for its cell-binding function. The finding that other autotransporters or large adhesin-encoding genes are linked to Aah homologue-encoding genes suggests that glycosylation may be important, if not essential, for the function of these proteins, as for AIDA-I. In conclusion, the results presented in this thesis bring new information about the autotransporter family and also give new insight into the mechanisms that are important for different aspects of the biogenesis of monomeric autotransporters.
26

Les autotransporteurs auto-associatifs d’Escherichia coli : de facteurs de virulence à déterminants sociaux

Côté, Jean-Philippe 07 1900 (has links)
Les autotransporteurs monomériques représentent le système de sécrétion le plus simple et le plus utilisé chez les bactéries à Gram négatif. Les autotransporteurs monomériques sont des protéines modulaires qui contiennent toute l’information pour leur sécrétion dans leur séquence. Les phénotypes associés à l’expression d’un autotransporteur peuvent être très variés et, souvent, les autotransporteurs sont des protéines multifonctionnelles. C’est le cas notamment des autotransporteurs AIDA-I, TibA et Ag43 d’Escherichia coli qui promouvoient l’adhésion et l’invasion de cellules épithéliales, l’auto-agrégation des bactéries et la formation de biofilm. Ces trois autotransporteurs ont d’ailleurs été regroupés dans une même famille, appelée les autotransporteurs auto-associatifs (SAATs). À cause de leur fonctionnalité, les SAATs sont considérés comme étant d’importants facteurs de virulence d’Escherichia coli. Toutefois, il existe plusieurs différences entre les SAATs qui ne sont pas bien comprises, si bien que leur rôle pour les bactéries n’est toujours pas bien compris. Nous avons donc d’abord caractérisé TibA, le membre des SAATs le moins bien étudié à l’aide d’une étude structure-fonction. Nous avons observé que TibA était une protéine modulaire et que son domaine fonctionnel était composé de deux modules : un module d’auto-agrégation en N-terminal et un module d’adhésion en C-terminal. En comparant nos résultats avec ceux obtenus pour les autres SAATs, nous avons réalisé que l’organisation des trois SAATs était très variée, c’est-à-dire que les trois SAATs sont composés de modules différents. Nous avons par ailleurs observé cet arrangement en modules lorsque nous avons analysé plusieurs séquences d’aidA, suggérant qu’un mécanisme d’échange et d’acquisition de modules était à la base de l’évolution des SAATs. Sans surprise, nous avons aussi observé que la famille des SAATs ne se limitait pas à AIDA-I, TibA et Ag43 et ne se limitait pas à Escherichia coli. La comparaison a aussi révélé l’importance du phénotype d’auto-agrégation dans la fonctionnalité des SAATs. Nous avons donc entrepris une étude du mécanisme d’auto-agrégation. Nos résultats on montré que l’auto-agrégation était le résultat d’une interaction directe SAAT/SAAT et ont mis en évidence un mécanisme similaire à celui utilisé par les cadhérines eucaryotes. De plus, nous avons observé que, comme les cadhérines, les SAATs étaient impliqués dans des interactions homophiliques; un SAAT interagit donc spécifiquement avec lui-même et non avec un différent SAAT. Finalement, les SAATs font parties des quelques protéines qui sont glycosylées chez Escherichia coli. Nous avons déterminé que le rôle de la glycosylation de TibA était de stabiliser la protéine et de lui donner la flexibilité nécessaire pour moduler sa conformation et, ainsi, être pleinement fonctionnelle. Globalement, nos résultats suggèrent que les SAATs sont des molécules « cadhérines-like » qui permettent la reconnaissance de soi chez les bactéries. Une telle habilité à discriminer entre le soi et le non-soi pourrait donc être utilisée par les bactéries pour organiser les communautés bactériennes. / Autotransporters are versatile virulence factors of Gram-negative bacteria and use one of the simplest and most widespread secretion system in bacteria. The name autotransporter originate from the observation that all the information needed for the secretion of the protein is encoded in its own sequence, meaning that autotransporters do not need a specialized secretion apparatus. Many autotransporters are multifunctional proteins and can perform a large variety of functions. The self-associating autotransporters (SAATs), represented by AIDA-I, TibA and Ag43, are such multifunctional proteins and can mediate the adhesion and invasion of epithelial cells, the auto-aggregation of bacteria and the formation of biofilm. Because of these functionalities, SAATs are considered important virulence factors of Escherichia coli. However, there are many differences between the SAATs and we still do not know their exact role for the bacteria. Therefore, we have realized a structure-function study of TibA, the least studied SAAT. Our study showed that TibA is a modular protein and that the functional domain of TibA is composed of two modules: an N-terminal module responsible for auto-aggregation and a C-terminal module responsible for adhesion. Our results showed that the organization of AIDA-I, TibA and Ag43 is different and that the SAATs represent different assemblies of modules. We also observed the modular organization when we analyzed various sequence of aidA, suggesting that the SAATs have evolved by a mechanism of domain shuffling. Not surprisingly, we have found new SAATs in Escherichia coli and in other proteobacteria. Our results also highlighted the importance of auto-aggregation in the functionality of the SAATs. We therefore assessed the mechanism of SAAT-mediated auto-aggregation of bacteria. Our results showed that SAATs mediate auto-aggregation of bacteria through direct SAAT/SAAT interactions and that these interactions were reminiscent of the interactions made by cadherin molecules in eukaryotes. We further observed that the SAATs were involved in homophilic interactions, as it is the case with cadherin molecules. SAATs are part of the few proteins that are glycosylated in Escherichia coli. We therefore characterized the glycosylation of TibA and found that glycosylation of TibA stabilized the protein and allowed the protein to modulate its conformation, resulting in a fully functional protein. Taken together, our results suggest that the SAATs may be cadherin-like molecules by bacteria in order to discriminate between self and non-self. Such an ability to discriminate self from non-self is rarely evoked in bacteria, but could play a role in the organization of multicellular communities.
27

Les bactéries exprimant AIDA-I interagissent avec l'apolipoprotéine A-I cellulaire

Létourneau, Jason 08 1900 (has links)
AIDA-I (adhesin involved in diffuse adherence) est une importante adhésine autotransporteur exprimée par certaines souches de Escherichia. coli impliquée dans la colonisation des porcelets sevrés causant la diarrhée post-sevrage et la maladie de l’œdème. Une précédente étude de notre laboratoire a identifié l’apolipoprotéine AI (ApoAI) du sérum porcin, la protéine structurale des lipoprotéines à haute densité, comme récepteur cellulaire putatif de AIDA-I. L’interaction entre ces deux protéines doit être caractérisée. Ici, nous montrons par ELISA que AIDA-I purifiée est capable d’interagir avec l’ApoAI humaine, mais également avec les apolipoprotéines B et E2. L’ApoAI est rencontrée sous deux formes, soit libre ou associée aux lipides. Nous montrons que la forme libre n’interagit pas avec les bactéries AIDA-I+ mais s’associe spécifiquement à l’ApoAI membranaire de cellules épithéliales HEp-2. Afin d’étudier le rôle de l’ApoAI dans l’adhésion des bactéries, nous avons infecté des cellules HEp-2 en présence d’anticorps dirigés contre l’ApoAI, mais l’adhésion des bactéries AIDA I+ n’a jamais été réduite. De plus, l’induction de l’expression de l’ApoAI par fénofibrate et GW7647 chez les cellules Caco 2 polarisée et Hep G2, n’a pas permis l’augmentation de l’adhésion cellulaire des E. coli exprimant AIDA-I. Notre étude suggère davantage que l’interaction entre AIDA-I et ApoAI n’intervient pas dans les mécanismes d’adhésion cellulaire. / The adhesin involved in diffuse adherence (AIDA-I) is an important autotransporter adhesin expressed by some strains of Escherichia coli and is involved in the intestinal colonisation of weaned piglets, causing the postweaning diarrhea and the edema disease. A previous study from our laboratory identified the apolipoprotein AI (ApoAI) from porcine serum, the structural protein of high density lipoproteins, as a putative receptor of AIDA-I. The interaction between these two proteins must be characterized. Here, we show that purified AIDA-I, using an ELISA assay, is able to bind the human ApoAI and the apolipoprotein B and E2. The ApoAI is found under two forms, either free or bound to lipid. We show that the free form of ApoAI does not interact with AIDA-I+ bacteria but specifically interact with membrane bound ApoAI on Hep-2 epithelial cells. To study the role of ApoAI in the adhesion of bacteria, we infected Hep-2 cells preincubated with antibodies to ApoAI. The adhesion of AIDA-I+ bacteria to the cells couldn’t be reduced. Additionally, the induction of ApoAI synthesis using fenofibrate and GW7647 on polarized Caco-2 or Hep G2 cells did not increase the adhesion of AIDA-I+ bacteria. Our study suggests that the interaction between AIDA-I and ApoAI is not involved in the cellular adhesion of the bacteria.
28

L’apolipoprotéine A-I interagit avec l’adhésine impliquée dans l’adhérence diffuse (AIDA-I) d’Escherichia coli : rôle lors du processus d’adhésion et d’invasion

René, Mélissa 05 1900 (has links)
L’adhésine impliquée dans l’adhérence diffuse (AIDA-I) est une adhésine bactérienne présente chez certaines souches d’Escherichia coli qui, associée aux toxines Stx2e ou STb, contribue à l’apparition de la maladie de l’œdème ou de la diarrhée post-sevrage chez les porcelets. AIDA-I est un autotransporteur qui confère des capacités d’autoaggrégation, de formation de biofilms et d’adhésion. L’objectif principal du projet de recherche consistait en la recherche de récepteur(s) potentiel(s) d’AIDA-I. Les bactéries pathogènes adhèrent aux cellules-cibles soit en liant directement des molécules à la surface cellulaire ou en utilisant des molécules intermédiaires qui permettent de diminuer la distance séparant la bactérie de la cellule-cible. Puisque le sérum est un fluide qui contient de nombreuses molécules, celui-ci a été utilisé comme matériel de départ pour l’isolement de récepteur(s) potentiels. Nous avons isolé un récepteur potentiel à partir du sérum porcin : l’apolipoprotéine A-I. L’interaction entre l’apolipoprotéine A-I et AIDA-I a été confirmée par ELISA et microscopie à fluorescence. La capacité à envahir les cellules épithéliales offre aux pathogènes la possibilité d’établir une niche intracellulaire qui les protègent contre les attaques du milieu extérieur. La présente étude a démontré que la présence d’AIDA-I en tant que seul facteur de virulence chez une souche de laboratoire permet de conférer la capacité d’envahir les cellules sans promouvoir la survie intracellulaire. L’étude de la souche sauvage 2787, exprimant AIDA-I en association avec d’autres facteurs de virulence, a démontré une différence significative pour les phénotypes d’invasion et de survie intracellulaire face à la souche de laboratoire exprimant AIDA-I. / The adhesin involved in diffuse adherence (AIDA-I) is a bacterial adhesin associated with some Escherichia coli strains that might, when associated with toxin Stx2e or STb, contribute to the development of edema disease or post-weaning diarrhea in piglets. AIDA-I is an autotransporter that mediates various phenotypes such as adhesion, autoaggregation and biofilm formation. The main aim of our project was to find potential receptor(s) for AIDA-I. Pathogens can either bind cell directly by targeting exposed cell surface molecules or use an intermediate molecule as a bridge to lessen the space separating them from their target cell. Serum is known to contain a wide range of molecules so it has been used as raw material for the isolation of a putative receptor for AIDA-I. We isolated a putative receptor for AIDA-I: the apolipoprotein A-I. The interaction between the apolipoprotein A-I and AIDA-I was confirmed by ELISA and fluorescent microscopy. The capacity to invade epithelial cell enables pathogens to create an intracellular niche that protects them against attacks from the extracellular environment. The present report has shown that the presence of AIDA-I as the sole virulence factor in a laboratory strain, enable bacteria to invade cultured cells but does not promote intracellular survival. Studies conducted on wild-type strain 2787, which express AIDA-I in association with other virulence factors, has shown a significant difference in invasion and intracellular survival phenotypes compared to the laboratory strain expressing AIDA-I.
29

L’apolipoprotéine A-I interagit avec l’adhésine impliquée dans l’adhérence diffuse (AIDA-I) d’Escherichia coli : rôle lors du processus d’adhésion et d’invasion

René, Mélissa 05 1900 (has links)
L’adhésine impliquée dans l’adhérence diffuse (AIDA-I) est une adhésine bactérienne présente chez certaines souches d’Escherichia coli qui, associée aux toxines Stx2e ou STb, contribue à l’apparition de la maladie de l’œdème ou de la diarrhée post-sevrage chez les porcelets. AIDA-I est un autotransporteur qui confère des capacités d’autoaggrégation, de formation de biofilms et d’adhésion. L’objectif principal du projet de recherche consistait en la recherche de récepteur(s) potentiel(s) d’AIDA-I. Les bactéries pathogènes adhèrent aux cellules-cibles soit en liant directement des molécules à la surface cellulaire ou en utilisant des molécules intermédiaires qui permettent de diminuer la distance séparant la bactérie de la cellule-cible. Puisque le sérum est un fluide qui contient de nombreuses molécules, celui-ci a été utilisé comme matériel de départ pour l’isolement de récepteur(s) potentiels. Nous avons isolé un récepteur potentiel à partir du sérum porcin : l’apolipoprotéine A-I. L’interaction entre l’apolipoprotéine A-I et AIDA-I a été confirmée par ELISA et microscopie à fluorescence. La capacité à envahir les cellules épithéliales offre aux pathogènes la possibilité d’établir une niche intracellulaire qui les protègent contre les attaques du milieu extérieur. La présente étude a démontré que la présence d’AIDA-I en tant que seul facteur de virulence chez une souche de laboratoire permet de conférer la capacité d’envahir les cellules sans promouvoir la survie intracellulaire. L’étude de la souche sauvage 2787, exprimant AIDA-I en association avec d’autres facteurs de virulence, a démontré une différence significative pour les phénotypes d’invasion et de survie intracellulaire face à la souche de laboratoire exprimant AIDA-I. / The adhesin involved in diffuse adherence (AIDA-I) is a bacterial adhesin associated with some Escherichia coli strains that might, when associated with toxin Stx2e or STb, contribute to the development of edema disease or post-weaning diarrhea in piglets. AIDA-I is an autotransporter that mediates various phenotypes such as adhesion, autoaggregation and biofilm formation. The main aim of our project was to find potential receptor(s) for AIDA-I. Pathogens can either bind cell directly by targeting exposed cell surface molecules or use an intermediate molecule as a bridge to lessen the space separating them from their target cell. Serum is known to contain a wide range of molecules so it has been used as raw material for the isolation of a putative receptor for AIDA-I. We isolated a putative receptor for AIDA-I: the apolipoprotein A-I. The interaction between the apolipoprotein A-I and AIDA-I was confirmed by ELISA and fluorescent microscopy. The capacity to invade epithelial cell enables pathogens to create an intracellular niche that protects them against attacks from the extracellular environment. The present report has shown that the presence of AIDA-I as the sole virulence factor in a laboratory strain, enable bacteria to invade cultured cells but does not promote intracellular survival. Studies conducted on wild-type strain 2787, which express AIDA-I in association with other virulence factors, has shown a significant difference in invasion and intracellular survival phenotypes compared to the laboratory strain expressing AIDA-I.

Page generated in 0.1045 seconds