• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 10
  • 6
  • 2
  • 1
  • 1
  • 1
  • Tagged with
  • 22
  • 22
  • 11
  • 7
  • 7
  • 6
  • 6
  • 5
  • 5
  • 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

Developing and Utilizing a Next-Generation Humanized Mouse Model for Investigating HIV and Tuberculosis

Lepard, Madeleine January 2022 (has links)
Infection & Immunity / Currently, there are 38 million people living with human immunodeficiency virus (HIV-1) worldwide and there were 680,000 HIV-related deaths in 2020 alone. The greatest cause of mortality in people living with HIV (PLHIV) is infection with opportunistic pathogens such as tuberculosis (TB), which accounts for one third of HIV-related deaths. PLHIV are 20 times more susceptible to TB and co-infection leads to significantly worsened outcomes in terms of both diseases. Humanized mouse (hu-mouse) models, which possess human immune cells for HIV to infect, have been useful for HIV research. Our aim is to create hu- mouse models of HIV, TB and co-infection to investigate disease progression, immune responses, therapeutics, prevention and vaccination. NOD-Rag1null-IL2rgnull (NRG) mice are highly immunocompromised mice that are traditionally used to generate hu-mouse models. We are also developing NRG mice that are transgenic for human HLA-DR4 and HLA-A2 (DRAG-A2) and similar mice have been reported to have improved immune responses. NRG and DRAG-A2 mice were humanized with hematopoietic stem cells obtained from human umbilical cord blood. DRAG-A2 mice had significantly higher engraftment success rates (defined as the percentage of mice with >10% hCD45+) as well as higher overall CD45+ leukocyte, CD4+ T cell, CD19+ B cell and CD14+ monocyte reconstitution in the blood compared to huNRGs. huNRG mice were permissive to infection with JR-CSF or NL4.3-Bal-Env HIV-1 intravaginally or systemically. huDRAG-A2 mice were also infected intravaginally with NL4.3-Bal-Env HIV-1. huDRAG-A2 mice, but not huNRGs, produced HIV-specific IgG, indicating improved immune responses. huNRG mice were infected intranasally with mCherry-Erdman, YFP-H37Rv or H37Rv Mtb. huDRAG-A2 mice were also infected with H37Rv. Human immune cell involvement and human-like granuloma formation was observed using flow cytometry and immunohistopathology. These findings show that the DRAG-A2 model may be optimal for investigating HIV, TB and co-infection, which continue to be serious global health concerns. / Thesis / Master of Science (MSc) / Human immunodeficiency virus (HIV) and tuberculosis (TB) are infectious diseases that affect millions of people worldwide every year. The greatest cause of death in people living with HIV is co-infection with TB and HIV-positive individuals are much more likely to get TB. Humanized mouse (hu-mouse) models possess human immune cells for HIV to infect and are useful for studying HIV. Our goal is to create hu-mouse models of HIV, TB and HIV/TB co-infection that will allow us to study how these diseases interact. We are currently developing a traditional hu-mouse model (known as NRG), as well as an improved next-generation model (known as DRAG-A2) with a more functional immune system. Both models have been successfully infected with HIV or TB. Only DRAG-A2 mice were able to make antibodies against HIV. The improved DRAG-A2 model will enable future studies on HIV, TB and co-infection, which continue to be understudied global problems.
2

Creating and Use of an New Experimental Preclinical HLA Transgenic Mice Model to Mapping HLA-restricted T Cells Epitopes for Polyepitopes Vaccine Design / Exploitation d’un modèle expérimentale préclinique de souris HLA transgénique pour l’identification des épitopes T HLA-restreint afin de concevoir des vaccins poly-epitopiques

Ru, Zhitao 21 February 2012 (has links)
Une nouvelle lignée homozygote de souris « humanisé » HLA transgéniques : HLA-A2.1+/+HLA-DP4+/+hCD4+/+mCD4-/-IAβ-/-β2m-/- (HLA-A2/DP4), a été obtenue en croisant entre les souris HLA transgéniques HLA-A2.1+/+β2m-/-(A2) et les souris HLA transgéniques HLA-DP4+/+hCD4+/+mCD4-/-IAβ-/-(DP4). Chez les souris HLA-A2/DP4, les réponses cellulaire T HLA-A2 restreint ou HLA-DP4 restreint contre l’antigène HBs du virus de l’Hépatite B suite à une immunisation avec le vaccin anti-AgHBs sont similaires à ceux observés chez les souris A2, chez les souris DP4, chez les humains infectés par le virus ou immunisés avec le même vaccin. Ces résultats montrent que les réponses cellulaires induites les souris HLA-A2/DP4 miment fidèlement les réponses homologues humaines. Ainsi, ces souris représentent un excellent modèle d’expérimentations précliniques animal pour évaluer ou comparer l’efficacité des réponses « humain » induites in vivo par des candidates vaccins. Le modèle facilitera également l'identification de nouvelles épitopes HLA-A2 et HLA-DP4 restreints, qui constituera de futurs réactives de suivi clinique des réponses contre l'infection chez les humains.En exploitant ces souris HLA-A2/DP4, nous avons identifié quatre nouveaux HLA-DP4-restricted épitopes issus de l'AgHBs et deux nouveaux HLA-A2 restreint épitopes dérivés de protéines M1. / A new homozygous humanized HLA transgenic mouse strain, HLA-A2.1+/+HLA-DP4+/+hCD4+/+mCD4-/-IAβ-/-β2m-/- (HLA-A2/DP4), was obtained by crossing the HLA transgenic HLA-A2.1+/+β2m-/-(A2) mice and HLA transgenic HLA-DP4+/+hCD4+/+mCD4-/-IAβ-/-(DP4) mice. In HLA-A2/DP4 mice, HLA-A2 restricted or HLA-DP4 restricted T cell responses against HBs antigen of hepatitis B virus after immunization with the HBsAg vaccine are similar to those induced in A2 mice, in DP4 mice, in HBV-infected or HBsAg-vaccinated humans. These results show that cellular responses induced in HLA-A2/DP4 mice faithfully mimic human responses counterparts. Thus, these mice represent an excellent animal model for preclinical experimentations to evaluate or compare the effectiveness of responses "human" induced in vivo by candidate vaccines. The model will also facilitate the identification of new epitopes HLA-A2 and HLA-DP4 restricted, which will be of future reactive for clinical monitoring response against infection in humans. By exploiting these HLA-A2/DP4 mice, we identified four new HLA-DP4-restricted epitopes from HBsAg and two new HLA-A2 restricted epitopes derived from protein M1.
3

Creating and use of an new experimental preclinical HLA transgenic mice model to mapping HLA-restricted T cells epitopes for polyepitopes vaccine design.

Ru, Zhitao 21 February 2012 (has links) (PDF)
A new homozygous humanized HLA transgenic mouse strain, HLA-A2.1+/+HLA-DP4+/+hCD4+/+mCD4-/-IAβ-/-β2m-/- (HLA-A2/DP4), was obtained by crossing the HLA transgenic HLA-A2.1+/+β2m-/-(A2) mice and HLA transgenic HLA-DP4+/+hCD4+/+mCD4-/-IAβ-/-(DP4) mice. In HLA-A2/DP4 mice, HLA-A2 restricted or HLA-DP4 restricted T cell responses against HBs antigen of hepatitis B virus after immunization with the HBsAg vaccine are similar to those induced in A2 mice, in DP4 mice, in HBV-infected or HBsAg-vaccinated humans. These results show that cellular responses induced in HLA-A2/DP4 mice faithfully mimic human responses counterparts. Thus, these mice represent an excellent animal model for preclinical experimentations to evaluate or compare the effectiveness of responses "human" induced in vivo by candidate vaccines. The model will also facilitate the identification of new epitopes HLA-A2 and HLA-DP4 restricted, which will be of future reactive for clinical monitoring response against infection in humans. By exploiting these HLA-A2/DP4 mice, we identified four new HLA-DP4-restricted epitopes from HBsAg and two new HLA-A2 restricted epitopes derived from protein M1.
4

Challenging Development of a Humanized Mouse Model for Evaluating the HTLV-1 Infection and Leukemogenic Process in vivo

Villaudy, Julien 22 December 2011 (has links) (PDF)
Human T-cell Leukemia Virus type 1 (HTLV-1) is the etiologic agent of the Adult T-cell Leukemia (ATL), an aggressive lymphoproliferation of activated CD4+ T cells. The lack of a reliable small animal model to reproduce in vivo the leukemogenic process associated with HTLV-1 infection has impaired the understanding of the early stages of this process as well as the discovery of effective therapeutic approaches. Recently, improvement in the models of humanized mouse models were achieved allowing the development of a human immune system in mice. Injection of human hematopoietic stem and progenitors cells purified from cord blood into Balb/c Rag2-/-γc-/- newborns allows the de novo production of human dendritic, B and T cells. We infected humanized mice with HTLV-1 producing cell lines resulting in infection of human cells within the mice and the development of lymphomas and leukemias. This infection also results in the alteration of the T-cell development within the thymus pushing the thymocytes toward a more mature phenotype. This small animal model recapitulating in vivo the HTLV-1 infection and its associated pathogenesis gave us the opportunity to study the evolution of the clonality of the virus among human cells in different lymphoid organs. Based on these observations, preliminary results on the use of a new therapeutic approach were obtained. We finally tried to adjust the humanization protocol in order to obtain better engraftment in this model.
5

Etude pré-clinique d'une stratégie de thérapie génique de l'infection par le VIH combinant l'expression de deux inhibiteurs d'entrée / Pre-clinical study of an HIV gene therapy strategy combining two entry inhibitors

Petit, Nicolas 30 September 2014 (has links)
Parmi les nouvelles stratégies thérapeutiques, la thérapie génique contre le VIH connaît actuellement un regain d'intérêt. La description du premier cas de guérison avéré après greffe de cellules souches mutantes pour le corécepteur majeur du VIH (CCR5) a relancé l'intérêt de la communauté pour développer des stratégies de thérapies géniques ciblant CCR5. Durant ma thèse, j'ai développé une stratégie basée sur l'inhibition de l'entrée du virus dans les lymphocytes T CD4 humains. Nos gènes thérapeutiques comprennent un inhibiteur de fusion membranaire (peptide C46) et un inhibiteur d'expression de CCR5. L'expertise de l'équipe dans les modèles de souris humanisées m'a permis de valider l'intérêt pré-clinique de cette stratégie dans un modèle de transfert de lymphocytes T modifiés. J'ai pu montrer que les lymphocytes T modifiés possédaient un avantage sélectif massif in vivo comparé à des cellules non protégées, ainsi qu’une protection totale contre la délétion induite par le VIH. De plus, dans une autre étude, j'ai montré que la dynamique virale dans les modèles de souris humanisées par transfert de CSH est plus proche des patients. Cette étude nous montre qu'il n'est pas nécessaire d'invoquer la réponse immune, pour expliquer les profils de réplication virale observés dans la phase primaire de l'infection. Ce résultat suggère donc qu'une partie de la dynamique virale est dépendante du nombre de cellules à infecter et du nombre de virions disponibles. L'ensemble des résultats de ma thèse a permis (i) de faire progresser notre compréhension de la dynamique virale, et (ii) de valider la combinaison d'inhibiteurs d'entrée dans un vecteur lentiviral. / Among new therapeutic strategies to fight the infection, gene therapy targeting CCR5 will be an asset. Indeed, the first patient to be cured from the infection received hematopoietic progenitors deprived of CCR5. This finding has ignited a flurry of research on genetic means to decrease CCR5 from the cell surface.During my PhD, I developed a gene therapy strategy based on entry inhibition of HIV in CD4 T cells. Our therapeutic genes are a membrane fusion inhibitor (the C46 peptide) and a CCR5 expression inhibitor, based on a super-agonist ligand. The experience of the team in humanized mice models allowed the validation of the strategy in vivo in a model of gene-modified T cell transfer. I first showed that the combination of the transgenes was able to prevent HIV infection in vitro in a dose dependent manner. Most importantly, I then showed that this protection conferred modified T cells with a huge selective advantage in vivo. This advantage was associated with a complete protection of CD4 T cells from HIV-induced deletion. Moreover, in another study, I showed that the viral dynamics in mouse models humanized by transferring HSC is closer to patients. This study also teaches us that it is not necessary to invoke the immune response to HIV, to explain the profiles of viral replication observed early after infection. This result suggests that part of the viral dynamic is dependent on the target/virus ratio. Together, results collected during my PhD thesis provides new information on viral dynamic and establish the interest of combining two viral entry inhibitors for gene therapy of HIV infection.
6

Développement d’un modèle préclinique de leucémogénèse expérimentale chez la souris humanisée / Construction of a preclinical model of leucemogenesis in humanized mice

Dupont, Salomé 12 December 2017 (has links)
Les modèles animaux actuellement disponibles pour l’étude des leucémies humaines ne sont pas adaptés pour le développement optimal de nouvelles thérapies ciblées. Au cours de ce projet, qui s’inscrit dans une double perspective fondamentale et industrielle, nous avons cherché à générer un modèle versatile de leucémogénèse humaine chez la souris humanisée BRGS (BALB/c Rag2-/- IL-2Rγc-/- SIRPα.NOD). Les animaux sont greffés avec des progéniteurs hématopoïétiques transduits par des vecteurs lentiviraux surexprimant les oncogènes MYC et BCL2 et placés sous le contrôle d’un promoteur ubiquitaire (EF1α ou SFFV). Le suivi longitudinal des animaux sur une période de 5 mois montre que seule la construction SFFV/Myc-T2A-Bcl2 entraîne la transformation des progéniteurs hématopoïétiques. Entre 12 à 14 semaines post-greffe, >90% des animaux développent des lympho-proliférations de type pro-B (CD19+CD10+CD9+CD20-cytIgM-) infiltrant principalement la rate et la moelle osseuse, et circulant en abondance dans le sang. Le caractère transmissible des tumeurs est validé par des greffes secondaires de tumeurs spléniques. Les cultures in vitro de progéniteurs hématopoïétiques suggèrent que l’émergence des blastes est liée à la réactivation d’un programme B latent dans les précurseurs T, dont le développement est bloqué. Nous avons développé en parallèle un modèle de tumeur autologue. L’ensemble de ces résultats valide le modèle de leucémogénèse humaine développé chez la souris humanisée BRGS et ouvre des perspectives pour la caractérisation fonctionnelle des mécanismes de leucémogénèse, et la validation préclinique de nouvelles stratégies anti-tumorales. / Existing animal models for the study of human leukemia are not accurate for the proper development of innovative, targeted therapies. The aim of this project, which contains both a fundamental and an industrial perspective, therefore was to develop a new, versatile model of human leukemogenesis in the BRGS (BALB/c Rag2-/- IL-2Rγc-/- SIRPα.NOD) humanized mouse. Animals are grafted with hematopoietic progenitors transduced with lentivirals vectors to allow overexpression of MYC and BCL2 proteins under the control of an ubiquitous promotor (EF1α or SFFV). Longitudinal monitoring of the animals over five months shows that only the SFFV/Myc-T2A-Bcl2 construction induces the transformation of humans hematopoietics progenitors. Between 12 and 14 weeks post-transplantation, more than 90% of the animals develop pro-B lympho-proliferations (CD19+CD10+CD9+CD20-cytIgM-), with tumor cells being mainly found in the spleen, the bone marrow and in blood. Tumor transferability is achievable through secondary transplantation in immunodeficient mouse recipients. In vitro culture of bone marrow T cell progenitors suggest that the blasts arise from these cells after reactivation of a latent B cell program with blockade of their T cell development. In parallel, we have also developed an autologous tumor model. Altogether, these results validate the human leukemogenesis model constructed here in humanized BRGS mice and provide attractive prospects regarding the functional characterization of leukemogenesis and a preclinical validation of new anti-tumor strategies.
7

Microscopic morphomolecular characterization of humanized mouse models of SARS-CoV-2 implanted with human fetal lung xenografts

Montanaro, Paige 24 November 2021 (has links)
INTRODUCTION: SARS-CoV-2 is a novel virus from the coronavirus family that emerged in the Hubei province of China in December 2019 and rapidly spread throughout the world. On March 11, 2020, the World Health Organization declared a global pandemic. Infection with SARS-CoV-2 causes coronavirus disease 19 (COVID19) which can be fatal. There is an obvious and pressing need for research surrounding SARS-CoV-2 that will aid in eradication of this pandemic. OBJECTIVE: The goal of this study was to absolve the dire need for small animal models of human disease that demonstrate hallmark pathological features of infection. Due to ethical and financial obstacles, the use of animals that closely resemble human immunity, such as non-human primates, is often not a viable option. For this reason, there is a push to develop small animal models that can mimic human disease responses, particularly those in viral infections that have a narrow species tropism. To achieve this in the context of the novel coronavirus, SARS-CoV-2, we studied various mouse models and their capacity to become infected with and mount an immune response to SARS-CoV-2. Our goal was to identify a model that sufficiently mimics severe COVID19 seen in humans as well as provide molecular insight into pathways that prevent the development of severe disease. METHODS: NRG-L and HIS-NRGF-L mice were subcutaneously implanted with human fetal lung xenografts and infected with SARS-CoV-2. Tissues were stained with H&E for histopathological scoring. NRG-L and HIS-NRGF-L tissues were fluorescently labeled using 2 different multiplex immunohistochemistry panels. Slides were digitized by a Vectra Polaris™ fluorescent whole slide scanner and digital analysis was completed using HALO™. Statistical analysis was conducted using GraphPad Prism™ 9.0.1. RESULTS: Infected NRG-L mice present extensive histopathological manifestations when compared to uninfected controls. Cumulative histology scores at both 2 and 7DPI were increased when compared to uninoculated fLX. Neutrophil influx, intra-airspace necrosis, capillary fibrin thrombi, and presence of syncytial cells were the most significant independent observations that contributed to the increased cumulative score. Together these findings indicate that fLX inoculated with SARS-CoV-2 faithfully recapitulate several features of diffuse alveolar damage (DAD) described in severe COVID-19. HIS-NRGF-L mice displayed decreased influx of neutrophils, intra-airway necrosis, and syncytial cells when compared to NRG-L fLX. Hemorrhage was decreased at both 2 and 7 DPI for HIS-NRGF-L fLX compared to NRG-L fLX. Cumulative histology scores were decreased in HIS-NRGF-L fLX at 7 DPI when compared to NRG-L fLX. Taken together these findings support the hypothesis that human myeloid and lymphoid infiltrates suppress or prevent the disparate host response observed in NRGL-L fLX that manifested in pronounced diffuse alveolar damage. CONCLUSION: Using digital image analysis of multiplex immunohistochemistry paired with semi-quantitative histopathological scoring, this study characterized important hallmark lesions observed in severe COVID19 as seen in small animal models. NRG-L and HIS-NRGF-L mice that are subcutaneously implanted with human fetal lung xenografts are susceptible to infection with SARS-CoV-2 and can produce severe and moderate disease phenotypes respectively. Co-engraftment with human fetal lung tissue and human immune system components in HIS-NRGF-L mice suppresses the divergent host response that is observed in NRG-L mice. For this reason, NRG-L mice engrafted with fLX are an ideal small animal model of severe COVID19, whereas HIS-NRGF-L mice severe as a valuable and informative model for deciphering molecular mechanisms driving severe COVID-19 that will serve as targets for therapeutic development.
8

The Neuroimmunological Consequences of Spinal Cord Injury

Carpenter, Randall Scott 02 October 2019 (has links)
No description available.
9

Induktion einer Endotoxämie in der humanisierten Maus

Scholbach, Johanna 24 February 2016 (has links) (PDF)
Die Sepsis ist ein gefürchtetes Krankheitsbild, das in hochentwickelten Industrienationen mit einer hohen Mortalität verknüpft ist und damit zu den häufigsten Todesursachen gehört. Die Pathomechanismen dieses komplexen und heterogenen Krankheitsbildes zu entdecken, gehört momentan zu den Hauptinteressengebieten der Sepsisforschung. Da die Interpretation klinischer Studien aufgrund der Heterogenität des Patientenguts schwierig ist, kommt der Entwicklung adäquater Tiermodelle eine entscheidende Bedeutung zu. Die hierbei gängigen Tiermodelle in Mäusen weisen jedoch Unzulänglichkeiten auf, die die Übertragung der in Tierexperimenten gewonnen Daten auf den klinischen Kontext nur teilweise ermöglichen. Eine Brücke kann hierbei das Tiermodell der humanisierten Maus schlagen, in der, durch Transplantation mit humanen hämatopoetischen Stammzellen, ein humanes Immunsystem reift. Die vorliegende Arbeit beschäftigt sich mit der Fragestellung, inwieweit die humanen Immunzellen in der humanisierten Maus in der Lage sind, auf LPS als Stimmulus zu reagieren. Darüberhinaus wird die Nutzung der Endotoxämie in der humanisierten Maus als alternatives Sepsismodell im Bezug zum klinischen Kontext untersucht. Hierbei ergab sich eine mögliche Nutzung des Endotoxämiemodells in der humanisierten Maus zur genaueren Erforschung des Zytokinmilieus, sowie neuer Surrogatmarker wie Pentraxin 3. Bezüglich der Reaktion einzelner immunologischer Subpopulationen und deren Bedeutung für die Klinik scheint eine Untersuchung an Modellen, die eine B- und T-Zell-Reifung nachvollziehen können und in der murine Residualzellen möglichst gering vorhanden sind, als sinnvoll.
10

Développement d'anticorps monoclonaux humains de type IgA dirigés contre la partie C-terminale de la protéine d'enveloppe gp41 du VIH-1 / Development of human monoclonal IgA antibodies directed against the C-terminal region of the gp41 envelope protein of HIV1

Benjelloun, Fahd 08 July 2013 (has links)
La transmission du Virus de l’Immunodéficience Humaine (VIH) par voie sexuelle représente le mode majoritaire de contamination (80%) (UNAIDS). Ce mode de contamination implique le passage du virus à travers les muqueuses et une interaction avec les cellules épithéliales et les cellules immunitaires présentes au sein de ces muqueuses (cellules dendritiques, macrophages ou lymphocytes). Les muqueuses représentent le principal site d'exposition de l’organisme aux antigènes de l’environnement. Les SIgA (IgA sécrétoires) présentes dans la lumière de ces muqueuses représentent la première ligne de défense immunitaire contre l’infection et la colonisation des muqueuses. Les IgA sont capables d’interagir avec les glycoprotéines (gp) exprimées à la surface du VIH et de bloquer l’infection et/ou la transcytose à travers l’épithélium muqueux. Nous avons pu étudier la prévalence des SIgA anti-gp41 et plus précisément anti-MPER présentes dans la salive parotidienne de personnes Exposées au VIH Séronégatives (ESN) et leur rôle dans l’inhibition de l’infection par le virus in vitro. Nous avons pu démontrer que ces sujets présentaient un taux plus important de SIgA anti-MPER neutralisantes. Ce premier travail nous a permis de valider la gp41 comme immunogène d’intérêt pour la génération de SIgA neutralisantes. Nous avons pu générer des IgA1 dans un modèle murin α1Kl chimérique capable de produire des anticorps IgA1 humanisés. L’immunisation de ces souris a permis la production de 6 anticorps monoclonaux spécifiques de la région MPER capables de reconnaître des épitopes conformationnels élargis, correspondant aux épitopes reconnus par le 2F5 et le 4E10. Les IgA1 présentaient de fortes capacités neutralisantes pour différentes souches de laboratoire et de souches primaires du VIH. Les études de caractérisation des fonctions antivirales de ces anticorps permettront de mieux définir le mode d’action de ces anticorps. A notre connaissance, ces IgA1 neutralisantes anti-MPER sont les premières décrites à ce jour dans la littérature. De par leur faible immunogénicité et leur faible autoréactivité, ces anticorps peuvent facilement être intégrés dans des approches thérapeutiques locales ou par sérothérapie passive pour la protection après administration de SHIV dans des modèles animaux comme le macaque. L’ensemble de mes travaux de thèse ont confirmé l’intérêt thérapeutique potentiel des SIgA dans la lutte contre le VIH et notamment celles dirigées contre la partie gp41 de l’enveloppe / Sexual transmission of the Human Immunodeficiency Virus is the major mode of contamination (80%) for this pathogen (UNAIDS). This mode of transmission involves a passage of the virus though the mucosa and an interaction with epithelial cells and immune cells present in the mucosa (dendritic cells, macrophages and lymphocytes). Mucosa represents the major site of exposure for the organism to environmental antigens. The IgA expressed in the lumen of mucosa are the first line of immune defence against infection and colonization of mucosa. IgA are able to interact with glycoproteins (gp) expressed on the surface of HIV and prevent infection and/or block epithelial transcytosis. In this study we have investigated the prevalence of SIgA anti-MPER present in the parotid saliva of Exposed to HIV but Seronegative individuals (ESN). This study has allowed us to validate gp41 as an immunogen of interest for the generation of neutralizing IgA. IgA1 were generated in a chimeric mice model α1Kl that produced humanized IgA1 type antibodies. Immunizations of these mice has led to the elicitation of six monoclonal antibodies specific to the MPER region able to recognize extended conformational neutralizing epitopes of 2F5 and 4E10, two broadly neutralizing monoclonal antibodies specific to MPER. Elicited IgA1 have potent neutralizing properties for both laboratory and primary HIV strains. Characterization studies of the antiviral functions of these antibodies will further define the mode of action of these antibodies. To our knowledge, these anti-MPER humanized monoclonal neutralizing IgA1 antibodies are the first of this type described to date in the literature. By their low immunogenicity and autoreactivity, these antibodies can be easily integrated into local therapeutic approaches or passive serotherapy for protection in animal models such as the macaque challenged with SHIV. All the results of my PhD work confirm the great interest of gp41-specific SIgA as therapeutic agents against HIV

Page generated in 0.0697 seconds