• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 7
  • 3
  • 1
  • 1
  • 1
  • Tagged with
  • 14
  • 14
  • 10
  • 9
  • 7
  • 7
  • 7
  • 7
  • 6
  • 5
  • 4
  • 4
  • 4
  • 4
  • 4
  • 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

The Influence of 1,25-Dihydroxyvitamin D3 on the Cross-Priming of Lymphocytic Choriomeningitis Virus Nucleoprotein

Kim, Julia 02 September 2011 (has links)
Biologically active 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) binds the vitamin D receptor (VDR) to exert its effect on target cells. VDR expression is found in a number of immune cells including professional antigen-presenting cells such as dendritic cells. It has been found that the actions of 1,25-(OH)2D3 on the immune system are mainly immunosuppressive. The cross-presentation pathway allows for exogenously derived antigens to be presented by pAPCs on MHC-I molecules to CD8+ T cells. CD8+ T cell activation results in the expansion of epitope-specific T cell populations that confer host protection. These epitopes can be organized into an immunodominance hierarchy. Previous work demonstrated that introducing LCMV-NP via the cross-priming pathway significantly alters the immunodominance hierarchy of a subsequent LCMV infection. Building upon these observations, our study assessed the effects of LCMV-NP cross priming in the presence of a single dose of 1,25-(OH)2D3. Treatment with 1,25-(OH)2D3 was found to have biological effects in our model system. In vitro pAPCs were demonstrated to up-regulate IL-10 and CYP24A1 mRNA, in addition to the transactivation of cellular VDR, as demonstrated by a relocalization to the nuclear region. Mice treated with 1,25-(OH)2D3 were found to produce up-regulated IL-10 and CYP24A1 transcripts. Expression of VDR was increased at both the transcript and protein level. Our results demonstrate that a single dose of 1,25-(OH)2D3 does not affect the cross-priming pathway in this system. Treatment with 1,25-(OH)2D3 did not influence the ability of differentiated pAPCs to phagocytose or cross-present exogenous antigen to epitope-specific CD8+ T cells. Furthermore, 1,25-(OH)2D3 did not alter cross-priming or the establishment of the LCMV immunodominance hierarchy in vivo. By confirming that 1,25-(OH)2D3 does not suppress cross-priming in our model, our study helps to expand the understanding of the immunomodulatory role of exogenous 1,25-(OH)2D3 on the outcome of virus infection. Collectively, our data supports the observation that the role of 1,25-(OH)2D3 in the immune system is not always associated with suppressive effects. / Thesis (Master, Microbiology & Immunology) -- Queen's University, 2011-08-29 14:53:18.766
2

Modulation of dendritic cell function and T cell immunity by bacterial lipopolysaccharide

Papadopoulos, George 14 June 2019 (has links)
Several Gram-negative bacteria modify their outer most surface structure, lipopolysaccharide (LPS), to evade immune surveillance and survive within the host. Many of these changes occur within the lipid A domain, a region that is recognized by the innate immune system via Toll-like receptor-4 (TLR4). One such pathogen, Porphyromonas gingivalis, orchestrates chronic inflammatory disease by disrupting immune homeostasis. P. gingivalis initially synthesizes a penta-acylated lipid A that functions as a weak TLR4 agonist but displays tetra-acylated forms that are either immunologically silent or TLR4 antagonists. The impact of lipid A modifications on downstream signaling and antigen-specific immunity are unclear. TLR4 signals from the plasma membrane through a MyD88-dependent pathway and intracellularly through a TRIF-dependent pathway. Here we show that expression of immunological silent or antagonistic lipid A enables P. gingivalis to evade TRIF-dependent signaling in dendritic cells (DCs). Evasion of TRIF signaling accelerated antigen degradation and impaired priming of pathogen-specific T cells. In contrast, a P. gingivalis strain expressing agonist lipid A potently activated TRIF signaling and delayed antigen degradation, thereby preserving peptides for optimal T cell activation. We propose that lipid A modifications control the endocytic activity of DCs and the efficiency at which microbe-specific T cells are primed. We next investigated the impact of purified P. gingivalis LPS on innate signaling and antigen presentation. All P. gingivalis LPS species induced a program of DC maturation that allowed for constitutive antigen uptake and cross-presentation. This was independent of TLR4 agonist activity and required CD14, a protein that transports TLR4 to endosomes where TRIF signaling can occur. Agonist LPS induced signaling through both MyD88 and TRIF and elicited T cell priming. Antagonistic LPS potently accelerated CD14 endocytosis and induced TRIF-biased signaling leading to comparable degree of cross-priming. Immunologically silent LPS promoted CD14 endocytosis but failed to activate signaling and induced T cell tolerance. Collectively, our results demonstrate that modification of lipid A structure enables Gram-negative bacteria to direct the host immune system towards tolerance or immunity. We propose that these findings can be harnessed for therapeutic modulation of the immune system to treat a variety of immune-mediated diseases. / 2021-06-14T00:00:00Z
3

Mechanisms of Molecular Chaperone Surface Binding and Endocytosis: Insights into the Molecular Basis for GRP94 Immune Function

Jockheck-Clark, Angela Roberta January 2010 (has links)
<p>Extracellular GRP94 can elicit both innate and adaptive immune responses by interacting with endocytic and signaling receptors on professional antigen presenting cells (pAPCs). CD91 was the first receptor proposed to facilitate GRP94-mediated immune responses. Using a GRP94 affinity matrix, a CD91 fragment was isolated from the detergent-solubilized membranes of a pAPC cell line. It was then demonstrated that CD91 ligands could inhibit GRP94-mediated peptide cross-presentation, suggesting that CD91 played a critical role in this process. While these studies implied that CD91 could function as a GRP94 endocytic receptor, later works suggested that CD91 may not recognize GRP94 at the cell surface. These opposing observations have lead to a significant controversy surrounding the identity of CD91 as an endocytic receptor for GRP94. Because the ability of CD91 to directly mediate GRP94 surface binding and uptake has not been established, the studies included in this dissertation have focused on evaluating the ability of CD91 to facilitate three processes that are necessary for GRP94-mediated peptide cross-presentation: surface binding, internalization, and processing.</p><p>These studies utilized a recombinantly-expressed N-terminal domain of GRP94 (GRP94.NTD), which was previously shown to have nearly identical biological activity to full length GRP94. The ability of CD91 to directly bind and internalize GRP94.NTD was examined using murine embryonic fibroblast (MEF) cell lines whose expression of CD91 was either reduced via siRNA, or eliminated by genetic disruption of the CD91 locus. Binding competition experiments were also conducted. Together, these studies reveal that CD91 does not directly interact with GRP94 at the cell surface. The ability of CD91 to directly facilitate GRP94 internalization was examined using various internalization and internalization competition assays. These studies demonstrated that GRP94.NTD and the CD91 ligand RAP were internalized through spatially and kinetically distinct pathways, that CD91 was not necessary for GRP94.NTD internalization, and that RAP did not inhibit GRP94 endocytosis. Together, these studies strongly suggest that CD91 does not directly facilitate GRP94 internalization. When these studies were extended to DC2.4 mouse dendritic cells, the CD91 ligand RAP reduced GRP94.NTD internalization/process by ~15%. This suggests that CD91 may indirectly facilitate GRP94 internalization in pAPC cell lines. Lastly, cross-presentation studies were utilized to examine the ability of various CD91 ligands to influence GRP94.NTD-mediated peptide cross-presentation through a post-uptake mechanism using the DC2.4/OT-1 system. Although it was discovered that DC2.4 cells can internalize and process GRP94.NTD/peptide complexes through fluid-phase endocytosis, CD91 ligands did not significantly inhibit GRP94-mediated peptide cross-presentation by DC2.4 cells. These studies demonstrate that CD91 does not play a primary role in GRP94-mediated peptide cross-presentation.</p><p>In the course of these studies, cell surface heparan sulfate proteoglycans (HSPGs) were identified as novel cell surface binding sites for GRP94.NTD on MEF cells. This conclusion was established using three distinct experimental approaches. GRP94.NTD surface binding was significantly decreased following heparin pre-treatment, following incubation with the sulfation inhibitor sodium chlorate, and following digestion with extracellular heparinase II. Conversely, these treatments did not significantly influence GRP94.NTD binding to RAW264.7 mouse macrophage-like cells. This suggested that GRP94.NTD-HSPG cell surface interactions may require the expression of a specific type of cell surface HSPG that is not expressed by RAW264.7 cells. However, additional studies strongly suggested that GRP94.NTD-HSPG cell surface interactions were mediated by the heparan sulfate-containing side chains rather than the presence of a specific cell surface HSPG core protein.</p><p>This dissertation focuses on the critical re-examination of CD91 functions in GRP94 surface binding, uptake, and cross-presentation. Together, these results clarify conflicting data on CD91 function in GRP94 surface binding and endocytosis. This dissertation also describes the identification of cell surface HSPGs as GRP94 binding sites on MEF cells. These studies extend the diversity of surface receptors that recognize of GRP94, and suggest that cell surface HSPG-dependent interactions may contribute to the biology of GRP94-elicited immune responses.</p> / Dissertation
4

Die Expression des Chemokinrezeptors XCR1 kennzeichnet kreuzpräsentierende dendritische Zellen der Maus und des Menschen

Bachem, Annabell 26 September 2013 (has links)
Bei der Kreuzpräsentation durch dendritische Zellen (DC) werden extrazelluläre Antigene in den MHC I-Präsentationsweg eingeschleust und CD8+ T-Zellen präsentiert. Bisher werden die kreuzpräsentierenden DC der Maus durch die Expression der Moleküle CD8 in der Milz und CD103 in der Peripherie abgegrenzt; der kreuzpräsentierende Subtyp primärer humaner DC war vor Beginn dieser Arbeit nicht bekannt. Innerhalb dieser Arbeit konnte erstmals eine durchflusszytometrische Färbung von XCR1 auf der Oberfläche von Zellen etabliert werden, wodurch demonstriert wurde, dass XCR1 auf 83 % der CD8+ und 4 % der CD8-CD4- DC der Milz exprimiert wird. Der Phänotyp der XCR1+ DC der Milz unterschied sich deutlich von dem der XCR1- DC. In Milzen von Batf3- und Irf 8-defizienten Mäusen konnten keine XCR1+ DC detektiert werden. Zudem wurde nach Applikation des Wachstumsfaktors Flt3 Ligand in C57BL/6 Mäusen der Anteil der XCR1+ DC signifikant erhöht. Diese Ergebnisse verdeutlichen, dass die Entwicklung der XCR1+ DC von diesen Faktoren abhängig ist. Funktionell waren die XCR1+ DC wesentlich effektiver in der Kreuzpräsentation von löslichen und zellassoziierten Antigenen. Damit kann XCR1 als Oberflächenmarker verwendet werden, um murine kreuzpräsentierende DC zu kennzeichnen. Um herauszufinden, ob XCR1+ DC auch im Menschen existieren, wurde die Expression von XCR1 auf Zellen des humanen peripheren Blutes anhand von qPCR und Durchflusszytometrie untersucht. Ausschließlich CD141+ DC exprimierten XCR1-mRNA und -Protein. Durch die Etablierung einer effektiven Sortierungsstrategie zur Isolierung aller DC-Subtypen konnte erstmals gezeigt werden, dass die CD141+ DC den einzigen effektiven kreuzpräsentierenden DC-Subtyp des Blutes darstellen. XCR1 ist somit auch im humanen System spezifisch auf kreuzpräsentierenden DC exprimiert. Zusammengefasst zeigen die Ergebnisse, dass es sich bei den XCR1+ DC der Maus und des Menschen um funktionelle Homologe handelt. / Cross-presentation by dendritic cells (DC) is a process, in which extracellular antigen is shunted into the MHC I presentation pathway and presented to CD8+ T cells. So far, murine cross-presenting DC were defined by the expression of the molecules CD8 in the spleen and CD103 in the periphery. However, cross-presenting DC have not been characterized in humans. In this work, a flow cytometric staining of XCR1 was established for the first time which allowed the detection of XCR1 on 83 % of CD8+ DC and 4 % of CD8-CD4- DC of the spleen. The phenotype of splenic XCR1+ DC differed markedly from XCR1- DC. Both, Batf3- and Irf 8-deficient mouse strains showed an absence of splenic XCR1+ DC. Furthermore, the frequency of XCR1+ DC was significantly increased in spleens of C57BL/6 mice treated with Flt3 ligand. These results demonstrate that the development of XCR1+ DC is dependent of these factors. To test the ability to cross-present antigen, all splenic conventional DC were sorted according to their expression of CD8 and XCR1. XCR1+ DC were most efficient in cross-presenting soluble and cell-associated antigen to CD8+ T cells. Therefore, XCR1 is the first surface marker that can be used to delineate murine cross-presenting DC and the development of this distinct DC population is strongly dependent on Batf3, IRF-8 and Flt3 ligand. To explore if XCR1+ DC also exist in men, cell populations of human peripheral blood were analysed for their XCR1-expression using qPCR and flow cytometric staining. Only CD141+ DC express XCR1 mRNA and protein. An efficient sorting strategy for the isolation of all DC subsets was established to compare their ability to cross-present soluble and cell-associated antigen. CD141+ DC were the only effective cross-presenting DC subtype. Therefore, XCR1 is also in the human a receptor expressed specifically on cross-presenting DC. In summary, the data show that the XCR1+ DC of mouse and men are functional homologues.
5

Pathways of paternal antigen presentation to initiate antigen-specific immune responses in pregnancy.

Moldenhauer, Lachlan January 2008 (has links)
The fetus and its placenta, collectively called the conceptus, are semi-allogeneic to the mother, as they express transplantation antigens of paternal origin. Foreign tissues generally experience immunological rejection by the host immune system; however in a normal healthy pregnancy the conceptus does not undergo immune attack. Emerging evidence indicates the conceptus avoids rejection through a number of mechanisms including the induction of active maternal immune tolerance specific for paternal antigens. However, the mechanisms responsible for establishing this tolerance remain undefined, including the timing of the first encounter with paternal antigen and the cellular processes by which paternal antigen is presented to the maternal immune system. Exposure to paternal transplantation antigens occurs in two waves: initially in the context of male seminal fluid at conception, and secondly after placental trophoblast invasion of maternal tissues in mid-gestation pregnancy. Therefore the aim of this research was to evaluate the female immune response to paternal antigens in seminal fluid and those associated with the conceptus. The mechanisms of antigen presentation, the impact of the cytokine environment and the consequences of T cell activation on pregnancy were also investigated. A transgenic system using ovalbumin (OVA) as the model paternal antigen was established. The transgenic Act-mOVA mouse expresses OVA constitutively and ubiquitously under a B-actin promoter and OVA was shown to be present in seminal fluid and in the fetal and placental tissue of sired progeny. The OVA-reactive CD8+ OT-I and CD4+ OT-II T cells were employed to gauge the relative amount of OVA antigen presented, with the strength of the maternal immune response quantified based upon the extent of T cell proliferation, as assessed by CFSE dye-dilution. Utilising bone marrow chimeric mice, it was demonstrated that upon insemination by an Act-mOVA male, seminal fluid-derived OVA was processed and indirectly presented by maternal bone marrow-derived antigen presenting cells to induce activation and proliferation of the CD8+ OT-I T cells within the uterinedraining para-aortic lymph nodes of the female. Likewise, OT-II T cells were responsive to MHC class II-restricted presentation of seminal fluid OVA. Post-implantation conceptus-derived OVA was detected within peripheral lymph nodes and the spleen where it was presented via the MHC class I and class II-restricted pathways to induce systemic proliferation of both OT-I and OT-II T cells. Furthermore, as gestation advanced the extent of OVA presentation and hence T cell proliferation intensified. Conceptus-derived OVA was still presented systemically until 20 days pp. The impact of the uterine cytokine environment was assessed to determine its influence on seminal OVA antigen processing and presentation. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a key factor in regulating the leukocyte population of the female reproductive tract. GM-CSF-deficient female mice were unable to process and present seminal fluid OVA as effectively or efficiently as their wildtype counterparts, as assessed by their reduced capacity to drive OT-I and OT-II T cell proliferation following insemination by an Act-mOVA male. Finally, with highly-reactive OVA-specific T cells activated in response to seminal and conceptus OVA antigen, it was of interest to determine the effect of OT-I T cell activation on fetal survival and pregnancy success. It was found that OT-I T cells activated in vivo to paternal OVA antigen in the context of seminal fluid and pregnancy were not deleterious to pregnancy outcomes. However the transfer of cytotoxic OT-I T cells generated in vitro in the presence of an IL-2 into female mice carrying OVA-expressing conceptuses was detrimental to fetal survival. Collectively these experiments demonstrated that the initial exposure to paternal antigen, and hence the first opportunity to develop paternal antigen-specific tolerance, occurs at insemination. Paternal antigen is presented to the maternal T cell repertoire throughout gestation and may play a role in maintaining immune tolerance during pregnancy. The processing and presentation of paternal-derived antigen is chiefly performed by female bone marrow-derived antigen presenting cells. The cytokine environment of the mated female reproductive tract is critical in allowing optimal antigen processing and presentation, to generate an immune response consistent with maternal immune tolerance of the conceptus. / Thesis (Ph.D.) - University of Adelaide, School of Paediatrics and Reproductive Health, 2008
6

Mise en évidence et caractérisation de la coopération entre les cellules NK et les cellules dendritiques humaines pour la présentation croisée d’antigènes / Characterization of NK/DC cooperation for Ag cross-presentation

Deauvieau, Florence 06 July 2011 (has links)
Les données de la littérature ont récemment souligné l’importance du dialogue réciproque qui s’instaure entre les cellules Natural Killer (NK) et les cellules dendritiques (DC) au cours des phases précoces de la réponse immune pour l’initiation des réponses T spécifiques. La présentation croisée d’antigènes (Ag), processus qui permet aux DC de présenter des Ag exprimés par d’autres cellules aux lymphocytes T CD8+, est requise pour le développement d’une immunité cellulaire spécifique dans la plupart des infections par des pathogènes intracellulaires et des tumeurs. L’étude des mécanismes physiologiques impliqués dans la régulation de cette fonction suscite donc un intérêt majeur. Ce travail a permis de mettre en lumière une coopération entre les cellules NK et les DC humaines pour la présentation croisée d’un Ag exprimé par des cibles tumorales. Dans ce contexte, nous avons montré que la lyse de ces cibles par les cellules NK n’est pas nécessaire à la capture de leurs Ag par les DC. Au contraire, la sécrétion d’IFN-γ et de TNF-α par les cellules NK activées au contact des cibles tumorales joue un rôle prépondérant dans l’induction de la présentation croisée d’Ag. Ainsi, nous avons identifié une nouvelle fonction « helper » des cellules NK à l’interface entre l’immunité innée et adaptative. Le ciblage de cette fonction pourrait offrir de nouvelles perspectives en immunothérapies anti-tumorales dont le but ultime est le développement d’une immunité cellulaire spécifique / Recent reports have demonstrated the importance of the reciprocal crosstalk between natural killer (NK) cells and dendritic cells (DC) occurring during early phase of immune response for shaping downstream T cell immunity. Antigen (Ag) cross-presentation, a process by which DC present Ag from neighboring cells to CD8+ T lymphocytes is a prerequisite for the developpment of specific cellular immunity against most intracellular pathogens and tumors. A more detailed understanding of the mechanisms that regulate this specific DC function is thus a major challenge for immunologists. Here, we highlight the cooperation between NK and DC for tumor cell-derived Ag cross-presentation. In this context, we show that the NK cell-mediated lysis of target cells is not required for Ag capture by DC. In contrast, both IFN-γ and TNF-α produced by NK cells upon recognition of tumor cells play a critical role in the induction of Ag cross-presentation. These findings define a novel « helper» function of NK cells bridging innate and adaptive immunity. This novel function could be harnessed in cancer immunotherapy for inducing Ag-specific cellular immunity
7

Effect of HIV antiretroviral drugs on antigen processing and epitope presentation by MHC-I to cytotoxic T cells / Effet des antirétroviraux sur la voie d’apprêtement des antigènes et la présentation directe ainsi que croisée des épitopes par les CMH-I

Kourjian, Georgio 30 June 2015 (has links)
L’apprêtement antigénique par les protéases intracellulaires et la présentation des épitopes sont essentiels pour la reconnaissance des cellules infectées par les lymphocytes CD8+. Ici nous avons montré que certains inhibiteurs de la protéase de la VIH (IPs) modulent l’activité de la protéasome et aminopeptidase impliqué dans l’apprêtement antigénique endogène et l’activité cathepsins importante dans l’apprêtement croisée. Deux IPs agissent directement sur les cathepsins et leurs régulateurs en inhibant les activités kinase, NOX2 et en régulant le pH phagolysosomal. Les IPs ont changé la dégradation des protéines viral et la production des épitopes de façon séquence- et cellule-spécifique, ont altéré la présentation direct et croisée des épitopes, et ont partiellement changé l’auto-peptidome des cellules primaires. La modulation par les drogues de l’apprêtement et la présentation des épitopes peut fournir une approche thérapeutique alternative pour moduler la reconnaissance immunitaire. / Antigen processing by intracellular proteases and peptidases and epitope presentation are critical for recognition of pathogen-infected cells by CD8+ T lymphocytes. Here we show that several HIV protease inhibitors (PIs) prescribed to HIV-infected persons variably modulate proteasome and aminopeptidase activities involved in endogenous antigen presentation and cathepsin activities involved in antigen cross-presentation. Two HIV PIs acted directly on cathepsins and on their regulators by inhibiting kinases, NOX2 and the regulation of phagolysosomal pH, subsequently enhancing cathepsin activities. HIV PIs modified HIV protein degradation and epitope production in a sequence- and cell-dependent manner, altered direct- and cross-presentation and T cell-mediated killing, and partly changed the self-peptidome of primary cells. Drug-induced modulation of antigen processing and peptidome may provide an alternate therapeutic approach to modulate immune recognition.
8

Caractérisation des cellules dendritiques humaines BDCA3high et de leur modulation par le microenvironnement tumoral / Characterization of BDCA3high human dendritic cells and their modulation by tumor microenvironment

Ollion, Vincent 24 September 2015 (has links)
Les cellules dendritiques {DC) jouent un rôle majeur dans l'induction de l'immunité anti-tumorale spécifique de l'antigène {Ag). Récemment, les DC BDCA3high humaines sont apparues comme étant homologues aux DC CD8a+ connues pour activer très efficacement les lymphocytes T CD8 par présentation croisée d'Ag chez la souris. Par ailleurs, ces deux populations de DC sont les productrices principales d'interféron-λ{IFN-λ), une cytokine récemment découverte et possédant des propriétés antivirales, antiprolifératives et anti-tumorales. Mon travail de thèse a permis de mieux caractériser la présentation croisée d'Ag cellulaire par les DC BDCA3high à l'aide d'un modèle in vitro et de mettre en lumière le rôle des cellules NK dans l'induction de ce processus. Ce travail a également aboutit à la mise en évidence des DC BDCA3high dans les tumeurs de sein et de révéler l'inhibition de leur sécrétion d'IFN-λ par des facteurs solubles présents dans le microenvironnement tumoral. L'ensemble de ces résultats devraient permettre de concevoir de nouvelles stratégies thérapeutiques basées sur le ciblage des DC BDCA3high / Dendritic cells {DC) play a major role in the induction of antigen {Ag) specific anti-tumoral immunity. Recently, human BDCA3high DC appeared to be homologous with CD8a+ DC known to activate very efficiently T CD8 lymphocyte by Ag cross-presentation in mice. Moreover, those two DC populations are the main producers of interferon-λ {IFN-λ), a recently discovered cytokine family with antiviral, anti-proliferative and anti-tumoral properties. My works participated to better characterize cell derived Ag cross-presentation by BDCA3high DC using an in vitro model and enlightened the role of NK cells in its induction. This works also end up in revealing the presence of BDCA3high DC in breast tumors and the inhibition of their IFN-λsecretion by soluble factor from tumor microenvironment. Altogether, those results should allow designing new anti-tumoral immunotherapeutic strategies based on BDCA3high DC targeting
9

UM171-induced ROS promote antigen cross-presentation of immunogenic peptides by bone marrow-derived mesenchymal stromal cells

Salame, Natasha 07 1900 (has links)
En raison de leur multipotence considérable, les cellules stromales mésenchymateuses (CSM) ont été énormément utilisées en clinique dans le contexte de la médecine régénérative. Pourtant, la stimulation des CSM avec de faibles concentrations d'interféron-gamma (IFN-gamma) déclenche une augmentation du complexe majeur d’histocompatibilité de classe I et II, et surtout une capacité de novo de présentation croisée des antigènes. Ainsi, malgré leurs propriétés immunosuppressives naturelles, les CSM peuvent être modulées pour devenir pro-inflammatoires. Comme le dérivé pyrimidoindole UM171 induit l’augmentation de l’expression de plusieurs gènes impliqués dans la présentation antigénique dans les cellules souches hématopoïétiques humaines, nous avons étudié son potentiel pour le déclenchement de la présentation antigénique par les CSM. L'analyse par cytométrie en flux a montré une élévation des niveaux de H2-kB après le traitement avec le médicament, en corrélation avec une augmentation de la présentation de l'antigène, démontrée par une activation plus importante de la lignée de cellules T B3Z spécifique au peptide SIINFEKL. Cette présentation croisée de novo d'un peptide immunogène ne résulte pas d'une augmentation de l'absorption ou de la digestion enzymatiques des protéines, mais plutôt de l'expression du gène Psmb8 induit par le médicament. Comme le traitement avec plusieurs antioxydants et inhibiteurs des complexes de la chaîne de transport des électrons a réduit de manière significative les effets observés, nous concluons que la présentation croisée médiée par UM171 est dépendante des ROS. Dans le contexte de la vaccination thérapeutique, l'immunisation avec des CSM traitées par UM171 chez des souris présentant des tumeurs EG.7 préétablies a permis d'obtenir un taux de survie de 40%. Dans l'ensemble, notre étude révèle une nouvelle approche pharmacologique pour modifier les CSM afin qu'elles deviennent des cellules présentatrices d'antigènes, ce qui permet de développer de nouveaux vaccins anticancéreux innovants et puissants. / Due to their considerable multipotency, mesenchymal stromal cells (MSCs) have been tremendously employed in the clinic for regenerative medicine. Yet, stimulation of MSCs with low concentrations of interferon-gamma (IFN-gamma) triggers an increase in the major histocompatibility complex I and II, and most importantly, a de novo antigen cross-presentation capacity. Thus, despite their natural immunosuppressive properties, MSCs can be modulated to become pro-inflammatory. As the pyrimidoindole derivative UM171 induces the upregulation of several antigen presentation-involved genes in human hematopoietic stem cells, we investigated its potential for inducing antigen presentation by MSCs. Flow cytometry analysis showed an upregulation in H2-kB levels after treatment with the drug, correlating with an increase in antigen presentation indicated by higher activation of the SIINFEKL-specific B3Z T cell line. This de novo cross-presentation of an immunogenic peptide did not result from an increase in protein uptake or processing, but rather stemmed from the drug-induced expression of the Psmb8 gene. As treatment with multiple antioxidants and inhibitors of the electron transport chain complexes significantly reduced the observed effects, we conclude that UM171-mediated cross-presentation is ROS-dependent. In the context of therapeutic vaccination, immunization with UM171-treated MSCs in mice with pre-established EG.7 tumors resulted in 40% survival. Overall, our study reveals a new pharmacological approach in modifying MSCs to become antigen presenting cells, hence allowing the development of future innovative and potent anti-tumoral vaccines.
10

A1-reprogrammed mesenchymal stromal cells as a therapeutic vaccine against solid tumors

Pereira Gonçalves, Marina 09 1900 (has links)
L'efficacité de la réponse antitumorale repose sur l'activité des cellules T cytotoxiques, qui peut être stimulée par des vaccins contenant des antigènes spécifiques aux tumeurs. Malgré le fait d'être les principales cellules présentatrices d'antigènes (CPA) responsables de l'activation des cellules TCD8, les cellules dendritiques (CD) ont rencontré des défis dans le développement de vaccins contre le cancer, notamment en ce qui concerne leur fabrication et efficacité. Pour combler ces problèmes, cette étude propose d’utiliser des cellules stromales mésenchymateuses (CSM) comme plateforme de vaccination alternative, en exploitant les avantages des CSMs en matière de fabrication, de sécurité et de plasticité. La plasticité remarquable des CSMs leur permet d'acquérir une capacité de présentation croisée sous des stimuli spécifiques. Étant donné que la présentation croisée est essentielle pour induire l'activation des cellules T contre les antigènes tumoraux, cette étude vise à convertir les CSMs en cellules présentatrice d‘antigènes en améliorant l'exportation des antigènes des endosomes vers le cytosol - une étape critique du processus. Dans cette démarche, nous avons examiné une librairie de molécules dérivés de l’Accum, une molécule initialement conçue pour favoriser la destruction de la membrane endosomale. Après avoir évalué leur potentiel à induire la présentation croisée, nous avons sélectionné la molécule A1 pour des investigations subséquentes. Les études mécanistiques ont démontré qu'A1 déclenchait des processus cellulaires essentiels favorisant une présentation croisée efficace, notamment une augmentation de la capture, dégradation et évasion des antigènes des endosomes ainsi que la production de d’espèces oxygénés réactifs. L'efficacité thérapeutique des CSMs reprogrammées par A1 (ARM) en tant que vaccin anticancéreux a été évaluée chez des souris ayant des tumeurs, en monothérapie et en combinaison avec l’anti-PD-1. La thérapie combinée ARM a induit une régression tumorale et a augmenté les taux de survie dans les modèles de tumeurs solides. En conclusion, cette étude présente une stratégie innovante pour transformer les CSM en cellules à présentation croisée en déclenchant l'échappement endosomal de l'antigène. Les cellules ARMs en association avec des inhibiteurs des points de contrôle immunitaire présentent un potentiel en tant que plateforme de vaccination contre les tumeurs solides. De plus, ces résultats soulignent l'importance de l'évasion des endosomes dans la présentation croisée d‘antigènes et ouvrent la voie à de nouvelles plateformes de vaccins contre le cancer. / The effectiveness of antitumoral response relies on cytotoxic T-cell activity, which can be stimulated through vaccines carrying tumor-specific antigens. Despite being the primary antigen-presenting cells (APCs) responsible for CD8 T-cell activation, dendritic cells (DCs) have encountered challenges in cancer vaccine development, particularly in manufacturing and efficiency. To address this gap, this study proposes mesenchymal stromal cells (MSCs) as an alternative vaccine platform, leveraging the advantages in manufacturing, safety profile, and plasticity of MSCs. The remarkable plasticity of MSCs enables them to acquire cross-presentation capacity under specific stimuli. Given that cross-presentation is pivotal for inducing T-cell activation against tumor antigens, this study aims to convert MSCs into antigen cross-presenting cells by enhancing the export of antigens from endosomes to the cytosol—a critical step in the process. In this pursuit, we screened a library of Accum and variant molecules designed to promote endosomal disruption. After evaluating their potential to induce cross-presentation, we selected the molecule A1 for further investigation. Mechanistic studies demonstrated that A1 triggers essential cellular processes supporting efficient cross-presentation, including enhanced antigen uptake, processing, endosomal escape, and reactive oxygen species production. The therapeutic efficacy of A1-reprogrammed MSCs (ARMs) as an anticancer vaccine was evaluated in tumor-bearing mice, as monotherapy and combined with anti-PD-1. In solid tumor models, ARMs combination therapy induced tumor regression and increased survival rates. In conclusion, this study presents an innovative strategy to transform MSCs into cross-presenting cells by triggering antigen endosomal escape. ARM cells in combination with immune checkpoint inhibitors hold potential as a vaccination platform against solid tumors. These findings underscore the importance of endosomal escape on antigen cross-presentation and pave the way for new cancer vaccine platforms.

Page generated in 0.512 seconds