• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 142
  • 82
  • 69
  • 18
  • 13
  • 11
  • 5
  • 4
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 392
  • 111
  • 87
  • 74
  • 51
  • 49
  • 43
  • 43
  • 38
  • 33
  • 29
  • 29
  • 26
  • 25
  • 25
  • 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.
61

Innate Immune Cell Phenotypes Are Dictated by Distinct Epigenetic Reprogramming

Adams, Kevin Douglas 01 December 2018 (has links)
The innate immune system is the first line of host defense against external exposures. During these initial encounters, antigen presenting cells - specifically monocytes and macrophages - modulate further inflammatory responses. Macrophages exist along a spectrum of phenotypic programs; on the inflammatory M1 end they enhance immune activity while on the anti-inflammatory M2 end they suppress further immune activation. Furthermore, within M2 macrophages there exist many subpopulations, namely M2a and M2d, each with specific roles during infection or exposure. We sought to compare the epigenetic profiles of these subpopulations of macrophages to determine key regulatory gene networks and factors that could be exploited for therapeutic benefit.While traditionally viewed as primitive and nonspecific, a growing body of clinical and experimental evidence argues the innate immune system develops memory as a result of previous exposures, allowing the innate system to respond with enhanced and broad immunological protection upon exposure to a secondary stimulus. This biological process of innate immunity has been termed trained immunity. Trained immunity shares many phenotypic and epigenetic characteristics with adaptive immune memory; however, one of the starkest distinctions is the propensity of trained immunity to develop against heterologous stimuli. Innate memory is not antigen specific, frequently protecting the host against unrelated organisms.
62

Impaired Phagocytosis Directs Human Monocyte Activation in Response to Fungal Derived β-glucan Particles

Camilli, Giorgio, Eren, Elif, Williams, David L., Aimanianda, Vishukumar, Meunier, Etienne, Quintin, Jessica 01 May 2018 (has links)
Recognition of the fungal cell wall carbohydrate β-glucan by the host receptor Dectin-1 elicits broad immunomodulatory responses, such as phagocytosis and activation of oxidative burst. These responses are essential for engulfing and killing fungal pathogens. Phagocytic monocytes are key mediators of these early host inflammatory responses to infection. Remarkably, whether phagocytosis of fungal β-glucan leads to an inflammatory response in human monocytes remains to be established. Here, we show that phagocytosis of heat-killed Candida albicans is essential to trigger inflammation and cytokine release. By contrast, inhibition of actin-dependent phagocytosis of particulate (1-3,1-6)-β-glucan induces a strong inflammatory signature. Sustained monocyte activation, induced by fungal β-glucan particles upon actin cytoskeleton disruption, relies on Dectin-1 and results in the classical caspase-1 inflammasome formation through NLRP3, generation of an oxidative burst, NF-κB activation, and increased inflammatory cytokine release. PI3K and NADPH oxidase were crucial for both cytokine secretion and ROS generation, whereas Syk signaling mediated only cytokine production. Our results highlight the mechanism by which phagocytosis tightly controls the activation of phagocytes by fungal pathogens and strongly suggest that actin cytoskeleton dynamics are an essential determinant of the host's susceptibility or resistance to invasive fungal infections.
63

Investigation of Novel lincRNAs SIMALR and RP11-184M15.1 Functions in Inflammatory and Resolving Human Macrophage

Cynn, Esther January 2022 (has links)
Long noncoding RNAs (lncRNAs) are emerging as novel regulators of macrophage biology and related inflammatory cardiovascular diseases. However, studies focused on lncRNAs in human macrophage subtypes, particularly human-specific lncRNAs that are not conserved in rodents, are limited. Through deep RNA-seq of human monocyte-derived macrophages, we identified SIMALR (suppressor of inflammatory macrophage apoptosis lincRNA), a human macrophage-specific long intergenic noncoding RNA (lincRNA), to be highly induced in the nucleus of LPS/IFNγ-stimulated macrophages. Treatment of LPS/IFNγ-stimulated THP1 human macrophages with SIMALR antisense oligonucleotides induced apoptosis of inflammatory macrophages, as shown by increased Annexin V+ macrophages, and increased protein expression of cleaved PARP, caspase 9, and caspase 3. Differential expression analysis of RNA-seq data from SIMALR knockdown versus control in human macrophages revealed Netrin 1 (NTN1), a known regulator of macrophage apoptosis, to be one of the top downregulated genes. NTN1 knockdown in LPS/IFNγ-stimulated THP1 macrophages induced apoptosis. This apoptotic phenotype was attenuated by treating LPS/IFNγ-stimulated macrophages with recombinant human NTN1 after SIMALR knockdown. Furthermore, NTN1 promoter-luciferase reporter activity was increased in HEK293T cells treated with lentiviral overexpression of SIMALR. NTN1 promoter activity is known to require HIF1α and RNA immunoprecipitation (RIP) showed that SIMALR binds HIF1α, suggesting that SIMALR may modulate HIF1α binding at the NTN1 promoter to regulate apoptosis of macrophages. In human translational studies, SIMALR was found to be upregulated in macrophages in unstable human atherosclerotic plaques, suggesting a possible mechanistic link between inflammation and cardiovascular diseases. In addition to SIMALR, through deep RNA-seq of human monocyte-derived macrophages, we identified RP11-184M15.1, a human macrophage-specific lincRNA, to be highly induced in the cytoplasm of IL-4-stimulated macrophage. Preliminary data showed that treatment of IL-4-stimulated THP1 human macrophages with RP11-184M15.1 small interfering RNA (siRNA) repressed apoptosis of resolving macrophages, as shown by decreased Annexin V+ macrophages, and reduced protein expression of cleaved PARP. Biotinylated RP11-184M15.1 pulldown coupled with mass spectrometry indicated an interaction between RP11-184M15.1 and zinc finger RNA-binding protein (ZFR). RIP corroborated the proposed interaction between RP11-184M15.1 and ZFR. RNAInter revealed mRNAs predicted to interact with ZFR, and some of those genes (e.g., ALYREF, CCNYL1) were also differentially expressed in RNA-seq data of control versus RP11-184M15.1 knockdown in IL-4-stimulated THP1 macrophages. qPCR validated that ALYREF and CCNYL1 expression are reduced with RP11-184M15.1 knockdown. In contrast, with ZFR siRNA, ALYREF and CCNYL1 mRNA expressions were elevated. Thus, a hypothesis to be further tested is that RP11-184M15.1 interacts with ZFR to regulate mRNA stability in IL-4-stimulated macrophages. Nuclear RNA export factor 1 (NXF1) was also validated by RIP to interact with RP11-184M15.1. NXF1 is a known interacting partner of ALYREF in the transcription-export (TREX) complex. With RP11-184M15.1 knockdown, the protein level of ALYREF decreased, and Ingenuity Pathway Analysis (IPA) of RNA-seq data of control versus RP11-184M15.1 knockdown revealed that THO complex subunit 5 homolog (THOC5), another component of the TREX complex, may be an upstream regulator. In addition, past studies have revealed that ALYREF and NXF1 are involved in nuclear export of inflammatory mRNAs and proinflammatory macrophage phenotype, respectively. With RP11-184M15.1 knockdown, there was decreased expression of inflammatory macrophage-associated genes. It may be possible that RP11-184M15.1 functions in mRNA export, along with NXF1 and ALYREF. In human translational studies, RP11-184M15.1 was found to be upregulated in macrophages in unstable human atherosclerotic plaques. Further work is needed to better understand the functions and molecular mechanism of RP11-184M15.1. In summary, we found that SIMALR may interact with HIF1α to regulate macrophage apoptosis via NTN1. Our preliminary work also revealed that RP11-184M15.1 may regulate apoptosis, mRNA stability and mRNA export in anti-inflammatory macrophages. Both lincRNAs may be upregulated in unstable human atherosclerotic plaques. By studying SIMALR and RP11-184M15.1, we were able to illustrate the importance of interrogating the functions of human-specific lincRNAs despite the lack of rodent models, and demonstrated roles in macrophage inflammation that may be relevant to human cardiovascular disease.
64

Étude moléculaire de la régulation de l'interféron alpha dans les monocytes humains infectés par le virus Epstein-Barr

Duval, Annick 11 April 2018 (has links)
Le virus Epstein-Barr (EBV) est un virus herpès oncogène causant la mononucléose infectieuse et est associé à différents types de cancer d'origine épithélial et lymphoïde. Nous avons précédemment démontré que EBV peut infecter et se répliquer dans les monocytes humains. Suite à cette infection, plusieurs fonctions importantes du monocyte s'en trouvent affectées, dont la phagocytose, la biosynthèse de prostaglandine E2 et la production de TNF-oc et d'IFN-a. Il est bien connu que l'IFN de type I (IFN-a/(3) est un puissant médiateur antiviral qui est rapidement sécrété par les cellules infectées lors des stades précoces de l'infection. OBJECTIFS Ce projet vise à caractériser le(s) mécanismes(s) affectant la production d'IFN-a par les monocytes humains infectés par le virus EBV en étudiant notamment la cascade d'activation des protéines JAKs et STATs. MÉTHODES Nous avons évalué l'effet de l'infection des monocytes humains par EBV sur l'activation des facteurs de transcription IRF-3 et IRF-7, sur les protéines de la cascade des JAKs-STATs Qakl, Tyk2, Statl et Stat2) ainsi que sur les protéines SOCS. RÉSULTATS L'activation des protéines IRF-3 et IRF-7 n'est pas affectée, puisqu'il a été possible d'observer une translocation nucléaire de ces facteurs de transcription suite à l'infection par EBV. Cependant, la boucle d'amplification de la voie des JAKs-STATs est supprimée suite à l'inhibition de la phosphorylation de Statl. Le mécanisme d'inhibition de la synthèse d'IFN-a par le virus EBV dans les monocytes humains implique l'activation de la protéine SOCS-3. CONCLUSION Ce projet permet d'approfondir nos connaissances sur les mécanismes immunosuppresseurs mis en œuvre par EBV afin de supprimer les fonctions monocytaires. / Epstein-Barr virus (EBV) is an oncogenic herpesvirus recognized as the causative agent of infectious mononucleosis and is associated with several human malignancies. We have recently provided evidences that human monocytes were permissive to EBV infection and replication. Following infection, EBV can affect various cellular functions of monocytes, such as phagocytosis, biosynthesis of prostaglandins E2 and production of TNF-a and IFN-a. The type I interferon System (IFN-a/p) represents an important elements of host defence against ail kinds of pathogens, mainly viruses. Following infection, virus-infected cells rapidly produce and secrete IFN-a/p. OBJECTIVES The present work aims to determine the mechanism affecting the IFN-a production by the EBV-infected monocytes, in particular by the study of the JAK-STAT pathway. METHODS We examined the effect of EBV infection in monocytes on IRF-3 and IRF-7 nuclear accumulation, on JAKs and STATs proteins activation (Jakl, Tyk2, Statl and Stat2), and on SOCS proteins expression. RESULTS The IRF-3 and IRF-7 activation is not affected, since it was possible to observe a nuclear translocation of these transcription factors following EBV infection. However, the positive-feedback loop of the JAK-STAT pathway was found to be affected by the inhibition of Statl phosphorylation. The mechanism of IFN-a inhibition in EBV-infected monocytes involves the SOCS-3 protein activation. CONCLUSION Further description of EBV inhibitory mechanisms on monocytes immune functions would certainly improve our understanding of the role of these cells in the early stages of EBV pathogenesis.
65

The Role of Liver-X-Receptor and Retinoid-X-Receptor in the Regulation of Tumour Necrosis Factor-a Expression and Production in Human Monocytes / Regulation of TNF-a in Monocytes by LXR and RXR

Landis, Mark 08 1900 (has links)
Liver X receptor (LXR) is a member of the nuclear hormone receptor superfamily that is activated by hydroxylated cholesterol derivatives referred to as oxysterols. It has also been shown to play a crucial role in regulating cholesterol trafficking and lipid metabolism in liver and macrophages. Furthermore, LXR. has also been directly implicated in the reduction of atherosclerosis in several murine models of the disease by virtue of its ability to promote reverse cholesterol efllux from intima-resident lipid-loaded macrophages. While roles for LXR in monocyte biology have focused primarily on cholesterol trafficking, evidence for other functions for the receptor outside of its traditional role as a mediator of cholesterol homeostasis is lacking. Presented herein is evidence that LXR also serves as a mediator of cytokine expression. This work has shown that treatment of human peripheral blood monocytes or monocytic THP-1 cells with the LXR ligand 22(R)-hydroxycholesterol (22R-HC), in combination with 9-cis-retinoic acid (9cRA), a ligand for the LXR. heterodimerization partner retinoid X receptor (RXR), results in the specific induction of the potent pro-apoptotic and pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-α.). Promoter analysis, inhibitor studies, and order-of-addition experiments demonstrated that TNF-α. induction by 22R-HC and 9cRA occurs by a novel two-step process. The initial step involves 22R-HC-dependent induction of TNF-α. mRNA, and intracellular accumulation of TNF-alpha protein, mediated by binding of LXRα/RXRα to an LXR response element at position -879 of the TNF-α promoter. Subsequent cell release of TNF-alpha protein occurs via a separable RXR-dependent step that requires de novo transcription and protein synthesis. Furthermore, the RXR-dependent secretory event can be mimicked by agents that induce monocytic differentiation like phorbol esters that culminate in RXR activation by a pathway that does not require exogenous ligand. In this context, RXR was also shown to be a down stream target of the protein kinase C (PKC) signal transduction cascade, that results in the activation of RXR and the induction of secretory factor(s) which facilitate secretion of LXR-derived TNF-α. These studies have provided evidence that should help to expand the currently known role for LXR in monocyte biology and have furthermore identified a new role for RXRs in promoting the secretion of soluble factors like cytokines. Furthermore, in light of reports that show LXR activity promotes a reduction in atherosclerosis, it stands to reason that this regulatory circuit of LXR-dependent production of TNF -α from monocytes would similarly contribute to the attenuation of atherosclerosis 𝘪𝘯 𝘷𝘪𝘷𝘰. / Thesis / Master of Science (MSc)
66

Monocyte Derived Dendritic Cells: Sentinels and Translators of Immune Response to Staphylococcus aureus

Bharathan, Mini 03 December 2010 (has links)
<i>Staphylococcus aureus</i> is a versatile opportunistic pathogen causing a wide spectrum of diseases in both humans and animals. My research focused on characterization of the immune responses of monocyte derived dendritic cells (DC) to <i>S. aureus</i>. We initially evaluated the potential of circulating monocytes to serve as precursors for DC during <i>S. aureus</i> infection. The CD14⁺ monocytes, when stimulated with irradiated (ISA) or live <i>S. aureus</i> (LSA), differentiated into CD11c<sup>high</sup> CD11b<sup>high</sup> DC (MonoDC) in an autocrine fashion. This was associated with the up- regulation of granulocyte-macrophage colony stimulating factor (GMCSF) and tumor necrosis factor-α (TNF-α) gene transcription. We continued our studies to identify the role of TNF-α in the LSA induced differentiation of monocyte to MonoDC. Blocking TNF-α reduced the expression of CD11c and increased the expression of CD14 on LSA stimulated monocyte derived MonoDC. Stimulated monocytes were able to secrete monocyte chemotactic protein-1 (MCP-1), a chemokine that recruits monocytes to the site of infection/injury and induces the expression of β₂ integrins on DC. Characterization of the response of DC derived from monocytes using GMCSF and IL-4 revealed that, intact <i>S. aureus</i> rather than its purified structural components were efficient in DC activation. In response to ISA or LSA stimulation, DC induced proliferation of T cells collected from the peripheral circulation of cows with a history of <i>S. aureus</i> mastitis. Subsequent characterization of the proliferating T cells identified the presence of memory T cells. Finally, we identified a unique population of DEC205⁺CD8<sup>a+</sup> in monocyte derived DC. We further elucidated the role of DC DEC205, a C-type lectin, in <i>S. aureus</i> uptake. Blocking of receptor mediated endocytosis resulted in reduced uptake of <i>S. aureus</i> by DC. Confocal microscopy confirmed a role for DEC205 in <i>S. aureus</i> internalization and delivery to endosomes. DEC205 DC upon stimulation with <i>S. aureus</i> displayed enhanced maturation and antigen presentation. In conclusion, monocyte derived DC can uptake <i>S. aureus</i> and elicit cell mediated immune responses. / Ph. D.
67

Dynamic Programming of Innate Immunity in Health and Disease

Yuan, Ruoxi 02 November 2016 (has links)
Whether innate immune cells may be adapted into potential memory states has becoming an important question in the field of immunity. Although previous conceptual paradigm failed to acknowledge this important question, emerging clinical and basic observations have started to shed intriguing clues to shake the previous dogma regarding innate immunity of being "simple", "raw", "first-line defense with no memory". We have aimed to further address this fundamental issue in this dissertation work, under the close guidance of Dr. Liwu Li. We have chosen to use the model system of Toll-Like-Receptor (TLR) signaling networks within primary monocytes. TLRs play fundamental roles in sensing pathogen-associated molecular patterns (PAMPs) and modulation of innate immunity. Lipopolysaccharide (LPS), an endotoxin found on the cell membrane of gram-negative bacteria, is the ligand of TLR4 and induces a range of inflammatory as well as anti-inflammatory responses. Higher dosages of LPS were known to cause robust yet transient expression of pro-inflammatory mediators. On the other hand, the effects of super-low dose LPS, commonly manifested in humans with adverse health conditions, have been largely ignored in the basic research field. Super-low dose LPS may skew host immune environment into a mild non-resolving pro-inflammatory state, which is a risk factor for inflammatory diseases such as atherosclerosis, compromised wound healing, and elevated risks for sepsis. Our central hypothesize is that monocytes may be adapted by super-low dose LPS into a non-resolving low-grade inflammatory state conducive for the pathogenesis of inflammatory diseases. We have employed both in vitro cell culture system as well as in vivo disease models to test this hypothesis. For the in vitro system, we have cultured primary murine monocytes with increasing signal strength of LPS. Monocyte phenotypes such as the expression of key inflammatory mediators including cytokines, chemokines, and cellular surface markers were studied. Potential molecular and cellular mechanisms were examined. We revealed a novel low-grade inflammatory monocyte phenotype termed ML adapted by super-low dose LPS, mediated through IRF5. For the in vivo system, we have employed both acute and chronic models of inflammation. For the chronic model, we have tested the effects of super-low dose LPS on monocyte polarization in vivo, as well as its contribution to the pathogenesis of atherosclerosis. Furthermore, we have tested the effects of programmed monocytes on wound healing. For the acute model, we have tested the effects of pre-conditioning with super-low dose LPS on the subsequence risks of sepsis elicited by cecal ligation and puncture. We have demonstrated aggravated atherosclerosis, compromised wound healing, and increased sepsis mortality in mice pre-conditioned with super-low dose LPS. Taken together, our findings reveal that monocytes can be differentially programmed into distinct states, depending on the signal strength of LPS. The differential programming and adaptation of monocytes can occur both in vitro and in vivo, and may bear profound pathological consequences. / Ph. D.
68

Dose-dependent effects of endotoxin on monocyte and the underlying mechanisms

Pradhan, Kisha 24 January 2022 (has links)
Monocytes are dynamic innate immune cells that respond differently based upon the dose and duration of an infection. While super low dose endotoxin is found in chronic inflammatory diseases such as atherosclerosis, exposure to high dose endotoxin leads to sepsis. However, clear characterization of monocytes and the underlying mechanisms in these disease conditions is lacking. To elucidate the missing information, we conducted two different projects. In the first project, we investigated the role of super low dose endotoxin in polarizing monocytes to a prolonged low-grade inflammatory state with no resolution, disrupting homeostasis. This low grade inflammatory phenotype was confirmed by sustained induction of inflammatory mediators CD40 and CD11a. In addition, low grade inflammatory monocytes influence neighboring T cells by suppressing T cell regulatory functions. Mechanistically, we showed that the non-resolving inflammatory phenotypes in monocytes is dependent on non-traditional TLR4 adaptor called TRAM. In the second project, we focused on the effects of high dose endotoxin on monocyte phenotypes. We reported that high dose endotoxin give rise to a mix of both immunosuppressive and pathogenic inflammatory monocytes, leading to monocyte exhaustion. While thorough research is conducted to study the immunosuppressive monocytes and underlying long term effects, role of pathogenic inflammatory monocytes is not well addressed. Monocyte exhaustion leads to elevated levels of CD38, an inflammatory mediator, elevated ROS levels, depleted NAD+ and mitochondrial respiration. STAT1 and KLF4 are critical transcription factors in sustaining exhausted phenotypes. Indeed, TRAM adaptor molecule also mediates this exhaustion as TRAM deletion restores monocyte health. Taken together, our work defines novel monocyte phenotypes and mechanism in super-low dose or high dose endotoxin environments. / Doctor of Philosophy / Healthy inflammatory response is represented by initial induction of inflammatory cells in the site of infection and pathogen clearance, followed by resolution of inflammation and damage repair. This balance between inflammation and resolution maintains immune homeostasis. Imbalances in this homeostasis can be a cause or effect of various disease conditions such as atherosclerosis and sepsis, for example. Despite rigorous research, these diseases are still prevalent and treatments are still lacking. It is essential to investigate inflammatory responses at a cellular level and understand how an immune cell responds to a given pathogen. Depending upon the intensity, dose and duration of a pathogen can dictate immune cell functions. Recent discoveries, including the research in our lab have reported that super low dose bacterial endotoxin exacerbates atherosclerosis. Mouse monocytes (innate immune cells) treated with super low dose endotoxin continuously induce mild but sustained inflammatory molecules but are unable to exhibit resolving mediators to dampen the inflammation and hence, monocyte homeostasis is disrupted. Homeostatic imbalance is also in seen in sepsis, when monocytes exposed to high dose bacterial endotoxin. Due to a repetitive exposure to high dose endotoxin, monocytes are unable to respond accurately, where they simultaneously exhibit inflammatory and anti-inflammatory mediators but in a dysregulated manner.
69

Contribution du réservoir macrophagique intestinal sous traitement antirétroviral au cours de l'infection par le Virus de l'Immunodéficience Simienne

Clain, Julien 01 February 2021 (has links)
Les cellules monocytaires et les macrophages sont des cibles du virus de l’immunodéficience humaine (VIH-1) et du virus de l’immunodéficience simienne (VIS). D’une part, il a été proposé que les monocytes sanguins sont essentiels dans la réponse immune intestinale. D’autre part, les cellules monocytaires circulants reconstituent et repeuplent la région intestinale, qui est la première cible du VIH-1 et du VIS. Par conséquent, les cellules monocytaires intestinales pourraient contribuer à la dissémination virale dans l’organisme mais aussi représenter un réservoir viral sous thérapie antirétrovirale (TAR). Dans ce contexte, nous avons abordé la contribution des monocytes intestinaux dans le maintien des réservoirs viraux (VRs) chez le macaque rhésus (MR) infecté par la souche SIVmac251. Nos résultats montrent que la TAR précoce contrôle efficacement l’infection virale des cellules monocytaires, aussi bien dans le sang que dans l’intestin. Parallèlement au rebond viral après l'interruption du traitement, nous avons observé l'infection précoce des monocytes, qui présentent non seulement de l'ADN viral, mais également de l'ARN viral. Ces résultats indiquent que les sous-ensembles de cellules monocytes ne peuvent pas être considérés comme des VRs majeurs sous TAR, mais peuvent contribuer à la production virale et à la dissémination du virus une fois que celui-ci est interrompu. / Monocytic cells and macrophages are targets of human immunodeficiency virus (HIV-1) and simian immunodeficiency virus (SIV). On the one hand, it has been proposed that blood monocytes are essential in the intestinal immune response. On the other hand, circulating monocytic cells reconstitute and repopulate the intestinal tract, which is the primary target of HIV-1 and SIV. Therefore, intestinal monocytic cells could contribute to the viral dissemination but also represent a viral reservoir under antiretroviral therapy (ART). In this context, we discussed the contribution of intestinal monocytes in the maintenance of viral reservoirs (VRs) in rhesus macaque (RM) infected with SIVmac251. Our results show that early ART effectively controls the viral infection of monocytic cells, both in the blood and in the intestine. Along with viral rebound after treatment interruption, we have observed early infection of monocytes, which not only display viral DNA, but also viral RNA. These results indicate that subsets of monocyte cells cannot be considered as major VRs under ART, but may contribute to viral production and viral dissemination once treatment is interrupted.
70

Etude de la réponse immunitaire innée au cours de l'infection à Orientia tsutsugamushi

Tantibhedhyangkul, Wiwit 03 July 2012 (has links)
Orientia tsutsugamushi, l'agent pathogène responsable du typhus des broussailles, est une bactérie cytosolique qui envahit l'endothélium et les monocytes/macrophages. La réponse immune à l'infection par O. tsutsugamushi reste à ce jour mal connue. L'objectif de cette thèse est de mieux comprendre la réponse des cellules de la réponse immune innée humaine à O. tsutsugamushi. Nous avons montré que O. tsutsugamushi se réplique dans les monocytes humains. En utilisant un microarray portant sur la totalité du génome, nous avons également montré que les bactéries vivantes induisent de profondes modifications du profil transcriptionnel des monocytes. C'est ainsi que l'expression des gènes codant l'interféron de type I et des gènes stimulés par l'interféron est fortement augmentée. Les monocytes infectés expriment plusieurs gènes codant des cytokines et des chimiokines inflammatoires, ce qui montre qu'ils sont polarisés vers un phénotype M1 (classically-activated phenotype). Les bactéries vivantes induisent également la sécrétion de l'interleukine-1&#946; et probablement l'activation des inflammasomes et de la caspase-1. O. tsutsugamushi affecte enfin l'expression des gènes associés à l'apoptose et induit la mort d'une partie des monocytes infectés. Nous avons en outre étudié le profil transcriptionnel de patients atteints d'un typhus des broussailles et avons trouvé une signature spécifique incluant la modulation de gènes de type M1 et de gènes stimulés par l'interféron. Nous avons finalement étudié la réponse des macrophages humains dérivés des monocytes à O. tsutsugamushi. / Orientia tsutsugamushi, the causative pathogen of scrub typhus, is a cytosolic bacterium that invades endothelium and monocytes/macrophages. So far, the knowledge of immune response to O. tsutsugamushi is still limited. The objective of this thesis is to better understand the response of human innate immune cells against this pathogen. We demonstrated that O. tsutsugamushi was able to replicate in human monocytes. Using whole genome microarrays, we showed that live O. tsutsugamushi induced robust changes in the transcriptional profiles of monocytes. First, type I interferons and interferon-stimulated genes were remarkably up-regulated. Second, infected monocytes expressed several inflammatory cytokine and chemokine genes, and were polarized toward the classically-activated M1 phenotype. Third, live bacteria induced interleukin-1&#946; secretion and likely inflammasome and caspase-1 activation. We also showed that O. tsutsugamushi altered the expression of apoptosis-related genes and induced cell death in monocytes. We extended our work to the study of the transcriptional profiles of patients with scrub typhus and found a specific signature in patients that included the modulation of M1-associated genes and interferon-stimulated genes. We finally studied the response of human monocyte-derived macrophages to O. tsutsugamushi. The transcriptional and functional responses of macrophages to O. tsutsugamushi were roughly similar to those observed in circulating monocytes including type I IFN response, pro-inflammatory cytokine gene expression and IL-1&#946; secretion.

Page generated in 0.0743 seconds