• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 14
  • 12
  • 12
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 55
  • 9
  • 8
  • 8
  • 8
  • 7
  • 7
  • 7
  • 6
  • 6
  • 6
  • 6
  • 5
  • 5
  • 5
  • 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.
31

Estrogen-Induced Modulation of Innate and Adaptive Immune Function

Masseoud, Feda N 30 April 2009 (has links)
Host defense against infection and disease relies on the reciprocal communication between the immune and neuroendocrine systems where sex hormones exert negative and positive feedback actions on immune functions. Indeed, sex hormones have been implicated in gender dimorphic immune response and in the potentiation of immune-related disorders. The female hormone estrogen plays a role as an immunomodulator and may exert immunosuppressive and immunostimulatory effects. Though many studies focus on estrogen’s role in immunity within the female reproductive tract and autoimmunity, the modulatory effects of estrogen on vaccine responses are largely unexplored. The insufficient efficacy of some vaccines in certain target populations, as for example the elderly population, is well recognized. Hormones fluctuate throughout an individual’s life, and females in particular undergo several necessary reproductive (pregnancy and menopause) and lifestyle (oral contraceptive use) changes which involve sex hormones. Vaccine efficacy might be influenced by endogenous estrogen levels or by exogenous estrogen administration. Therefore, in the pursuit of improved vaccine efficacy, it is necessary to consider such hormonal factors and their contribution to immune status. We have studied estrogen’s role in modulation of vaccine responses using a mouse ovariectomy model where exogenous estrogen delivery can be controlled. Our studies included two different types of vaccines, a bacterial toxoid formulation and a bacterial secreted protein formulation. Results from these studies indicate that estrogen enhances vaccine-specific antibody production by likely supporting a general TH2 pathway and also modulates expression of genes encoding molecules critical in innate immune signaling and required for development of proper adaptive immune responses and antigen clearance through antibody-mediated mechanisms. The level at which estrogen modulates antibody responses appears to be dependent on the route of vaccine administration. The enhancement of specific humoral responses may involve mechanisms involving TLR2 and antibody Fc receptor expression on macrophages, cells that link innate and adaptive immune responses. Advances in our understanding of the relationship between sex hormones and the immune system may provide new insights into the mechanisms by which hormones act and thus may be exploited to guide the design of future vaccine strategies.
32

Investigating the Roles of Mast Cells and Innate Activators in Oral Tolerance

Tunis, Matthew C. 26 June 2012 (has links)
Oral tolerance is the state of immunologic non-responsiveness that is established following oral antigen consumption. Failures of oral tolerance can result in food allergy. The mechanisms regulating oral tolerance are not well understood, but similar mechanisms may control tolerance to foods and commensal microbes in the intestine. The specific roles of many pattern recognition receptors (PRRs) and innate cells have not been examined in the context of oral tolerance. Mast cells are innate sentinel cells positioned at mucosal surfaces, and have been identified as key regulators of peripheral tolerance to allografts. Toll-like receptor 2 (TLR2) is a PRR involved in bacterial responses and the regulation of intestinal inflammation. We evaluated the impact of mast cells, TLR2, immunoglobulin E (IgE)-mediated mast cell activation, TLR2 activation, and histamine receptor blockade in the development of oral tolerance in mice. Models of tolerance to ovalbumin, peanut butter, and cow’s milk were established. Oral tolerance was assessed in wild type, TLR2-deficient, or mast cell-deficient mice and was measured primarily by analysis of antigen-specific antibody levels after a systemic antigen challenge. The development of antigen-specific Tregs was also assessed. We observed that neither mast cells nor TLR2 were necessary for oral tolerance induction. Moreover, IgE-mediated mast cell activation and antihistamine treatment did not significantly alter oral tolerance induction. TLR2 activators, notably Pam3CSK4, were administered orally concurrent with food antigen and were found to impair oral tolerance to a later systemic antigen challenge. When Pam3CSK4 was administered as an oral adjuvant with ovalbumin, a profound selective enhancement of the IgA response to oral challenge was observed. These results highlight an important differential regulation of oral tolerance by TLR2. Oral TLR2 activation selectively promotes IgA responses to antigen upon repeated oral challenge but prevents the maintenance of oral tolerance upon a systemic challenge. Taken together these results suggest that mast cells are not essential regulators of oral tolerance, but TLR2 is involved in regulating IgA and IgE responses during oral and systemic challenges. These findings inform mechanisms of commensal tolerance and have implications for the potential therapeutic manipulation of oral tolerance to foods.
33

Rôle des calpaïnes dans le vieillissement et la réponse anti-tumorale / Role of calpain in aging and anti-tumor response

Hanouna, Guillaume 08 November 2016 (has links)
Les calpaïnes 1 et 2 sont des protéases à cystéine ubiquitaires et la calpastatine est leur inhibiteur naturel, également ubiquitaire. Les calpaïnes sont impliquées dans le développement de la réponse inflammatoire via l’activation par protéolyse partielle de plusieurs substrats (activation de NF-κB par le clivage de I-κBα, remodelage du cytosquelette des cellules inflammatoires, clivage de la protéine chaperonne HSP 90…).Il a été précédemment démontré que les calpaïnes favorisent le vieillissement neuronal. Nous avons pu montrer dans un modèle murin que l’inhibition in vivo des calpaïnes par la surexpression de calpastatine limite le vieillissement notamment rénal et vasculaire. L’inflammation liée au vieillissement ou « inflammaging » est considérablement réduite par l’inhibition spécifique des calpaïnes. Ceci est dû, au moins en partie, à l’effet des calpaïnes sur la production de cytokines pro-inflammatoires et sur la maturation de l’interleukine-1.Si les calpaïnes intracellulaires exercent un rôle pro-inflammatoire, les calpaïnes externalisées ont un effet anti-inflammatoire via le clivage de TLR2. Les calpaïnes peuvent en effet être excrétées hors des cellules via les transporteurs ABCA1. Dans le cadre d’un modèle murin de mélanome, nous avons pu montrer que l’inhibition des seules calpaïnes extracellulaires par la surexpression de calpastatine extracellulaire préserve TLR2 et limite ainsi la progression de la tumeur.Les calpaïnes intra- et extracellulaires sont des médiateurs majeurs de la réponse inflammatoire et modulent « l’inflammaging » ainsi que la réponse immune anti-tumorale. / Calpain 1 and 2 are cysteine proteases and calpastatin is their natural inhibitor. Calpains and calpastatin are ubiquitous. Calpains are involved in inflammatory response development via activation by partial proteolysis of several substrates (NF-kappaB activation by I-kappaBalpha cleavage, remodeling of inflammatory cells cytoskeleton, cleavage of chaperone protein HSP90 ... ). It has been previously shown that calpains promote neuronal aging. We have shown in a mouse model that inhibition of calpain by calpastatin overexpression limits renal and vascular aging. The inflammation associated with aging or "inflammaging" is considerably reduced by specific inhibition of calpain. This is due, at least in part, to calpain effect on production of pro-inflammatory cytokines and in maturation of interleukin-1 alpha. If intracellular calpains are pro-inflammatory, secreted calpains have an anti-inflammatory effect via cleavage of TLR2. Calpains can indeed be excreted out of the cells via the transporter ABCA1. In the context of a mouse model of melanoma, we have shown that inhibition of extracellular calpain by only extracellular calpastatin overexpression preserves TLR2 and thus limit the progression of the tumor.Calpains intra- and extracellular are major mediators of inflammatory response and modulate the "inflammaging" and the anti-tumor immune response.
34

Un nouveau défaut héréditaire chez l’homme : déficit en TIRAP

Israel, Laura 21 November 2013 (has links)
Les maladies infectieuses constituent le principal groupe de maladies humaines communément considérées comme étant d’origine environnementale. Cependant, une question fondamentale dans ce domaine est la variabilité interindividuelle : pourquoi, au sein d’une même population exposée aux mêmes pathogènes, seule une minorité des individus infectés par un pathogène donné va développer une maladie dont l’expression clinique peut varier de l’état asymptomatique à la mort pour les formes les plus sévères. Le microbe est donc nécessaire mais non suffisant pour expliquer ces divergences. Selon la théorie génétique des maladies infectieuses, une proportion d’enfants ayant une ou plusieurs infections sévères mais étant par ailleurs sains, présenterait une immunodéficience primaire Mendélienne à l’origine d’une réponse immunitaire altérée spécifique du pathogène responsable. En considérant la susceptibilité accrue à des infections sévères par des bactéries pyogènes, cette hypothèse a conduit à la découverte, dans les années 2000, de plusieurs immunodéficiences primaires dans la voie de signalisation NF-kB, avec des mutations dans les gènes NEMO/IKBKG, IKBA, IRAK4 et MyD88 chez des enfants présentant des infections sévères par bactéries pyogènes, notamment Streptococcus pneumoniae et Staphylococcus aureus, et conduisant à une altération des voies Toll-interleukine-1 récepteur (TIR)-NF-kB. Mon travail de thèse a principalement porté sur l’étude d’une patiente aujourd’hui âgée de 5 ans. Elle est issue d’une famille consanguine et a souffert d’une pneumonie sévère à l’âge de 3 mois à la suite d’une infection par une souche PVL+ de S. aureus. .J’ai identifié chez elle un défaut autosomique récessif complet en TIRAP. Cependant, sept autres membres de la famille âgés entre 16 et 50 ans sont homozygotes pour la même mutation et n’ont jamais souffert d’infection sévère. TIRAP est un adaptateur des récepteurs TLR2 et TLR4 et la mutation rare R121W affecte un acide aminé très conservé dans son domaine TIR. L’allèle mutant de TIRAP induit une expression normale de l’ARN messager ainsi que de la protéine mais demeure non fonctionnel. Les réponses après stimulation par différents agonistes du TLR2 tels que le PAM2CSK4, le PAM3CSK4, et le FSL-1 ainsi que par le LPS, agoniste du TLR4 sont altérées dans les fibroblastes, les granulocytes et les monocytes des individus homozygotes pour la mutation R121W. Cependant, la réponse des cellules sanguines totales à l’acide lipoteichoïque (LTA) purifié de S. aureus, un autre agoniste du TLR2 est altérée uniquement chez la patiente. J’ai pu montrer que ce défaut est dû à une absence d’anticorps dirigés contre le LTA dans le plasma de la patiente. L’effet combiné de la mutation du gène TIRAP et de l’absence d’anticorps anti-LTA pourrait expliquer la survenue de l’infection sévère par S. aureus chez le cas index. L’ensemble de ce travail décrit pour la première fois un défaut héréditaire en TIRAP chez l’homme et fournit ainsi une meilleure compréhension du rôle de cette protéine dans les réponses en aval des TLRs. Il suggère également que, chez l’homme, la voie dépendante de TIRAP en aval du TLR2 est importante pour le contrôle des infections par S. aureus, au moins chez les enfants présentant un défaut de production d’anticorps anti-LTA ; cependant, TIRAP serait un gène en partie redondant dans l’immunité antibactérienne. / We describe a kindred comprising eight individuals with autosomal recessive complete deficiency of TIRAP (also called MAL), an adaptor downstream of TLR2 and TLR4. The 4-year-old proband suffered at three month of age from a life-threatening pneumonia caused by Staphylococcus aureus. Seven adult relatives, aged between 16 and 50 years, homozygous for the same TIRAP mutation never suffered from any serious infections. The rare missense R121W mutation affects a highly conserved amino acid in the TIR domain of TIRAP. The mutant TIRAP allele is expressed but displays loss of function. Responses to a variety of TLR2 agonists, including PAM2CSK4, PAM3CSK4, and FSL-1, and to the TLR4 agonist lipopolysaccharide (LPS), were impaired in fibroblasts, granulocytes, and monocytes of all TIRAP R121W homozygous individuals tested. Interestingly, the whole blood response to staphylococcal lipoteichoic acid (LTA), another TLR2 agonist, was impaired only in the index case. This defective response was due to a lack of anti-LTA antibodies in the patient’s plasma. The combined effect of the TIRAP R121W mutation and the absence of anti-LTA antibody provide an explanation for severe staphylococcal disease in the index case. These results provide the first description of human inherited TIRAP deficiency and help to delineate the role of human TIRAP in TLR responses. They suggest that human TIRAP-dependent TLR2 immunity is important for the control of S. aureus infection, at least in children lacking anti-LTA antibodies, but that TIRAP is otherwise redundant in host defense.
35

Mechanistic And Functional Insights Into Mycobacterium Bovis BCG Triggered TLR2 Signaling : Implications For Immune Evasion Strategies

Ghorpade, Devram Sampat 07 1900 (has links) (PDF)
Mycobacteria are multifaceted pathogens capable of causing both acute disease as well as an asymptomatic latent infection. Host immune responses during mycobacterial infection involve potent cell effector functions including that of CD4+, CD8+ and γδT cells, macrophages and dendritic cells (DCs). Further, the critical regulators of protective immunity to mycobacterial infection include IFN-γ, IL-12, IL-23, TNF-α, lymphotoxins, CD40, nitric oxide and reactive oxygen species. However, the success of mycobacterial infection often relies in its ability to evade immune surveillance mechanisms mediated by sentinels of host immunity by modulating host signal transduction pathways and expression of immunoregulatory molecules. Therefore, the key to control mycobacterial growth and limit pathogenesis lies in the understanding the interactions between Mycobacterium and primary responders like macrophages and DCs. In this scenario, the role of pattern recognition receptors (PPRs) in orchestrating host immune responses assumes central importance. The cell surface receptors play crucial role in influencing overall immune responses. Of the PRRs, the Toll-like receptors (TLRs) form key immune surveillance mechanisms in recognition as well as control of mycobacterial infection. Among them, TLR2 is the primary interacting receptor on antigen presenting cells that recognize the invading mycobacteria. Mycobacterial cell wall constituents such as LAM, LM, PIM and 19-kDa protein have been shown to activate TLR2 signaling leading to proinflammatory responses. Recent reports have suggested that PE_PGRS antigens of M. tuberculosis interact with TLR2. For example, RV0754, Rv0978c, RV1917c have been implicated in modulation of human DCs. The 19-kDa lipoprotein, LpqH (Rv3763) and LprG (Rv1411c) utilize TLR2 signaling to inhibit macrophage responsiveness to IFN-γ triggered MHC class II expression and mycobacterial antigen presentation. Interestingly, recognition and amplification of pathogenic-specific signaling events play important roles in not only discriminating the invading microbes, but also in regulating explicit immune responses. In this context, integration of key signaling centers, which modulate host immunity to pathogenic mycobacterial infections, remains unexplored. In accordance to above observations, signal transduction pathways downstream to TLRs play a critical role in modulation of battery of host cells genes in terms of expression and production of immune modulatory cytokines and chemokines, recruitment of cellular machineries to site of infections etc. This suggests the decisive role for TLRs in modulation of host cell fate decisions. However, during the ensuing immunity to invading pathogens, beside TLR signaling pathways, various other signaling molecules are thought to execute specific functions in divergent cellular contexts. Recent studies from our laboratory have clearly demarcated a novel cross talk of TLR2-NOTCH1 and TLR2-Wnt signaling pathways during mycobacterial infections. The current study primary focuses on the broad range of cross talk of TLR2 and Sonic hedgehog (SHH) signaling pathways and its functional significance. The present investigation demonstrates that M. bovis BCG, a vaccine strain, triggers a robust activation of SHH signaling in macrophages compared to infection with diverse Gram-positive or Gram-negative microbes. This observation was further evidenced by the heightened SHH signaling signatures during in vivo scenario in cells /tissues from pulmonary tuberculosis (TB) individuals as well as tuberculous meningitis (TBM) patients. Furthermore, we show that the sustained TNF-α secretion by macrophages upon infection with M. bovis BCG is a critical necessity for SHH activation. Significantly, perturbation studies implicate a vital role for M. bovis BCG stimulated TLR2/PI3K/PKC/MAPK/NF-κB axis to induce TNF-α, that contributes to enhance SHH signaling. The TNF-α driven SHH signaling downregulates M. bovis BCG induced TLR2 signaling events leading to modulation of battery of genes that regulate various functions of macrophages genes like Vegf-a, Socs-3, Cox-2, Mmp-9 and M1/M2 genes. Importantly, utilizing whole-genome microRNA (miRNA) profiling, roles for specific miRNAs were identified as the molecular regulators that bring about the negative-feedback loop comprising TLR2-SHH signaling events. Thus, the current study illustrates how SHH signaling tightly regulates the kinetics and strengths of M. bovis BCG specific TLR2 responses, emphasizing a novel role for SHH signaling in host immune responses to mycobacterial infections. As described, variety of host factors contributes for ensuing effective host defenses and modulation of host cell fate decisions. Interestingly, avirulent pathogenic mycobacteria, including the vaccine strain M. bovis BCG, unlike virulent M. tuberculosis, cause extensive apoptosis of infected macrophages, which suggests a significant contribution of the apoptosis process to the initiation and subsequent amplification of innate as well as adaptive immune responses. Among various cues that could lead to apoptosis of host cells, the initiation of the apoptotic machinery by posttranscriptional mechanisms assumes significant importance. Among posttranscriptional control mechanisms, miRNAs are suggested to regulate several biological processes including immune responses. Various effectors of host immunity are known to be regulated by several miRNAs, and a prominent one among them, miRNA-155 (miR-155), often exhibits crucial roles during innate or adaptive immune responses. In this perspective, we identified a novel role of miR-155 during M. bovis BCG induced apoptosis of macrophages. The genetic and signaling perturbations data suggested that miR-155 regulates PKA signaling by directly targeting a negative regulator of PKA, protein kinase inhibitor alpha (PKI-α). Enhanced activation of PKA signaling resulted in induced expression of the apoptotic genes as well as Caspase-3 cleavage and Cytochrome c translocation. Thus, augmented PKA signaling by M. bovis BCG-driven miR-155 dictates cell fate decisions of infected macrophages, emphasizing a novel role for miR-155 in host immunity to mycobacterial infections. In perspective of these studies, important directives are often comprised of sequential and coordinated activation of TLR and NLR-driven signal transduction pathways, thus exhibiting foremost influence in determining the overall strength of the innate immune responses. As described, TLR2 exhibits dominant role in sensing various agonists including pathogen-associated molecular patterns (PAMPs) of microbes at the cell surface and generally considered as major effectuator of proinflammatory responses. Interestingly, NLRs like NOD1 or NOD2 often act in contrary, thus regulating anti-inflammatory responses as well as polarization of T cells towards skewed Th2 phenotype. This presents an interesting conundrum to functionality of DCs or macrophages in terms of effector functions during rapidly evolving immunological processes including effects originating from immunosuppressive effectors such as CTLA-4 or TGF-. DCs like macrophages are important sentinels of innate immunity, possesses array of PRRs that include TLRs and NOD-like receptors (NLRs). Signaling events associated with innate sensors like TLRs and NLRs often act as regulatory circuits that modulate the overall functions of DCs in terms of maturation process, cytokine or chemokine production, receptor expression, migration to secondary lymphoid organs for antigen presentation for effectuating Th polarization. TLR2, while acting as sensors for extracellular cues or endocytic network, drives signaling events in response to recognition of PAMPs including mycobacterial antigens like ESAT-6, PE_PGRS antigens, while NOD1 and NOD2 operate as cytosolic sensors initiating signaling pathways upon recognition of diaminopimelic acid (DAP) and muramyl dipeptide (MDP), components of bacterial peptidoglycan. Thus, TLRs or NOD receptors could trigger similar or contrasting immune responses by cooperative or non-cooperative sensing, consequently exhibiting immense complexity during combinatorial triggering of host DCs-PRR repertoire. In view of these observations, our current investigation comprehensively demonstrated that maturation process of human DCs were cooperatively regulated by signaling cascades initiated by engagements of TLR2, NOD1 and NOD2 receptors. Importantly, combined triggering of TLR2 and NOD receptors abolished the TGF-β or CTLA-4-mediated impairment of human DCs maturation, which required critical participation of NOTCH1-PI3K signaling cohorts. Thus, our data delineated the novel insights in modulation of macrophages and DCs effector functions by mycobacterial TLR2 or NOD agonists and broaden our understanding on the signal dynamics and integration of multiple signals from PRRs during mycobacterial infections. Altogether, our findings establish the understanding of conceptual frame work in fine tuning of TLR2 responses by SHH signaling as well as potential co-operativity among TLRs and NODs to modulate NOTCH1 dependent DCs maturation. Importantly, our study provides mechanistic and functional insights into various molecular regulators of macrophage cell fate decisions like miR-31. miR-150 and miR-155, which can fuel the search for attractive and effective drug targets and novel therapeutics to combat diseases of the hour like tuberculosis.
36

Modulation der membranären Lipidzusammensetzung von Makrophagen durch mehrfach ungesättigte Fettsäuren und deren Bedeutung für die TLR2-Signalkaskade

Hellwing, Christine 07 November 2019 (has links)
In der vorliegenden Arbeit wurde der Einfluss mehrfach ungesättigter Fettsäuren (PUFA) auf das Phospho- und Sphingolipidmuster der Non-raft- und Lipid raft-Bereiche in Membranen von RAW264.7-Makrophagen massenspektrometrisch untersucht. Außerdem wurden mittels Fluoreszenzmikroskopie die Auswirkungen einer PUFA-Zugabe auf die Lokalisation des immunologisch bedeutsamen Mustererkennungs-Rezeptor TLR2 und seinen Ko-Rezeptoren TLR1 und TLR6 untersucht.
37

Myeloid Differentiation Factor 88 and Interleukin-1R1 Signaling Contribute to Resistance to Coccidioides Immitis

Viriyakosol, Suganya, Walls, Lorraine, Okamoto, Sharon, Raz, Eyal, Williams, David L., Fierer, Joshua 01 June 2018 (has links)
Rodents are a natural host for the dimorphic pathogenic fungi Coccidioides immitis and Coccidioides posadasii, and mice are a good model for human infection. Humans and rodents both express Dectin-1 and Toll-like receptor 2 (TLR2) on myeloid cells, and those receptors collaborate to maximize the cytokine/chemokine responses to spherules (the tissue form of the fungi) and to formalin-killed spherules (FKS). We showed that Dectin-1 is necessary for resistance to pulmonary coccidioidomycosis, but the importance of TLR2 in vivo is uncertain. Myeloid differentiation factor 88 (MyD88) is the adapter protein for TLR2 and -4, interleukin-1R1 (IL-1R1), and IL-18R1. MyD88/TRIF -/- and MyD88 -/- mice were equally susceptible to C. immitis infection, in contrast to C57BL/6 (B6) controls. Of the four surface receptors, only IL-1R1 was required for resistance to C. immitis, partially explaining the susceptibility of MyD88 -/- mice. We also found that FKS stimulated production of IL-1Ra by bone marrow-derived dendritic cells (BMDCs), independent of MyD88 and Dectin-1. There also was a very high concentration of IL-1Ra in the lungs of infected B6 mice, supporting the potential importance of this regulatory IL-1 family protein in the largely ineffective response of B6 mice to coccidioidomycosis. These results suggest that IL-1R1 signaling is important for defense against C. immitis infection.
38

ENTERIC PARASITE INFECTION-INDUCED ALTERATION OF THE GUT MICROBIOTA REGULATES INTESTINAL GOBLET CELL BIOLOGY AND MUCIN PRODUCTION VIA TLR2 SIGNALLING

Yousefi, Yeganeh January 2022 (has links)
In the gastrointestinal (GI) tract, goblet cells are the major source of mucins, the main structural components of the mucus layer, which functions as the front line of innate defense. The GI tract contains trillions of commensal microbes, and these microbes can manipulate mucin production by activating different signalling cascades initiated by pattern recognition receptors (PRRs), including bacterial sensing Toll-like receptors (TLRs). In addition, sterile α motif pointed domain-containing ETS transcription factor (SPDEF) is a transcription factor that modulates goblet cell differentiation and positively regulates mucin production. During helminth infections, due to the co-existence of parasites and microbiota in close proximity of goblet cells in the gut, it is likely that helminth-microbiota interactions play an important role in mucin production. Indeed, goblet cell hyperplasia and increased mucin production are observed in many enteric helminth infections, including Trichuris muris, and these processes play key roles in host infection clearance. However, it should be noted that the role of microbiota within this axis is not yet understood. Here, we hypothesize T. muris-induced altered microbiota modulates goblet cell differentiation and mucin production via SPDEF-mediated transcriptional regulation and TLR2 signalling. C57BL/6 mice were gavaged with ~300 T. muris eggs. Mice were sacrificed 36 days post-infection. Microbiota from these T. muris-infected and non-infected mice were transferred into two groups of germ-free (GF) mice. Microbiota analysis revealed that treatments in both experiments (infection with T. muris and microbiota transfer from T. muris-infected mice into GF mice) significantly account for the among-sample variations in the composition of the gut microbiota between groups (p <= 0.001). In GF mice, transfer of T. muris-infected microbiota significantly increased goblet cell numbers and TLR2 expression as well as upregulated Muc2 expression compared to MSc Thesis –Yousefi Y; McMaster University – Medical Sciences v GF mice with non-infected microbiota. Antibiotic-treated (ABX-treated) TLR2 knockout (KO) mice after receiving microbiota from T. muris-infected mice showed significantly decreased expression of Muc2 and Muc5ac compared to ABX-treated wild-type (WT) mice receiving the same microbiota. To investigate whether SPDEF is a driving factor for Muc2 production in response to T. muris microbiota stimulation, we next transferred T. muris-infected microbiota into antibiotic-treated SPDEF KO and WT mice. We observed a slight, though not significant, the influence of SPDEF on the stimulation of mucin production by T. muris microbiota. These findings reveal important interactions among parasites, resident microbiota, and host in relation to goblet cell response in the gut. In addition, this study provides new information on TLR2-based innate signalling in the regulation of goblet cell biology and mucin productio / Thesis / Master of Science (MSc)
39

Studies on the exaggerated inflammatory response caused by streptococcus suis at systemic and central nervous system levels

Domínguez Punaro, María de la Cruz 04 1900 (has links)
Streptococcus suis de type 2 est un microorganisme pathogène d’importance chez le porc. Il est la cause de différentes pathologies ayant comme caractéristique commune la méningite. C’est également un agent émergeant de zoonose : des cas cliniques humains ont récemment été rapportés en Asie. Cependant, la pathogénèse de S. suis n’est pas encore complètement élucidée. Jusqu’à présent, la réponse pro-inflammatoire initiée par S. suis n’a été étudiée qu’in vitro. L’étude du choc septique et de la méningite requiert toujours des modèles expérimentaux appropriés. Au cours de cette étude, nous avons développé un modèle in vivo d’infection chez la souris qui utilise la voie d’inoculation intra-péritonéale. Ce modèle a servi à l’étude de la réponse pro-inflammatoire associée à ce pathogène, tant au niveau systémique qu’au niveau du système nerveux central (SNC). Il nous a également permis de déterminer si la sensibilité aux infections à S. suis pouvait être influencée par des prédispositions génétiques de l’hôte. Le modèle d’infection par S. suis a été mis au point sur des souris de lignée CD1. Les résultats ont démontré une bactériémie élevée pendant les trois jours suivant l’infection. Celle-ci était accompagnée d’une libération rapide et importante de différentes cytokines pro-inflammatoires (TNF-α, IL-6, IL-12p40/p70, IFN-ɣ) et de chémokines (KC, MCP-1 and RANTES), qui ont entraîné un choc septique et la mort de 20 % des animaux. Ensuite, pour confirmer le rôle de l’inflammation sur la mortalité et pour déterminer si les caractéristiques génétiques de l’hôte pouvaient influencer la réponse inflammatoire et l’issue de la maladie, le modèle d’infection a été étendu à deux lignées murines consanguines différentes considérées comme résistante : la lignée C57BL/6 (B6), et sensible : la lignée A/J. Les résultats ont démontré une importante différence de sensibilité entre les souris A/J et les souris B6, avec un taux de mortalité atteignant 100 % à 20 h post-infection (p.i.) pour la première lignée et de seulement 16 % à 36 h p.i. pour la seconde. La quantité de bactéries dans le sang et dans les organes internes était similaire pour les deux lignées. Donc, tout comme dans la lignée CD1, la bactériémie ne semblait pas être liée à la mort des souris. La différence entre les taux de mortalité a été attribuée à un choc septique non contrôlé chez les souris A/J infectées par S. suis. Les souris A/J présentaient des taux exceptionnellement élevés de TNF-α, IL-12p40/p70, IL-1β and IFN- γ, significativement supérieurs à ceux retrouvés dans la lignée B6. Par contre, les niveaux de chémokines étaient similaires entre les lignées, ce qui suggère que leur influence est limitée dans le développement du choc septique dû à S. suis. Les souris B6 avaient une production plus élevée d’IL-10, une cytokine anti-inflammatoire, ce qui suppose que la cascade cytokinaire pro-inflammatoire était mieux contrôlée, entraînant un meilleur taux de survie. Le rôle bénéfique potentiel de l’IL-10 chez les souris infectées par S. suis a été confirmé par deux approches : d’une part en bloquant chez les souris B6 le récepteur cellulaire à l’IL-10 (IL-10R) par un anticorps monoclonal anti-IL-10R de souris et d’autre part en complémentant les souris A/J avec de l’IL-10 de souris recombinante. Les souris B6 ayant reçu le anticorps monoclonal anti-IL-10R avant d’être infectées par S. suis ont développé des signes cliniques aigus similaires à ceux observés chez les souris A/J, avec une mortalité rapide et élevée et des taux de TNF-α plus élevés que les souris infectées non traitées. Chez les souris A/J infectées par S. suis, le traitement avec l’IL-10 de souris recombinante a significativement retardé l’apparition du choc septique. Ces résultats montrent que la survie au choc septique dû à S. suis implique un contrôle très précis des mécanismes pro- et anti-inflammatoires et que la réponse anti-inflammatoire doit être activée simultanément ou très rapidement après le début de la réponse pro-inflammatoire. Grâce à ces expériences, nous avons donc fait un premier pas dans l’identification de gènes associés à la résistance envers S. suis chez l’hôte. Une des réussites les plus importantes du modèle d’infection de la souris décrit dans ce projet est le fait que les souris CD1 ayant survécu à la septicémie présentaient dès 4 jours p.i. des signes cliniques neurologiques clairs et un syndrome vestibulaire relativement similaires à ceux observés lors de méningite à S. suis chez le porc et chez l’homme. L’analyse par hybridation in situ combinée à de l’immunohistochimie des cerveaux des souris CD1 infectées a montré que la réponse inflammatoire du SNC débutait avec une augmentation significative de la transcription du Toll-like receptor (TLR)2 et du CD14 dans les microvaisseaux cérébraux et dans les plexus choroïdes, ce qui suggère que S. suis pourrait se servir de ces structures comme portes d’entrée vers le cerveau. Aussi, le NF-κB (suivi par le système rapporteur de l’activation transcriptionnelle de IκBα), le TNF-α, l’IL-1β et le MCP-1 ont été activés, principalement dans des cellules identifiées comme de la microglie et dans une moindre mesure comme des astrocytes. Cette activation a également été observée dans différentes structures du cerveau, principalement le cortex cérébral, le corps calleux, l’hippocampe, les plexus choroïdes, le thalamus, l’hypothalamus et les méninges. Partout, cette réaction pro-inflammatoire était accompagnée de zones extensives d’inflammation et de nécrose, de démyélinisation sévère et de la présence d’antigènes de S. suis dans la microglie. Nous avons mené ensuite des études in vitro pour mieux comprendre l’interaction entre S. suis et la microglie. Pour cela, nous avons infecté des cellules microgliales de souris avec la souche sauvage virulente (WT) de S. suis, ainsi qu’avec deux mutants isogéniques, un pour la capsule (CPS) et un autre pour la production d’hémolysine (suilysine). Nos résultats ont montré que la capsule était un important mécanisme de résistance à la phagocytose pour S. suis et qu’elle modulait la réponse inflammatoire, en dissimulant les composants pro-inflammatoires de la paroi bactérienne. Par contre, l’absence d’hémolysine, qui est un facteur cytotoxique potentiel, n’a pas eu d’impact majeur sur l’interaction de S. suis avec la microglie. Ces études sur les cellules microgliales ont permis de confirmer les résultats obtenus précédemment in vivo. La souche WT a induit une régulation à la hausse du TLR2 ainsi que la production de plusieurs médiateurs pro-inflammatoires, dont le TNF-α et le MCP-1. S. suis a induit la translocation du NF-kB. Cet effet était plus rapide dans les cellules stimulées par le mutant déficient en CPS, ce qui suggère que les composants de la paroi cellulaire représentent de puissants inducteurs du NF-kB. De plus, la souche S. suis WT a stimulé l’expression de la phosphotyrosine, de la PKC et de différentes cascades liées à l’enzyme mitogen-activated protein kinase (MAPK). Cependant, les cellules microgliales infectées par le mutant déficient en CPS ont montré des profils de phosphorylation plus forts et plus soutenus que celles infectées par le WT. Finalement, la capsule a aussi modulé l’expression de l’oxyde nitrique synthétase inductible (iNOS) induite par S. suis et par la production subséquente d’oxyde nitrique par la microglie. Ceci pourrait être lié in vivo à la neurotoxicité et à la vasodilatation. Nous pensons que ces résultats contribueront à une meilleure compréhension des mécanismes sous-tendant l’induction de l’inflammation par S. suis, ce qui devrait permettre, d’établir éventuellement des stratégies plus efficaces de lutte contre la septicémie et la méningite. Enfin, nous pensons que ce modèle expérimental d’infection chez la souris pourra être utilisé dans l’étude de la pathogénèse d’autres bactéries ayant le SNC pour cible. / Streptococcus suis serotype 2 is an important swine pathogen responsible for diverse infections, meningitis being its most striking feature. In addition, it is an emerging agent of zoonosis, which has gained worldwide attention due to important outbreaks in Asia. Understanding the pathogenesis of S. suis infections still represents a challenge. Up to present, the pro-inflammatory response due to S. suis has only been studied in vitro, and there is still a great need of appropriate experimental models for both septic shock and meningitis. In the present study, we successfully developed an in vivo model of S. suis infection in adult mice infected by the intraperitoneal route. This model served to investigate the pro-inflammatory events that take place at both the systemic and Central Nervous System (CNS) levels associated with this important pathogen. In addition, this model was useful to determine if susceptibility to S. suis infection may be influenced by the genetic background of the host. The mouse model of S. suis infection was standardized in CD1 mice. Results showed sustained bacteremia during the 3 days post-infection (p.i.), accompanied by a quick and substantial release of different pro-inflammatory cytokines (TNF-α, IL-6, IL-12p40/p70, IFN-ɣ) and chemokines (KC, MCP-1 and RANTES) that lead to septic shock and 20% mortality in mice. Once the hallmark of the septic phase of S. suis infection was established in CD1 mice, research continued with the objective to confirm the role of inflammation in mortality and to determine if the genetic background of the host may influence the inflammatory response toward this pathogen and the further outcome of the disease. For this, the mouse model of S. suis infection was used with two genetically different inbred mouse strains, this is, C57BL/6 (B6) and A/J mice, which are considered as the prototype of Th1-type and Th2-type mice, respectively. Results demonstrated a striking susceptibility to S. suis infection in A/J mice in comparison to B6 mice, with 100% mortality in the former mice strain at 20 h p.i., and 16 % mortality at 36 h p.i. for the latter. Very interestingly, and similarly to CD1 mice, bacteremia did not seem to be responsible for the death of mice, as both mice strains presented similar amounts of bacteria in blood and organs. Thus, it was postulated that the higher mortality in S. suis-infected A/J mice was due to uncontrolled septic shock. In fact, A/J mice presented very high levels of TNF-α, IL-12p40/p70, IL-1β and IFN-ɣ, that significantly exceeded those found in B6 mice. Remarkably, chemokine levels were similar between strains, suggesting their limited participation in the development of septic shock by S. suis. A greater survival of B6 mice was partially related to a better regulation of the pro-inflammatory cytokine cascade, as they showed a higher production of the anti-inflammatory cytokine IL-10 than A/J mice. The potential beneficial role of the IL-10 in mice infected with S. suis was confirmed using two approaches: the first, by blockage of the cell receptor of IL-10 (IL-10R) with an anti-mouse IL-10R monoclonal antibody (Mab) in B6 mice and the second by administrating recombinant mouse (rm)IL-10 (rmIL-10) to A/J mice. B6 mice that received the IL-10R MAb treatment before challenge with S. suis developed a clinical acute disease similar to that observed with A/J mice, with a striking and rapid increase in mortality and higher levels of TNF-α in comparison to those of infected mice that did not receive the treatment. Controversially, treatment with rmIL-10 significantly delayed the onset of septic shock in A/J mice infected with S. suis. These results show that survival from S. suis septic shock requires a tight regulation of pro- and anti-inflammatory mechanisms, and that the latter should be activated at the same time or soon after the onset of the pro-inflammatory response. This part of the study may represent a first step in the identification of host genes associated with resistance against S. suis. One of the most important achievements of the mouse model of infection described in this project is the development of distinct clinical signs of neurological disease in CD1 mice from 4 days p.i. Indeed, in CD1 mice that survived sepsis due to S. suis infection, clinical signs of neurological disease and vestibular syndrome, which are quite similar to those observed in clinical cases of S. suis meningitis in both pigs and humans, were observed. Studies of the brains of infected CD1 mice using in situ hybridization combined with immunocytochemistry, demonstrated that the CNS inflammatory response began with a significant increase in the transcription of Toll-like receptor (TLR)2 and CD14 initially in the brain microvasculature and choroid plexuses, suggesting that S. suis may use these structures as portals of entry to the brain. There also was activation of NF-κB (as indicated by transcriptional activation of IκBα as a reporter system) and TNF-α, IL-1β and MCP-1, mainly in cells identified as microglia and to a lesser extent in astrocytes. These signals reached different brain structures, mainly the brain cortex, corpus callosum, hippocampus, choroid plexuses, thalamus, hypothalamus and meninges. All of these pro-inflammatory events were associated with extensive areas of inflammation and necrosis, severe demyelination and presence of antigens of S. suis inside microglia. In vitro studies were conducted in order to better understand the interactions of S. suis and microglia. For this, mouse microglia were infected with a virulent wild type (WT) strain of S. suis. Two isogenic mutants deficient in capsule (CPS) or hemolysin production (suilysin, SLY) respectively, were also included for comparative purposes. The CPS was important for S. suis resistance to phagocytosis, and it also modulated the inflammatory response by hiding pro-inflammatory components from the bacterial cell wall. On the other hand, the absence of SLY, a potential cytotoxic factor, did not have a major impact on S. suis interactions with microglia. Studies with microglia helped to confirm previous findings in vivo in mice, as the WT S. suis strain induced the up-regulation of TLR2 and the production of several pro-inflammatory mediators, including TNF-α and MCP-1. As observed in mice, S. suis induced NF-kB translocation, which was more rapid for cells stimulated with the CPS-deficient mutant, suggesting that bacterial cell wall components are potent inducers of NF-kB. Moreover, WT S. suis promoted phosphotyrosine, PKC and different mitogen-activated protein kinase (MAPK) events. However, microglia infected with the CPS-deficient mutant showed overall stronger and more sustained phosphorylation profiles. Finally, the CPS also modulated S. suis-induced inducible nitrogen oxide synthase (iNOS) expression and further nitric oxide production in microglia, which could be related to neurotoxicity and vasodilatation in vivo. We are confident that our results may help to more fully understand the mechanisms underlying S. suis induction of inflammation, leading to the design of more efficient anti-inflammatory strategies for sepsis and meningitis. Finally, we believe this experimental model of infection in mice could also be useful for studying the pathogenesis of infections of the CNS, due to other bacteria.
40

Aspectos moleculares da gênese e progressão de lesões periapicais induzidas experimentalmente em camundongos / Molecular aspects of genesis and progression of induced apical periodontitis in mice

Barreiros, Driely 18 July 2017 (has links)
O conhecimento dos eventos biológicos que ocorrem no periápice dos dentes com necrose pulpar se torna importante para compreender o desenvolvimento das lesões periapicais. Muitas são as moléculas e mediadores que participam na instalação da lesão periapical, a partir da infecção bacteriana que ocorre no interior dos canais radiculares. Assim, o objetivo do presente trabalho foi avaliar moléculas do sistema imune inato, da osteoclastogênese e metaloproteinases em lesões periapicais (LP) induzidas experimentalmente em camundongos knockout e wild type. Para esse objetivo, o presente estudo foi dividido em dois trabalhos distintos. O primeiro teve como objetivo avaliar a expressão de metaloproteinase 2 (MMP2) e metaloproteinase 9 (MMP9) durante a progressão da LP em camundongos knockout para TLR2 (TLR2 KO) e MyD88 (MyD88 KO), em comparação com camundongos wild type (WT). O segundo estudo avaliou a correlação da expressão gênica e imunomarcação de RANK, RANKL, OPG, TLR2 e MyD88 durante a progressão da LP em camundongos WT. No primeiro estudo lesões periapicais foram induzidas em molares inferiores de 54 camundongos TLR2 KO, MyD88 KO e WT (n=18/grupo). Após 7, 21 e 42 dias, os animais foram eutanaziados e as mandíbulas foram dissecadas e submetidas a processamento histotécnico. Os cortes histológicos foram submetidos a imunohistoquímica e posteriormente foi avaliada presença ou ausência de MMP2 e MMP9 nos diferentes grupos. No segundo estudo, 35 camundongos WT foram utilizados. As lesões periapicais foram induzidas nos primeiros molares inferiores de ambos os lados. Após 0 (G0), 7 (G7), 21 (G21) e 42 (G42) dias, os animais foram anestesiados e eutanasiados para que as mandíbulas fossem dissecadas e divididas ao meio.O lado direito das mandíbulas foi para o processamento histotécnico, para posterior marcação de RANK, RANKL, OPG, TLR2 e MyD88, por meio da imuno-histoquímica do lado esquerdo da mandíbula foi utilizado para a extração de RNA, para a determinação da expressão gênica de RANK (Tnfrsf11a), RANKL (Tnfrsf11), OPG (Tnfrsf11b), TLR2 (Tlr2) e MyD88 (Myd88) utilizando quantificação em Tempo Real da Reação da Polimerase em Cadeia (qRT-PCR). Para ambos os estudos, testes paramétricos e não paramétricos foram realizados com nível de significância de 5%. Foi possível observar, no primeiro estudo, que nos períodos iniciais da progressão da lesão periapical, houve um aumento na imunomarcação de MMP9 nos camundongos TLR2 KO e MyD88 KO, quando comparados aos WT, diferente da MMP2 que não se observou nenhum aumento na imunomarcação. No entanto, aos 42 dias observou-se uma redução da imunomarcação de MMP2 e um aumento da MMP9 nos camundongos TLR2 KO. Adicionalmente, no segundo estudo, foi possível observar um aumento da imunomarcação para RANK, RANKL, OPG, TLR2 e MyD88 durante a progressão da lesão periapical (p<0,05). O aumento da expressão de Tnfrsf11 foi diferente entre os grupos G0 e G42, e G21 e G42 (p=0,006). No entanto, a expressão de Tnfrsf11b foi diferente entre os grupos G0 e G7, G7, G21 e G42, sendo possível observar uma diminuição dessa expressão ao longo do tempo (p<0,001). Tlr2 foi mais expresso entre os grupos G0 e G42 (p=0,03). E a expressão da molécula Myd88 foi estatisticamente significante entre os grupos G0 e G7, G21 e G42 (p=0,01). A razão Tnfrsf11/Tnfrsf11b aumentou durante a progressão da lesão periapical (p=0,002). Também foi possível observar uma correlação moderada entre Myd88 e Rankl (r=0,42; p=0,03) e entre Myd88 e Tlr2 (r=0,48; p<0,0001). Após as metodologias empregadas e os dados analisados, concluímos que a produção de MMP2 e MMP9 foi modulada por TLR2 e Myd88 durante a progressão da lesão periapical. Alem disso, podemos sugerir que existe uma correlação positiva entre o sistema RANK/RANKL/OPG e as proteínas do sistema imune inato, TLR2 e MyD88, durante a perda óssea decorrente da infecção bacteriana dos canais radiculares e posterior progressão da lesão periapical. / Knowledge of the biological events occurring inteeth apex with pulp necrosis becomes important to understand the development of periapical lesions. There are manymolecules and mediators that participate in the installation of the periapical lesion, from the bacterial infection that occurs inside the root canals. Thus, the aim of the present study was to evaluate molecules of the innate immune system, osteoclastogenesis and metalloproteinases in experimentally apical periodontitis (AP) induced in knockout and wild type mice. For this purpose, the present study was divided into two distinct studies. The first one aimed to evaluate the expression of metalloproteinases 2 (MMP2) and metalloproteinases 9 (MMP9) during the progression of AP in TLR2 knockout mice (TLR2 KO) and MyD88 knockout mice (MyD88 KO), compared to wild type mice (WT). The second study evaluated the correlation of gene expression and immunostaining of RANK, RANKL, OPG, TLR2 and MyD88 during LP progression in WT mice. In the first study AP were induced in lower molars of 54 TLR2 KO, MyD88 KO and WT mice (n = 18 / group). After 7, 21 and 42 days, the animals were euthanized and the jaws were dissected and submitted to histotechnical processing. The histological sections were submitted to immunohistochemistry and subsequently the presence or absence of MMP2 and MMP9 in the different groups was evaluated. In the second study, 35 WT mice were used. Periapical lesions were induced in the lower first molars on both sides. After 0 (G0) to 7 (G7), 21 (G21) and 42 (G42) days, the animals were anesthetized and euthanized so that the jaws were dissected and divided in half. The right side of the jaws was for the histotechnic processing, for subsequent imunostaining of RANK, RANKL, OPG, TLR2 and MyD88, through immunohistochemistry and the left side of the jaws was used for the extraction of RNA, for the determination of expression of RANK (Tnfrsf11a), RANKL (Tnfrsf11), OPG (Tnfrsf11b), TLR2 (Tlr2) and MyD88 (Myd88) using Quantification Real Time of Polymerase Chain Reaction (qRT-PCR). For both studies, parametric and non-parametric tests were performed with significance level of 5%. It was possible to observe in the first study that in the initial periods of AP progression there was an increase in MMP9 immunostaining in TLR2 KO and MyD88 KO mice when compared to WT, different from MMP2 that no increase in immunostaining was observed. However, at 42 days there was a reduction in MMP2 immunostaining and an increase of MMP9 in TLR2 KO mice was observed. Additionally, in the second study, it was possible to observe an increase in the immunostaining for RANK, RANKL, OPG, TLR2 and MyD88 during periapical lesion progression (p <0.05). The increase in Tnfrsf11 expression was different between groups G0 and G42, and G21 and G42 (p = 0.006). However, the expression of Tnfrsf11b was different between the G0 and G7, G7, G21 and G42 groups, and a decrease in expression over time (p <0.001) was observed. Tlr2 was more expressed between the G0 and G42 groups (p = 0.03). And the expression of the Myd88 molecule was statistically significant between the G0 and G7, G21 and G42 groups (p = 0.01). The Tnfrsf11 / Tnfrsf11b ratio increased during the AP progression (p = 0.002). It was also possible to observe a moderate correlation between Myd88 and Rankl (r = 0.42, p = 0.03) and between Myd88 and Tlr2 (r = 0.48, p <0.0001). After the methodologies used and the data analyzed, we conclude that the production of MMP2 and MMP9 was modulated by TLR2 and Myd88 during the AP progression. In addition, we can suggest that there is a positive correlation between the RANK / RANKL / OPG system and the proteins of the innate immune system, TLR2 and MyD88, during bone loss due to bacterial infection of the root canals and subsequent progression of the apical periodontitis.

Page generated in 0.0205 seconds