981 resultados para Plasmacytoid dendritic cell


Relevância:

90.00% 90.00%

Publicador:

Resumo:

The immunopathophysiologic development of systemic autoimmunity involves numerous factors through complex mechanisms that are not fully understood. In systemic lupus erythematosus, type I IFN (IFN-I) produced by plasmacytoid dendritic cells (pDCs) critically promotes the autoimmunity through its pleiotropic effects on immune cells. However, the host-derived factors that enable abnormal IFN-I production and initial immune tolerance breakdown are largely unknown. Previously, we found that amyloid precursor proteins form amyloid fibrils in the presence of nucleic acids. Here we report that nucleic acid-containing amyloid fibrils can potently activate pDCs and enable IFN-I production in response to self-DNA, self-RNA, and dead cell debris. pDCs can take up DNA-containing amyloid fibrils, which are retained in the early endosomes to activate TLR9, leading to high IFNα/β production. In mice treated with DNA-containing amyloid fibrils, a rapid IFN response correlated with pDC infiltration and activation. Immunization of nonautoimmune mice with DNA-containing amyloid fibrils induced antinuclear serology against a panel of self-antigens. The mice exhibited positive proteinuria and deposited antibodies in their kidneys. Intriguingly, pDC depletion obstructed IFN-I response and selectively abolished autoantibody generation. Our study reveals an innate immune function of nucleic acid-containing amyloid fibrils and provides a potential link between compromised protein homeostasis and autoimmunity via a pDC-IFN axis.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

A comprehensive understanding of the complex, autologous cellular interactions and regulatory mechanisms that occur during normal dendritic cell (DC)-stimulated immune responses is critical to optimizing DC-based immunotherapy. We have found that mature, immunogenic human monocyte-derived DCs (moDCs) up-regulate the immune-inhibitory enzyme, indoleamine 2,3-dioxygenase (IDO). Under stringent autologous culture conditions without exogenous cytokines, mature moDCs expand regulatory T cells (Tregs) by an IDO-dependent mechanism. The priming of resting T cells with autologous, IDO-expressing, mature moDCs results in up to 10-fold expansion of CD4(+)CD25(bright)Foxp3(+)CD127(neg) Tregs. Treg expansion requires moDC contact, CD80/CD86 ligation, and endogenous interleukin-2. Cytofluorographically sorted CD4(+) CD25(bright)Foxp3(+) Tregs inhibit as much as 80% to 90% of DC-stimulated autologous and allogeneic T-cell proliferation, in a dose-dependent manner at Treg:T-cell ratios of 1:1, 1:5, and as low as 1:25. CD4(+)CD25(bright)Foxp3(+) Tregs also suppress the generation of cytotoxic T lymphocytes specific for the Wilms tumor antigen 1, resulting in more than an 80% decrease in specific target cell lysis. Suppression by Tregs is both contact-dependent and transforming growth factor-beta-mediated. Although mature moDCs can generate Tregs by this IDO-dependent mechanism to limit otherwise unrestrained immune responses, inhibition of this counter-regulatory pathway should also prove useful in sustaining responses stimulated by DC-based immunotherapy.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Introduction: Recently, mesenchymal stem cells (MSC) of perivascular origin have been identified in several organs not including the heart. Using a novel cell isolation protocol, we have isolated cells sharing common characteristics from mouse hearts and pancreas. The aim of the present study was to characterize these cells in vitro.Methods: Cells were isolated from neonatal and adult mouse hearts and pancreas and cultured for more than 6 months. Surface marker expression was analyzed by flow cytometry and immunocytochemistry. Cell differentiation was tested using multiple differentiation media. Insulin production by pancreas-derived cells was tested by dithizone staining.Results: Cells showing a similar, distinctive morphology were obtained from the heart and pancreas after 4-8 weeks of culture. Cells from the two organs also showed a very similar immunophenotype, characterized by expression of c-kit (stem cell factor receptor), CD44, the common leukocyte marker CD45, and the monocytic markers CD11b and CD14. A significant proportion of cardiac and pancreatic cells expressed NG2, a marker for pericytes and other vascular cells. A significant proportion of cardiac, but not of pancreatic cells expressed stem cell antigen-1 (Sca-1). However, cells did not express T, B or dendritic cell markers. Cells of both cardiac and pancreatic origin spontaneously formed "spheres" (spherical cell aggregates similar to "neurospheres" formed by neural stem cells) in vitro. Cardiosphere formation was enhanced by TNF-alpha. Several cardiospheres (but no "pancreatospheres") derived from neonatal (but not adult) cells showed spontaneous rhythmic contractions, thus demonstrating cardiac differentiation (this was confirmed by immunostaining for alpha-sarcomeric actinin). Beating activity was enhanced by low serum conditions. Cells from both organs formed adipocytes, osteocytes and osteocytes under appropriate conditions, the typical differentiation pattern of MSCs. Pancreas-derived cells also formed dithizonepositive insulin-producing cells.Conclusions: We have defined cardiac and pancreatic cell populations that share a common morphology, growth characteristics, and a unique immunophenotype. Expression of perivascular and monocytic markers, along with stem/priogenitor cell markers by these cells suggests a relationship with pericytes-mesoangioblasts and so-called multipotent monocytes. Cells show MSC-typical growth and differentiation patterns, together with tissue-specific differentiation potential: cardiomyocytes for cardiac-derived cells and insulinproducing cells for pancreas-derived cells.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Although the importance of the NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome in health and disease is well appreciated, a precise characterization of NLRP3 expression is yet undetermined. To this purpose, we generated a knock-in mouse in which the Nlrp3 coding sequence was substituted for the GFP (enhanced GFP [egfp]) gene. In this way, the expression of eGFP is driven by the endogenous regulatory elements of the Nlrp3 gene. In this study, we show that eGFP expression indeed mirrors that of NLRP3. Interestingly, splenic neutrophils, macrophages, and, in particular, monocytes and conventional dendritic cells showed robust eGFP fluorescence, whereas lymphoid subsets, eosinophils, and plasmacytoid dendritic cells showed negligible eGFP levels. NLRP3 expression was highly inducible in macrophages, both by MyD88- and Trif-dependent pathways. In vivo, when mice were challenged with diverse inflammatory stimuli, differences in both the number of eGFP-expressing cells and fluorescence intensity were observed in the draining lymph node. Thus, NLRP3 levels at the site of adaptive response initiation are controlled by recruitment of NLRP3-expressing cells and by NLRP3 induction.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Adjuvants are vaccine additives that stimulate the immune system without having any specific antigenic effect of itself. In this study we show that alum adjuvant induces the release of IL-1beta from macrophages and dendritic cells and that this is abrogated in cells lacking various NALP3 inflammasome components. The NALP3 inflammasome is also required in vivo for the innate immune response to OVA in alum. The early production of IL-1beta and the influx of inflammatory cells into the peritoneal cavity is strongly reduced in NALP3-deficient mice. The activation of adaptive cellular immunity to OVA-alum is initiated by monocytic dendritic cell precursors that induce the expansion of Ag-specific T cells in a NALP3-dependent way. We propose that, in addition to TLR stimulators, agonists of the NALP3 inflammasome should also be considered as vaccine adjuvants.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Dendritic cells (DCs) are the most efficient antigen presenting cells, they provide co-stimulation, are able to secrete various proinflammatory cytokines and therefore play a pivotal role in shaping adaptive immune responses. Moreover, they are important for the promotion and maintenance of central and peripheral tolerance through several mechanisms like the induction of anergy or apoptosis in effector T cells or by promoting regulatory T cells. The murine CD8α+ (MuTu) dendritic cell line was previously derived and described in our laboratory. The MuTu cell line has been shown to maintain phenotypical and functional characteristics of endogenous CD8α+ DCs. They are able to cross-present exogenous antigens to CD8+ T cells and produce interleukin (IL-) 12 upon engagement of Toll like receptors. The cell line constitutes an infinite source of homogenous, phenotypically well-defined dendritic cells. This allows us to investigate the role and potential of specific molecules in the induction as well as regulation of immune responses by DCs in a rational and standardized way. In a first project the MuTu dendritic cell line was transduced in order to stably express the immunosuppressive molecules IL-10, IL-35 or the active form of TGF-β (termed IL-10+DC, IL-35+DC or actTGFβ+DC). We investigated the capability of these potentially suppressive or tolerogenic dendritic cell lines to induce immune tolerance and explore the mechanisms behind tolerance induction. The expression of TGF-β by the DC line did not affect the phenotype of the DCs itself. In contrast, IL-10+ and IL-35+DCs were found to exhibit lower expression of co-stimulatory molecules and MHC class I and II, as well as reduced secretion of pro-inflammatory cytokines upon activation. In vitro co-culture with IL-35+, IL10+ or active TGFβ+ DCs interfered with function and proliferation of CD4+ and CD8+ T cells. Furthermore, IL-35 and active TGF-β expressing DC lines induced regulatory phenotype on CD4+ T cells in vitro without or with expression of Foxp3, respectively. In different murine cancer models, vaccination with IL-35 or active TGF-β expressing DCs resulted in faster tumor growth. Interestingly, accelerated tumor growth could be observed when IL-35-expressing DCs were injected into T cell-deficient RAG-/- mice. IL-10expressing DCs however, were found to rather delay tumor growth. Besides the mentioned autocrine effects of IL-35 expression on the DC line itself, we surprisingly observed that the expression of IL-35 or the addition of IL-35 containing medium enhances neutrophil survival and induces proliferation of endothelial cells. Our findings indicate that the cytokine IL-35 might not only be a potent regulator of adaptive immune responses, but it also implies IL-35 to mediate diverse effects on an array of cellular targets. This abilities make IL-35 a promising target molecule not only for the treatment of auto-inflammatory disease but also to improve anti-cancer immunotherapies. Indeed, by applying active TGFβ+ in murine autoimmune encephalitis we were able to completely inhibit the development of the disease, whereas IL-35+DCs reduced disease incidence and severity. Furthermore, the preventive transfer of IL-35+DCs delayed rejection of transplanted skin to the same extend as the combination of IL-10/actTGF-β expressing DCs. Thus, the expression of a single tolerogenic molecule can be sufficient to interfere with the adequate activation and function of dendritic cells and of co-cultured T lymphocytes. The respective mechanisms of tolerance induction seem to be different for each of the investigated molecule. The application of a combination of multiple tolerogenic molecules might therefore evoke synergistic effects in order to overcome (auto-) immunity. In a second project we tried to improve the immunogenicity of dendritic cell-based cancer vaccines using two different approaches. First, the C57BL/6 derived MuTu dendritic cell line was genetically modified in order to express the MHC class I molecule H-2Kd. We hypothesized that the expression of BALB/c specific MHC class I haplotype (H-2Kd) should allow the priming of tumor-specific CD8+ T cells by the otherwise allogeneic dendritic cells. At the same time, the transfer of these H-2Kd+ DCs into BALB/c mice was thought to evoke a strong inflammatory environment that might act as an "adjuvant", helping to overcome tumor induced immune suppression. Using this so called "semi-allogeneic" vaccination approach, we could demonstrate that the delivery of tumor lysate pulsed H-2Kd+ DCs significantly delayed tumor growth when compared to autologous or allogeneic vaccination. However, we were not able to coherently elucidate the cellular mechanisms underlying the observed effect. Second, we generated MuTu DC lines which stably express the pro-inflammatory cytokines IL-2, IL-12 or IL-15. We investigated whether the combination of DC vaccination and local delivery of pro-inflammatory cytokines might enhance tumor specific T cell responses. Indeed, we observed an enhanced T cell proliferation and activation when they were cocultured in vitro with IL-12 or IL-2-expressing DCs. But unfortunately we could not observe a beneficial or even synergistic impact on tumor development when cytokine delivery was combined with semi-allogeneic DC vaccination.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

T cells play a critical role in tumor immune surveillance as evidenced by extensive mouse-tumor model studies as well as encouraging patient responses to adoptive T cell therapies and dendritic cell vaccines. It is well established that the interplay of tumor cells with their local cellular environment can trigger events that are immunoinhibitory to T cells. More recently it is emerging that the tumor vasculature itself constitutes an important barrier to T cells. Endothelial cells lining the vessels can suppress T cell activity, target them for destruction, and block them from gaining entry into the tumor in the first place through the deregulation of adhesion molecules. Here we review approaches to break this tumor endothelial barrier and enhance T cell activity.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Interleukin 17-producing helper T cells (TH17 cells) have a major role in protection against infections and in mediating autoimmune diseases, yet the mechanisms involved are incompletely understood. We found that interleukin 26 (IL-26), a human TH17 cell-derived cytokine, is a cationic amphipathic protein that kills extracellular bacteria via membrane-pore formation. Furthermore, TH17 cell-derived IL-26 formed complexes with bacterial DNA and self-DNA released by dying bacteria and host cells. The resulting IL-26-DNA complexes triggered the production of type I interferon by plasmacytoid dendritic cells via activation of Toll-like receptor 9, but independently of the IL-26 receptor. These findings provide insights into the potent antimicrobial and proinflammatory function of TH17 cells by showing that IL-26 is a natural human antimicrobial that promotes immune sensing of bacterial and host cell death.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Neutrophil extracellular traps (NETs) formation is a cell death mechanism characterized by the extrusion of DNA fibers associated to antimicrobial peptides such as LL37. Beside their antimicrobial role, NETs are highly immunogenic by their ability to activate plasmacytoid dendritic cells (pDCs). In this context, LL37 binds to NET-DNA, leading to endosomal Toll¬like-receptor (TLR) 9 binding, resulting in Interferon alpha (IFNa) production by pDCs. Uncontrolled pDC activation by NETs is an important player in the pathogenesis of autoimmune disease such as Lupus Erythematosus (LE); however the regulation of NET- driven pDC activation is poorly characterized. Olfactomedin 4 (OLFM4) is a granule protein present in a subset of circulating neutrophils and was shown to bear anti-inflammatory properties in a mouse model, raising the possibility that it may regulate neutrophil-induced inflammation. Therefore, in this project, we aimed at deciphering the mechanism by which OLFM4 may regulate inflammation induced by NET-activated pDC and its relevance in the pathogenesis of Lupus Erythematosus (LE). First, we show that OLFM4 directly interacted with LL37 in neutrophils, impairing LL37/DNA complexes formation and pDC activation to produce IFNa. Then, by using an in vivo model of acute inflammation depending on NET- driven activation of pDCs, we observed that the absence of Olfm4 led to uncontrolled type I IFN production, confirming the regulatory role of neutrophil-derived OLFM4. Beyond controlling NET-induced inflammation, we also show that OLFM4 could inhibit pDC activation mediated by DNA-containing immune complexes (ICs), suggesting that OLFM4 holds anti¬inflammatory properties in the context of LE. Of note, we identified a previously unknown population of OLFM4hi9h neutrophils in healthy individuals that may belong to the immunosuppressive subset of granulocytic myeloid-derived suppressor cells (g-MDSCs). Strikingly, we observed a decreased frequency of OLFM4h'9h cells among inflammatory Low density granulocytes (LDGs) neutrophils in LE patients, suggesting that a disequilibrium between pro- and anti-inflammatory neutrophils may participate to the disease pathogenesis. Altogether, this study demonstrates that OLFM4 is involved in the resolution of inflammation. -- La NETose (formation de Neutrophil Extracellular Traps, NETs) est une réponse à un stimulus inflammatoire caractérisée par l'expulsion de l'ADN lié à des peptides antimicrobiens comme le LL37, induisant la mort de la cellule. Les NETs possèdent des propriétés antibactériennes et sont pro-inflammatoires via leur capacité à activer les cellules dendritiques plasmacytoïdes (pDCs). Dans ce contexte, les complexes ADN/LL37 libérés lient le récepteur Toll-like 9 des pDCs, induisant la production d'Interféron alpha (IFNa). La production incontrôlée d'IFNa par les pDCs est impliquée dans la pathogenèse du Lupus Erythemateux (LE), cependant la régulation de l'activation des pDCs reste mal connue. L'Oflactomédine 4 (OLFM4) est une protéine produite par une sous-population de neutrophiles, avec des propriétés anti-inflammatoires possibles. Le but de ce projet était d'identifier les mécanismes par lesquels l'OLFM4 pourrait réguler l'inflammation induite par les NETs et sa relevance dans la pathogenèse du LE. Tout d'abord, nous avons montré que l'OLFM4 interagissait avec le LL37, empêchant la production des complexes ADN/LL37 qui activent les pDCs. Nous avons vérifié notre hypothèse in vivo en utilisant un modèle murin d'inflammation locale dépendant des pDCs et des NETs. Dans ce contexte, le déficit en Olfm4 était associé à une production accrue d'IFNa, confirmant le rôle de l'OLFM4 dans le contrôle de l'inflammation. De plus, l'OLFM4 pouvait également inhiber l'activation des pDCs induite par des complexes immuns, suggérant que l'OLFM4 serait aussi anti-inflammatoire dans le contexte du LE. Ensuite, nous avons identifié une nouvelle population de neutrophiles OLFM4h'9h chez les sujets sains qui pourraient appartenir au sous-type anti¬inflammatoire des g-MDSCs (granulocytic myeloid-derived suppressor cells). Nous avons observé une diminution de ces cellules parmi les neutrophiles pro-inflammatoires LDGs (Low Density Granulocytes) dans le LE suggérant qu'un déséquilibre entre les sous-types de neutrophiles pourrait participer à l'inflammation excessive de cette maladie. Ces travaux mettent en évidence l'implication de l'OLFM4 dans la résolution de l'inflammation et suggèrent qu'une expression altérée de l'OLFM4 pourrait participer à la pathogenèse du LE. -- Les neutrophils constituent la majorité des globules blancs circulants et sont rapidement mobilisés depuis le sang dans un organe lésé en cas d'infection ou de blessure. Ils représentent la première ligne de défense du système immunitaire. Ils sont indispensables dans la défense contre les infections par leur capacité à tuer les bactéries, par exemple en produisant des peptides antimicrobiens (AMPs) qui fonctionnent comme des antibiotiques naturels. De plus, les neutrophiles recrutent les autres membres du système immunitaire qui sont nécessaires à l'éradication complète des microbes et à la réparation des tissus. Les nombreux outils permettant aux neutrophiles de contrôler les infections ne sont cependant pas sans danger pour les tissus. En effet, diverses molécules comme les AMPs peuvent induire des dommages tissulaires substantiels en participant au développement d'une inflammation chronique. Ceci est particulièrement le cas lorsque les neutrophiles meurent par un processus nommé NETose. Dans ce contexte, la cellule subit une dissolution de sa membrane suivie de l'expulsion de son ADN associé à des AMPs. Ces complexes formés d'ADN et d'AMPs induisent la production de cytokines pro-inflammatoires dont l'Interféron alpha (IFNa). Certaines maladies auto-immunes comme le lupus érythémateux sont associées à un excès de NETose produit par les neutrophiles et à un excès d'IFNa qui participe au développement de la maladie. Dans cette thèse, nous avons montré que l'Olfactomédine 4 (OLFM4), une protéine produite par les neutrophiles eux-mêmes, est un inhibiteur de cette inflammation. Nous avons démontré que TOLFM4 empêchait la formation des complexes ADN/AMPs, réduisant par là la production d'IFNa in vitro et in vivo. Finalement, nos recherches ont suggéré que l'OLFM4 pourrait être insuffisamment produite chez les patients souffrant de lupus, ce qui pourrait participer à l'inflammation chronique associée à la maladie.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

NOD-like receptors (NLR) are a family of cytosolic pattern recognition receptors that include many key drivers of innate immune responses. NLRP12 is an emerging member of the NLR family that is closely related to the well-known inflammasome scaffold, NLRP3. Since its discovery, various functions have been proposed for NLRP12, including the positive regulation of dendritic cell (DC) and neutrophil migration and the inhibition of NF-κB and ERK signalling in DC and macrophages. We show here that NLRP12 is poorly expressed in murine macrophages and DC, but is strongly expressed in neutrophils. Using myeloid cells from WT and Nlrp12(-/)(-) mice, we show that, contrary to previous reports, NLRP12 does not suppress LPS- or infection-induced NF-κB or ERK activation in myeloid cells, and is not required for DC migration in vitro. Surprisingly, we found that Nlrp12 deficiency caused increased rather than decreased neutrophil migration towards the chemokine CXCL1 and the neutrophil parasite Leishmania major, revealing NLRP12 as a negative regulator of directed neutrophil migration under these conditions.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

BACKGROUND: Transmission of mucosal pathogens relies on their ability to bind to the surfaces of epithelial cells, to cross this thin barrier, and to gain access to target cells and tissues, leading to systemic infection. This implies that pathogen-specific immunity at mucosal sites is critical for the control of infectious agents using these routes to enter the body. Although mucosal delivery would ensure the best onset of protective immunity, most of the candidate vaccines are administered through the parenteral route. OBJECTIVE: The present study evaluates the feasibility of delivering the chemically bound p24gag (referred to as p24 in the text) HIV antigen through secretory IgA (SIgA) in nasal mucosae in mice. RESULTS: We show that SIgA interacts specifically with mucosal microfold cells present in the nasal-associated lymphoid tissue. p24-SIgA complexes are quickly taken up in the nasal cavity and selectively engulfed by mucosal dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin-positive dendritic cells. Nasal immunization with p24-SIgA elicits both a strong humoral and cellular immune response against p24 at the systemic and mucosal levels. This ensures effective protection against intranasal challenge with recombinant vaccinia virus encoding p24. CONCLUSION: This study represents the first example that underscores the remarkable potential of SIgA to serve as a carrier for a protein antigen in a mucosal vaccine approach targeting the nasal environment.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Les mécanismes qui régulent le processus de guérison de la peau lésée ne sont pas entièrement compris. Nous avons précédemment montré que les cellules dendritiques plasmocytoïdes (pDCs) sont normalement absentes de la peau saine mais infiltrent rapidement la peau humaine ainsi que celle des souris après une blessure cutanée. Après avoir infiltré la peau, ces pDCs sont capables de détecter les acides nucléiques par l'expression des récepteurs de type Toll 7 et 9 ce qui les active à produire de 1' interféron (IFN) de type I. Ce processus est primordial pour la re- épithélisation des blessures cutanées. Cependant, les mécanismes conduisant à l'infiltration et à 1'activation des pDCs restent inconnus. Dans notre projet, nous montrons que la chimiokine CxcllO est responsable de l'infiltration des pDCs. De façon importante, nous démontrons que les neutrophiles qui infiltrent également la peau lésée sont la source majeure de cette chimiokine. La déplétion des neutrophiles abolit d'ailleurs le recrutement des pDCs confirmant ainsi que CxcllO produit par les neutrophiles est responsable de l'infiltration des pDCs dans la peau endommagée. De façon intéressante, nous avons trouvé que CxcllO en plus de son activité chimiotactique, est capable de former des complexes avec l'ADN et d'activer ainsi les pDCs à produire de l'IFN de type I. De plus, nous avons observé que les neutrophiles qui infiltrent la peau forment des Neutrophil Extracellular Traps (NETs). Ces NETs sont constitués de filaments extracellulaires d'ADN recouverts par de nombreuses protéines principalement d'origine granulaire. D'une manière frappante, le blocage de la NETose ou l'utilisation de souris déficientes pour la formation de NETs altère le recrutement et l'activation des pDCs ainsi que la réponse inflammatoire qui en découle ainsi que le processus de re-epithélisation qui s'ensuit. En prenant en compte toutes ces données, nos résultats démontrent que suite à une blessure de la peau, les neutrophiles par la production de CxcllO contrôlent l'infiltration des pDCs dans la peau lésée et par la formation de NETs, promeuvent l'activation des pDCs. Notre étude fournit donc de nouvelles informations sur les mécanismes de guérison de la peau et ouvre de nouvelles perspectives thérapeutiques quant à la réparation tissulaire de la peau soit dans le but de l'amplifier ou de l'inhiber. -- The mechanisms that regulate healing of the injured skin are not well understood. We have previously shown that plasmacytoid dendritic cells (pDCs) are normally absent from the healthy skin, but rapidly infiltrate both murine and human skin upon injury. Upon skin infiltration, pDCs sense nucleic acids via TLR7/TLR9 and are activated to produce type I interferon (IFN), a process that is crucial for re-epithelialisation of skin wounds. However, the mechanisms that drive pDCs recruitment and activation in injured skin remain unclear. We show that CxcllO is responsible for pDCs infiltration. Importantly, we demonstrate that skin infiltrating neutrophils are the major source of this chemokine. Neutrophils depletion completely abrogated pDCs recruitment confirming that CxcllO- driven pDCs recruitment is controlled by neutrophils. Interestingly, CxcllO was also found to form complexes with DNA and to activate pDCs to produce Type I IFN in addition to its chemotactic activity. Moreover, we observed that infiltrating neutrophils release Neutrophils Extracellular Traps (NETs) composed of DNA filaments decorated with neutrophils-derived proteins. Strikingly, blocking NETosis or using mice deficient for NETs production impaired pDCs recruitment and activation as well as the subsequent inflammatory response and the re-epithelialisation process. Altogether, these data demonstrate that upon skin injury, neutrophils control pDCs infiltration into the injured skin by the release of CxcllO and via the production of NETs, they allow complex formation between CxcllO and NET-DNA leading to pDCs activation. Our findings provide new insights into the mechanisms of wound healing and open new avenues for potential therapeutic interventions to boost or inhibit wound repair in the skin.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

Lyme borreliosis is a tick-transmitted infection caused by the spirochete bacterium Borrelia burgdorferi sensu lato. The tick injects bacteria into host skin, where a first line defence, mainly the complement system, neutrophils, dendritic cells and macrophages are ready to attack foreign intruders. However, in the case of Lyme borreliosis, the original immune response in the skin is untypically mild among bacterial infections. A further untypical feature is the ability of B. burgdorferi to disseminate to distant organs, where, in some patients, symptoms appear after years after the original infection. This study aimed at uncovering some of the immune evasion mechanisms utilized by B. burgdorferi against the complement system, neutrophils and dendritic cells. B. burgdorferi was shown to inhibit chemotaxis of human neutrophils towards nformyl- methyl-leucyl-phenylalanine (fMLP). Outer surface protein B (OspB) of B. burgdorferi was shown to promote resistance to the attack of the complement system and neutrophil phagocytosis at low complement concentrations. B. burgdorferi was shown to inhibit migration of dendritic cells in vitro towards CCL19 and CCL21 and also in an in vivo model. This effect was shown to be due to the absence of CD38 on the borrelia-stimulated dendritic cell surface. A defect in p38 mitogen-activated-protein-kinase (p38) signaling was linked to defective CD38 expression. A defect in CD38 expression on B. burgdorferi-stimulated neutrophils was also observed. In this study, a number of novel immune evasion strategies utilized by B burgdorferi were chracterized. However, further studies are needed as other immune evasion mechanisms await to be uncovered.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

A dendritic cell (DC)-based vaccine strategy could reduce the risk of recurrence and improve the survival of breast cancer patients. However, while therapy-induced apoptosis of hepatocellular and colorectal carcinoma cells can enhance maturation and antigen presentation of DCs, whether this effect occurs in breast cancer is currently unknown. In the present study, we investigated the effect of doxorubicin (ADM)-induced apoptotic MCF-7 breast cancer cells on the activation of DCs. ADM-induced apoptotic MCF-7 cells could effectively induce immature DC (iDC) maturation. The mean fluorescence intensity (MFI) of DC maturity marker CD83 was 23.3 in the ADM-induced apoptotic MCF-7 cell group compared with 8.5 in the MCF-7 cell group. The MFI of DC co-stimulatory marker CD86 and HLA-DR were also increased after iDCs were treated with ADM-induced apoptotic MCF-7 cells. Furthermore, the proliferating autologous T-lymphocytes increased from 14.2 to 40.3% after incubated with DCs induced by apoptotic MCF-7 cells. The secretion of interferon-γ by these T-lymphocytes was also increased. In addition, cell-cell interaction between apoptotic MCF-7 cells and iDCs, but not soluble factors released by apoptotic MCF-7 cells, was crucial for the maturation of iDCs. These findings constitute a novel in vitro DC-based vaccine strategy for the treatment of breast cancer by ADM-induced apoptotic MCF-7 cells.

Relevância:

90.00% 90.00%

Publicador:

Resumo:

La greffe de sang de cordon est de plus en plus utilisée et a permis de traiter avec succès chez l’enfant des déficits immunitaires ainsi que des hémopathies malignes comme les leucémies. Malgré d’importants avantages tels que l’absence de risque pour le donneur ou la plus faible incidence de maladie du greffon contre l’hôte (GvHD), utiliser le sang de cordon comporte certains inconvénients. En effet, une reconstitution immunitaire retardée, des infections opportunistes en plus grand nombre et un risque de rechute sont des complications qui peuvent survenir et engendrer un risque pour le pronostic vital du patient. Par conséquent, de nouvelles stratégies d’immunothérapies doivent être envisagées. Dans le cadre de ce travail, nous nous sommes particulièrement intéressés aux cellules dendritiques plasmacytoides (pDC) dont les fonctions sont importantes pour l’initiation des réponses immunitaires innée et adaptative et particulièrement pour leur capacité à activer les cellules NK. Afin d’élucider le rôle et l’impact de ces cellules dans les greffes de sang de cordon, le nombre et la fonction des pDC et des NK a été suivi longitudinalement chez des patients ayant subi une greffe de sang de cordon comparativement à des patients transplantés avec de la moelle osseuse. Nous avons ainsi démontré que les pDC et les NK apparaissent précocement suite à une greffe de sang de cordon et que ces cellules sont fonctionnelles. Ces résultats mettent donc en lumière que ces cellules pourraient être de bons outils pour l’établissement d’une immunothérapie après greffe de sang de cordon. De plus, la caractérisation fonctionnelle des pDC du greffon de sang de cordon a permis de révéler une plus faible production d’IFN-α par les pDC, comparativement aux pDC de sang d’adulte. Cette différence pourrait jouer un rôle dans la plus faible incidence de GvHD après les greffes de sang de cordon. Dans le but de préciser les mécanismes moléculaires de régulation négative de la production d’IFN-α par les pDC de sang de cordon, nous avons étudié les protéines de la voie de signalisation TLR9-IRF7. L’expression similaire de l’ARN du TLR9, MyD88, IRAK1 et IRF7 contraste avec la plus faible expression des protéines correspondantes. De plus, l’expression des MicroARNs miR-146a et miR-155 est plus élevé dans les pDC de sang de cordon comparativement aux pDC de sang d’adultes. Ensemble, ces données pointent une régulation négative post-transcriptionnelle de la voie TLR9-IRF7 qui pourrait expliquer la plus faible production d’IFN-α des pDC du sang de cordon. L’ensemble des ces travaux suggère que les pDC pourraient représenter une cible de choix dans le développement de nouvelles approches thérapeutiques dans les greffes de sang de cordon.