Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
PLoS Pathog ; 19(7): e1011159, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37486946

RESUMO

NK cells are important mediators of innate immunity and play an essential role for host protection against infection, although their responses to bacteria are poorly understood. Recently NK cells were shown to display memory properties, as characterized by an epigenetic signature leading to a stronger secondary response. Although NK cell memory could be a promising mechanism to fight against infection, it has not been described upon bacterial infection. Using a mouse model, we reveal that NK cells develop specific and long-term memory following sub-lethal infection with the extracellular pathogen Streptococcus pneumoniae. Memory NK cells display intrinsic sensing and response to bacteria in vitro, in a manner that is enhanced post-bacterial infection. In addition, their transfer into naïve mice confers protection from lethal infection for at least 12 weeks. Interestingly, NK cells display enhanced cytotoxic molecule production upon secondary stimulation and their protective role is dependent on Perforin and independent of IFNγ. Thus, our study identifies a new role for NK cells during bacterial infection, opening the possibility to harness innate immune memory for therapeutic purposes.


Assuntos
Células Matadoras Naturais , Streptococcus pneumoniae , Imunidade Inata , Perforina
2.
Immunology ; 167(2): 197-211, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35758054

RESUMO

Type 2-high asthma is a chronic inflammatory disease of the airways which is increasingly prevalent in countries where helminth parasite infections are rare, and characterized by T helper 2 (Th2)-dependent accumulation of eosinophils in the lungs. Regulatory cytokines such as TGF-ß can restrain inflammatory reactions, dampen allergic Th2 responses, and control eosinophil activation. The murine helminth parasite Heligmosomoides polygyrus releases a TGF-ß mimic (Hp-TGM) that replicates the biological and functional properties of TGF-ß despite bearing no structural similarity to the mammalian protein. Here, we investigated if Hp-TGM could alleviate allergic airway inflammation in mice exposed to Alternaria alternata allergen, house dust mite (HDM) extract or alum-adjuvanted ovalbumin protein (OVA). Intranasal administration of Hp-TGM during Alternaria exposure sharply reduced airway and lung tissue eosinophilia along with bronchoalveolar lavage fluid IL-5 and lung IL-33 cytokine levels at 24 h. The protective effect of Hp-TGM on airway eosinophilia was also obtained in the longer T-cell mediated models of HDM or OVA sensitisation with significant inhibition of eotaxin-1, IL-4 and IL-13 responses depending on the model and time-point. Hp-TGM was also protective when administered parenterally either when given at the time of allergic sensitisation or during airway allergen challenge. This project has taken the first steps in identifying the role of Hp-TGM in allergic asthma and highlighted its ability to control lung inflammation and allergic pathology. Future research will investigate the mode of action of Hp-TGM against airway allergic eosinophilia, and further explore its potential to be developed as a biotherapeutic in allergic asthma.


Assuntos
Asma , Eosinofilia , Helmintos , Alérgenos/farmacologia , Animais , Asma/tratamento farmacológico , Asma/patologia , Líquido da Lavagem Broncoalveolar/química , Quimiocina CCL11 , Citocinas/metabolismo , Eosinofilia/tratamento farmacológico , Eosinofilia/patologia , Interleucina-13 , Interleucina-33 , Interleucina-4 , Interleucina-5 , Pulmão , Mamíferos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina , Fator de Crescimento Transformador beta
3.
Mucosal Immunol ; 15(6): 1243-1256, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35288645

RESUMO

Macrophage migration inhibitory factor (MIF) is a key innate immune mediator with chemokine- and cytokine-like properties in the inflammatory pathway. While its actions on macrophages are well-studied, its effects on other cell types are less understood. Here we report that MIF is required for expansion of intestinal tuft cells during infection with the helminth Nippostrongylus brasiliensis. MIF-deficient mice show defective innate responses following infection, lacking intestinal epithelial tuft cell hyperplasia or upregulation of goblet cell RELMß, and fail to expand eosinophil, type 2 innate lymphoid cell (ILC2) and macrophage (M2) populations. Similar effects were observed in MIF-sufficient wild-type mice given the MIF inhibitor 4-IPP. MIF had no direct effect on epithelial cells in organoid cultures, and MIF-deficient intestinal stem cells could generate tuft cells in vitro in the presence of type 2 cytokines. In vivo the lack of MIF could be fully compensated by administration of IL-25, restoring tuft cell differentiation and goblet cell expression of RELM-ß, demonstrating its requirement upstream of the ILC2-tuft cell circuit. Both ILC2s and macrophages expressed the MIF receptor CXCR4, indicating that MIF may act as an essential co-factor on both cell types to activate responses to IL-25 in helminth infection.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Infecções por Strongylida , Camundongos , Animais , Fatores Inibidores da Migração de Macrófagos/genética , Imunidade Inata , Linfócitos , Nippostrongylus
4.
Nat Microbiol ; 6(2): 257-269, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33349663

RESUMO

Streptococcus pneumoniae is a natural colonizer of the human respiratory tract and an opportunistic pathogen. Although epithelial cells are among the first to encounter pneumococci, the cellular processes and contribution of epithelial cells to the host response are poorly understood. Here, we show that a S. pneumoniae serotype 6B ST90 strain, which does not cause disease in a murine infection model, induces a unique NF-κB signature response distinct from an invasive-disease-causing isolate of serotype 4 (TIGR4). This signature is characterized by activation of p65 and requires a histone demethylase KDM6B. We show, molecularly, that the interaction of the 6B strain with epithelial cells leads to chromatin remodelling within the IL-11 promoter in a KDM6B-dependent manner, where KDM6B specifically demethylates histone H3 lysine 27 dimethyl. Remodelling of the IL-11 locus facilitates p65 access to three NF-κB sites that are otherwise inaccessible when stimulated by IL-1ß or TIGR4. Finally, we demonstrate through chemical inhibition of KDM6B with GSK-J4 inhibitor and through exogenous addition of IL-11 that the host responses to the 6B ST90 and TIGR4 strains can be interchanged both in vitro and in a murine model of infection in vivo. Our studies therefore reveal how a chromatin modifier governs cellular responses during infection.


Assuntos
Montagem e Desmontagem da Cromatina , Interações Hospedeiro-Patógeno/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/patogenicidade , Células A549 , Células Epiteliais Alveolares , Animais , Benzazepinas/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos , Células Epiteliais/microbiologia , Regulação da Expressão Gênica , Humanos , Interleucina-11/genética , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , NF-kappa B/farmacologia , Infecções Pneumocócicas/enzimologia , Infecções Pneumocócicas/genética , Regiões Promotoras Genéticas , Pirimidinas/farmacologia
5.
Cell Rep ; 30(12): 4016-4026.e4, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32209465

RESUMO

Pathogenic bacteria can alter host gene expression through post-translational modifications of histones. We show that a natural colonizer, Streptococcus pneumoniae, induces specific histone modifications, including robust dephosphorylation of histone H3 on serine 10 (H3S10), during infection of respiratory epithelial cells. The bacterial pore-forming toxin pneumolysin (PLY), along with the pyruvate oxidase SpxB responsible for H2O2 production, play important roles in the induction of this modification. The combined effects of PLY and H2O2 trigger host signaling that culminates in H3S10 dephosphorylation, which is mediated by the host cell phosphatase PP1. Strikingly, S. pneumoniae infection induces dephosphorylation and subsequent activation of PP1 catalytic activity. Colonization of PP1 catalytically deficient cells results in impaired intracellular S. pneumoniae survival and infection. Interestingly, PP1 activation and H3S10 dephosphorylation are not restricted to S. pneumoniae and appear to be general epigenomic mechanisms favoring intracellular survival of pathogenic bacteria.


Assuntos
Histonas/metabolismo , Interações Hospedeiro-Patógeno , Fosfoproteínas Fosfatases/metabolismo , Infecções Pneumocócicas/enzimologia , Streptococcus pneumoniae/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Inflamação/genética , Camundongos Endogâmicos C57BL , Fosforilação , Fosfosserina/metabolismo , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/genética , Estreptolisinas/metabolismo , Transcrição Gênica
6.
Cell Rep ; 29(12): 3933-3945.e3, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31851924

RESUMO

Natural killer (NK) cells are unique players in innate immunity and, as such, an attractive target for immunotherapy. NK cells display immune memory properties in certain models, but the long-term status of NK cells following systemic inflammation is unknown. Here we show that following LPS-induced endotoxemia in mice, NK cells acquire cell-intrinsic memory-like properties, showing increased production of IFNγ upon specific secondary stimulation. The NK cell memory response is detectable for at least 9 weeks and contributes to protection from E. coli infection upon adoptive transfer. Importantly, we reveal a mechanism essential for NK cell memory, whereby an H3K4me1-marked latent enhancer is uncovered at the ifng locus. Chemical inhibition of histone methyltransferase activity erases the enhancer and abolishes NK cell memory. Thus, NK cell memory develops after endotoxemia in a histone methylation-dependent manner, ensuring a heightened response to secondary stimulation.


Assuntos
Endotoxemia/imunologia , Infecções por Escherichia coli/imunologia , Histonas/metabolismo , Imunidade Inata/imunologia , Memória Imunológica/imunologia , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Animais , Endotoxemia/metabolismo , Endotoxemia/microbiologia , Endotoxemia/patologia , Elementos Facilitadores Genéticos , Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Histonas/genética , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Interferon gama/metabolismo , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/microbiologia , Células Matadoras Naturais/patologia , Masculino , Camundongos
7.
mBio ; 10(6)2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848284

RESUMO

Listeria monocytogenes is a pathogenic bacterium causing potentially fatal foodborne infections in humans and animals. While the mechanisms used by Listeria to manipulate its host have been thoroughly characterized, how the host controls bacterial virulence factors remains to be extensively deciphered. Here, we found that the secreted Listeria virulence protein InlC is monoubiquitinated by the host cell machinery on K224, restricting infection. We show that the ubiquitinated form of InlC interacts with the intracellular alarmin S100A9, resulting in its stabilization and in increased reactive oxygen species production by neutrophils in infected mice. Collectively, our results suggest that posttranslational modification of InlC exacerbates the host response upon Listeria infection.IMPORTANCE The pathogenic potential of Listeria monocytogenes relies on the production of an arsenal of virulence determinants that have been extensively characterized, including surface and secreted proteins of the internalin family. We have previously shown that the Listeria secreted internalin InlC interacts with IκB kinase α to interfere with the host immune response (E. Gouin, M. Adib-Conquy, D. Balestrino, M.-A. Nahori, et al., Proc Natl Acad Sci USA, 107:17333-17338, 2010, https://doi.org/10.1073/pnas.1007765107). In the present work, we report that InlC is monoubiquitinated on K224 upon infection of cells and provide evidence that ubiquitinated InlC interacts with and stabilizes the alarmin S100A9, which is a critical regulator of the immune response and inflammatory processes. Additionally, we show that ubiquitination of InlC causes an increase in reactive oxygen species production by neutrophils in mice and restricts Listeria infection. These findings are the first to identify a posttranscriptional modification of an internalin contributing to host defense.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Listeria/fisiologia , Listeriose/metabolismo , Listeriose/microbiologia , Calgranulina B/metabolismo , Suscetibilidade a Doenças , Células Epiteliais , Humanos , Ubiquitinação
8.
PLoS Pathog ; 15(5): e1007811, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31107928

RESUMO

Leptospira interrogans are pathogenic spirochetes responsible for leptospirosis, a worldwide reemerging zoonosis. Many Leptospira serovars have been described, and prophylaxis using inactivated bacteria provides only short-term serovar-specific protection. Therefore, alternative approaches to limit severe leptospirosis in humans and morbidity in cattle would be welcome. Innate immune cells, including macrophages, play a key role in fighting infection and pathogen clearance. Recently, it has been shown that functional reprograming of innate immune cells through the activation of pattern recognition receptors leads to enhanced nonspecific antimicrobial responses upon a subsequent microbial encounter. This mechanism is known as trained immunity or innate immune memory. We have previously shown that oral treatment with Lactobacillus plantarum confers a beneficial effect against acute leptospirosis. Here, using a macrophage depletion protocol and live imaging in mice, we established the role of peritoneal macrophages in limiting the initial dissemination of leptospires. We further showed that intraperitoneal priming of mice with CL429, a TLR2 and NOD2 agonist known to mimic the modulatory effect of Lactobacillus, alleviated acute leptospiral infection. The CL429 treatment was characterized as a training effect since i.) it was linked to peritoneal macrophages that produced ex vivo more pro-inflammatory cytokines and chemokines against 3 different pathogenic serovars of Leptospira, independently of the presence of B and T cells, ii.) it had systemic effects on splenic cells and bone marrow derived macrophages, and iii.) it was sustained for 3 months. Importantly, trained macrophages produced more nitric oxide, a potent antimicrobial compound, which has not been previously linked to trained immunity. Accordingly, trained macrophages better restrict leptospiral survival. Finally, we could use CL429 to train ex vivo human monocytes that produced more cytokines upon leptospiral stimulation. In conclusion, host-directed treatment using a TLR2/NOD2 agonist could be envisioned as a novel prophylactic strategy against acute leptospirosis.


Assuntos
Memória Imunológica/imunologia , Leptospira interrogans/imunologia , Leptospirose/prevenção & controle , Macrófagos Peritoneais/imunologia , Proteína Adaptadora de Sinalização NOD2/agonistas , Bibliotecas de Moléculas Pequenas/farmacologia , Receptor 2 Toll-Like/agonistas , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Humanos , Memória Imunológica/efeitos dos fármacos , Leptospirose/imunologia , Leptospirose/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
9.
Nat Commun ; 9(1): 2636, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29980664

RESUMO

Invasive aspergillosis (IA) is a severe infection that can occur in severely immunocompromised patients. Efficient immune recognition of Aspergillus is crucial to protect against infection, and previous studies suggested a role for NOD2 in this process. However, thorough investigation of the impact of NOD2 on susceptibility to aspergillosis is lacking. Common genetic variations in NOD2 has been associated with Crohn's disease and here we investigated the influence of these  genetic variations on the anti-Aspergillus host response. A NOD2 polymorphism reduced the risk of IA after hematopoietic stem-cell transplantation. Mechanistically, absence of NOD2 in monocytes and macrophages increases phagocytosis leading to enhanced fungal killing, conversely, NOD2 activation reduces the antifungal potential of these cells. Crucially, Nod2 deficiency results in resistance to Aspergillus infection in an in vivo model of pulmonary aspergillosis. Collectively, our data demonstrate that genetic deficiency of NOD2 plays a protective role during Aspergillus infection.


Assuntos
Aspergilose/genética , Resistência à Doença , Proteína Adaptadora de Sinalização NOD2/deficiência , Proteína Adaptadora de Sinalização NOD2/genética , Animais , Aspergilose/etiologia , Aspergilose/microbiologia , Aspergillus , Citocinas/metabolismo , Feminino , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Lectinas Tipo C , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Seios Paranasais/patologia , Fagocitose , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco
10.
Microbes Infect ; 20(3): 156-165, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29287986

RESUMO

Pathogens or endogenous molecules can reprogram innate immunity. This process can take the form of priming or tolerance depending on the activating signal, and favors enhanced resistance to infection and other insults, by modulating inflammation. Similarly to their organ-specific properties, reprogramming of macrophages and NK cells, is also compartmentalized.


Assuntos
Endotoxinas/imunologia , Imunidade Inata/imunologia , Memória Imunológica/imunologia , Especificidade de Órgãos/imunologia , Animais , Citocinas/imunologia , Citocinas/metabolismo , Endotoxinas/metabolismo , Humanos , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Transdução de Sinais/imunologia
11.
Virulence ; 8(8): 1744-1752, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-28594271

RESUMO

Cyclosporin A (CsA) is widely used as an immunosuppressive agent for organ transplant recipients. CsA inhibits calcineurin, which is highly conserved in mammals and fungi, and thus affects both types of organism. In mammals, the immunosuppressive effect of CsA is via hampering T cell activation. In fungi, the growth inhibitory effect of CsA is via interference with hyphal growth. The aim of this study was to determine whether CsA renders mice susceptible to invasive pulmonary aspergillosis (IPA) and whether it can protect immunosuppressed mice from infection. We therefore examined both the antifungal and the immunosuppressive activity of CsA in immunosuppressed and in immunocompetent mice infected with Aspergillus fumigatus to model IPA. We found that daily injections of CsA could not produce an antifungal effect sufficient to rescue immunosuppressed mice from lethal IPA. However, a 100% survival rate was obtained in non-immunosuppressed mice receiving daily CsA, indicating that CsA did not render the mice vulnerable to IPA. The lymphocyte subset was significantly suppressed by CsA, while the myeloid subset was not. Therefore, we speculate that CsA does not impair the host defense against IPA since the myeloid cells are preserved.


Assuntos
Aspergillus fumigatus/fisiologia , Ciclosporina/administração & dosagem , Imunossupressores/administração & dosagem , Aspergilose Pulmonar Invasiva/tratamento farmacológico , Células Mieloides/imunologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Humanos , Hospedeiro Imunocomprometido/efeitos dos fármacos , Aspergilose Pulmonar Invasiva/imunologia , Aspergilose Pulmonar Invasiva/microbiologia , Aspergilose Pulmonar Invasiva/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/citologia
12.
Front Immunol ; 8: 1777, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29326692

RESUMO

One of the major life-threatening infections for which severely immunocompromised patients are at risk is invasive aspergillosis (IA). Despite the current treatment options, the increasing antifungal resistance and poor outcome highlight the need for novel therapeutic strategies to improve outcome of patients with IA. In the current study, we investigated whether and how the intracellular pattern recognition receptor NOD1 is involved in host defense against Aspergillus fumigatus. When exploring the role of NOD1 in an experimental mouse model, we found that Nod1-/- mice were protected against IA and demonstrated reduced fungal outgrowth in the lungs. We found that macrophages derived from bone marrow of Nod1-/- mice were more efficiently inducing reactive oxygen species and cytokines in response to Aspergillus. Most strikingly, these cells were highly potent in killing A. fumigatus compared with wild-type cells. In line, human macrophages in which NOD1 was silenced demonstrated augmented Aspergillus killing and NOD1 stimulation decreased fungal killing. The differentially altered killing capacity of NOD1 silencing versus NOD1 activation was associated with alterations in dectin-1 expression, with activation of NOD1 reducing dectin-1 expression. Furthermore, we were able to demonstrate that Nod1-/- mice have elevated dectin-1 expression in the lung and bone marrow, and silencing of NOD1 gene expression in human macrophages increases dectin-1 expression. The enhanced dectin-1 expression may be the mechanism of enhanced fungal killing of Nod1-/- cells and human cells in which NOD1 was silenced, since blockade of dectin-1 reversed the augmented killing in these cells. Collectively, our data demonstrate that NOD1 receptor plays an inhibitory role in the host defense against Aspergillus. This provides a rationale to develop novel immunotherapeutic strategies for treatment of aspergillosis that target the NOD1 receptor, to enhance the efficiency of host immune cells to clear the infection by increasing fungal killing and cytokine responses.

13.
J Immunol ; 197(6): 2444-54, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27521338

RESUMO

Systemic inflammatory response syndrome is a whole-body reaction to a triggering insult that often results in life-threatening illness. Contributing to the development of this inflammatory cascade are numerous cellular partners, among which NK cells were shown to play a key role. Accumulating evidence points to organ-specific properties of systemic inflammation and NK cells. However, little is known about compartment-specific activation of NK cells during systemic inflammatory response syndrome or the relative contribution of NK cell-intrinsic properties and microenvironmental cues. In this study, we undertook a sequential characterization of NK responses in the spleen, lungs, bone marrow, peritoneum, and blood using a mouse model of endotoxemia. We report that, despite similar systemic dynamics of NK cell responses, expression of activation markers (CD69 and CD25) and effector molecules (IFN-γ, granzyme B, and IL-10) display organ-specific thresholds of maximum activation. Using adoptive transfers of spleen and lung NK cells, we found that these cells have the capacity to quickly adapt to a new environment and adjust their response levels to that of resident NK cells. This functional adaptation occurs without significant alterations in phenotype and independently of subpopulation-specific trafficking. Thus, using a dynamic in vivo-transfer system, to our knowledge our study is the first to report the compartmentalization of NK cells responses during systemic inflammation and to show that NK cell-intrinsic properties and microenvironmental cues are involved in this process, in a sequential manner.


Assuntos
Microambiente Celular , Inflamação/imunologia , Células Matadoras Naturais/imunologia , Transferência Adotiva , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/genética , Antígenos de Diferenciação de Linfócitos T/imunologia , Células da Medula Óssea/imunologia , Citotoxicidade Imunológica , Granzimas/imunologia , Inflamação/sangue , Inflamação/fisiopatologia , Interferon gama/imunologia , Interleucina-10/imunologia , Subunidade alfa de Receptor de Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Células Matadoras Naturais/fisiologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Leucócitos/imunologia , Pulmão/citologia , Pulmão/imunologia , Camundongos , Peritônio/citologia , Peritônio/imunologia , Baço/citologia , Baço/imunologia
14.
Sci Rep ; 6: 26490, 2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27215684

RESUMO

Hypoxia as a result of pulmonary tissue damage due to unresolved inflammation during invasive pulmonary aspergillosis (IPA) is associated with a poor outcome. Aspergillus fumigatus can exploit the hypoxic microenvironment in the lung, but the inflammatory response required for fungal clearance can become severely disregulated as a result of hypoxia. Since severe inflammation can be detrimental to the host, we investigated whether targeting the interleukin IL-1 pathway could reduce inflammation and tissue hypoxia, improving the outcome of IPA. The interplay between hypoxia and inflammation was investigated by in vivo imaging of hypoxia and measurement of cytokines in the lungs in a model of corticosteroid immunocompromised and in Cxcr2 deficient mice. Severe hypoxia was observed following Aspergillus infection in both models and correlated with development of pulmonary inflammation and expression of hypoxia specific transcripts. Treatment with IL-1 receptor antagonist reduced hypoxia and slightly, but significantly reduced mortality in immunosuppressed mice, but was unable to reduce hypoxia in Cxcr2(-/-) mice. Our data provides evidence that the inflammatory response during invasive pulmonary aspergillosis, and in particular the IL-1 axis, drives the development of hypoxia. Targeting the inflammatory IL-1 response could be used as a potential immunomodulatory therapy to improve the outcome of aspergillosis.


Assuntos
Corticosteroides/efeitos adversos , Citocinas/metabolismo , Proteína Antagonista do Receptor de Interleucina 1/administração & dosagem , Aspergilose Pulmonar Invasiva/tratamento farmacológico , Receptores de Interleucina-8B/deficiência , Animais , Anidrase Carbônica IX/metabolismo , Hipóxia Celular/efeitos dos fármacos , Modelos Animais de Doenças , Hospedeiro Imunocomprometido , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Aspergilose Pulmonar Invasiva/etiologia , Aspergilose Pulmonar Invasiva/imunologia , Camundongos , Receptores de Interleucina-1/antagonistas & inibidores , Receptores de Interleucina-8B/genética , Resultado do Tratamento
15.
PLoS Negl Trop Dis ; 10(5): e0004716, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27182703

RESUMO

A critical role for intracellular TLR9 has been described in recognition and host resistance to Leishmania parasites. As TLR9 requires endolysosomal proteolytic cleavage to achieve signaling functionality, we investigated the contribution of different proteases like asparagine endopeptidase (AEP) or cysteine protease cathepsins B (CatB), L (CatL) and S (CatS) to host resistance during Leishmania major (L. major) infection in C57BL/6 (WT) mice and whether they would impact on TLR9 signaling. Unlike TLR9-/-, which are more susceptible to infection, AEP-/-, CatL-/- and CatS-/- mice are as resistant to L. major infection as WT mice, suggesting that these proteases are not individually involved in TLR9 processing. Interestingly, we observed that CatB-/- mice resolve L. major lesions significantly faster than WT mice, however we did not find evidence for an involvement of CatB on either TLR9-dependent or independent cytokine responses of dendritic cells and macrophages or in the innate immune response to L. major infection. We also found no difference in antigen presenting capacity. We observed a more precocious development of T helper 1 responses accompanied by a faster decline of inflammation, resulting in resolution of footpad inflammation, reduced IFNγ levels and decreased parasite burden. Adoptive transfer experiments into alymphoid RAG2-/-γc-/- mice allowed us to identify CD3+ T cells as responsible for the immune advantage of CatB-/- mice towards L. major. In vitro data confirmed the T cell intrinsic differences between CatB-/- mice and WT. Our study brings forth a yet unappreciated role for CatB in regulating T cell responses during L. major infection.


Assuntos
Catepsina B/deficiência , Catepsina B/metabolismo , Leishmania major , Leishmaniose Cutânea/imunologia , Subpopulações de Linfócitos T/imunologia , Receptor Toll-Like 9/metabolismo , Transferência Adotiva , Animais , Apresentação de Antígeno , Complexo CD3/análise , Complexo CD3/imunologia , Catepsina B/genética , Catepsina L/deficiência , Catepsina L/genética , Catepsinas/deficiência , Catepsinas/genética , Células Dendríticas/imunologia , Endopeptidases/deficiência , , Inflamação/imunologia , Interferon gama/biossíntese , Leishmania major/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Carga Parasitária , Transdução de Sinais , Células Th1/imunologia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia
16.
PLoS One ; 11(4): e0153829, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27078879

RESUMO

Phagocytes restrict the germination of Aspergillus fumigatus conidia and prevent the establishment of invasive pulmonary aspergillosis in immunecompetent mice. Here we report that immunecompetent mice recovering from a primary A. fumigatus challenge are protected against a secondary lethal challenge. Using RAGγc knock-out mice we show that this protection is independent of T, B and NK cells. In protected mice, lung phagocytes are recruited more rapidly and are more efficient in conidial phagocytosis and killing. Protection was also associated with an enhanced expression of CXCR2 and Dectin-1 on bone marrow phagocytes. We also show that protective lung cytokine and chemokine responses are induced more rapidly and with enhanced dynamics in protected mice. Our findings support the hypothesis that following a first encounter with a non-lethal dose of A. fumigatus conidia, the innate immune system is primed and can mediate protection against a secondary lethal infection.


Assuntos
Aspergilose/imunologia , Aspergillus fumigatus/imunologia , Fagócitos/imunologia , Esporos Fúngicos/imunologia , Animais , Aspergilose/microbiologia , Aspergillus fumigatus/fisiologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/microbiologia , Quimiocinas/imunologia , Quimiocinas/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Resistência à Doença/imunologia , Citometria de Fluxo , Interações Hospedeiro-Patógeno/imunologia , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/microbiologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagócitos/metabolismo , Fagócitos/microbiologia , Receptores de Interleucina-8B/imunologia , Receptores de Interleucina-8B/metabolismo , Esporos Fúngicos/fisiologia
17.
Future Microbiol ; 11(2): 293-314, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26849633

RESUMO

Severe sepsis remains a worldwide threat, not only in industrialized countries, due to their aging population, but also in developing countries where there still are numerous cases of neonatal and puerperal sepsis. Tools for early diagnosis, a prerequisite for rapid and appropriate antibiotic therapy, are still required. In this review, we highlight some recent developments in our understanding of the associated systemic inflammatory response that help deciphering pathophysiology (e.g., epigenetic, miRNA, regulatory loops, compartmentalization, apoptosis and synergy) and discuss some of the consequences of sepsis (e.g., immune status, neurological and muscular alterations). We also emphasize the challenge to better define animal models and discuss past failures in clinical investigations in order to define new efficient therapies.


Assuntos
Pesquisa Biomédica , Sepse , Animais , Biomarcadores , Países Desenvolvidos , Países em Desenvolvimento , Modelos Animais de Doenças , Humanos , Sistema Imunitário/microbiologia , Inflamação , Camundongos , Sepse/diagnóstico , Sepse/fisiopatologia , Sepse/terapia
18.
Hum Vaccin Immunother ; 12(4): 1009-26, 2016 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-26618392

RESUMO

Millions of seasonal and pandemic influenza vaccine doses containing oil-in-water emulsion adjuvant have been administered in order to enhance and broaden immune responses and to facilitate antigen sparing. Despite the enactment of a Global Action Plan for Influenza Vaccines and a multi-fold increase in production capabilities over the past 10 years, worldwide capacity for pandemic influenza vaccine production is still limited. In developing countries, where routine influenza vaccination is not fully established, additional measures are needed to ensure adequate supply of pandemic influenza vaccines without dependence on the shipment of aid from other, potentially impacted first-world countries. Adaptation of influenza vaccine and adjuvant technologies by developing country influenza vaccine manufacturers may enable antigen sparing and corresponding increases in global influenza vaccine coverage capacity. Following on previously described work involving the technology transfer of oil-in-water emulsion adjuvant manufacturing to a Romanian vaccine manufacturing institute, we herein describe the preclinical evaluation of inactivated split virion H5N1 influenza vaccine with emulsion adjuvant, including immunogenicity, protection from virus challenge, antigen sparing capacity, and safety. In parallel with the evaluation of the bioactivity of the tech-transferred adjuvant, we also describe the impact of concurrent antigen manufacturing optimization activities. Depending on the vaccine antigen source and manufacturing process, inclusion of adjuvant was shown to enhance and broaden functional antibody titers in mouse and rabbit models, promote protection from homologous virus challenge in ferrets, and facilitate antigen sparing. Besides scientific findings, the operational lessons learned are delineated in order to facilitate adaptation of adjuvant technologies by other developing country institutes to enhance global pandemic influenza preparedness.


Assuntos
Adjuvantes Imunológicos , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza , Transferência de Tecnologia , Avaliação Pré-Clínica de Medicamentos , Emulsões/química , Humanos , Virus da Influenza A Subtipo H5N1/fisiologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Óleos , Pandemias/prevenção & controle , Romênia , Vírion/fisiologia , Inativação de Vírus
19.
PLoS One ; 9(4): e95378, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24740379

RESUMO

Proteasomes play a fundamental role in intracellular protein degradation and therewith regulate a variety of cellular processes. Exposure of cells to (pro)inflammatory cytokines upregulates the expression of three inducible catalytic proteasome subunits, the immunosubunits, which incorporate into newly assembled proteasome complexes and alter the catalytic activity of the cellular proteasome population. Single gene-deficient mice lacking one of the three immunosubunits are resistant to dextran sulfate sodium (DSS)-induced colitis development and, likewise, inhibition of one single immunosubunit protects mice against the development of DSS-induced colitis. The observed diminished disease susceptibility has been attributed to altered cytokine production and CD4+ T-cell differentiation in the absence of immunosubunits. To further test whether the catalytic activity conferred by immunosubunits plays an essential role in CD4+ T-cell function and to distinguish between the role of immunosubunits in effector T-cells versus inflamed tissue, we used a T-cell transfer-induced colitis model. Naïve wt or immunosubunit-deficient CD4+ T-cells were adoptively transferred into RAG1-/- and immunosubunit-deficient RAG1-/- mice and colitis development was determined six weeks later. While immunosubunit expression in recipient mice had no effect on colitis development, transferred immunosubunit-deficient T- cells were more potent in inducing colitis and produced more proinflammatory IL17 than wt T-cells. Taken together, our data show that modifications in proteasome-mediated proteolysis in T-cells, conferred by lack of immunosubunit incorporation, do not attenuate but enhance CD4+ T-cell-induced inflammation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Colite/imunologia , Expressão Gênica/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Subunidades Proteicas/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/transplante , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Feminino , Deleção de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-17/biossíntese , Interleucina-17/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/genética , Subunidades Proteicas/genética
20.
J Am Assoc Lab Anim Sci ; 51(4): 469-74, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23043813

RESUMO

We used whole-body plethysmography to investigate the effect of restraint, ear marking, tail vein and retroorbital blood sampling, and tail clipping on respiration in Balb/c × TCR-HA +/- F1 hybrid mice (F1h). Baseline values of breathing parameters were determined. During the experiment, mice experienced a procedure and then plethysmographic recordings were obtained immediately and at 4, 24, and 48 h afterward. Baseline breathing parameters showed significant differences between sexes. Restraint affected minute volume differently than did handling in male mice and to a lesser extent in female mice. Ear marking significantly changed minute volume compared with handling but not restraint in male mice and in the opposite manner in female mice. Tail vein blood sampling changed minute volume in a significant manner compared with restraint but not compared with handling in both sexes. Retroorbital blood sampling significantly changed minute volume compared with values for both handling and restraint in male mice but only compared with handling in female mice. Tail clipping modified minute volume significantly compared with handling in male mice and compared with restraint in both sexes. Analysis of data showed that routine procedures affect minute volume in mice depending on invasiveness of maneuver and in a sex-biased manner for as long as 24 h after the procedure. Our experiment shows that procedures performed on laboratory mice can change respiratory parameters and can be investigated by plethysmography.


Assuntos
Camundongos Endogâmicos BALB C/fisiologia , Pletismografia Total/veterinária , Respiração , Manejo de Espécimes/veterinária , Animais , Coleta de Amostras Sanguíneas/efeitos adversos , Coleta de Amostras Sanguíneas/veterinária , DNA/análise , Feminino , Masculino , Camundongos , Camundongos Transgênicos/classificação , Camundongos Transgênicos/genética , Restrição Física/fisiologia , Restrição Física/veterinária , Manejo de Espécimes/efeitos adversos , Cauda/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...