Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
Add more filters

Publication year range
1.
Parasite Immunol ; 40(2)2018 02.
Article in English | MEDLINE | ID: mdl-29117626

ABSTRACT

Cerebral malaria (CM) is one complication of Plasmodium parasite infection that can lead to strong inflammatory immune responses in the central nervous system (CNS), accompanied by lung inflammation and anaemia. Here, we focus on the role of the innate immune response in experimental cerebral malaria (ECM) caused by blood-stage murine Plasmodium berghei ANKA infection. While T cells are important for ECM pathogenesis, the role of innate lymphoid cells (ILCs) is only emerging. The role of ILCs and non-lymphoid cells, such as neutrophils and platelets, contributing to the host immune response and leading to ECM and human cerebral malaria (HCM) is reviewed.


Subject(s)
Cytokines/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Animals , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Protein Kinase C-theta/immunology
2.
Parasite Immunol ; 37(9): 479-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26079128

ABSTRACT

African trypanosomes infect humans and animals throughout the African continent. These parasites maintain chronic infections by various immune evasion strategies. While antigenic variation of their surface coat is the most studied strategy linked to evading the host humoral response, African trypanosomes also induce impaired B-cell lymphopoiesis, the destruction of the splenic B-cell compartment and abrogation of protective memory responses. Here we investigate the mechanism of follicular B-cell destruction. We show that during infection follicular B cells undergo apoptosis, correlating to enhanced Fas death receptor surface expression. Investigation of various type 1 cytokine knockout mice indicates a crucial role of IFN-γ in the early onset of FoB cell destruction. Indeed, both IFN-γ(-/-) and IFN-γR(-/-) mice are protected from trypanosomosis-associated FoB cell depletion, exhibiting an inhibition of B-cell apoptosis as well as a reduced activation of FoB cells during the first week post-infection. The data presented herein offer new insights into B-cell dysfunctioning during experimental African trypanosome infections.


Subject(s)
B-Lymphocyte Subsets/immunology , Interferon-gamma/immunology , Trypanosomiasis, African/immunology , Animals , Antigenic Variation , Female , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interferon/genetics , Spleen/cytology , Spleen/immunology , Interferon gamma Receptor
3.
J Antimicrob Chemother ; 66(5): 1100-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21393169

ABSTRACT

OBJECTIVES: To investigate the in vivo efficacy in a murine pulmonary infection model of panobacumab (KBPA101), a human IgM monoclonal antibody directed against the O-polysaccharide moiety of Pseudomonas aeruginosa serotype O11, and to describe the anti-inflammatory effects in the lung as a consequence of the treatment. METHODS: We established an experimental murine model of acute pneumonia by intranasal administration of P. aeruginosa serotype O11. Mice were treated, after infection, with a single intravenous injection of panobacumab and panobacumab lung bioavailability was assessed. Inflammatory parameters such as pro-inflammatory cytokine production and neutrophil recruitment in broncho-alveolar lavage fluid (BALF) were measured and bacterial load in the lung was analysed. RESULTS: Panobacumab plays a significant role in addition to the host innate immune response, leading to improved control of pulmonary infection. The IgM antibody is able to reach the broncho-alveolar space and reduce the pulmonary bacterial load as well as lung inflammation in a dose-dependent manner. Furthermore, panobacumab treatment leads to enhanced neutrophil recruitment in BALF while reducing the host-derived production of pro-inflammatory mediators and lung injury. CONCLUSIONS: These data provide evidence that panobacumab, an IgM-based immunotherapeutic, is highly efficacious in controlling acute lung infection by enhancing the natural innate immune response.


Subject(s)
Antibodies, Bacterial/administration & dosage , Antibodies, Monoclonal/administration & dosage , Immunoglobulin M/administration & dosage , Immunotherapy/methods , Pneumonia, Bacterial/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Disease Models, Animal , Female , Injections, Intravenous , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/pathogenicity , Treatment Outcome
4.
Allergy ; 66(8): 1047-57, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21443539

ABSTRACT

BACKGROUND: Inflammasome activation with the production of IL-1ß received substantial attention recently in inflammatory diseases. However, the role of inflammasome in the pathogenesis of asthma is not clear. Using an adjuvant-free model of allergic lung inflammation induced by ovalbumin (OVA), we investigated the role of NLRP3 inflammasome and related it to IL-1R1 signaling pathway. METHODS: Allergic lung inflammation induced by OVA was evaluated in vivo in mice deficient in NLRP3 inflammasome, IL-1R1, IL-1ß or IL-1α. Eosinophil recruitment, Th2 cytokine, and chemokine levels were determined in bronchoalveolar lavage fluid, lung homogenates, and mediastinal lymph node cells ex vivo. RESULTS: Allergic airway inflammation depends on NLRP3 inflammasome activation. Dendritic cell recruitment into lymph nodes, Th2 lymphocyte activation in the lung and secretion of Th2 cytokines and chemokines are reduced in the absence of NLRP3. Absence of NLRP3 and IL-1ß is associated with reduced expression of other proinflammatory cytokines such as IL-5, IL-13, IL-33, and thymic stromal lymphopoietin. Furthermore, the critical role of IL-1R1 signaling in allergic inflammation is confirmed in IL-1R1-, IL-1ß-, and IL-1α-deficient mice. CONCLUSION: NLRP3 inflammasome activation leading to IL-1 production is critical for the induction of a Th2 inflammatory allergic response.


Subject(s)
Asthma/etiology , Carrier Proteins/immunology , Inflammasomes/immunology , Adjuvants, Immunologic , Aluminum , Animals , Asthma/pathology , Interleukin-1/biosynthesis , Interleukin-1beta , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Ovalbumin , Pneumonia , Receptors, Interleukin-1/metabolism , Signal Transduction/immunology , Th2 Cells/immunology
5.
J Exp Med ; 185(2): 329-39, 1997 Jan 20.
Article in English | MEDLINE | ID: mdl-9016881

ABSTRACT

The transgenic (tg) expression of interleukin (IL)-4 under the control of a major histocompatibility complex (MHC) class I promoter leads to B cell hyperactivity in mice, characterized by increased B cell surface MHC class II and CD23 expression, elevated responsiveness of the B cells to polyclonal ex vivo stimulation, and increased immunoglobulin (Ig)G1 and IgE serum levels. Tg mice develop anemia, glomerulonephritis with complement and immune deposition in the glomeruli, and show increased production of autoantibodies. Treatment of IL-4 tg mice with anti-IL-4 neutralizing antibodies protected the mice from disease development, showing that IL-4 was responsible for the observed disorders. Deletion of superantigen responsive autoreactive T cells in the IL-4 tg mice was normal and treatment of mutant mice with deleting anti-CD4 antibodies failed to ablate the onset of autoimmune-like disease, suggesting that CD4+ T cells were not the primary cause of the disorders. Furthermore, the deletion of B cells reacting against MHC class I molecules was also normal in the IL-4 tg mice. Therefore the most likely explanation for the increased production of autoantibodies and the autoimmunelike disorders is that IL-4 acts directly on autoreactive B cells by expanding them in a polyclonal manner. Taken together our results show that inappropriate multi-organ expression of IL-4 in vivo leads to autoimmune-type disease in mice.


Subject(s)
Autoimmune Diseases/genetics , Interleukin-4/genetics , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , Autoimmune Diseases/immunology , B-Lymphocytes/immunology , Female , Interleukin-4/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , Superantigens/immunology
6.
J Exp Med ; 187(4): 469-77, 1998 Feb 16.
Article in English | MEDLINE | ID: mdl-9463397

ABSTRACT

During immune responses the initial activation of B cells takes place in T cell zones of periarteriolar lymphoid sheaths (PALS) of the splenic white pulp. After initial activation, B cells migrate into the primary follicles and, in association with follicular dendritic cells (FDCs), undergo clonal expansion and differentiation giving rise to germinal centers (GCs). Peanut agglutinin binding (PNA+) cells of the GC differentiate further into memory or plasma cells. Here we report that in tumor necrosis factor receptor 1-deficient mice (TNFR1(-/-)), the location of B cells was altered and that plasma cells were abnormally distributed in the splenic PALS. In contrast to lymphotoxin alpha-deficient mice (LTalpha-/-), bone marrow or fetal liver transplantation did not correct the abnormal organization of the spleen, location of B cells, the lack of an FDC network, nor the antibody response in TNFR1(-/-) mice. These results argue for a crucial role of TNFR1 expression on nonhematopoietic cells for the maintenance of the splenic architecture and proper B cell location. In addition, the lack in development of an FDC network after adoptive transfer suggests that either FDCs are not of bone marrow origin or that they depend on signals from nonhematopoietic cells for maturation.


Subject(s)
Antigens, CD/physiology , B-Lymphocytes/metabolism , Receptors, Tumor Necrosis Factor/physiology , Spleen/cytology , Animals , Antigens, CD/biosynthesis , Dendritic Cells/metabolism , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Rats , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor, Type I
7.
J Exp Med ; 183(5): 2367-72, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8642347

ABSTRACT

Using mice double deficient for tumor necrosis factor (TNF) and lymphotoxin alpha (LT alpha), we demonstrated that TNF and/or LT alpha are necessary for development of a normal splenic microarchitecture and for isotype switch after immunization with sheep red blood cells (SRBC). In the present study, we extended these observations by determining which TNF receptor (TNFR) is involved in morphological and functional differentiation of the spleen. Spleen morphology and antibody response were investigated in wild-type, TNFR1-/-, TNFR2-/- and TNF/LT alpha-/- mice immunized with SRBC. TNF/LT alpha-/- mice, which have a complete disruption of the TNF/LT alpha signaling system including the LT beta-receptor pathway, displayed an abnormal microarchitecture, and isotype switch did not take place. TNFR1-/- and TNFR2-/- mice displayed a normal spleen microarchitecture and mounted an IgM and IgG antibody response to SRBC. However, the IgG production in TNFR1-/- mice was minimal, with citers leveling off 6 d after immunization. In this strain, immunofluorescence revealed a lack of follicular dendritic cells (FDC) network, detected with FDC-M1 as well as anti-CR1, and a lack of germinal centers, detected with peanut agglutinin. In conclusion, whereas normal splenic microarchitecture and isotype switch might require the LT beta receptor, differentiation of FDC network, development of germinal centers, and full IgG response depend on signaling via TNFR1.


Subject(s)
Antibody Formation , Dendritic Cells/cytology , Dendritic Cells/immunology , Lymphotoxin-alpha/genetics , Receptors, Tumor Necrosis Factor/physiology , Animals , Cell Differentiation , Crosses, Genetic , Fluorescent Antibody Technique , Immunoglobulin Class Switching , Immunoglobulin G/blood , Immunoglobulin G/classification , Immunoglobulin M/blood , Lymphotoxin-alpha/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Signal Transduction , Species Specificity , Spleen/cytology , Spleen/immunology
8.
J Exp Med ; 179(5): 1437-44, 1994 May 01.
Article in English | MEDLINE | ID: mdl-8163930

ABSTRACT

Antibody neutralization studies have established interferon gamma (IFN-gamma) as a critical mediator of endotoxic shock. The advent of IFN-gamma receptor negative (IFN gamma R-/-) mutant mice has enabled a more direct assessment of the role of IFN-gamma in endotoxin (lipopolysaccharide [LPS]-induced shock. We report that IFN gamma R-/- mice have an increased resistance to LPS-induced toxicity, this resistance manifesting well before the synthesis and release of LPS-induced IFN-gamma. LPS-induced lymphopenia, thrombocytopenia, and weight loss seen in wild-type mice were attenuated in IFN gamma R-/- mice. IFN gamma R-/- mice tolerated 100-1,000 times more LPS than the minimum lethal dose for wild-type mice in a D-galactosamine (D-GalN)/LPS model. Serum tumor necrosis factor (TNF) levels were 10-fold reduced in mutant mice given LPS or LPS/D-GalN. Bone marrow and splenic macrophages from IFN gamma R-/- mice had a four- to sixfold decreased LPS-binding capacity which correlated with similar reduction in CD14. Serum from mutant mice reduced macrophage LPS binding by a further 50%, although LPS binding protein was only 10% reduced. The expression of TNF receptor I (p55) and II (p75) was identical between wild-type and mutant mice. Thus, depressed TNF synthesis, diminished expression of CD14, and low plasma LPS-binding capacity, in addition to blocked IFN-gamma signaling in the mutant mice, likely to combine to manifest in the resistant phenotype of IFN gamma R-/- mice to endotoxin.


Subject(s)
Receptors, Interferon/deficiency , Shock, Septic/immunology , Animals , Body Weight , Endotoxins , Immunity, Innate , Lipopolysaccharides/toxicity , Mice , Molecular Sequence Data , Receptors, Interferon/immunology , Tumor Necrosis Factor-alpha/analysis , Interferon gamma Receptor
9.
J Exp Med ; 181(5): 1893-8, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7722464

ABSTRACT

Interleukin (IL)-12 synergizes with other cytokines to stimulate the proliferation and differentiation of early hematopoietic progenitors in vitro. However, in vivo administration of IL-12 decreases peripheral blood counts and bone marrow hematopoiesis. Here, we used interferon (IFN) gamma receptor-deficient (IFN gamma R-/-) mice to investigate whether the in vivo inhibition of hematopoiesis by IL-12 is indirectly mediated by IL-12-induced IFN-gamma. IL-12 administered for 4 d (1 microgram/mouse per day) resulted in lower peripheral blood counts and a 2-fold decrease in bone marrow cellularity in wild-type mice, but not in IFN gamma R-/- mice. Bone marrow hematopoietic progenitors were decreased after IL-12 treatment in wild-type mice, but rather increased in IFN gamma R-/- mice. Splenic cellularity was 2.3-fold higher after IL-12 administration in wild-type mice, largely due to natural killer (NK) cell and macrophage infiltration together with some extramedullary hematopoiesis. In IFN gamma R-/- mice, spleen cellularity was less increased, there were fewer infiltrating NK cells, but a strong extramedullary hematopoiesis. Thus, alterations mediated by IL-12-induced IFN-gamma include reduction in bone marrow cellularity and hematopoietic progenitors, as well as pronounced splenomegaly, largely caused by NK cell infiltration. In the absence of IFN-gamma signaling, IL-12 promotes hematopoiesis, consistent with its in vitro activities.


Subject(s)
Hematopoiesis/drug effects , Interferon-gamma/physiology , Interleukin-12/antagonists & inhibitors , Animals , Bone Marrow/drug effects , Mice , Receptors, Interferon/analysis , Interferon gamma Receptor
10.
Neuropathol Appl Neurobiol ; 36(4): 300-11, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20070537

ABSTRACT

AIMS: Copper and manganese levels are altered in mice both lacking PrPc and prion-infected brains. The aim of this study was to analyse the effects of manganese and copper imbalance on neuronal apoptosis in a scrapie-infected Tga20 mouse model. METHODS: Immunoreactivities for the apoptotic proteins Bax and active caspase-3 were evaluated in nine regions of the brain of scrapie-infected and control Tga20 mice treated with one of several diets: depleted cooper (-Cu), loaded manganese (+Mn), depleted copper/loaded manganese (-Cu+Mn) and regular diet. Immunohistochemical determination of NeuN was used to detect possible neuronal loss. RESULTS: Intracellular Bax detection was significantly decreased in animals fed with modified diets, particularly in those treated with copper-depleted diets. A decrease in active caspase-3 was primarily observed in animals fed with enhanced manganese diets. Our results show that the -Cu, -Cu+Mn and +Mn diets protected against apoptosis in scrapie-infected mice. However, NeuN immunolabelling quantification revealed that no diet was sufficient to arrest neuronal death. CONCLUSIONS: With regard to apoptosis induction, the response of Tga20 mice to prion infection was similar to that reported for other mice models. Our results demonstrate the neuroprotective effects of -Cu, -Cu+Mn and +Mn diets in a murine model of scrapie. However, neuronal death induced by infection with prions seems to be independent of apoptosis marker signalling. Moreover, copper-modified diets were neuroprotective against the possible toxicity of the prion transgene in Tga20 control and infected mice even though manganese supplementation could not counteract this toxicity.


Subject(s)
Apoptosis/physiology , Brain/metabolism , Copper/metabolism , Manganese/metabolism , Neurons/metabolism , Scrapie/metabolism , Animals , Caspase 3 , Copper/administration & dosage , Copper/deficiency , DNA-Binding Proteins , Diet , Disease Models, Animal , Manganese/administration & dosage , Manganese/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Scrapie/diet therapy , bcl-2-Associated X Protein/metabolism
11.
Phytother Res ; 24(5): 680-5, 2010 May.
Article in English | MEDLINE | ID: mdl-19827027

ABSTRACT

BACKGROUND: The herbal Petasites hybridus (butterbur) extract (Ze 339, PET) is known to have leukotriene inhibiting properties, and therefore might inhibit allergic diseases. METHODS: The effect of PET was investigated in ovalbumin (OVA) immunized BALB/c mice given intranasally together with antigen challenge in the murine model of allergic airway disease (asthma) with the analysis of the inflammatory and immune parameters in the lung. RESULTS: PET given with the antigen challenge inhibited the allergic response. PET inhibited airway hyperresponsiveness (AHR) and eosinophil recruitment into the bronchoalveolar lavage (BAL) fluid upon allergen challenge, but had no effect in the saline control mice. Eosinophil recruitment was further assessed in the lung by eosinophil peroxidase (EPO) activity at a concentration of 100 microg PET. Microscopic investigations revealed less inflammation, eosinophil recruitment and mucus hyperproduction in the lung with 100 microg PET. Diminution of AHR and inflammation was associated with reduced IL-4, IL-5 and RANTES production in the BAL fluid with 30 microg PET, while OVA specific IgE and eotaxin serum levels remained unchanged. CONCLUSION: PET, which has been reported to inhibit leukotriene activity, reduced allergic airway inflammation and AHR by inhibiting the production of the Th2 cytokines IL-4 and IL-5, and RANTES.


Subject(s)
Asthma/drug therapy , Bronchial Hyperreactivity/drug therapy , Petasites/chemistry , Phytotherapy , Plant Extracts/pharmacology , Th2 Cells/drug effects , Animals , Asthma/immunology , Asthma/physiopathology , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL5/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Eosinophil Peroxidase/immunology , Eosinophils/immunology , Immunoglobulin E/blood , Interleukin-4/immunology , Interleukin-5/immunology , Mice , Mice, Inbred BALB C , Mucus/immunology , Ovalbumin , Th2 Cells/immunology
12.
Gut ; 58(6): 771-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19174417

ABSTRACT

OBJECTIVE: Yersinia pseudotuberculosis causes ileitis and mesenteric lymphadenitis by mainly invading the Peyer's patches that are positioned in the terminal ileum. Whereas toll-like-receptor 2 (TLR2) controls mucosal inflammation by detecting certain microbiota-derived signals, its exact role in protecting Peyer's patches against bacterial invasion has not been defined. DESIGN: Wild-type, Tlr2-, Nod2- and MyD88-deficient animals were challenged by Y pseudotuberculosis via the oral or systemic route. The role of microbiota in conditioning Peyer's patches against Yersinia through TLR2 was assessed by delivering, ad libitum, exogenous TLR2 agonists in drinking water to germ-free and streptomycin-treated animals. Bacterial eradication from Peyer's patches was measured by using a colony-forming unit assay. Expression of cryptdins and the c-type lectin Reg3 beta was quantified by quantitative reverse transcriptase polymerase chain reaction analysis. RESULTS: Our data demonstrated that Tlr2-deficient mice failed to limit Yersinia dissemination from the Peyer's patches and succumbed to sepsis independently of nucleotide-binding and oligomerisation domain 2 (NOD2). Recognition of both microbiota-derived and myeloid differentiation factor 88 (MyD88)-mediated elicitors was found to be critically involved in gut protection against Yersinia-induced lethality, while TLR2 was dispensable to systemic Yersinia infection. Gene expression analyses revealed that optimal epithelial transcript level of the anti-infective Reg3 beta requires TLR2 activation. Consistently, Yersinia infection triggered TLR2-dependent Reg3 beta expression in Peyer's patches. Importantly, oral treatment with exogenous TLR2 agonists in germ-free animals was able to further enhance Yersinia-induced expression of Reg3 beta and to restore intestinal resistance to Yersinia. Lastly, genetic ablation of Reg3 beta resulted in impaired clearance of the bacterial load in Peyer's patches. CONCLUSIONS: TLR2/REG3 beta is thus an essential component in conditioning epithelial defence signalling pathways against bacterial invasion.


Subject(s)
Peyer's Patches/microbiology , Proteins/metabolism , Signal Transduction/physiology , Toll-Like Receptor 2/metabolism , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis , Animals , Cell Line , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Female , Gene Deletion , Gene Expression Profiling/methods , Germ-Free Life , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Pancreatitis-Associated Proteins , Peyer's Patches/metabolism , Peyer's Patches/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction/methods , Toll-Like Receptor 2/genetics
13.
Infect Immun ; 77(12): 5359-68, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19822651

ABSTRACT

The course and outcome of infection with mycobacteria are determined by a complex interplay between the immune system of the host and the survival mechanisms developed by the bacilli. Recent data suggest a regulatory role of histamine not only in the innate but also in the adaptive immune response. We used a model of pulmonary Mycobacterium tuberculosis infection in histamine-deficient mice lacking histidine decarboxylase (HDC(-/-)), the histamine-synthesizing enzyme. To confirm that mycobacterial infection induced histamine production, we exposed mice to M. tuberculosis and compared responses in C57BL/6 (wild-type) and HDC(-/-) mice. Histamine levels increased around fivefold above baseline in infected C57BL/6 mice at day 28 of infection, whereas only small amounts were detected in the lungs of infected HDC(-/-) mice. Blocking histamine production decreased both neutrophil influx into lung tissue and the release of proinflammatory mediators, such as interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-alpha), in the acute phase of infection. However, the accumulation and activation of CD4(+) T cells were augmented in the lungs of infected HDC(-/-) mice and correlated with a distinct granuloma formation that contained abundant lymphocytic infiltration and reduced numbers of mycobacteria 28 days after infection. Furthermore, the production of IL-12, gamma interferon, and nitric oxide, as well as CD11c(+) cell influx into the lungs of infected HDC(-/-) mice, was increased. These findings indicate that histamine produced after M. tuberculosis infection may play a regulatory role not only by enhancing the pulmonary neutrophilia and production of IL-6 and TNF-alpha but also by impairing the protective Th1 response, which ultimately restricts mycobacterial growth.


Subject(s)
Histamine/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Granuloma/microbiology , Granuloma/pathology , Histidine Decarboxylase/deficiency , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Nitric Oxide/metabolism
14.
Mucosal Immunol ; 12(1): 290, 2019 01.
Article in English | MEDLINE | ID: mdl-30504789

ABSTRACT

The original version of this Article omitted the author Dr Mathias Chamaillard from the l'Institut de Pasteur, Lille, France. This has been corrected in both the PDF and HTML versions of the Article.

15.
Tuberculosis (Edinb) ; 88(3): 221-34, 2008 May.
Article in English | MEDLINE | ID: mdl-17950671

ABSTRACT

TNF is required for protection against virulent and non-virulent mycobacterial infections. Here we compared the effect of Tm-TNF and sTNF, two different molecular forms of TNF, in virulent and non-virulent murine challenge models. Using non-virulent Mycobacterium bovis BCG intranasal infection we established that immunity is durably compromised in Tm-TNF mice, with augmented bacilli burden, leading to chronic but non-lethal infection. Acute infection by a virulent Mycobacterium tuberculosis low-dose aerosol challenge was controlled in Tm-TNF mice with bacilli burdens equivalent to that in WT mice and pulmonary pathology characterised by the formation of well-defined, bactericidal granulomas. Protective immunity was however compromised in Tm-TNF mice during the chronic phase of M. tuberculosis infection, with increased lung bacterial growth and inflammatory cell activation, dissolution of granulomas associated with dispersed iNOS expression, increased pulmonary IFNgamma and IL-10 expression but decreased IL-12 production, followed by death. In conclusion, membrane TNF is sufficient to control non-virulent, M. bovis BCG infection, and acute but not chronic infection with virulent M. tuberculosis.


Subject(s)
Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/prevention & control , Tumor Necrosis Factor-alpha/immunology , Acute Disease , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Cytokines/biosynthesis , Disease Susceptibility , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mycobacterium bovis/growth & development , Mycobacterium bovis/isolation & purification , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/isolation & purification , Nitric Oxide Synthase Type II/biosynthesis , Tuberculoma/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology , Virulence
16.
Clin Exp Allergy ; 38(10): 1668-79, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18631348

ABSTRACT

BACKGROUND: Epidemiological and experimental data suggest that bacterial lipopolysaccharides (LPS) can either protect from or exacerbate allergic asthma. Lipopolysaccharides trigger immune responses through toll-like receptor 4 (TLR4) that in turn activates two major signalling pathways via either MyD88 or TRIF adaptor proteins. The LPS is a pro-Type 1 T helper cells (Th1) adjuvant while aluminium hydroxide (alum) is a strong Type 2 T helper cells (Th2) adjuvant, but the effect of the mixing of both adjuvants on the development of lung allergy has not been investigated. OBJECTIVE: We determined whether natural (LPS) or synthetic (ER-803022) TLR4 agonists adsorbed onto alum adjuvant affect allergen sensitization and development of airway allergic disease. To dissect LPS-induced molecular pathways, we used TLR4-, MyD88-, TRIF-, or IL-12/IFN-gamma-deficient mice. METHODS: Mice were sensitized with subcutaneous injections of ovalbumin (OVA) with or without TLR4 agonists co-adsorbed onto alum and challenged with intranasally with OVA. The development of allergic lung disease was evaluated 24 h after last OVA challenge. RESULTS: Sensitization with OVA plus LPS co-adsorbed onto alum impaired in dose-dependent manner OVA-induced Th2-mediated allergic responses such as airway eosinophilia, type-2 cytokines secretion, airway hyper-reactivity, mucus hyper production and serum levels of IgE or IgG1 anaphylactic antibodies. Although the levels of IgG2a, Th1-affiliated isotype increased, investigation into the lung-specific effects revealed that LPS did not induce a Th1 pattern of inflammation. Lipopolysaccharides impaired the development of Th2 immunity, signaling via TLR4 and MyD88 molecules and via the IL-12/IFN-gamma axis, but not through TRIF pathway. Moreover, the synthetic TLR4 agonists that proved to have a less systemic inflammatory response than LPS also protected against allergic asthma development. CONCLUSION: Toll-like receptor 4 agonists co-adsorbed with allergen onto alum down-modulate allergic lung disease and prevent the development of polarized T cell-mediated airway inflammation.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Asthma/prevention & control , Lipopolysaccharides/administration & dosage , Toll-Like Receptor 4/agonists , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/immunology , Allergens/immunology , Animals , Antibodies/blood , Asthma/immunology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Cytokines/analysis , Cytokines/immunology , Disease Models, Animal , Female , Interferon-gamma/immunology , Interleukin-12/deficiency , Interleukin-12/immunology , Interleukin-12/metabolism , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/immunology , Ovalbumin/immunology , Phospholipids/pharmacology , Toll-Like Receptor 4/immunology
17.
Mucosal Immunol ; 11(3): 811-819, 2018 05.
Article in English | MEDLINE | ID: mdl-29139477

ABSTRACT

Flavones represent a class of polyphenols that are found in many plant-derived food sources. Herein, we provide evidence that the anti-inflammatory and antiproliferative effect of the flavone apigenin relies on the regulation of the gut microbiota by the NOD-like receptor family pyrin domain containing 6 (Nlrp6). When challenged by dextran sulfate sodium (DSS) in drinking water, mice were protected against colitis upon cohousing with apigenin-treated animals. In contrast, the protective effect was lost in the absence of Nlrp6. Sequencing of the 16S ribosomal RNA gene revealed a shift in the composition of the gut microbiota in apigenin-treated mice that was not observed in the absence of Nlrp6. Equally important, we find that the antiproliferative effect of apigenin was dominantly transmitted after cohousing, while being compromised in Nlrp6-deficient mice. In contrast, the symptoms of colitis were alleviated upon apigenin administration even in the absence of either caspase-1/11 or Asc. Collectively, these data indicate that apigenin modulated an inflammasome-independent mechanism by which Nlrp6 reprograms the gut microbiota for protecting mice against colitis. Our study highlights a modulation of the Nlrp6 signaling pathway by a prominent constituent of the human diet that may point toward improved ways to treat inflammatory bowel diseases.


Subject(s)
Apigenin/administration & dosage , Colitis/prevention & control , Diet , Flavones/administration & dosage , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/diet therapy , Receptors, Cell Surface/metabolism , Animals , Colitis/chemically induced , Dextran Sulfate , Housing, Animal , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Ribosomal, 16S/genetics , Receptors, Cell Surface/genetics , Signal Transduction
18.
Mucosal Immunol ; 11(4): 1181-1190, 2018 07.
Article in English | MEDLINE | ID: mdl-29728643

ABSTRACT

Upon oral infection with Toxoplasma gondii cysts (76 K strain) tachyzoites are released into the intestinal lumen and cross the epithelial barrier causing damage and acute intestinal inflammation in C57BL/6 (B6) mice. Here we investigated the role of microbiota and IL-22 in T.gondii-induced small intestinal inflammation. Oral T.gondii infection in B6 mice causes inflammation with IFNγ and IL-22 production. In IL-22-deficient mice, T.gondii infection augments the Th1 driven inflammation. Deficiency in either IL-22bp, the soluble IL-22 receptor or Reg3γ, an IL-22-dependent antimicrobial lectin/peptide, did not reduce inflammation. Under germ-free conditions, T.gondii-induced inflammation was reduced in correlation with parasite load. But intestinal inflammation is still present in germ-free mice, at low level, in the lamina propria, independently of IL-22 expression. Exacerbated intestinal inflammation driven by absence of IL-22 appears to be independent of IL-22 deficiency associated-dysbiosis as similar inflammation was observed after fecal transplantation of IL-22-/- or WT microbiota to germ-free-WT mice. Our results suggest cooperation between parasite and intestinal microbiota in small intestine inflammation development and endogenous IL-22 seems to exert a protective role independently of its effect on the microbiota. In conclusion, IL-22 participates in T.gondii induced acute small intestinal inflammation independently of microbiota and Reg3γ.


Subject(s)
Gastrointestinal Microbiome/immunology , Inflammation/immunology , Interleukins/metabolism , Intestines/immunology , Toxoplasma/physiology , Toxoplasmosis/immunology , Animals , Cells, Cultured , Disease Progression , Interleukins/genetics , Intestines/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis-Associated Proteins/genetics , Pancreatitis-Associated Proteins/metabolism , Parasite Load , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Interleukin-22
19.
Cancer Gene Ther ; 14(9): 802-14, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17589432

ABSTRACT

Immunization with mRNA encoding tumor antigen is an emerging vaccine strategy for cancer. In this paper, we demonstrate that mice receiving systemic injections of MART1 mRNA histidylated lipopolyplexes were specifically and significantly protected against B16F10 melanoma tumor progression. The originality of this work concerns the use of a new tumor antigen mRNA formulation as vaccine, which allows an efficient protection against the growth of a highly aggressive tumor model after its delivery by intravenous route. Synthetic melanoma-associated antigen MART1 mRNA was formulated with a polyethylene glycol (PEG)ylated derivative of histidylated polylysine and L-histidine-(N,N-di-n-hexadecylamine)ethylamide liposomes (termed histidylated lipopolyplexes). Lipopolyplexes comprised mRNA/polymer complexes encapsulated by liposomes. The tumor protective effect was induced with MART1 mRNA carrying a poly(A) tail length of 100 adenosines at an optimal dose of 12.5 microg per mouse. MART1 mRNA lipopolyplexes elicited a cellular immune response characterized by the production of interferon-gamma and the induction of cytotoxic T lymphocytes. Finally, the anti-B16 response was enhanced using a formulation containing both MART1 mRNA and MART1-LAMP1 mRNA encoding the antigen targeted to the major histocompatibility complex class II compartments by the lysosomal sorting signal of LAMP1 protein. Our results provide a basis for the development of mRNA histidylated lipopolyplexes for cancer vaccine.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/administration & dosage , Histidine/metabolism , Melanoma, Experimental/pathology , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/genetics , RNA, Messenger/genetics , Animals , Antigens, Neoplasm/metabolism , Cancer Vaccines/genetics , Disease Progression , MART-1 Antigen , Melanoma, Experimental/immunology , Mice , Microscopy, Electron, Transmission , Neoplasm Proteins/metabolism , RNA, Messenger/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Transcription, Genetic
20.
Mucosal Immunol ; 10(3): 695-704, 2017 05.
Article in English | MEDLINE | ID: mdl-27731323

ABSTRACT

The complement subunit C1q was recently identified as a marker for monocyte-derived regulatory dendritic cells supporting the differentiation of interleukin (IL)-10-secreting CD4+ T cells with a suppressive activity. Furthermore, C1q expression is upregulated in peripheral blood mononuclear cells of allergic patients in the course of successful allergen immunotherapy. Herein, we investigated a potential direct role of C1q in downregulating allergic inflammation. In mice with ovalbumin (OVA) or birch pollen (BP)-induced allergic asthma, C1q is as efficacious as dexamethasone to reduce both airway hyperresponsiveness (AHR), eosinophil, and ILC2 infiltrates in bronchoalveolar lavages, as well as allergen-specific T helper 2 cells in the lungs. Administration of C1q does not expand IL-10+/Foxp3+ regulatory T cells in the lungs, spleen, or in the blood. Depletion of plasmacytoid dendritic cells (pDCs) abrogates the capacity of C1q to reduce AHR and eosinophilic infiltrates in OVA-sensitized mice. Also C1q treatment inhibits the activation of human and mouse pDCs by CpGs, thereby demonstrating a critical role for pDCs in the anti-inflammatory activity of C1q. We conclude that regulatory dendritic cells can mediate a potent direct anti-inflammatory activity via the expression and/or secretion of molecules such as C1q, independently of their capacity to expand the pool of regulatory T cells.


Subject(s)
Complement C1q/metabolism , Dendritic Cells/immunology , Eosinophils/immunology , Hypersensitivity/immunology , Lung/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Betula , Cells, Cultured , Complement C1q/administration & dosage , Female , Humans , Interleukin-10/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Plant Extracts , Pollen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL