Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Immunology ; 155(4): 477-490, 2018 12.
Article in English | MEDLINE | ID: mdl-30098206

ABSTRACT

The excessive inflammation often present in patients with severe dengue infection is considered both a hallmark of disease and a target for potential treatments. Interleukin-33 (IL-33) is a pleiotropic cytokine with pro-inflammatory effects whose role in dengue has not been fully elucidated. We demonstrate that IL-33 plays a disease-exacerbating role during experimental dengue infection in immunocompetent mice. Mice infected with dengue virus serotype 2 (DENV2) produced high levels of IL-33. DENV2-infected mice treated with recombinant IL-33 developed markedly more severe disease compared with untreated mice as assessed by mortality, granulocytosis, liver damage and pro-inflammatory cytokine production. Conversely, ST2-/- mice (deficient in IL-33 receptor) infected with DENV2 developed significantly less severe disease compared with wild-type mice. Furthermore, the increased disease severity and the accompanying pathology induced by IL-33 during dengue infection were reversed by the simultaneous treatment with a CXCR2 receptor antagonist (DF2156A). Together, these results indicate that IL-33 plays a disease-exacerbating role in experimental dengue infection, probably driven by CXCR2-expressing cells, leading to elevated pro-inflammatory response-mediated pathology. Our results also indicate that IL-33 is a potential therapeutic target for dengue infection.


Subject(s)
Dengue Virus/immunology , Interleukin-33/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Recombinant Proteins/pharmacology , Animals , Dengue/immunology , Dengue/virology , Disease Progression , Interleukin-1 Receptor-Like 1 Protein/deficiency , Interleukin-1 Receptor-Like 1 Protein/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sulfonamides/pharmacology
2.
Front Immunol ; 8: 123, 2017.
Article in English | MEDLINE | ID: mdl-28243234

ABSTRACT

Recent in silico studies suggested that the transcription cofactor LIM-only protein FHL2 is a major transcriptional regulator of mouse natural killer (NK) cells. However, the expression and role of FHL2 in NK cell biology are unknown. Here, we confirm that FHL2 is expressed in both mouse and human NK cells. Using FHL2-/- mice, we found that FHL2 controls NK cell development in the bone marrow and maturation in peripheral organs. To evaluate the importance of FHL2 in NK cell activation, FHL2-/- mice were infected with Streptococcus pneumoniae. FHL2-/- mice are highly susceptible to this infection. The activation of lung NK cells is altered in FHL2-/- mice, leading to decreased IFNγ production and a loss of control of bacterial burden. Collectively, our data reveal that FHL2 is a new transcription cofactor implicated in NK cell development and activation during pulmonary bacterial infection.

3.
PLoS Pathog ; 11(2): e1004607, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25659095

ABSTRACT

Cerebral malaria (CM) is a complex parasitic disease caused by Plasmodium sp. Failure to establish an appropriate balance between pro- and anti-inflammatory immune responses is believed to contribute to the development of cerebral pathology. Using the blood-stage PbA (Plasmodium berghei ANKA) model of infection, we show here that administration of the pro-Th2 cytokine, IL-33, prevents the development of experimental cerebral malaria (ECM) in C57BL/6 mice and reduces the production of inflammatory mediators IFN-γ, IL-12 and TNF-α. IL-33 drives the expansion of type-2 innate lymphoid cells (ILC2) that produce Type-2 cytokines (IL-4, IL-5 and IL-13), leading to the polarization of the anti-inflammatory M2 macrophages, which in turn expand Foxp3 regulatory T cells (Tregs). PbA-infected mice adoptively transferred with ILC2 have elevated frequency of M2 and Tregs and are protected from ECM. Importantly, IL-33-treated mice deleted of Tregs (DEREG mice) are no longer able to resist ECM. Our data therefore provide evidence that IL-33 can prevent the development of ECM by orchestrating a protective immune response via ILC2, M2 macrophages and Tregs.


Subject(s)
Interleukin-33/immunology , Macrophages/immunology , Malaria, Cerebral/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Adoptive Transfer , Animals , Coculture Techniques , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immunity, Innate , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology , Real-Time Polymerase Chain Reaction
4.
Eur J Immunol ; 45(5): 1354-65, 2015 May.
Article in English | MEDLINE | ID: mdl-25682948

ABSTRACT

Cerebral malaria, a severe complication of Plasmodium falciparum infection, can be modeled in murine Plasmodium berghei ANKA (PbA) infection. PbA-induced experimental cerebral malaria (ECM) is CD8(+) T-cell mediated, and influenced by TH 1/TH 2 balance. Here, we show that IL-33 expression is increased in brain undergoing ECM and we address the role of the IL-33/ST2 pathway in ECM development. ST2-deficient mice were resistant to PbA-induced neuropathology. They survived >20 days with no ECM neurological sign and a preserved cerebral microcirculation, while WT mice succumbed within 10 days with ECM, brain vascular leakage, distinct microvascular pathology obstruction, and hemorrhages. Parasitemia and brain parasite load were similar in ST2-deficient and WT mice. Protection was accompanied by reduced brain sequestration of activated CD4(+) T cells and perforin(+) CD8(+) T cells. While IFN-γ and T-cell-attracting chemokines CXCL9 and CXCL10 were not affected in the absence of functional ST2 pathway, the local expression of ICAM-1, CXCR3, and LT-α, crucial for ECM development, was strongly reduced, and this may explain the diminished pathogenic T-cell recruitment and resistance to ECM. Therefore, IL-33 is induced in PbA sporozoite infection, and the pathogenic T-cell responses with local microvascular pathology are dependent on IL-33/ST2 signaling, identifying IL-33 as a new actor in ECM development.


Subject(s)
Malaria, Cerebral/etiology , Plasmodium berghei , Receptors, Interleukin/metabolism , Animals , Brain/immunology , Brain/parasitology , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Female , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/metabolism , Lymphocyte Activation , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium berghei/immunology , Plasmodium berghei/pathogenicity , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics
5.
Nat Commun ; 5: 4575, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25099390

ABSTRACT

Th9 cells protect hosts against helminthic infection but also mediate allergic disease. Here we show that nitric oxide (NO) promotes Th9 cell polarization of murine and human CD4(+) T cells. NO de-represses the tumour suppressor gene p53 via nitrosylation of Mdm2. NO also increases p53-mediated IL-2 production, STAT5 phosphorylation and IRF4 expression, all essential for Th9 polarization. NO also increases the expression of TGFßR and IL-4R, pivotal to Th9 polarization. OVA-sensitized mice treated with an NO donor developed more severe airway inflammation. Transferred Th9 cells induced airway inflammation, which was exacerbated by NO and blocked by anti-IL-9 antibody. Nos2(-/-) mice had less Th9 cells and developed attenuated eosinophilia during OVA-induced airway inflammation compared with wild-type mice. Our data demonstrate that NO is an important endogenous inducer of Th9 cells and provide a hitherto unrecognized mechanism for NO-mediated airway inflammation via the expansion of Th9 cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Inflammation/pathology , Interleukin-9/metabolism , Nitric Oxide/chemistry , Animals , Cell Separation , Cells, Cultured , Eosinophilia/metabolism , Flow Cytometry , Humans , Inflammation/chemically induced , Interferon Regulatory Factors/metabolism , Interleukin-2/metabolism , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type II/metabolism , STAT5 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism
6.
J Allergy Clin Immunol ; 134(6): 1422-1432.e11, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24985397

ABSTRACT

BACKGROUND: The initiation and regulation of pulmonary fibrosis are not well understood. IL-33, an important cytokine for respiratory diseases, is overexpressed in the lungs of patients with idiopathic pulmonary fibrosis. OBJECTIVES: We aimed to determine the effects and mechanism of IL-33 on the development and severity of pulmonary fibrosis in murine bleomycin-induced fibrosis. METHODS: Lung fibrosis was induced by bleomycin in wild-type or Il33r (St2)(-/-) C57BL/6 mice treated with the recombinant mature form of IL-33 or anti-IL-33 antibody or transferred with type 2 innate lymphoid cells (ILC2s). The development and severity of fibrosis was evaluated based on lung histology, collagen levels, and lavage cytology. Cytokine and chemokine levels were quantified by using quantitative PCR, ELISA, and cytometry. RESULTS: IL-33 is constitutively expressed in lung epithelial cells but is induced in macrophages by bleomycin. Bleomycin enhanced the production of the mature but reduced full-length form of IL-33 in lung tissue. ST2 deficiency, anti-IL-33 antibody treatment, or alveolar macrophage depletion attenuated and exogenous IL-33 or adoptive transfer of ILC2s enhanced bleomycin-induced lung inflammation and fibrosis. These pathologic changes were accompanied, respectively, by reduced or increased IL-33, IL-13, TGF-ß1, and inflammatory chemokine production in the lung. Furthermore, IL-33 polarized M2 macrophages to produce IL-13 and TGF-ß1 and induced the expansion of ILC2s to produce IL-13 in vitro and in vivo. CONCLUSIONS: IL-33 is a novel profibrogenic cytokine that signals through ST2 to promote the initiation and progression of pulmonary fibrosis by recruiting and directing inflammatory cell function and enhancing profibrogenic cytokine production in an ST2- and macrophage-dependent manner.


Subject(s)
Interleukins/immunology , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Receptors, Interleukin/immunology , Animals , Fibrosis , Interleukin-1 Receptor-Like 1 Protein , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-33 , Interleukins/genetics , Lymphocytes/immunology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology
7.
J Immunol ; 192(5): 2442-8, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24470502

ABSTRACT

CD4(+) T cells have long been grouped into distinct helper subsets on the basis of their cytokine-secretion profile. In recent years, several subsets of innate lymphoid cell have been described as key producers of these same Th-associated cytokines. However, the functional relationship between Th cells and innate lymphoid cells (ILCs) remains unclear. We show in this study that lineage-negative ST2(+)ICOS(+)CD45(+) type 2 ILCs and CD4(+) T cells can potently stimulate each other's function via distinct mechanisms. CD4(+) T cell provision of IL-2 stimulates type 2 cytokine production by type 2 ILCs. By contrast, type 2 ILCs modulate naive T cell activation in a cell contact-dependent manner, favoring Th2 while suppressing Th1 differentiation. Furthermore, a proportion of type 2 ILCs express MHC class II and can present peptide Ag in vitro. Importantly, cotransfer experiments show that type 2 ILCs also can boost CD4(+) T cell responses to Ag in vivo.


Subject(s)
Antigens, Differentiation/immunology , Cell Differentiation/immunology , Cytokines/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Innate/physiology , Th2 Cells/immunology , Animals , Antigens, Differentiation/genetics , Cell Differentiation/genetics , Cytokines/genetics , Histocompatibility Antigens Class II/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology
8.
J Leukoc Biol ; 94(6): 1317-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23975892

ABSTRACT

IPF is a chronic, progressive pulmonary disease, leading to respiratory failure. In search of mechanisms of IPF, we used the bleomycin-induced lung-injury model in mice, which causes acute inflammation that may progress to chronic lung inflammation and fibrosis. Here, we asked whether CXCL6/GCP-2, a member of the CXC chemokine superfamily, may be involved in IPF development. First, we reported an increase of CXCL6 levels in BALF from patients with IPF, as well as in the lung of mice, 24 h after bleomycin administration. To investigate whether CXCL6 played a role in experimental bleomycin-induced pulmonary fibrosis, we treated mice with an anti-mCXCL6 mAb that has been shown to inhibit neutrophil chemotaxis in vitro. CXCL6 antibody blockade attenuated acute inflammation with a reduced pulmonary neutrophil influx, IL-1ß, CXCL1, and TIMP-1 production. In the later phase (14 days after bleomycin exposure), lymphocyte recruitment and fibrosis markers, such as collagen and TIMP-1, were diminished, as well as collagen deposition and fibrotic lesion the lung. Therefore, the data suggest that CXCL6 contributes to experimental pulmonary fibrosis, and CXCL6 inhibition might be used to reduce lung toxicity associated with bleomycin treatment.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Neutralizing/pharmacology , Bleomycin/adverse effects , Chemokine CXCL6/antagonists & inhibitors , Pneumonia/immunology , Pulmonary Fibrosis/immunology , Animals , Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neutralizing/immunology , Bleomycin/pharmacology , Cell Migration Inhibition/drug effects , Cell Migration Inhibition/immunology , Chemokine CXCL6/immunology , Chemokine CXCL6/metabolism , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/chemically induced , Pneumonia/drug therapy , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/pathology
9.
J Immunol ; 191(1): 164-70, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23720815

ABSTRACT

NO is a free radical with pleiotropic functions. We have shown earlier that NO induces a population of CD4(+)CD25(+)Foxp3(-) regulatory T cells (NO-Tregs) that suppress the functions of CD4(+)CD25(-) effector T cells in vitro and in vivo. We report in this study an unexpected finding that NO-Tregs suppressed Th17 but not Th1 cell differentiation and function. In contrast, natural Tregs (nTregs), which suppressed Th1 cells, failed to suppress Th17 cells. Consistent with this observation, NO-Tregs inhibited the expression of retinoic acid-related orphan receptor γt but not T-bet, whereas nTregs suppressed T-bet but not retinoic acid-related orphan receptor γt expression. The NO-Treg-mediated suppression of Th17 was partially cell contact-dependent and was associated with IL-10. In vivo, adoptively transferred NO-Tregs potently attenuated experimental autoimmune encephalomyelitis. The disease suppression was accompanied by a reduction of Th17, but not Th1 cells in the draining lymph nodes, and a decrease in the production of IL-17, but an increase in IL-10 synthesis. Our results therefore demonstrate the differential suppressive function between NO-Tregs and nTregs and indicate specialization of the regulatory mechanism of the immune system.


Subject(s)
Cell Differentiation/immunology , Growth Inhibitors/physiology , Nitric Oxide/physiology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Cell Communication/immunology , Cells, Cultured , Female , Growth Inhibitors/pharmacology , Immune Tolerance/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/pharmacology , Primary Cell Culture , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Th17 Cells/cytology
10.
Eur J Immunol ; 43(6): 1529-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23505056

ABSTRACT

Dengue virus (DENV), a mosquito-borne flavivirus, is a public health problem in many tropical countries. IL-22 and IL-17A are key cytokines in several infectious and inflammatory diseases. We have assessed the contribution of IL-22 and IL-17A in the pathogenesis of experimental dengue infection using a mouse-adapted DENV serotype 2 strain (P23085) that causes a disease that resembles severe dengue in humans. We show that IL-22 and IL-17A are produced upon DENV-2 infection in immune-competent mice. Infected IL-22(-/-) mice had increased lethality, neutrophil accumulation and pro-inflammatory cytokines in tissues, notably IL-17A. Viral load was increased in spleen and liver of infected IL-22(-/-) mice. There was also more severe liver injury, as seen by increased transaminases levels and tissue histopathology. γδ T cells and NK cells are sources of IL-17A and IL-22, respectively, in liver and spleen. We also show that DENV-infected HepG2 cells treated with rhIL-22 had reduced cell death and decreased IL-6 production. IL-17RA(-/-) mice were protected upon infection and IL-17A-neutralizing-Ab-treatment partially reversed the phenotype observed in IL-22(-/-) -infected mice. We suggest that disrupting the balance between IL-22 and IL-17A levels may represent an important strategy to reduce inflammation and tissue injury associated with severe dengue infection.


Subject(s)
Dengue Virus/immunology , Dengue/immunology , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Liver/metabolism , Neutrophils/immunology , Animals , Apoptosis/drug effects , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hep G2 Cells , Humans , Inflammation/genetics , Interleukin-17/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukins/genetics , Interleukins/immunology , Liver/immunology , Liver/pathology , Liver/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/virology , Receptors, Interleukin-17/genetics , Viral Load/genetics , Interleukin-22
11.
J Allergy Clin Immunol ; 130(5): 1159-1166.e6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22738676

ABSTRACT

BACKGROUND: The IL-1 family cytokine IL-33 is involved in the induction of airway inflammation in allergic patients and after viral infection. Several cell types, including CD4(+) T(H)2 cells and the recently described type 2 innate lymphoid cells (ILCs), are targets for IL-33, yet the mechanisms by which this cytokine modulates their activation are not clear. OBJECTIVES: Our goal was to investigate a role for mammalian target of rapamycin (mTOR) signaling in the activation of T(H)2 and ILC responses and the induction of airway inflammation by IL-33. METHODS: We biochemically determined the effect of IL-33 on mTOR activation in T(H)2 cells and ILCs and examined the effect of this signaling pathway in vivo using a murine model of IL-33-induced lung inflammation. RESULTS: We found that IL-33 induces mTOR activation through p110δ phosphoinositide 3-kinase and that blockade of the mTOR pathway inhibited IL-33-induced IL-5 and IL-13 production by T(H)2 cells and ILCs. Furthermore, use of a ribosomal protein S6 kinase 1 inhibitor implicated a role for ribosomal protein S6 kinase 1 in IL-33-induced mTOR-dependent cytokine production. Intranasal administration of IL-33 to wild-type mice induced airway inflammation, whereas adoptive transfer of wild-type ILCs to IL-33 receptor-deficient (St2(-/-)) mice recapitulated this response. Importantly, coadministration of the mTOR inhibitor rapamycin reduced IL-33-dependent ILC, macrophage, and eosinophil accumulation; cytokine secretion; and mucus deposition in the airways. CONCLUSION: These data reveal a hitherto unrecognized role of mTOR signaling in IL-33-driven, ILC-dependent inflammation in vivo and suggest that manipulation of this pathway might represent a target for therapeutic intervention for airway inflammation.


Subject(s)
Interleukins/administration & dosage , Pneumonia/drug therapy , Pneumonia/immunology , TOR Serine-Threonine Kinases/metabolism , Th2 Cells/drug effects , Animals , Cells, Cultured , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interleukin-1 Receptor-Like 1 Protein , Interleukin-13/metabolism , Interleukin-33 , Interleukin-5/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Pneumonia/chemically induced , Receptors, Interleukin/genetics , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Th2 Cells/immunology , Th2 Cells/transplantation
12.
J Mol Cell Biol ; 4(1): 3-10, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22147847

ABSTRACT

Allergic asthma has increased dramatically in prevalence and severity over the last three decades. Both clinical and experimental data support an important role of Th2 cell response in the allergic response. Recent investigations revealed that airway exposure to allergen in sensitized individuals causes the release of ATP and uric acid, activating the NLRP3 inflammasome complex and cleaving pro-IL-1ß to mature IL-1ß through caspase-1. The production of pro-IL-1ß requires a toll-like receptor (TLR) 4 signal which is provided by the allergen. IL-1ß creates a pro-inflammatory milieu with the production of IL-6 and chemokines which mobilize neutrophils and enhance Th17 cell differentiation in the lung. Here, we review our results showing that NLRP3 inflammasome activation is required to develop allergic airway inflammation in mice and that IL-17 and IL-22 production by Th17 cells plays a critical role in established asthma. Therefore, inflammasome activation leading to IL-1ß production contributes to the control of allergic asthma by enhancing Th17 cell differentiation.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1/immunology , Pneumonia/immunology , Th17 Cells/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , Cell Differentiation , Inflammasomes/metabolism , Interleukin-1/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-1beta/immunology , Interleukins/immunology , Interleukins/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Pneumonia/metabolism , Signal Transduction , Th2 Cells/immunology , Toll-Like Receptor 4/immunology , Uric Acid/metabolism , Interleukin-22
13.
Eur J Immunol ; 41(6): 1675-86, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21469105

ABSTRACT

IL-33, a new member of the IL-1 family cytokine, is involved in Th2-type responses in a wide range of diseases and signals through the ST2 receptor expressed on many immune cells. Since the effects of IL-33 on DCs remain controversial, we investigated the ability of IL-33 to modulate DC functions in vitro and in vivo. Here, we report that IL-33 activates myeloid DCs to produce IL-6, IL-1b, TNF, CCL17 and to express high levels of CD40, CD80 OX40L and CCR7. Importantly, IL-33-activated DCs prime naive lymphocytes to produce the Th2 cytokines IL-5 and IL-13, but not IL-4. In vivo, IL-33 exposure induces DC recruitment and activation in the lung. Using an OVA-induced allergic lung inflammation model, we demonstrate that the reduced airway inflammation in ST2-deficient mice correlates with the failure in DC activation and migration to the draining LN. Finally, we show that adoptive transfer of IL-33-activated DCs exacerbates lung inflammation in a DC-driven model of allergic airway inflammation. These data demonstrate for the first time that IL-33 activates DCs during antigen presentation and thereby drives a Th2-type response in allergic lung inflammation.


Subject(s)
Cytokines/metabolism , Dendritic Cells/metabolism , Interleukins/metabolism , Receptors, Interleukin/metabolism , Respiratory Hypersensitivity/immunology , Th2 Cells/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Differentiation , Cell Movement , Cells, Cultured , Cytokines/genetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/immunology , Lung/immunology , Lung/metabolism , Lung/pathology , Lymphocyte Activation , Mice , Mice, Knockout , Myeloid Progenitor Cells/pathology , Pneumonia , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Th1-Th2 Balance , Th2 Cells/immunology , Th2 Cells/pathology
14.
Am J Respir Crit Care Med ; 183(9): 1153-63, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21297073

ABSTRACT

RATIONALE: IL-22 has both proinflammatory and antiinflammatory properties. Its role in allergic lung inflammation has not been explored. OBJECTIVES: To investigate the expression and roles of IL-22 in the onset and resolution of experimental allergic asthma and its cross-talk with IL-17A. METHODS: IL-22 expression was assessed in patient samples and in the lung of mice immunized and challenged with ovalbumin. IL-22 functions in allergic airway inflammation were evaluated using mice deficient in IL-22 or anti-IL-22 neutralizing antibodies. Moreover, the effects of recombinant IL-22 and IL-17A neutralizing antibodies were investigated. MEASUREMENTS AND MAIN RESULTS: Increased pulmonary IL-22 expression is found in the serum of patients with asthma and mice immunized and challenged with ovalbumin. Allergic lung inflammation is IL-22 dependent because eosinophil recruitment, Th2 cytokine including IL-13 and IL-33, chemokine production, airway hyperreactivity, and mucus production are drastically reduced in mice deficient in IL-22 or by IL-22 antibody neutralization during immunization of wild-type mice. By contrast, IL-22 neutralization during antigen challenge enhanced allergic lung inflammation with increased Th2 cytokines. Consistent with this, recombinant IL-22 given with allergen challenge protects mice from lung inflammation. Finally, IL-22 may regulate the expression and proinflammatory properties of IL-17A in allergic lung inflammation. CONCLUSIONS: IL-22 is required for the onset of allergic asthma, but functions as a negative regulator of established allergic inflammation. Our study reveals that IL-22 contributes to the proinflammatory properties of IL-17A in experimental allergic asthma.


Subject(s)
Asthma/immunology , Interleukin-17/immunology , Interleukins/immunology , Animals , Asthma/blood , Chemokines/immunology , Cytokines/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Eosinophils/immunology , Flow Cytometry , Humans , Interleukins/blood , Mice , Mice, Knockout , Th2 Cells/immunology , Interleukin-22
15.
PLoS One ; 5(12): e15680, 2010 Dec 29.
Article in English | MEDLINE | ID: mdl-21206747

ABSTRACT

Dengue virus (DENV), a mosquito-borne flavivirus, is a public health problem in many tropical countries. Recent clinical data have shown an association between levels of different chemokines in plasma and severity of dengue. We evaluated the role of CC chemokine receptors CCR1, CCR2 and CCR4 in an experimental model of DENV-2 infection in mice. Infection of mice induced evident clinical disease and tissue damage, including thrombocytopenia, hemoconcentration, lymphopenia, increased levels of transaminases and pro-inflammatory cytokines, and lethality in WT mice. Importantly, infected WT mice presented increased levels of chemokines CCL2/JE, CCL3/MIP-1α and CCL5/RANTES in spleen and liver. CCR1⁻/⁻ mice had a mild phenotype with disease presentation and lethality similar to those of WT mice. In CCR2⁻/⁻ mice, lethality, liver damage, levels of IL-6 and IFN-γ, and leukocyte activation were attenuated. However, thrombocytopenia, hemoconcentration and systemic TNF-α levels were similar to infected WT mice. Infection enhanced levels of CCL17/TARC, a CCR4 ligand. In CCR4⁻/⁻ mice, lethality, tissue injury and systemic inflammation were markedly decreased. Despite differences in disease presentation in CCR-deficient mice, there was no significant difference in viral load. In conclusion, activation of chemokine receptors has discrete roles in the pathogenesis of dengue infection. These studies suggest that the chemokine storm that follows severe primary dengue infection associates mostly to development of disease rather than protection.


Subject(s)
Dengue Virus/metabolism , Dengue/metabolism , Receptors, CCR1/metabolism , Receptors, CCR2/metabolism , Receptors, CCR4/metabolism , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/metabolism , Interferon-gamma/metabolism , Interleukin-6/metabolism , Ligands , Male , Mice , Mice, Inbred C57BL , Peroxidase/metabolism
16.
Nucleic Acids Res ; 35(11): 3551-60, 2007.
Article in English | MEDLINE | ID: mdl-17483519

ABSTRACT

The repair of DNA double-strand breaks (DSB) requires processing of the broken ends to complete the ligation process. Recently, it has been shown that DNA polymerase mu (polmu) and DNA polymerase lambda (pollambda) are both involved in such processing during non-homologous end joining in vitro. However, no phenotype was observed in animal models defective for either polmu and/or pollambda. Such observations could result from a functional redundancy shared by the X family of DNA polymerases. To avoid such redundancy and to clarify the role of polmu in the end joining process, we generated cells over-expressing the wild type as well as an inactive form of polmu (polmuD). We observed that cell sensitivity to ionizing radiation (IR) was increased when either polmu or polmuD was over-expressed. However, the genetic instability in response to IR increased only in cells expressing polmuD. Moreover, analysis of intrachromosomal repair of the I-SceI-induced DNA DSB, did not reveal any effect of either polmu or polmuD expression on the efficiency of ligation of both cohesive and partially complementary ends. Finally, the sequences of the repaired ends were specifically affected when polmu or polmuD was over-expressed, supporting the hypothesis that polmu could be involved in the repair of a DSB subset when resolution of junctions requires some gap filling.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Directed DNA Polymerase/physiology , Animals , Base Sequence , CHO Cells , Cell Line , Chromosome Aberrations , Cricetinae , Cricetulus , DNA/chemistry , Deoxyribonucleases, Type II Site-Specific/metabolism , Humans , Molecular Sequence Data , Radiation, Ionizing , Saccharomyces cerevisiae Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...