Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33443169

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by joint destruction and severe morbidity. Cigarette smoking (CS) can exacerbate the incidence and severity of RA. Although Th17 cells and the Aryl hydrocarbon receptor (AhR) have been implicated, the mechanism by which CS induces RA development remains unclear. Here, using transcriptomic analysis, we show that microRNA-132 is specifically induced in Th17 cells in the presence of either AhR agonist or CS-enriched medium. miRNA-132 thus induced is packaged into extracellular vesicles produced by Th17 and acts as a proinflammatory mediator increasing osteoclastogenesis through the down-regulation of COX2. In vivo, articular knockdown of miR-132 in murine arthritis models reduces the number of osteoclasts in the joints. Clinically, RA patients express higher levels of miR-132 than do healthy individuals. This increase is further elevated by cigarette smoking. Together, these results reveal a hitherto unrecognized mechanism by which CS could exacerbate RA and further advance understanding of the impact of environmental factors on the pathogenesis of chronic inflammatory diseases.


Subject(s)
Arthritis, Rheumatoid/genetics , MicroRNAs/genetics , Osteogenesis/physiology , Adult , Aged , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cigarette Smoking/adverse effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Middle Aged , Osteoclasts/metabolism , Osteogenesis/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Smoke , Th17 Cells/drug effects , Th17 Cells/metabolism , Tobacco Smoke Pollution/adverse effects
2.
Proc Natl Acad Sci U S A ; 115(28): 7404-7409, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29954866

ABSTRACT

Cerebral malaria (CM) is a serious neurological complication caused by Plasmodium falciparum infection. Currently, the only treatment for CM is the provision of antimalarial drugs; however, such treatment by itself often fails to prevent death or development of neurological sequelae. To identify potential improved treatments for CM, we performed a nonbiased whole-brain transcriptomic time-course analysis of antimalarial drug chemotherapy of murine experimental CM (ECM). Bioinformatics analyses revealed IL33 as a critical regulator of neuroinflammation and cerebral pathology that is down-regulated in the brain during fatal ECM and in the acute period following treatment of ECM. Consistent with this, administration of IL33 alongside antimalarial drugs significantly improved the treatment success of established ECM. Mechanistically, IL33 treatment reduced inflammasome activation and IL1ß production in microglia and intracerebral monocytes in the acute recovery period following treatment of ECM. Moreover, treatment with the NLRP3-inflammasome inhibitor MCC950 alongside antimalarial drugs phenocopied the protective effect of IL33 therapy in improving the recovery from established ECM. We further showed that IL1ß release from macrophages was stimulated by hemozoin and antimalarial drugs and that this was inhibited by MCC950. Our results therefore demonstrate that manipulation of the IL33-NLRP3 axis may be an effective therapy to suppress neuroinflammation and improve the efficacy of antimalarial drug treatment of CM.


Subject(s)
Antimalarials/pharmacology , Brain/parasitology , Drug Delivery Systems/methods , Interleukin-33/metabolism , Malaria, Cerebral/drug therapy , Malaria, Falciparum/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plasmodium falciparum/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Hemeproteins/metabolism , Interleukin-1beta/biosynthesis , Interleukin-33/antagonists & inhibitors , Macrophages/metabolism , Macrophages/pathology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Transcriptome/drug effects
3.
Crit Care ; 23(1): 113, 2019 Apr 08.
Article in English | MEDLINE | ID: mdl-30961634

ABSTRACT

BACKGROUND: Neutrophil extracellular traps (NETs) are innate defense mechanisms that are also implicated in the pathogenesis of organ dysfunction. However, the role of NETs in pediatric sepsis is unknown. METHODS: Infant (2 weeks old) and adult (6 weeks old) mice were submitted to sepsis by intraperitoneal (i.p.) injection of bacteria suspension or lipopolysaccharide (LPS). Neutrophil infiltration, bacteremia, organ injury, and concentrations of cytokine, NETs, and DNase in the plasma were measured. Production of reactive oxygen and nitrogen species and release of NETs by neutrophils were also evaluated. To investigate the functional role of NETs, mice undergoing sepsis were treated with antibiotic plus rhDNase and the survival, organ injury, and levels of inflammatory markers and NETs were determined. Blood samples from pediatric and adult sepsis patients were collected and the concentrations of NETs measured. RESULTS: Infant C57BL/6 mice subjected to sepsis or LPS-induced endotoxemia produced significantly higher levels of NETs than the adult mice. Moreover, compared to that of the adult mice, this outcome was accompanied by increased organ injury and production of inflammatory cytokines. The increased NETs were associated with elevated expression of Padi4 and histone H3 citrullination in the neutrophils. Furthermore, treatment of infant septic mice with rhDNase or a PAD-4 inhibitor markedly attenuated sepsis. Importantly, pediatric septic patients had high levels of NETs, and the severity of pediatric sepsis was positively correlated with the level of NETs. CONCLUSION: This study reveals a hitherto unrecognized mechanism of pediatric sepsis susceptibility and suggests that NETs represents a potential target to improve clinical outcomes of sepsis.


Subject(s)
Extracellular Traps/microbiology , Sepsis/therapy , Animals , Bacterial Load/methods , Brazil , Disease Models, Animal , Mice , Mice, Inbred C57BL/blood , Mice, Inbred C57BL/microbiology , Multiple Organ Failure/etiology , Multiple Organ Failure/pathology , Sepsis/mortality , Sepsis/pathology
4.
Proc Natl Acad Sci U S A ; 113(19): E2705-13, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27091974

ABSTRACT

Alzheimer's disease (AD) is a devastating condition with no known effective treatment. AD is characterized by memory loss as well as impaired locomotor ability, reasoning, and judgment. Emerging evidence suggests that the innate immune response plays a major role in the pathogenesis of AD. In AD, the accumulation of ß-amyloid (Aß) in the brain perturbs physiological functions of the brain, including synaptic and neuronal dysfunction, microglial activation, and neuronal loss. Serum levels of soluble ST2 (sST2), a decoy receptor for interleukin (IL)-33, increase in patients with mild cognitive impairment, suggesting that impaired IL-33/ST2 signaling may contribute to the pathogenesis of AD. Therefore, we investigated the potential therapeutic role of IL-33 in AD, using transgenic mouse models. Here we report that IL-33 administration reverses synaptic plasticity impairment and memory deficits in APP/PS1 mice. IL-33 administration reduces soluble Aß levels and amyloid plaque deposition by promoting the recruitment and Aß phagocytic activity of microglia; this is mediated by ST2/p38 signaling activation. Furthermore, IL-33 injection modulates the innate immune response by polarizing microglia/macrophages toward an antiinflammatory phenotype and reducing the expression of proinflammatory genes, including IL-1ß, IL-6, and NLRP3, in the cortices of APP/PS1 mice. Collectively, our results demonstrate a potential therapeutic role for IL-33 in AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Brain/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Interleukin-33/administration & dosage , Alzheimer Disease/diagnosis , Animals , Brain/drug effects , Cognition Disorders/diagnosis , Cytokines/metabolism , Female , Male , Mice , Mice, Transgenic , Neuroprotective Agents/administration & dosage , Treatment Outcome
5.
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
6.
J Autoimmun ; 90: 49-58, 2018 06.
Article in English | MEDLINE | ID: mdl-29426578

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune arthropathy characterized by chronic articular inflammation. Methotrexate (MTX) remains the first-line therapy for RA and its anti-inflammatory effect is associated with the maintenance of high levels of extracellular adenosine (ADO). Nonetheless, up to 40% of RA patients are resistant to MTX treatment and this is linked to a reduction of CD39 expression, an ectoenzyme involved in the generation of extracellular ADO by ATP metabolism, on circulating regulatory T cells (Tregs). However, the mechanism mediating the reduction of CD39 expression on Tregs is unknown. Here we demonstrated that the impairment in TGF-ß signalling lead to the reduction of CD39 expression on Tregs that accounts for MTX resistance. TGF-ß increases CD39 expression on Tregs via the activation of TGFBRII/TGFBRI, SMAD2 and the transcription factor CREB, which is activated in a p38-dependent manner and induces CD39 expression by promoting ENTPD1 gene transcription. Importantly, unresponsive patients to MTX (UR-MTX) show reduced expression of TGFBR2 and CREB1 and decreased levels of p-SMAD2 and p-CREB in Tregs compared to MTX-responsive patients (R-MTX). Furthermore, RA patients carrying at least one mutant allele for rs1431131 (AT or AA) of the TGFBR2 gene are significantly (p = 0.0006) associated with UR-MTX. Therefore, we have uncovered a molecular mechanism for the reduced CD39 expression on Tregs, and revealed potential targets for therapeutic intervention for MTX resistance.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Arthritis, Rheumatoid/immunology , Receptor, Transforming Growth Factor-beta Type II/genetics , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/metabolism , Adenosine Triphosphate/metabolism , Adult , Aged , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Resistance , Female , Gene Expression Regulation , Gene Frequency , Humans , Male , Methotrexate/therapeutic use , Middle Aged , Polymorphism, Single Nucleotide , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/genetics , Smad2 Protein/metabolism
7.
Proc Natl Acad Sci U S A ; 112(8): 2509-14, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675517

ABSTRACT

Rheumatoid arthritis (RA) is an inflammatory autoimmune disease characterized by joint destruction and severe morbidity. Methotrexate (MTX) is the standard first-line therapy of RA. However, about 40% of RA patients are unresponsive to MTX treatment. Regulatory T cells (Tregs, CD4(+)CD25(+)FoxP3(+)) are thought to play an important role in attenuating RA. To investigate the role of Tregs in MTX resistance, we recruited 122 RA patients (53 responsive, R-MTX; 69 unresponsive, UR-MTX) and 33 healthy controls. Three months after MTX treatment, R-MTX but not UR-MTX showed higher frequency of peripheral blood CD39(+)CD4(+)CD25(+)FoxP3(+) Tregs than the healthy controls. Tregs produce adenosine (ADO) through ATP degradation by sequential actions of two cell surface ectonucleotidases: CD39 and CD73. Tregs from UR-MTX expressed a lower density of CD39, produced less ADO, and had reduced suppressive activity than Tregs from R-MTX. In a prospective study, before MTX treatment, UR-MTX expressed a lower density of CD39 on Tregs than those of R-MTX or control (P < 0.01). In a murine model of arthritis, CD39 blockade reversed the antiarthritic effects of MTX treatment. Our results demonstrate that MTX unresponsiveness in RA is associated with low expression of CD39 on Tregs and the decreased suppressive activity of these cells through reduced ADO production. Our findings thus provide hitherto unrecognized mechanism of immune regulation in RA and on mode of action of MTX. Furthermore, our data suggest that low expression of CD39 on Tregs could be a noninvasive biomarker for identifying MTX-resistant RA patients.


Subject(s)
Antigens, CD/metabolism , Apyrase/metabolism , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Drug Resistance/immunology , Methotrexate/therapeutic use , T-Lymphocytes, Regulatory/immunology , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Biomarkers/metabolism , Drug Resistance/drug effects , Humans , Lymphocyte Count , Methotrexate/pharmacology , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects , Th1 Cells/immunology , Th17 Cells/immunology
8.
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
9.
FASEB J ; 30(1): 54-65, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26310268

ABSTRACT

Neuropathic pain from injury to the peripheral and CNS represents a major health care issue. We have investigated the role of IL-33/IL-33 receptor (ST2) signaling in experimental models of neuropathic pain in mice. Chronic constriction injury (CCI) of the sciatic nerve induced IL-33 production in the spinal cord. IL-33/citrine reporter mice revealed that oligodendrocytes are the main cells expressing IL-33 within the spinal cord together with a minor expression by neurons, microglia. and astrocytes. CCI-induced mechanical hyperalgesia was reduced in IL-33R (ST2)(-/ -) mice compared with wild-type (WT) mice. Intrathecal treatment of WT mice with soluble IL-33 receptor (IL-33 decoy receptor) markedly reduced CCI-induced hyperalgesia. Consistent with these observations, intrathecal injection of IL-33 enhanced CCI hyperalgesia and induced hyperalgesia in naive mice. IL-33-mediated hyperalgesia during CCI was dependent on a reciprocal relationship with TNF-α and IL-1ß. IL-33-induced hyperalgesia was markedly attenuated by inhibitors of PI3K, mammalian target of rapamycin, MAPKs (p38, ERK, and JNK), NF-κB, and also by the inhibitors of glial cells (microglia and astrocytes). Furthermore, targeting these signaling pathways and cells inhibited IL-33-induced TNF-α and IL-1ß production in the spinal cord. Our study, therefore, reveals an important role of oligodendrocyte-derived IL-33 in neuropathic pain.


Subject(s)
Alarmins/metabolism , Hyperalgesia/metabolism , Interleukin-33/metabolism , Neuralgia/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , Animals , Astrocytes/metabolism , Mice, Knockout , Microglia/metabolism , Pain Threshold/physiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Spinal Cord/physiopathology
10.
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
SELECTION OF CITATIONS
SEARCH DETAIL