Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
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.
Sci Adv ; 6(28): eaaz8272, 2020 07.
Article in English | MEDLINE | ID: mdl-32695877

ABSTRACT

Fibrotic disorders represent common complex disease pathologies that are therapeutically challenging. Inflammation is associated with numerous fibrotic pathogeneses; however, its role in the multifaceted mechanisms of fibrosis remains unclear. IL-13 is implicated in aberrant responses involved in fibrotic disease, and we aimed to understand its role in the inflammatory processes of a common fibrotic disorder, Dupuytren's disease. We demonstrated T-cells produced IFN-g, which induced IL-13 secretion from mast cells and up-regulated IL-13Ra1 on fibroblasts, rendering them more reactive to IL-13. Consequently, diseased myofibroblasts demonstrated enhanced fibroproliferative effects upon IL-13 stimulation. We established IFN-g and IL-13 responses involved STAT dependent pathways, and STAT targeting (tofacitinib) could inhibit IL-13 production from mast cells, IL-13Ra1 up-regulation in fibroblasts and fibroproliferative effects of IL-13 on diseased myofibroblasts. Accordingly, utilizing Dupuytren's as an accessible human model of fibrosis, we propose targeting STAT pathways may offer previously unidentified therapeutic approaches in the management of fibrotic disease.

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.
Shock ; 52(5): e100-e106, 2019 11.
Article in English | MEDLINE | ID: mdl-30724784

ABSTRACT

Neutrophils and inflammatory monocytes control sepsis by migration to the site of infection via their chemokine receptors. CCR5 is a chemokine receptor that is not expressed on neutrophils and inflammatory monocytes under homeostatic conditions. However, it has been demonstrated that CCR5 can become expressed on these cells during different models of inflammation. In the present study, we investigated if CCR5 is also expressed on neutrophil and inflammatory monocytes during sepsis, exerting an important role in the migration of these cells to the infectious focus. Using cecal ligation and puncture model to induce polymicrobial sepsis, we demonstrated that the expression of CCR5 is induced on CD11bLy6GLy6C inflammatory monocytes, but not on neutrophils (CD11bLy6GLy6C). Furthermore, CCR5 plays an important role for the migration of the inflammatory monocytes to infection focus during sepsis. CCR5-expressing inflammatory monocytes migrate from the bone marrow to the circulation and then into the site of infection, where they phagocytize and kill the bacteria. Consequently, CCR5 mice showed increased systemic inflammatory response and mortality compared to wild-type mice. These data therefore demonstrate a hitherto unrecognized protective role of CCR5 in sepsis.


Subject(s)
Bone Marrow Cells/immunology , Cell Movement/immunology , Monocytes/immunology , Receptors, CCR5/immunology , Sepsis/immunology , Animals , Bone Marrow Cells/pathology , Cell Movement/genetics , Mice , Mice, Knockout , Monocytes/pathology , Receptors, CCR5/genetics , Sepsis/genetics , Sepsis/pathology
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.
Front Immunol ; 9: 962, 2018.
Article in English | MEDLINE | ID: mdl-29867945

ABSTRACT

The ST2 receptor is a member of the Toll/IL-1R superfamily and interleukin-33 (IL-33) is its agonist. Recently, it has been demonstrated that IL-33/ST2 axis plays key roles in inflammation and immune mediated diseases. Here, we investigated the effect of ST2 deficiency in Staphylococcus aureus-induced septic arthritis physiopathology. Synovial fluid samples from septic arthritis and osteoarthritis individuals were assessed regarding IL-33 and soluble (s) ST2 levels. The IL-33 levels in samples from synovial fluid were significantly increased, whereas no sST2 levels were detected in patients with septic arthritis when compared with osteoarthritis individuals. The intra-articular injection of 1 × 107 colony-forming unity/10 µl of S. aureus American Type Culture Collection 6538 in wild-type (WT) mice induced IL-33 and sST2 production with a profile resembling the observation in the synovial fluid of septic arthritis patients. Data using WT, and ST2 deficient (-/-) and interferon-γ (IFN-γ)-/- mice showed that ST2 deficiency shifts the immune balance toward a type 1 immune response that contributes to eliminating the infection due to enhanced microbicide effect via NO production by neutrophils and macrophages. In fact, the treatment of ST2-/- bone marrow-derived macrophage cells with anti-IFN-γ abrogates the beneficial phenotype in the absence of ST2, which confirms that ST2 deficiency leads to IFN-γ expression and boosts the bacterial killing activity of macrophages against S. aureus. In agreement, WT cells achieved similar immune response to ST2 deficiency by IFN-γ treatment. The present results unveil a previously unrecognized beneficial effect of ST2 deficiency in S. aureus-induced septic arthritis.


Subject(s)
Arthritis, Infectious/immunology , Arthritis, Infectious/microbiology , Interleukin-1 Receptor-Like 1 Protein/genetics , Staphylococcal Infections/immunology , Synovial Fluid/immunology , Animals , Female , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-33/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis, Knee/immunology , Staphylococcus aureus
7.
Arthritis Res Ther ; 20(1): 119, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29884199

ABSTRACT

BACKGROUND: Epidemiologic studies have highlighted the association of environmental factors with the development and progression of autoimmune and chronic inflammatory diseases. Among the environmental factors, smoking has been associated with increased susceptibility and poor prognosis in rheumatoid arthritis (RA). However, the immune and molecular mechanism of smoking-induced arthritis aggravation remains unclear. The transcription factor aryl hydrocarbon receptor (AHR) regulates the generation of Th17 cells, CD4 T cells linked the development of autoimmune diseases. AHR is activated by organic compounds including polycyclic aromatic hydrocarbons (PAHs), which are environmental pollutants that are also present in cigarette smoke. In this study, we investigated the role of AHR activation in the aggravation of experiment arthritis induced by exposure to cigarette smoke. METHODS: Mice were exposed to cigarette smoke during the developmental phase of antigen-induced arthritis and collagen-induced arthritis to evaluate the effects of smoking on disease development. Aggravation of articular inflammation was assessed by measuring neutrophil migration to the joints, increase in articular hyperalgesia and changes in the frequencies of Th17 cells. In vitro studies were performed to evaluate the direct effects of cigarette smoke and PAH on Th17 differentiation. We also used mice genetically deficient for AHR (Ahr KO) and IL-17Ra (Il17ra KO) to determine the in vivo mechanism of smoking-induced arthritis aggravation. RESULTS: We found that smoking induces arthritis aggravation and increase in the frequencies of Th17 cells. The absence of IL-17 signaling (Il17ra KO) conferred protection to smoking-induced arthritis aggravation. Moreover, in vitro experiments showed that cigarette smoke can directly increase Th17 differentiation of T cells by inducing AHR activation. Indeed, Ahr KO mice were protected from cigarette smoke-induced arthritis aggravation and did not display increase in TH17 frequencies, suggesting that AHR activation is an important mechanism for cigarette smoke effects on arthritis. Finally, we demonstrate that PAHs are also able to induce arthritis aggravation. CONCLUSIONS: Our data demonstrate that the disease-exacerbating effects of cigarette smoking are AHR dependent and environmental pollutants with AHR agonist activity can induce arthritis aggravation by directly enhancing Th17 cell development.


Subject(s)
Arthritis, Experimental/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Smoke/adverse effects , Th17 Cells/metabolism , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/genetics , Azo Compounds/pharmacology , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Pyrazoles/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Receptors, Aryl Hydrocarbon/genetics , Receptors, Interleukin-17/genetics , Receptors, Interleukin-17/metabolism , Nicotiana/chemistry
8.
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
9.
Shock ; 49(6): 682-689, 2018 06.
Article in English | MEDLINE | ID: mdl-29589840

ABSTRACT

Sepsis is a systemic inflammatory response as a result of uncontrolled infections. Neutrophils are the first cells to reach the primary sites of infection, and chemokines play a key role in recruiting neutrophils. However, in sepsis chemokines could also contribute to neutrophil infiltration to vital organs leading to multiple organ failure. ACKR2 is an atypical chemokine receptor, which can remove and degrade inflammatory CC chemokines. The role of ACK2 in sepsis is unknown. Using a model of cecal ligation and puncture (CLP), we demonstrate here that ACKR2 deficient () mice exhibited a significant reduction in the survival rate compared with similarly treated wild-type (WT) mice. However, neutrophil migration to the peritoneal cavity and bacterial load were similar between WT and ACKR2 mice during CLP. In contrast, ACKR2 mice showed increased neutrophil infiltration and elevated CC chemokine levels in the lung, kidney, and heart compared with the WT mice. In addition, ACKR2 mice also showed more severe lesions in the lung and kidney than those in the WT mice. Consistent with these results, WT mice under nonsevere sepsis (90% survival) had higher expression of ACKR2 in these organs than mice under severe sepsis (no survival). Finally, the lungs from septic patients showed increased number of ACKR2 cells compared with those of nonseptic patients. Our data indicate that ACKR2 may have a protective role during sepsis, and the absence of ACKR2 leads to exacerbated chemokine accumulation, neutrophil infiltration, and damage to vital organs.


Subject(s)
Multiple Organ Failure/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Receptors, Chemokine/metabolism , Sepsis/metabolism , Animals , Disease Models, Animal , Female , Male , Mice , Multiple Organ Failure/pathology , Neutrophils/pathology , Sepsis/pathology
10.
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
11.
Nat Commun ; 8: 14919, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28374774

ABSTRACT

Patients who survive sepsis can develop long-term immune dysfunction, with expansion of the regulatory T (Treg) cell population. However, how Treg cells proliferate in these patients is not clear. Here we show that IL-33 has a major function in the induction of this immunosuppression. Mice deficient in ST2 (IL-33R) develop attenuated immunosuppression in cases that survive sepsis, whereas treatment of naive wild-type mice with IL-33 induces immunosuppression. IL-33, released during tissue injury in sepsis, activates type 2 innate lymphoid cells, which promote polarization of M2 macrophages, thereby enhancing expansion of the Treg cell population via IL-10. Moreover, sepsis-surviving patients have more Treg cells, IL-33 and IL-10 in their peripheral blood. Our study suggests that targeting IL-33 may be an effective treatment for sepsis-induced immunosuppression.


Subject(s)
Immune Tolerance/immunology , Interleukin-33/immunology , Sepsis/immunology , T-Lymphocytes, Regulatory/immunology , Aged , Animals , Female , Humans , Immune Tolerance/genetics , Interleukin-1 Receptor-Like 1 Protein/deficiency , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-33/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Sepsis/genetics , Sepsis/metabolism , T-Lymphocytes, Regulatory/metabolism
12.
JCI Insight ; 2(7): e91868, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28405622

ABSTRACT

In each influenza season, a distinct group of young, otherwise healthy individuals with no risk factors succumbs to life-threatening infection. To better understand the cause for this, we analyzed a broad range of immune responses in blood from a unique cohort of patients, comprising previously healthy individuals hospitalized with and without respiratory failure during one influenza season, and infected with one specific influenza A strain. This analysis was compared with similarly hospitalized influenza patients with known risk factors (total of n = 60 patients recruited). We found a sustained increase in a specific subset of proinflammatory monocytes, with high TNF-α expression and an M1-like phenotype (independent of viral titers), in these previously healthy patients with severe disease. The relationship between M1-like monocytes and immunopathology was strengthened using murine models of influenza, in which severe infection generated using different models (including the high-pathogenicity H5N1 strain) was also accompanied by high levels of circulating M1-like monocytes. Additionally, a raised M1/M2 macrophage ratio in the lungs was observed. These studies identify a specific subtype of monocytes as a modifiable immunological determinant of disease severity in this subgroup of severely ill, previously healthy patients, offering potential novel therapeutic avenues.


Subject(s)
Influenza, Human/immunology , Macrophages/immunology , Monocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Adult , Aged , Animals , Female , Humans , Influenza A Virus, H5N1 Subtype , Influenza, Human/pathology , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype , Viral Load , Young Adult
13.
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
14.
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
15.
Nat Commun ; 6: 6774, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25857925

ABSTRACT

MicroRNA (miRNA) has the potential for cross-regulation and functional integration of discrete biological processes during complex physiological events. Utilizing the common human condition tendinopathy as a model system to explore the cross-regulation of immediate inflammation and matrix synthesis by miRNA we observed that elevated IL-33 expression is a characteristic of early tendinopathy. Using in vitro tenocyte cultures and in vivo models of tendon damage, we demonstrate that such IL-33 expression plays a pivotal role in the transition from type 1 to type 3 collagen (Col3) synthesis and thus early tendon remodelling. Both IL-33 effector function, via its decoy receptor sST2, and Col3 synthesis are regulated by miRNA29a. Downregulation of miRNA29a in human tenocytes is sufficient to induce an increase in Col3 expression. These data provide a molecular mechanism of miRNA-mediated integration of the early pathophysiologic events that facilitate tissue remodelling in human tendon after injury.


Subject(s)
Fibroblasts/metabolism , Interleukin-33/genetics , MicroRNAs/genetics , Tendinopathy/genetics , Tendons/metabolism , Animals , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation , Genes, Reporter , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-1beta/pharmacology , Interleukin-33/metabolism , Luciferases/genetics , Luciferases/metabolism , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Tendinopathy/metabolism , Tendinopathy/pathology , Tendons/drug effects , Tendons/pathology , Tumor Necrosis Factor-alpha/pharmacology
16.
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
17.
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
18.
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
19.
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
20.
J Autoimmun ; 50: 107-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24491821

ABSTRACT

Regulatory B cells (Breg) have attracted increasing attention for their roles in maintaining peripheral tolerance. Interleukin 33 (IL-33) is a recently identified IL-1 family member, which leads a double-life with both pro- and anti-inflammatory properties. We report here that peritoneal injection of IL-33 exacerbated inflammatory bowel disease in IL-10-deficient (IL-10(-/-)) mice, whereas IL-33-treated IL-10-sufficient (wild type) mice were protected from the disease induction. A phenotypically unconventional subset(s) (CD19(+)CD25(+)CD1d(hi)IgM(hi)CD5(-)CD23(-)Tim-1(-)) of IL-10 producing Breg-like cells (Breg(IL-33)) was identified responsible for the protection. We demonstrated further that Breg(IL-33) isolated from these mice could suppress immune effector cell expansion and functions and, upon adoptive transfer, effectively blocked the development of spontaneous colitis in IL-10(-/-) mice. Our findings indicate an essential protective role, hence therapeutic potential, of Breg(IL-33) against mucosal inflammatory disorders in the gut.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Colitis/immunology , Gastric Mucosa/drug effects , Interleukin-10/immunology , Interleukins/pharmacology , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/transplantation , Colitis/genetics , Colitis/pathology , Female , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gene Expression , Injections, Intraperitoneal , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-33 , Interleukins/immunology , Lymphocyte Activation , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...