Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Front Immunol ; 14: 1215364, 2023.
Article in English | MEDLINE | ID: mdl-37415987

ABSTRACT

Background: Interleukin-18 is a proinflammatory cytokine, the activity of which is regulated by its natural inhibitor, IL-18 binding protein (IL-18BP). Elevated circulating levels of IL-18 have been observed in patients with systemic juvenile idiopathic arthritis (sJIA) and adult-onset Still's disease (AOSD), two conditions associated with dysregulated innate immune responses. This study examines the expression and function of IL-18 and IL-18BP in K/BxN serum transfer arthritis (STA), a model that is uniquely dependent on innate immune responses. Methods: Naïve and serum transfer-induced arthritis (STA) wild-type (WT) mice were used to examine the articular levels of IL-18 and IL-18BP mRNA by RT-qPCR. The cellular sources of IL-18BP in the joints were determined by using Il18bp-tdTomato reporter knock-in mice. The incidence and severity of arthritis, including mRNA levels of different cytokines, were compared in IL-18BP or IL-18 knock-out (KO) mice and their WT littermates. Results: IL-18 and IL-18BP mRNA levels were significantly increased in arthritic as compared to normal joints. Synovial neutrophils, macrophages, and endothelial cells represented the cellular sources of IL-18BP in arthritic joints, whereas IL-18BP production was limited to endothelial cells in non-inflamed joints. The incidence and severity of arthritis were similar in IL-18BP KO and IL-18 KO compared to their WT littermates. Transcript levels of different inflammatory cytokines were not different in the two KO mouse lines compared to WT mice. Conclusion: Although IL-18 and IL-18BP levels were increased in arthritic joints, our results show that the IL-18/IL-18BP balance is not involved in the regulation of STA.


Subject(s)
Interleukin-18 , Still's Disease, Adult-Onset , Animals , Mice , Interleukin-18/genetics , Interleukin-18/metabolism , Endothelial Cells/metabolism , Cytokines , RNA, Messenger
2.
Methods Mol Biol ; 2691: 263-277, 2023.
Article in English | MEDLINE | ID: mdl-37355553

ABSTRACT

The interleukin (IL)-18 cytokine plays an important driver role in a range of autoimmune and inflammatory diseases, as well as cancer. IL-18 is a potent inducer of interferon gamma (IFN-γ), and the bioactivity of IL-18 is regulated by its natural soluble inhibitor, IL-18-binding protein (IL-18BP), which is present at high concentrations in the circulation. Many cell types have been described to secrete IL-18BP, constitutively or under the influence of IFN-γ, thus generating a negative feedback loop for IL-18. Therefore, solely measuring total IL-18 protein levels does not allow to evaluate its biological activity, especially in the context of systemic inflammatory diseases or other circumstances where IL-18BP is present (e.g., samples containing plasma, cells constitutively expressing IL-18BP). Considering there is a critical need to accurately measure the protein levels of both mature, biologically active IL-18 and IL-18BP as biomarkers of disease activity in patients and also stratification for potential anti-IL-18 therapy, in this chapter we provide the latest techniques to measure mature, free, and bioactive IL-18 and IL-18BP in different samples.


Subject(s)
Cytokines , Interferon-gamma , Humans
3.
J Immunol ; 210(11): 1790-1803, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37074208

ABSTRACT

IL-18 is a pleiotropic immunoregulatory cytokine of the IL-1 family. IL-18 has been identified as a potent IFN-γ inducer in synergy with IL-12 and IL-15 and thus as a powerful Th1 cell-polarizing cytokine. IL-18 activity is regulated by its naturally occurring soluble inhibitor IL-18 binding protein (IL-18BP), the production of which is stimulated by IFN-γ in a negative feedback loop. Circulating levels of IL-18BP are elevated, and unbound bioactive free IL-18 is thus not detectable in the circulation in physiologic conditions. However, emerging evidence indicates that the IL-18/IL-18BP balance could be dysregulated in macrophage activation syndrome (MAS), as mirrored by the presence of free IL-18 in the circulation of patients with MAS. Herein, we sought to identify IL-18BP-producing cells in a murine CpG-induced MAS model using IL-18BP knock-in tdTomato reporter mice. Endothelial cells, tissue-resident macrophages, and neutrophils appeared as major cellular sources of IL-18BP. We also identified extramedullary and medullary early erythroid progenitors as IL-18BP-producing cells in an IFN-γ-dependent manner. This finding suggests a novel regulation of IL-18 activity by erythroid precursors, which are likely involved in the prevention of the negative effects of IL-18 on erythropoiesis. Indeed, coherent in vivo and in vitro results indicate that IL-18 indirectly impairs erythropoiesis while favoring myelopoiesis and thus contributes to anemia associated with MAS and potentially with other IL-18-driven inflammatory diseases. In conclusion, IL-18BP production by endothelial cells, neutrophils, macrophages, and erythroid precursors attenuates the anemia associated with murine CpG-induced MAS.


Subject(s)
Anemia , Macrophage Activation Syndrome , Animals , Mice , Carrier Proteins , Cytokines/metabolism , Endothelial Cells/metabolism , Interleukin-18/metabolism
4.
Immunol Cell Biol ; 101(5): 444-457, 2023 05.
Article in English | MEDLINE | ID: mdl-36967659

ABSTRACT

Helicobacter pylori (H. pylori) infection can trigger chronic gastric inflammation perpetuated by overactivation of the innate immune system, leading to a cascade of precancerous lesions culminating in gastric cancer. However, key regulators of innate immunity that promote H. pylori-induced gastric pathology remain ill-defined. The innate immune cytosolic DNA sensor absent in melanoma 2 (AIM2) contributes to the pathogenesis of numerous autoimmune and chronic inflammatory diseases, as well as cancers including gastric cancer. We therefore investigated whether AIM2 contributed to the pathogenesis of Helicobacter-induced gastric disease. Here, we reveal that AIM2 messenger RNA and protein expression levels are elevated in H. pylori-positive versus H. pylori-negative human gastric biopsies. Similarly, chronic Helicobacter felis infection in wild-type mice augmented Aim2 gene expression levels compared with uninfected controls. Notably, gastric inflammation and hyperplasia were less severe in H. felis-infected Aim2-/- versus wild-type mice, evidenced by reductions in gastric immune cell infiltrates, mucosal thickness and proinflammatory cytokine and chemokine release. In addition, H. felis-driven proliferation and apoptosis in both gastric epithelial and immune cells were largely attenuated in Aim2-/- stomachs. These observations in Aim2-/- mouse stomachs correlated with decreased levels of inflammasome activity (caspase-1 cleavage) and the mature inflammasome effector cytokine, interleukin-1ß. Taken together, this work uncovers a pathogenic role for the AIM2 inflammasome in Helicobacter-induced gastric disease, and furthers our understanding of the host immune response to a common pathogen and the complex and varying roles of AIM2 at different stages of cancerous and precancerous gastric disease.


Subject(s)
Felis , Helicobacter , Precancerous Conditions , Stomach Neoplasms , Animals , Humans , Mice , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Felis/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Helicobacter/metabolism , Inflammasomes/metabolism , Inflammation/pathology , Precancerous Conditions/pathology
5.
Front Immunol ; 14: 1117742, 2023.
Article in English | MEDLINE | ID: mdl-36875067

ABSTRACT

Background: The pathogenesis of rheumatoid arthritis (RA) is believed to initiate at mucosal sites. The so-called 'mucosal origin hypothesis of RA' postulates an increased intestinal permeability before disease onset. Several biomarkers, including lipopolysaccharide binding protein (LBP) and intestinal fatty acid binding protein (I-FABP), have been proposed to reflect gut mucosa permeability and integrity, while serum calprotectin is a new inflammation marker proposed in RA. Methods: We analyzed serum samples of individuals genetically at increased risk of RA in a nested-case-control study. Participants from a longitudinal cohort of first-degree relatives of RA patients (SCREEN-RA cohort) were divided into three pre-clinical stages of RA, based on the presence of risk factors for subsequent RA onset: 1) low-risk healthy asymptomatic controls; 2) intermediate-risk individuals without symptoms, but with RA-associated auto-immunity; 3) high-risk individuals with clinically suspect arthralgias. Five patients with newly diagnosed RA were also sampled. Serum LBP, I-FABP and calprotectin were measured using commercially available ELISA kits. Results: We included 180 individuals genetically at increased risk for RA: 84 asymptomatic controls, 53 individuals with RA-associated autoimmunity and 38 high risk individuals. Serum LBP, I-FAPB or calprotectin concentrations did not differ between individuals in different pre-clinical stages of RA. Conclusion: Based on the serum biomarkers LBP, I-FABP and calprotectin, we could not detect any evidence for intestinal injury in pre-clinical stages of RA.


Subject(s)
Arthritis, Rheumatoid , Humans , Case-Control Studies , Risk Factors , Biomarkers , Leukocyte L1 Antigen Complex
6.
J Immunol ; 205(4): 1167-1175, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32651219

ABSTRACT

IL-18 binding protein (IL-18BP) acts as a naturally occurring IL-18 decoy receptor. If the balance between IL-18 and IL-18BP is dysregulated, abnormal levels of free bioactive IL-18 are detected, such as in the sera of Il-18bp knockout (KO) mice with CpG-induced macrophage activation syndrome. To determine the cellular sources of Il-18bp in vivo, we selectively depleted Il-18bp expression in either radiosensitive or radioresistant cells using bone marrow transfer between wild-type (WT) and Il-18bp KO mice. Following repeated CpG injections, Il-18bp KO (donor)→ Il-18bp KO (recipient) chimeric mice exhibited more severe disease, with an enhanced Ifn-γ signature and circulating free Il-18 levels, in comparison with WT→WT chimeras. Interestingly, the phenotype of KO→WT and WT→KO mice did not differ from that of WT→WT mice. Consistent with this finding, serum Il-18bp levels were similar in these three groups of mice. The contribution of radioresistant and radiosensitive cells to Il-18bp production varied markedly according to the organ examined, with a major contribution of radiosensitive cells in the spleen as opposed to a major contribution of radioresistant cells in the lung. Finally, Ifn-γ blockade abrogated the CpG-induced but not the constitutive Il-18bp production. Our results demonstrate that circulating Il-18bp is induced in response to Ifn-γ during CpG-induced macrophage activation syndrome and is present at high levels in the circulation to prevent the deleterious systemic effects of Il-18.


Subject(s)
Carrier Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18/metabolism , Macrophage Activation Syndrome/metabolism , Animals , Female , Interferon-gamma/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/metabolism
7.
Life Sci Alliance ; 3(6)2020 06.
Article in English | MEDLINE | ID: mdl-32345660

ABSTRACT

IL-36R signaling plays an important role in the pathogenesis of psoriasis. We ought to assess the specific function of IL-36R in keratinocytes for the pathology of Aldara-induced psoriasis-like dermatitis. Il36r ΔK mice presenting deletion of IL-36R in keratinocytes were similarly resistant to Aldara-induced ear inflammation as Il36r -/- mice, but acanthosis was only prevented in Il36r -/- mice. FACS analysis revealed that IL-36R signaling in keratinocytes is mandatory for early neutrophil infiltration in Aldara-treated ears. RNASeq and qRT-PCR experiments demonstrated the crucial role of IL-36R signaling in keratinocytes for induction of IL-23, IL-17, and IL-22 at early time points. Taken together, our results demonstrate that IL-36R signaling in keratinocytes plays a major role in the induction of Aldara-induced psoriasis-like dermatitis by triggering early production of IL-23/IL-17/IL-22 cytokines and neutrophil infiltration.


Subject(s)
Drug Eruptions/immunology , Interleukin-23/biosynthesis , Keratinocytes/immunology , Otitis Externa/immunology , Psoriasis/immunology , Receptors, Interleukin-1/deficiency , Signal Transduction/genetics , Administration, Cutaneous , Animals , Drug Eruptions/etiology , Drug Eruptions/metabolism , Female , Gene Deletion , Imiquimod/administration & dosage , Imiquimod/adverse effects , Interleukin-17/biosynthesis , Interleukins/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Otitis Externa/chemically induced , Psoriasis/chemically induced , Receptors, Interleukin-1/genetics , Signal Transduction/immunology , Interleukin-22
8.
J Immunol ; 204(4): 967-979, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31932497

ABSTRACT

The inflammatory effects of IL-1α/ß are controlled by IL-1R antagonist (IL-1Ra). One IL-1Ra isoform is secreted, whereas three other isoforms (intracellular IL-1Ra [icIL-1Ra] 1, 2, and 3) are supposed to remain intracellular because of the absence of a signal peptide. In contrast to the well-characterized function of the secreted isoform, the biological role of the intracellular isoforms remains largely unclear. icIL-1Ra1 represents the major isoform in keratinocytes. We created icIL-1Ra1-/- mice and investigated the role of icIL-1Ra1 in Aldara (5% imiquimod)-induced psoriasis-like skin inflammation. Naive icIL-1Ra1-/- mice bred habitually and exhibited a normal phenotype. icIL-1Ra1 deficiency aggravated Aldara-induced skin inflammation, as demonstrated by increased ear thickness and increased mRNA levels of key proinflammatory cytokines. No intracellular effect of icIL-1Ra1 could be detected in isolated keratinocytes using RNA-sequencing analysis; however, Aldara treatment led to caspase 1/11-, caspase 8-, and RIPK3-independent keratinocyte cell death accompanied by the release of both icIL-1Ra1 and IL-1α. Furthermore, blocking IL-1α attenuated the clinical severity of Aldara-induced ear thickening in icIL-1Ra1-/- mice. Our data suggest that upon keratinocyte damage icIL-1Ra1 acts extracellularly as an antagonist of the alarmin IL-1α to immediately counteract its inflammatory effects.


Subject(s)
Alarmins/antagonists & inhibitors , Apoptosis/immunology , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/antagonists & inhibitors , Psoriasis/immunology , Alarmins/immunology , Alarmins/metabolism , Animals , Disease Models, Animal , Female , Humans , Imiquimod/immunology , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Keratinocytes/metabolism , Male , Mice , Mice, Knockout , Protein Isoforms/immunology , Protein Isoforms/metabolism , Psoriasis/diagnosis , Psoriasis/pathology , Severity of Illness Index , Signal Transduction/immunology , Skin/cytology , Skin/immunology , Skin/pathology
9.
PLoS One ; 13(3): e0194667, 2018.
Article in English | MEDLINE | ID: mdl-29554104

ABSTRACT

The IL-1 cytokine family includes eleven members, among which Il-36α, ß and γ, IL-36Ra and IL-38. The IL-36 cytokines are involved in the pathogenesis of psoriasis. IL-38 is also expressed in the skin and was previously proposed to act as an IL-36 antagonist. In this study, we thus examined expression and function of Il-38 in a mouse model of imiquimod (IMQ)-induced skin inflammation. Il-38 mRNA was detected in the epidermis and in primary mouse keratinocytes, but not in dermal fibroblasts. At the peak of IMQ-induced inflammation, skin Il-38 mRNA levels were reduced, whereas Il-36ra mRNA expression increased. The severity of IMQ-induced skin inflammation, as assessed by recording ear thickness and histological changes, was similar in Il-38 KO and WT littermate control mice, while, in contrast, Il-36ra-deficient mice displayed more severe skin pathology than their WT littermates. Il-38-deficiency had no impact on IMQ-induced expression of proinflammatory mediators in the skin in vivo, on the basal expression of various cytokines or chemokines by cultured primary keratinocytes and dermal fibroblasts in vitro, or on the response of these cells to Il-36ß. Finally, after cessation of topical IMQ application, the resolution of skin inflammation was also not altered in Il-38 KO mice. In conclusion, Il-38-deficiency did not impact the development or resolution of IMQ-induced skin inflammation. Our observations further suggest that endogenous Il-38 does not exert Il-36 inhibitory activity in this model, or in cultured skin cells. A potential anti-inflammatory function of Il-38 in mouse skin thus still remains to be demonstrated.


Subject(s)
Aminoquinolines , Dermatitis/genetics , Drug Eruptions/genetics , Inflammation/chemically induced , Interleukin-1/genetics , Interleukins/genetics , Skin/metabolism , Animals , Cells, Cultured , Dermatitis/metabolism , Dermatitis/pathology , Disease Models, Animal , Drug Eruptions/metabolism , Drug Eruptions/pathology , Female , Imiquimod , Inflammation/genetics , Inflammation/pathology , Interleukin-1/metabolism , Interleukins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index , Skin/pathology
10.
Blood ; 131(13): 1430-1441, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29295842

ABSTRACT

The term macrophage activation syndrome (MAS) defines a severe, potentially fatal disorder characterized by overwhelming inflammation and multiorgan involvement. Interleukin-18 (IL-18) is a proinflammatory cytokine belonging to the IL-1 family, the activity of which is regulated by its endogenous inhibitor IL-18 binding protein (IL-18BP). Elevated IL-18 levels have been reported in patients with MAS. Herein, we show that on repeated toll-like receptor 9 (TLR9) stimulation with unmethylated cytosine guanine dinucleotide containing single-stranded DNA (CpG), IL-18BP-/- mice display severe MAS manifestations, including increased weight loss, splenomegaly, anemia, thrombocytopenia, hyperferritinemia, and bone marrow hemophagocytosis as compared with wild-type mice. Serum-free IL-18 was detected in CpG-treated IL-18BP-/- mice only. Levels of interferon-γ (IFN-γ) and of IFN-γ signature genes, such as the chemokine Cxcl9 or the transcription factor CIIta, were significantly increased in IL-18BP-/- mice. Blocking IL-18 receptor signaling attenuated the severity of MAS and IFN-γ responses in IL-18BP-/- mice. Blocking IFN-γ had comparable effects to IL-18 inhibition on most MAS manifestations. Our data indicate that endogenous IL-18BP exerts a protective role in CpG-induced MAS and that IL-18, which acts upstream of IFN-γ, is involved in the severity of MAS.


Subject(s)
Interleukin-18/immunology , Macrophage Activation Syndrome/immunology , Signal Transduction/immunology , Toll-Like Receptor 9/immunology , Animals , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-18/genetics , Macrophage Activation Syndrome/chemically induced , Macrophage Activation Syndrome/genetics , Macrophage Activation Syndrome/pathology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Oligodeoxyribonucleotides/adverse effects , Oligodeoxyribonucleotides/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/genetics , Trans-Activators/genetics , Trans-Activators/immunology
11.
J Immunol ; 198(7): 2916-2926, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28235865

ABSTRACT

The biological activity of IL-1 is tightly regulated by the specific receptor antagonist (IL-1Ra) and the decoy receptor IL-1 receptor type 2 (IL-1R2). The role of IL-1Ra has been well demonstrated in IL-1Ra-deficient mice. In contrast, the role of endogenous IL-1R2 remains widely unknown. To define the functional role of endogenous IL-1R2 in the K/BxN serum transfer arthritis model and in IL-1ß- or LPS-induced systemic inflammation in vivo, IL-1R2-/- mice were created and compared with wild type mice. IL-1R2-/- mice bred habitually and exhibited a normal phenotype. IL-1R2 deficiency aggravated arthritis severity and increased mRNA levels for key cytokines and chemokines such as IL-6, IL-1ß, Cxcl-1, and Cxcl-2 significantly in ankles. There was no effect of IL-1R2 deficiency on the cell-autonomous cytokine response to IL-1ß in the tested cell types, i.e., neutrophils, macrophages, and fibroblasts, but IL-1R2 deficiency on neutrophils increased the IL-1-induced response of fibroblasts in trans. Furthermore, IL-1ß induced shedding of IL-1R2 in vivo. Inflammatory responses to IL-1ß and LPS-induced mortality were not different in IL-1R2-/- compared with wild type mice. Our data demonstrate that the decoy receptor IL-1R2 plays an important inhibitory role in local IL-1- and neutrophil-dependent tissue inflammation as shown in the K/BxN serum transfer arthritis model. In contrast to IL-1Ra, IL-1R2 appears to be less crucial for systemic responses to acute administration of IL-1 or LPS.


Subject(s)
Arthritis, Experimental/immunology , Inflammation/immunology , Receptors, Interleukin-1 Type II/immunology , Animals , Immunohistochemistry , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction
12.
J Immunol ; 197(6): 2239-49, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27527592

ABSTRACT

Human and mouse neonates exhibit limited vaccine responses characterized by predominant Th2 and limited Th1 responses. Because IL-36 exerts a synergic adjuvant effect with IL-12, enhancing Th1 polarization in adult (AD) mice, we administered IL-36ß to neonatal (1-wk old) and AD control mice at the time of immunization with tetanus toxoid adsorbed to aluminum hydroxide (TT/Alum). Unexpectedly, the combination of IL-36ß with TT/Alum, which was well tolerated in AD mice, proved toxic and even lethal in neonates. This neonatal toxicity was associated with high Il36r mRNA expression in neonatal liver, resulting in increased cytokine production. Liver Il36r mRNA expression decreased with the termination of fetal liver hematopoiesis, and this decrease correlated with a complete protection from TT/Alum/IL-36ß-induced mortality. The combination of IL-36ß and TT/Alum induced the rapid production of TNF-α and IFN-γ by liver myeloid and lymphoid cells, respectively. These responses were less marked when IL-36ß was used alone, with no adverse effect. The toxicity of IL-36ß + TT/Alum was abrogated by the administration of a neutralizing anti-TNF-α Ab, confirming causality. In conclusion, liver myeloid cells in neonatal mice are an important source of proinflammatory cytokines that may lead to TNF-α-mediated toxicity and even lethality.


Subject(s)
Interleukin-1/toxicity , Liver/immunology , Myeloid Cells/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Animals, Newborn , Cytokines/biosynthesis , Mice , Mice, Inbred C57BL , Tetanus Toxoid/toxicity
13.
Immun Inflamm Dis ; 3(3): 239-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26417439

ABSTRACT

Interleukin (IL)-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor. Previous work demonstrated that the systemic administration of recombinant IL-33 reduces the development of atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice by inducing a Th1-to-Th2 shift. The objective of our study was to examine the role of endogenous IL-33 and ST2 in atherosclerosis. ApoE(-/-), IL-33(-/-)ApoE(-/-), and ST2(-/-)ApoE(-/-) mice were fed with a cholesterol-rich diet for 10 weeks. Additionally, a group of ApoE(-/-) mice was injected with a neutralizing anti-ST2 or an isotype control antibody during the period of the cholesterol-rich diet. Atherosclerotic lesion development was measured by Oil Red O staining in the thoracic-abdominal aorta and the aortic sinus. There were no significant differences in the lipid-staining area of IL-33(-/-)ApoE(-/-), ST2(-/-)ApoE(-/-), or anti-ST2 antibody-treated ApoE(-/-) mice, compared to ApoE(-/-) controls. The absence of IL-33 signaling had no major and consistent impact on the Th1/Th2 cytokine responses in the supernatant of in vitro-stimulated lymph node cells. In summary, deficiency of the endogenously produced IL-33 and its receptor ST2 does not impact the development of atherosclerosis in ApoE-deficient mice.

14.
J Leukoc Biol ; 94(4): 791-802, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23817563

ABSTRACT

The proinflammatory activities of IL-1 are tightly controlled at different levels. IL-1R2 acts as a decoy receptor and has been shown to regulate the biological effects of IL-1 in vitro and in vivo. However, little is known about its natural expression in the mouse in physiologic and pathologic conditions. In this study, we examined IL-1R2 mRNA and protein expression in isolated cells and tissues in response to different stimulatory conditions. Data obtained using ex vivo CD11b(+)Ly6G(+) peripheral blood cells and in vitro-differentiated CD11b(+)Ly6G(+) BMG indicated that neutrophils are the major source of constitutively expressed IL-1R2 in the mouse. The expression of IL-1R2 on BMG and ex vivo Ly6G(+) peripheral blood cells was highly up-regulated by HC. IL-1R2 pull-down experiments showed that mouse rIL-1ß binds to BMG IL-1R2, whereas binding of IL-1Ra could not be detected. Furthermore, LPS treatment induced shedding of IL-1R2 from the neutrophil membrane in vitro and in vivo, executed mainly by ADAM17. Finally, in in vivo models of inflammation, including thioglycolate-induced acute peritonitis and acute lung injury, infiltrating Ly6G(+) neutrophils, expressed IL-1R2. Our data show that in the mouse, neutrophils mainly express the decoy receptor IL-1R2 under naïve and inflammatory conditions. These data suggest that neutrophils may contribute to the resolution of acute inflammation.


Subject(s)
Inflammation/metabolism , Neutrophils/metabolism , Receptors, Interleukin-1 Type II/metabolism , Acute Disease , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Hydrocortisone/pharmacology , Inflammation/pathology , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Protein Binding/drug effects , Up-Regulation/drug effects
15.
Cytokine ; 63(2): 135-44, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23684408

ABSTRACT

The cytokine interleukin-1α (IL-1α) is synthesized as a 31kDa peptide that lacks a leader peptide and is not secreted by the conventional secretory pathway. A distinctive characteristic of pro-IL-1α is the presence of a nuclear localization sequence in its amino-terminal moiety that allows its translocation to the nucleus. However no nuclear function(s) of the endogenous pro-IL-1α has been reported to date. In the present study, we used murine macrophages that produce IL-1α in response to pro-inflammatory stimuli, to gain further insight into the biology of the endogenous IL-1α protein in innate immune cells. We show that endogenous IL-1α is essentially found as a chromatin-associated nuclear protein in LPS-stimulated macrophages. In contrast to IL-1ß, IL-1α was not released upon inflammasome activation unless significant cell damage occurred. IL-1ß mRNA and protein levels were specifically decreased in IL-1α deficient macrophages after LPS stimulation. However, overexpression of human pro-IL-1α did not rescue this defective IL-1ß production, suggesting that this finding might be related to the insertion of the targeting construct into the IL-1 locus, rather than to a specific nuclear function of pro-IL-1α. Finally, by using both genomic and proteomic approaches, we could not identify a nuclear function of IL-1α. Taken together, these observations suggest that in macrophages IL-1α primarily acts as an alarmin that is rapidly released upon cell damage to activate early mechanisms of host defense.


Subject(s)
Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Animals , Cell Line , Cell Nucleus , Chromatin/metabolism , Humans , Immunity, Innate , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/biosynthesis , Lipopolysaccharides , Macrophage Activation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Arthritis Res Ther ; 15(2): R38, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23452551

ABSTRACT

INTRODUCTION: Interleukin (IL)-36 refers to three related IL-1 family cytokines, IL-36α, IL-36ß, and IL-36γ, that bind to the IL-36 receptor (IL-36R). IL-36 exerts proinflammatory effects in skin and lung and stimulates T cell responses. In the present study, we examined the expression and function of IL-36R and its ligands in experimental arthritis. METHODS: Collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and K/BxN serum transfer-induced arthritis were induced according to standard protocols. Messenger RNA levels for IL-36R and its ligands in the joints of mice with CIA were determined by RT-qPCR. Mice with CIA were injected with a blocking monoclonal anti-IL-36R, a blocking anti-IL-1RI, or their isotype-matched control antibodies at the time of arthritis onset. Anti-IL-36R or control antibodies were also injected at the time of AIA induction. Finally, IL-36R-deficient mice were examined in AIA and serum transfer-induced arthritis. The development and severity of arthritis were assessed by clinical and histological scoring. RESULTS: IL-36R, IL-36Ra and IL-36γ mRNA were detected in the joints of mice with CIA, but their levels did not correlate with arthritis severity. As opposed to anti-IL-1RI antibody treatment, the injection of an anti-IL-36R antibody was devoid of effect on the development and severity of CIA. The severity of joint inflammation and structural damage in AIA was also unaltered by anti-IL-36R antibody treatment. Finally, the severity of AIA and K/BxN serum transfer-induced arthritis was similar in IL-36R-deficient and wild-type mice. CONCLUSIONS: The development and severity of experimental arthritis are independent of IL-36R signaling.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Receptors, Interleukin-1/immunology , Signal Transduction , Animals , Arthritis, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-1/metabolism , Signal Transduction/immunology
17.
Arthritis Res Ther ; 15(1): R13, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23324173

ABSTRACT

INTRODUCTION: Interleukin (IL)-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor. Previous work suggested implication of the IL-33/ST2 axis in the pathogenesis of human and mouse arthritis. Here, we directly investigated the role of endogenous IL-33 in K/BxN serum transfer-induced arthritis by using IL-33 knockout (KO) mice. METHODS: Arthritis was induced by injection of complete K/BxN serum or purified IgG. Disease severity was monitored by clinical and histological scoring. RESULTS: K/BxN serum transfer induced pronounced arthritis with similar incidence and severity in IL-33 KO and wild-type (WT) mice. In contrast, disease development was significantly reduced in ST2 KO mice. IL-33 expression in synovial tissue was comparable in arthritic WT and ST2 KO mice, and absent in IL-33 KO mice. Transfer of purified arthritogenic IgG instead of complete K/BxN serum also resulted in similar arthritis severity in IL-33 KO and WT mice, excluding a contribution of IL-33 contained in the serum of donor mice to explain this result. We investigated additional potential confounding factors, including purity of genetic background, but the mechanisms underlying reduced arthritis in ST2 KO mice remained unclear. CONCLUSIONS: The data obtained with IL-33 KO mice indicate that endogenous IL-33 is not required for the development of joint inflammation in K/BxN serum transfer-induced arthritis. On the contrary, arthritis severity was reduced in ST2 KO mice. This observation might relate to IL-33 independent effects of ST2, and/or reveal the existence of confounding variables affecting the severity of joint inflammation in these KO strains.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Interleukins/immunology , Animals , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Genotype , Immunohistochemistry , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Real-Time Polymerase Chain Reaction , Receptors, Interleukin/deficiency , Receptors, Interleukin/immunology
18.
Blood ; 120(17): 3478-87, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22968459

ABSTRACT

The interleukin-1 (IL-1) superfamily of cytokines comprises a set of pivotal mediators of inflammation. Among them, the action of IL-36 cytokines in immune responses has remained elusive. In a recent study, we demonstrated a direct effect of IL-36 on immune cells. Here we show that, among T cells, the IL-36 receptor is predominantly expressed on naive CD4(+) T cells and that IL-36 cytokines act directly on naive T cells by enhancing both cell proliferation and IL-2 secretion. IL-36ß acts in synergy with IL-12 to promote Th1 polarization and IL-36 signaling is also involved in mediating Th1 immune responses to Bacillus Calmette-Guerin infection in vivo. Our findings point toward a critical function of IL-36 in the priming of Th1 cell responses in vitro, and in adaptive immunity in a model of mycobacterial infection in vivo.


Subject(s)
Mycobacterium bovis/immunology , Receptors, Interleukin-1/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Tuberculosis/metabolism , Tuberculosis/veterinary , Adaptive Immunity , Animals , Cell Differentiation , Cell Proliferation , Interleukin-1/immunology , Interleukin-1/pharmacology , Interleukin-12/immunology , Interleukin-12/pharmacology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Primary Cell Culture , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Signal Transduction/genetics , Th1 Cells/cytology , Th1 Cells/microbiology , Tuberculosis/genetics , Tuberculosis/immunology
19.
Eur J Immunol ; 42(5): 1294-303, 2012 May.
Article in English | MEDLINE | ID: mdl-22539301

ABSTRACT

Interleukin-1 receptor antagonist (IL-1Ra) is a specific IL-1 inhibitor that possesses anti-inflammatory activities. Several studies in human and mouse suggested a protective role for IL-1Ra in liver inflammation, and we previously demonstrated that hepatocytes produce high levels of IL-1Ra in response to inflammatory challenge in vitro and in vivo. In the present study, we investigated the production and the biological function of hepatocyte-derived IL-1Ra in concanavalin A (ConA)-induced hepatitis in mice. We show that the injured liver produces large amounts of IL-1Ra and that secreted and intracellular IL-1Ra isoforms are produced with different kinetics during the course of hepatitis. By using hepatocyte-specific IL-1Ra-deficient mice (IL-1Ra(ΔH)), we demonstrate that hepatocytes represent the major cellular source of local IL-1Ra. Most interestingly, hepatic necrosis and inflammation were increased in IL-1Ra(ΔH) as compared with wild-type mice during the late phase of the disease, leading to a delayed resolution of hepatitis in IL-1Ra(ΔH) mice. In conclusion, our results show that the local production of IL-1Ra by hepatocytes contributes to the resolution of hepatitis.


Subject(s)
Hepatitis/immunology , Hepatocytes/immunology , Interleukin 1 Receptor Antagonist Protein/immunology , Animals , Concanavalin A/toxicity , Cytokines/analysis , Hepatitis/genetics , Hepatitis/pathology , Hepatocytes/drug effects , Interleukin 1 Receptor Antagonist Protein/genetics , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Necrosis/immunology
20.
Ann Rheum Dis ; 71(2): 281-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22072017

ABSTRACT

OBJECTIVES: To define the cell type (myeloid vs other cells) specific effect of interleukin 1 (IL-1) receptor antagonist (IL-1Ra) deficiency on the acute inflammatory phase of arthritis. METHODS: Arthritis was induced by K/BxN serum transfer in wild-type (WT), IL-1Ra-deficient (IL-1Ra(-/-)) and conditional knockout mice. In the latter, IL-1Ra production was specifically targeted in myeloid cells (IL-1Ra(ΔM)) or in both hepatocytes and myeloid cells (IL-1Ra(ΔH+M)). Arthritis severity was clinically evaluated and ankle sections were scored for synovial inflammation and cartilage erosion. Quantitative RT-PCR, western blot and immunohistochemical analyses measured expression, localisation and cellular sources of the different IL-1Ra isoforms in arthritic joints. RESULTS: Total and myeloid cell-specific IL-1Ra deficiency was associated with increased arthritis severity, although disease incidence was similar to that of WT mice. Increased clinical scores were associated with exacerbated synovial inflammation. All IL-1Ra isoforms, except for intracellular (ic)IL-1Ra2, were expressed in arthritic joints of WT mice. In contrast, production of secreted (s)IL-1Ra and icIL-1Ra3 isoforms was markedly decreased in arthritic joints of both IL-1Ra(ΔM) and IL-1Ra(ΔH+M) mice. Immunohistochemical and western blot analyses suggested that the icIL-1Ra1 isoform is produced primarily by synovial fibroblasts. CONCLUSION: Myeloid cell-derived IL-1Ra, including both sIL-1Ra and icIL-1Ra3 isoforms, controls articular inflammation during the acute phase of K/BxN serum transfer-induced arthritis.


Subject(s)
Arthritis, Experimental/metabolism , Interleukin 1 Receptor Antagonist Protein/physiology , Myeloid Cells/metabolism , Acute Disease , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Disease Progression , Female , Interleukin 1 Receptor Antagonist Protein/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms/deficiency , Protein Isoforms/physiology , Serum , Severity of Illness Index , Synovitis/metabolism , Synovitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...