Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Eur Heart J ; 39(23): 2158-2167, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29905812

ABSTRACT

Aim: Rheumatoid arthritis (RA) is associated with an approximately two-fold elevated risk of cardiovascular (CV)-related mortality. Patients with RA present with systemic inflammation including raised circulating myeloid cells, but fail to display traditional CV risk-factors, particularly dyslipidaemia. We aimed to explore if increased circulating myeloid cells is associated with impaired atherosclerotic lesion regression or altered progression in RA. Methods and results: Using flow cytometry, we noted prominent monocytosis, neutrophilia, and thrombocytosis in two mouse models of RA. This was due to enhanced proliferation of the haematopoietic stem and progenitor cells (HSPCs) in the bone marrow and the spleen. HSPCs expansion was associated with an increase in the cholesterol content, due to a down-regulation of cholesterol efflux genes, Apoe, Abca1, and Abcg1. The HSPCs also had enhanced expression of key myeloid promoting growth factor receptors. Systemic inflammation was found to cause defective cellular cholesterol metabolism. Increased myeloid cells in mice with RA were associated with a significant impairment in lesion regression, even though cholesterol levels were equivalent to non-arthritic mice. Lesions from arthritic mice exhibited a less stable phenotype as demonstrated by increased immune cell infiltration, lipid accumulation, and decreased collagen formation. In a progression model, we noted monocytosis, enhanced monocytes recruitment to lesions, and increased plaque macrophages. This was reversed with administration of reconstituted high-density lipoprotein (rHDL). Furthermore, RA patients have expanded CD16+ monocyte subsets and a down-regulation of ABCA1 and ABCG1. Conclusion: Rheumatoid arthritis impairs atherosclerotic regression and alters progression, which is associated with an expansion of myeloid cells and disturbed cellular cholesterol handling, independent of plasma cholesterol levels. Infusion of rHDL prevented enhanced myelopoiesis and monocyte entry into lesions. Targeting cellular cholesterol defects in people with RA, even if plasma cholesterol is within the normal range, may limit vascular disease.


Subject(s)
Arthritis, Rheumatoid/metabolism , Atherosclerosis/metabolism , Cholesterol/metabolism , Hematopoietic Stem Cells/metabolism , Monocytes/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Adult , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Arthritis, Rheumatoid/immunology , Atherosclerosis/genetics , Atherosclerosis/immunology , Disease Models, Animal , Down-Regulation , Female , Hematopoiesis, Extramedullary/immunology , Humans , Leukocytosis , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Mice , Middle Aged , Monocytes/immunology , Myelopoiesis/immunology , Neutrophils , RNA, Messenger/metabolism , Thrombocytosis
2.
Cytokine ; 72(2): 135-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25647268

ABSTRACT

Macrophage migration inhibitory factor (MIF) enhances activation of leukocytes, endothelial cells and fibroblast-like synoviocytes (FLS), thereby contributing to the pathogenesis of rheumatoid arthritis (RA). A MIF promoter polymorphism in RA patients resulted in higher serum MIF concentration and worsens bone erosion; controversially current literature reported an inhibitory role of MIF in osteoclast formation. The controversial suggested that the precise role of MIF and its putative receptor CD74 in osteoclastogenesis and RA bone erosion, mediated by locally formed osteoclasts in response to receptor activator of NF-κB ligand (RANKL), is unclear. We reported that in an in vivo K/BxN serum transfer arthritis, reduced clinical and histological arthritis in MIF(-/-) and CD74(-/-) mice were accompanied by a virtual absence of osteoclasts at the synovium-bone interface and reduced osteoclast-related gene expression. Furthermore, in vitro osteoclast formation and osteoclast-related gene expression were significantly reduced in MIF(-/-) cells via decreasing RANKL-induced phosphorylation of NF-κB-p65 and ERK1/2. This was supported by a similar reduction of osteoclastogenesis observed in CD74(-/-) cells. Furthermore, a MIF blockade reduced RANKL-induced osteoclastogenesis via deregulating RANKL-mediated NF-κB and NFATc1 transcription factor activation. These data indicate that MIF and CD74 facilitate RANKL-induced osteoclastogenesis, and suggest that MIF contributes directly to bone erosion, as well as inflammation, in RA.


Subject(s)
Arthritis, Rheumatoid/physiopathology , Macrophage Migration-Inhibitory Factors/deficiency , Macrophage Migration-Inhibitory Factors/physiology , Osteoclasts/physiology , Animals , Antigens, Differentiation, B-Lymphocyte/physiology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Bone Resorption , Cells, Cultured , Disease Models, Animal , Histocompatibility Antigens Class II/physiology , Macrophage Migration-Inhibitory Factors/pharmacology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NFATC Transcription Factors/physiology , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Synovial Membrane/cytology
3.
J Immunol ; 190(3): 997-1007, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23267026

ABSTRACT

Annexin A1 (AnxA1) is recognized as an endogenous anti-inflammatory molecule. However, its effects on the adaptive immune response and, in particular, on T cells remain unclear. In this study, we investigated the actions of AnxA1 in three distinct models of T cell-mediated inflammation. In contact hypersensitivity, collagen-induced arthritis, and inflammation induced by OT-II TCR transgenic T cells responding to OVA, AnxA1 deficiency significantly increased Ag-induced T cell proliferation and the resultant level of inflammation. In the contact hypersensitivity model, this was associated with increased adhesion of CD4(+) T cells, CD8(+) T cells, and neutrophils in the dermal microvasculature, as well as increased T cell expression of RORγt and IL-17A. In collagen-induced arthritis, deficiency of endogenous AnxA1 increased susceptibility to arthritis and Ag-specific T cell activation. Deficiency of AnxA1 also increased OVA-induced cutaneous delayed-type hypersensitivity and IFN-γ and IL-17 release. Transfer experiments using CD4(+) T cells from AnxA1(-/-) mice demonstrated that the absence of AnxA1 solely in T cells resulted in increased inflammatory responses in wild-type recipients. Similarly, experiments using AnxA1(-/-) OT-II CD4(+) T cells demonstrated that the absence of AnxA1 in T cells was sufficient to induce increased Ag-specific CD4(+) T cell proliferation in vivo, augment T cell production of IFN-γ, IL-17, TNF, and IL-6, and increase Akt, ERK, and p38 activation. Together, these findings indicate that T cell-expressed AnxA1 functions to attenuate T cell-driven inflammatory responses via T cell-intrinsic effects on intracellular signaling, proliferation, and Th1/Th17 cytokine release.


Subject(s)
Annexin A1/deficiency , CD4-Positive T-Lymphocytes/immunology , Inflammation/immunology , Animals , Annexin A1/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Enzyme Activation/immunology , Gene Expression Regulation/immunology , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Inflammation/pathology , Interleukin-17/biosynthesis , Interleukin-17/genetics , Lymphocyte Activation , Lymphokines/biosynthesis , Lymphokines/genetics , Lymphokines/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Ovalbumin/immunology , Ovalbumin/toxicity , Oxazolone/immunology , Oxazolone/toxicity , Peptide Fragments/immunology , Peptide Fragments/toxicity , Signal Transduction/immunology , Specific Pathogen-Free Organisms
4.
J Immunol ; 191(1): 424-33, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23729444

ABSTRACT

Glucocorticoid-induced leucine zipper (GILZ) is an anti-inflammatory protein first identified in T lymphocytes. We recently observed that GILZ is highly expressed in synovial endothelial cells in rheumatoid arthritis. However, the function of GILZ in endothelial cells is unknown. To investigate the actions of GILZ in this cell type, we induced GILZ expression in HUVECs via transient transfection. GILZ overexpression significantly reduced the capacity of TNF-stimulated HUVECs to support leukocyte rolling, adhesion, and transmigration. These effects were associated with decreased expression of E-selectin, ICAM-1, CCL2, CXCL8, and IL-6. Experiments in a human microvascular endothelial cell line demonstrated that TNF-inducible NF-κB activity was significantly inhibited by overexpression of GILZ. Exogenous GILZ inhibited TNF-induced NF-κB p65 DNA binding, although this occurred in the absence of an effect on p65 nuclear translocation, indicating that the mechanism of action of exogenous GILZ in endothelial cells differs from that reported in other cell types. GILZ overexpression also inhibited TNF-induced activation of p38, ERK, and JNK MAPKs, as well as increased expression of the MAPK inhibitory phosphatase, MKP-1. In contrast, silencing endogenous GILZ in glucocorticoid-treated HUVECs did not alter their capacity to support leukocyte interactions. These data demonstrate that exogenous GILZ exerts inhibitory effects on endothelial cell adhesive function via a novel pathway involving modulation of NF-κB p65 DNA binding and MAPK activity. Induction of GILZ expression in endothelial cells may represent a novel therapeutic modality with the potential to inhibit inflammatory leukocyte recruitment.


Subject(s)
Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , MAP Kinase Signaling System/immunology , Transcription Factor RelA/metabolism , Transcription Factors/genetics , Transendothelial and Transepithelial Migration/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Communication/immunology , Cell Line , Cell Migration Inhibition/genetics , Cell Migration Inhibition/immunology , Endothelium, Vascular/physiology , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes/immunology , Leukocytes/metabolism , MAP Kinase Signaling System/genetics , Microcirculation/genetics , Microcirculation/immunology , Primary Cell Culture , Random Allocation , Transcription Factors/biosynthesis , Transcription Factors/physiology , Transendothelial and Transepithelial Migration/genetics
5.
Arthritis Rheum ; 65(5): 1203-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23335223

ABSTRACT

OBJECTIVE: Glucocorticoid-induced leucine zipper (GILZ) has effects on inflammatory pathways that suggest it to be a key inhibitory regulator of the immune system, and its expression is exquisitely sensitive to induction by glucocorticoids. We undertook this study to test our hypothesis that GILZ deficiency would exacerbate experimental immune-mediated inflammation and impair the effects of glucocorticoids on inflammation and, correspondingly, that exogenous GILZ would inhibit these events. METHODS: GILZ(-/-) mice were generated using the Cre/loxP system, and responses were studied in delayed-type hypersensitivity (DTH), antigen-induced arthritis (AIA), K/BxN serum-transfer arthritis, and lipopolysaccharide (LPS)-induced cytokinemia. Therapeutic expression of GILZ via administration of recombinant adeno-associated virus expressing the GILZ gene (GILZ-rAAV) was compared to the effects of glucocorticoid in collagen-induced arthritis (CIA). RESULTS: Increased T cell proliferation and DTH were observed in GILZ(-/-) mice, but neither AIA nor K/BxN serum-transfer arthritis was affected, and GILZ deficiency did not affect LPS-induced cytokinemia. Deletion of GILZ did not impair the effects of exogenous glucocorticoids on CIA or cytokinemia. In contrast, overexpression of GILZ in joints significantly inhibited CIA, with an effect similar to that of dexamethasone. CONCLUSION: Despite effects on T cell activation, GILZ deficiency had no effect on effector pathways of arthritis and was unexpectedly redundant with effects of glucocorticoids. These findings do not support the hypothesis that GILZ is central to the actions of glucocorticoids, but the efficacy of exogenous GILZ in CIA suggests that further evaluation of GILZ in inflammatory disease is required.


Subject(s)
Arthritis, Experimental/therapy , Hypersensitivity, Delayed/therapy , Transcription Factors/genetics , Adenoviridae/genetics , Animals , Arthritis, Experimental/genetics , Cell Proliferation , Dexamethasone/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Gene Targeting , Genetic Therapy/methods , Glucocorticoids/pharmacology , Hypersensitivity, Delayed/genetics , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , Transcription Factors/deficiency , Transduction, Genetic
6.
Arthritis Rheum ; 63(4): 960-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21452319

ABSTRACT

OBJECTIVE: Macrophage migration inhibitory factor (MIF) facilitates multiple aspects of inflammatory arthritis, the pathogenesis of which has been significantly linked to the activity of neutrophils. The effects of MIF on neutrophil recruitment are unknown. This study was undertaken to investigate the contribution of MIF to the regulation of neutrophil chemotactic responses. METHODS: K/BxN serum-transfer arthritis was induced in wild-type (WT), MIF(-/-) , and monocyte chemotactic protein 1 (MCP-1; CCL2)-deficient mice as well as in WT mice treated with monoclonal antibodies to cytokine-induced neutrophil chemoattractant (anti-KC). Leukocyte trafficking in vivo was examined using intravital microscopy, and neutrophil function in vitro was examined using migration chambers and assessment of MAP kinase activation. RESULTS: K/BxN serum-transfer arthritis was markedly attenuated in MIF(-/-) mice, with reductions in the clinical and histologic severity of arthritis and the synovial expression of KC and interleukin-1. Arthritis was also reduced by anti-KC antibody treatment, but not in MCP-1-deficient mice. In vivo, neutrophil recruitment responses to KC were reduced in MIF(-/-) mice. Similarly, MIF(-/-) mouse neutrophils exhibited reduced chemotactic responses to KC in vitro, despite displaying unaltered chemokine receptor expression. Reduced chemotactic responses of MIF(-/-) mouse neutrophils were associated with reduced phosphorylation of p38 and ERK MAP kinases. CONCLUSION: These findings suggest that MIF promotes neutrophil trafficking in inflammatory arthritis via facilitation of chemokine-induced migratory responses and MAP kinase activation. Therapeutic MIF inhibition could limit synovial neutrophil recruitment.


Subject(s)
Arthritis, Experimental/physiopathology , Chemotaxis, Leukocyte/physiology , Macrophage Migration-Inhibitory Factors/physiology , Neutrophils/pathology , Adaptive Immunity/physiology , Animals , Arthritis, Experimental/pathology , Cell Movement/drug effects , Chemokine CXCL1/pharmacology , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Macrophage Migration-Inhibitory Factors/deficiency , Macrophage Migration-Inhibitory Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects
7.
Arthritis Rheum ; 62(9): 2651-61, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20496421

ABSTRACT

OBJECTIVE: Glucocorticoid-induced leucine zipper (GILZ) is a glucocorticoid-induced protein, the reported molecular interactions of which suggest that it functions to inhibit inflammation. However, the role of endogenous GILZ in the regulation of inflammation in vivo has not been established. This study was undertaken to examine the expression and function of GILZ in vivo in collagen-induced arthritis (CIA), a murine model of rheumatoid arthritis (RA), and in RA synoviocytes. METHODS: GILZ expression was detected in mouse and human synovium by immunohistochemistry and in cultured cells by real-time polymerase chain reaction and permeabilization flow cytometry. GILZ function was assessed in vivo by small interfering RNA (siRNA) silencing using cationic liposome-encapsulated GILZ or control nontargeting siRNA and was assessed in vitro using transient overexpression. RESULTS: GILZ was readily detectable in the synovium of mice with CIA and was up-regulated by therapeutic doses of glucocorticoids. Depleting GILZ expression in vivo increased the clinical and histologic severity of CIA and increased synovial expression of tumor necrosis factor and interleukin-1 (IL-1), without affecting the levels of circulating cytokines or anticollagen antibodies. GILZ was highly expressed in the synovium of patients with active RA and in cultured RA synovial fibroblasts, and GILZ overexpression in synovial fibroblasts inhibited IL-6 and IL-8 release. CONCLUSION: Our findings indicate that GILZ functions as an endogenous inhibitor of chronic inflammation via effects on cytokine expression and suggest that local modulation of GILZ expression could be a beneficial therapeutic strategy.


Subject(s)
Arthritis, Experimental/metabolism , Glucocorticoids/physiology , Inflammation Mediators/physiology , Transcription Factors/metabolism , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Cells, Cultured , Cytokines/metabolism , Dexamethasone/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Silencing , Glucocorticoids/pharmacology , Humans , Male , Mice , Mice, Inbred DBA , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology , Transcription Factors/genetics , Transfection
8.
Nat Commun ; 11(1): 5794, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33188181

ABSTRACT

Necrotizing enterocolitis (NEC) is a severe, currently untreatable intestinal disease that predominantly affects preterm infants and is driven by poorly characterized inflammatory pathways. Here, human and murine NEC intestines exhibit an unexpected predominance of type 3/TH17 polarization. In murine NEC, pro-inflammatory type 3 NKp46-RORγt+Tbet+ innate lymphoid cells (ILC3) are 5-fold increased, whereas ILC1 and protective NKp46+RORγt+ ILC3 are obliterated. Both species exhibit dysregulation of intestinal TLR repertoires, with TLR4 and TLR8 increased, but TLR5-7 and TLR9-12 reduced. Transgenic IL-37 effectively protects mice from intestinal injury and mortality, whilst exogenous IL-37 is only modestly efficacious. Mechanistically, IL-37 favorably modulates immune homeostasis, TLR repertoires and microbial diversity. Moreover, IL-37 and its receptor IL-1R8 are reduced in human NEC epithelia, and IL-37 is lower in blood monocytes from infants with NEC and/or lower birthweight. Our results on NEC pathomechanisms thus implicate type 3 cytokines, TLRs and IL-37 as potential targets for novel NEC therapies.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/immunology , Adaptive Immunity , Animals , Animals, Newborn , Biomarkers/metabolism , Enterocolitis, Necrotizing/blood , Enterocolitis, Necrotizing/pathology , Homeostasis , Humans , Immunity, Innate , Infant, Newborn , Inflammation Mediators/metabolism , Interleukin-1 , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Toll-Like Receptors/metabolism
9.
Arthritis Rheumatol ; 71(1): 50-62, 2019 01.
Article in English | MEDLINE | ID: mdl-30009417

ABSTRACT

OBJECTIVE: The production of class-switched high-affinity autoantibodies derived from organized germinal centers (GCs) is a hallmark of many autoimmune inflammatory diseases, including rheumatoid arthritis (RA). TANK-binding kinase 1 (TBK-1) is a serine/threonine kinase involved in the maturation of GC follicular helper T (Tfh) cells downstream of inducible costimulator signaling. We undertook this study to assess the therapeutic potential of TBK-1 inhibition using the small-molecule inhibitor WEHI-112 in antibody-dependent models of inflammatory arthritis. METHODS: Using the models of collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and K/BxN serum-transfer-induced arthritis (STIA), we determined the effectiveness of WEHI-112 at inhibiting clinical and histologic features of arthritis in C57BL/6 and DBA/1 mice. We used immunohistochemistry to characterize GC populations during CIA development, and we used enzyme-linked immunosorbent assays to determine levels of Ig autoantibodies in WEHI-112-treated mice compared to vehicle-treated mice. RESULTS: WEHI-112, a tool compound that is semiselective for TBK-1 but that also has activity against IKKε and JAK2, abolished TBK-1-dependent activation of interferon (IFN) regulatory factor 3 and inhibited type I IFN responses in vitro. In vivo, treatment with WEHI-112 selectively abrogated clinical and histologic features of established, antibody-dependent CIA, but had minimal effects on an antibody-independent model of AIA or on K/BxN STIA. In keeping with these findings, WEHI-112 reduced arthritogenic type II collagen-specific IgG1 and IgG2b antibody production. Furthermore, WEHI-112 altered the GC Tfh cell phenotype and GC B cell function in CIA. CONCLUSION: We report that TBK-1 inhibition using WEHI-112 abrogated antibody-dependent CIA. As WEHI-112 failed to inhibit non-antibody-driven joint inflammation, we conclude that the major effect of this compound was most likely the targeting of TBK-1-mediated mechanisms in the GC reaction. This approach may have therapeutic potential in RA and in other GC-associated autoantibody-driven inflammatory diseases.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Autoantibodies/drug effects , Germinal Center/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/drug effects , Animals , Autoantibodies/immunology , Azetidines/pharmacology , Collagen Type II , Cyclobutanes/pharmacology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Freund's Adjuvant , Germinal Center/immunology , Immunohistochemistry , Immunologic Factors , In Vitro Techniques , Interferon Regulatory Factor-3/drug effects , Interferon Regulatory Factor-3/immunology , Interferon Type I/drug effects , Interferon Type I/immunology , Janus Kinase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Morpholines/pharmacology , Purines , Pyrazoles , Serum Albumin, Bovine , Sulfonamides/pharmacology , T-Lymphocytes, Helper-Inducer/immunology
10.
Int Immunopharmacol ; 61: 140-149, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29879657

ABSTRACT

The effects of formyl peptide receptors (FPRs) on effector T cells and inflammation-causing tissue-resident cells are not well known. Here, we explored the effect of FPR activation on efferent T cell responses in models of rheumatoid arthritis (RA) and on the expansion of fibroblast-like synoviocytes (FLS). Compound 43 (Cpd43; FPR1/2 agonist) was administered to mice with collagen-induced arthritis (CIA) or antigen-induced arthritis (AIA) after disease onset. Joint inflammation/damage and immunity were assessed. FLS were cultured with Cpd43 to test its effects on cell apoptosis and proliferation. To explore the effects of endogenous FPR2 ligands on FLS proliferation, FLS FPR2 was blocked or Annexin A1 (AnxA1) expression silenced. Cpd43 reduced arthritis severity in both models. In CIA, Cpd43 decreased CD4 T cell proliferation and survival and increased the production of the protective cytokine, IFNγ, in lymph nodes. In AIA, Cpd43 increased CD4 apoptosis and production of the anti-inflammatory IL-4, while augmenting the proportion of splenic regulatory T cells and their expression of IL-2Rα. In both models, Cpd43 increased CD4 IL-17A production, without affecting humoral immunity. FPR2 inhibitors reversed Cpd43-mediated effects on AIA and T cell immunity. Cpd43 decreased TNF-induced FLS proliferation and augmented FLS apoptosis in association with intracellular FPR2 accumulation, while endogenous AnxA1 and FPR2 reduced FLS proliferation via the ERK and NFκB pathways. Overall, FPR activation inhibits the expansion of arthritogenic effector CD4 T cells and FLS, and reduces joint injury in experimental arthritis. This suggests the therapeutic potential of FPR ligation for the treatment of RA.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Immunologic Factors/therapeutic use , Receptors, Formyl Peptide/agonists , Synoviocytes/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Humans , Immunologic Factors/pharmacology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , NF-kappa B/metabolism , Signal Transduction , Synoviocytes/physiology
11.
Theranostics ; 8(3): 846-859, 2018.
Article in English | MEDLINE | ID: mdl-29344311

ABSTRACT

Mesenchymal stem cells (MSC) are highly immunosuppressive cells able to reduce chronic inflammation through the active release of mediators. Recently, we showed that glucocorticoid-induced leucine zipper (Gilz) expression by MSC is involved in their therapeutic effect by promoting the generation of regulatory T cells. However, the mechanisms underlying this pivotal role of Gilz remain elusive. Methods and Results In this study, we have uncovered evidence that Gilz modulates the phenotype and function of Th1 and Th17 cells likely by upregulating the level of Activin A and NO2 secreted by MSC. Adoptive transfer experiments sustained this Gilz-dependent suppressive effect of MSC on Th1 and Th17 cell functions. In immunoregulatory MSC, obtained by priming with IFN-γ and TNF-α, Gilz was translocated to the nucleus and bound to the promoters of inos and Activin ßA to induce their expression. The increased expression of Activin A directly impacted on Th17 cells fate by repressing their differentiation program through the activation of Smad3/2 and enhancing IL-10 production. Conclusion Our results reveal how Gilz controls inos and Activin ßA gene expression to ultimately assign immunoregulatory status to MSC able to repress the pathogenic Th17 cell differentiation program and uncover Activin A as a novel mediator of MSC in this process.


Subject(s)
Activins/metabolism , Cell Differentiation , Mesenchymal Stem Cells/immunology , Th17 Cells/immunology , Transcription Factors/metabolism , Activins/genetics , Animals , Cells, Cultured , Cross-Priming , Interferon-gamma/metabolism , Interleukin-10/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Smad Proteins/metabolism , Th17 Cells/cytology , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
J Leukoc Biol ; 101(4): 901-911, 2017 04.
Article in English | MEDLINE | ID: mdl-27881605

ABSTRACT

The interleukin (IL)-1 family member IL-37 is one of few anti-inflammatory cytokines, and it is capable of countering a broad spectrum of proinflammatory assaults. Although it is known that leukocytes are a major source of IL-37, knowledge on IL-37 production and secretion in specific immune cell types remains limited. Thus, we investigated IL-37 mRNA expression as well as protein production and secretion in human PBMCs. In PBMCs stimulated with agonists of Toll-like receptors (TLRs) 1-6 and 9, IL1F7 (the IL-37-encoding gene) was induced up to 9-fold, peaked at 6-8 h and returned to steady-state at 72 h. LPS-induced IL1F7 expression comprised isoforms b and c but not a and e Flow cytometry revealed that among IL-37+ PBMCs, monocytes predominated (81-91%), but T cells (6-8%) and myeloid dendritic cells (mDCs, 1-2%) also contributed to the IL-37+ leukocyte pool. Monocytes and mDCs, but not T cells, were capable of secreting IL-37. Whereas monocytes and mDCs secreted IL-37 upon LPS stimulation, only mDCs also released IL-37 at steady-state. Among monocyte subsets, IL-37 was LPS inducible and secreted only in classical and, although less pronounced, in intermediate monocytes; secretion was observed as early as 3 h after stimulation. Overall, our data suggest that constitutive IL-37 secretion by mDCs may serve to maintain an anti-inflammatory milieu at steady state, whereas IL-37 is stored in monocytes to be available for rapid release upon inflammatory encounters, thus acting as a novel anti-inflammatory alarmin. These insights may prove important to advancing towards clinical use the protective functions of one of the most powerful anti-inflammatory mediators so far discovered.


Subject(s)
Dendritic Cells/metabolism , Interleukin-1/metabolism , Monocytes/metabolism , Adult , Dendritic Cells/drug effects , Flow Cytometry , Humans , Kinetics , Ligands , Lipopolysaccharides/pharmacology , Monocytes/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Toll-Like Receptors/metabolism
13.
Sci Immunol ; 2(8)2017 Feb 10.
Article in English | MEDLINE | ID: mdl-28783685

ABSTRACT

Dysregulation of the inflammatory response underlies numerous diseases. Although most interleukin-1 family cytokines are proinflammatory, human interleukin-37 (IL-37) is a powerful, broad-spectrum inhibitor of inflammation and immunity. We determined the crystal structure of IL-37 to establish the anti-inflammatory mechanism of this key cytokine in view of developing IL-37-based therapies. We found that two ß-trefoil fold IL-37 molecules form a head-to-head dimer that is stable in solution. IL-37 variants mutated to convert the cytokine into an obligate monomer were up to 13-fold more effective than the dimer in suppressing proinflammatory events both in primary human blood cells and in vivo in murine endotoxic shock. Therapeutic exploitation of the powerful anti-inflammatory properties of monomeric IL-37 may prove beneficial in treating a wide range of inflammatory and autoimmune disorders.

14.
Arthritis Rheumatol ; 66(8): 2059-70, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24782327

ABSTRACT

OBJECTIVE: Glucocorticoids remain a mainstay in the treatment of rheumatoid arthritis (RA). Dose-dependent adverse effects highlight the need for therapies that regulate glucocorticoid sensitivity to enable dosage reduction. Macrophage migration inhibitory factor (MIF) is a proinflammatory protein that has been implicated in the pathogenesis of RA; it impairs glucocorticoid sensitivity via MAPK phosphatase 1 (MKP-1) inhibition. The intracellular protein glucocorticoid-induced leucine zipper (GILZ) mimics the effects of glucocorticoids in models of RA, but whether it represents a target for the modulation of glucocorticoid sensitivity remains unknown. We undertook this study to investigate whether GILZ is involved in the regulation of glucocorticoid sensitivity by MIF. METHODS: GILZ expression was studied in the presence and absence of MIF, and the role of GILZ in the MIF-dependent regulation of the glucocorticoid sensitivity mediator MKP-1 was studied at the level of expression and function. RESULTS: GILZ expression was significantly inhibited by endogenous MIF, both basally and during responses to glucocorticoid treatment. The effects of MIF on GILZ were dependent on the expression and Akt-induced nuclear translocation of the transcription factor FoxO3A. GILZ was shown to regulate the expression of MKP-1 and consequent MAPK phosphorylation and cytokine release. CONCLUSION: MIF exerts its effects on MKP-1 expression and MAPK activity through inhibitory effects on GILZ. These findings suggest a previously unsuspected interaction between MIF and GILZ and identify GILZ as a potential target for the therapeutic regulation of glucocorticoid sensitivity.


Subject(s)
Glucocorticoids/pharmacology , Leucine Zippers/drug effects , Leucine Zippers/physiology , Macrophage Migration-Inhibitory Factors/physiology , Animals , Cells, Cultured , Dual Specificity Phosphatase 1/drug effects , Dual Specificity Phosphatase 1/physiology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/physiology
15.
J Control Release ; 167(1): 85-91, 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23353808

ABSTRACT

This study is the first to demonstrate in canines the ability of silica-lipid hybrid (SLH) microparticles to enhance the bioavailability and efficacy of a poorly water-soluble drug after oral administration. Spray-dried SLH microparticles comprising Capmul MCM (mono-diglycerides of C8/C12 fatty acids) and silica nanoparticles (Aerosil® 380) were shown to significantly enhance the fasted state oral bioavailability of celecoxib (CEL) (6.5 fold, relative to an aqueous suspension and more than 2-fold higher relative to the fed state) after oral administration to beagle dogs. Comparable bioavailability was observed between the SLH microparticle formulation and a conventional Capmul lipid solution, however, plasma concentrations were observed to be higher (Cmax, 1.1±0.06 vs. 0.8±0.03µg/mL) (p≤0.05) with the SLH microparticle system. The enhanced bioavailability of CEL observed with the SLH microparticles was reflected in a subsequent efficacy study conducted in an adjuvant-induced arthritis model in the rat. Reduced clinical and histological severity was observed at a dose of 3mg/kg/day, with the progression of arthritic symptoms and tissue damage reduced to a similar degree to that of a higher dose administered at 5mg/kg/day and prepared in an aqueous suspension., The enhanced bioavailability and improved efficacy observed with the SLH microparticles were attributed to the maintenance of CEL in a solubilised form during digestion of the lipid vehicle. We hypothesise that the presence of silica in the formulation may have contributed to the prevention of drug precipitation in the intestinal lumen by providing an alternative binding site for CEL to adsorb to prior to re-solubilisation and absorption. The study highlights the potential utility of novel SLH microparticle formulations as stable dry powders that possess the properties of a lipid-based formulation for the enhanced delivery and efficacy of poorly water-soluble drugs.


Subject(s)
Arthritis, Infectious/drug therapy , Caprylates/chemistry , Cyclooxygenase 2 Inhibitors/administration & dosage , Glycerides/chemistry , Pyrazoles/administration & dosage , Silicon Dioxide/chemistry , Sulfonamides/administration & dosage , Animals , Arthritis, Infectious/pathology , Biological Availability , Celecoxib , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacokinetics , Dogs , Female , Male , Mycobacterium tuberculosis , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Rats , Rats, Sprague-Dawley , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Treatment Outcome
16.
PLoS One ; 8(3): e59149, 2013.
Article in English | MEDLINE | ID: mdl-23516608

ABSTRACT

Spermatogonia stem cell (SSC) self-renewal and differentiation are tightly regulated processes that ensure a continued production of mature sperm throughout male adulthood. In the present study, we investigated the role of glucocorticoid-induced leucine zipper (GILZ) in maintenance of the male germline and spermatogenesis. GILZ was detectable in germ cells of wild type mice on the day of birth, suggesting a role for GILZ in prospermatogonia and SSC pool formation. Gilz KO mice were generated and adult males were azoospermic and sterile. During the first wave of spermatogenesis in Gilz KO mice, spermatogenesis arrested part way through pachytene of meiosis I. Subsequent waves resulted in a progressive depletion of germ cells through apoptosis to ultimately produce a Sertoli cell-only phenotype. Further, in contrast to wild type littermates, PLZF(+) cells were detected in the peri-luminal region of Gilz KO mice at day 6 post-natal, suggesting a defect in prospermatogonia migration in the absence of GILZ. At age 30 days, transient accumulation of PLZF(+) cells in a subset of tubules and severely compromised spermatogenesis were observed in Gilz KO mice, consistent with defective SSC differentiation. GILZ deficiency was associated with an increase in FOXO1 transcriptional activity, which leads to activation of a selective set of FOXO1 target genes, including a pro-apoptotic protein, BIM. On the other hand, no evidence of a heightened immune response was observed. Together, these results suggest that GILZ suppresses FOXO1 nuclear translocation, promotes SSC differentiation over self-renewal, and favours germ cell survival through inhibition of BIM-dependent pro-apoptotic signals. These findings provide a mechanism for the effects of GILZ on spermatogenesis and strengthen the case for GILZ being a critical molecule in the regulation of male fertility.


Subject(s)
Forkhead Transcription Factors/metabolism , Stem Cells/metabolism , Testis/metabolism , Animals , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatogenesis/genetics , Spermatogenesis/physiology , Spermatogonia/metabolism , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL