Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Eur J Immunol ; 48(9): 1506-1521, 2018 09.
Article in English | MEDLINE | ID: mdl-30004580

ABSTRACT

The production of IL-10, a potent immunosuppressive cytokine, must be strictly regulated to ensure a balanced immune response. IFN-γ, a key cytokine in multiple immune processes and pathologies, is known as an inhibitor of IL-10 production by monocytes and macrophages, but also has some regulatory functions. In the present study, we explored the role of IFN-γ on Toll-like receptor (TLR)-induced IL-10 production in murine peritoneal and spleen cells and in human peripheral blood mononuclear cells. IFN-γ inhibited IL-10 production induced by TLR2, TLR3, TLR4 and TLR7/8 agonists, but stimulated IL-10 production when cells were triggered with CpG oligodeoxynucleotides, a specific TLR9 agonist. The stimulatory effect of IFN-γ on TLR9-induced IL-10 was restricted to B cells. In line with the increased IL-10, B cells stimulated with CpG and IFN-γ profoundly inhibited CD4 T cell proliferation. Further research into the mechanisms involved, revealed that the mitogen-activated protein kinases p38 and JNK are essential players in this stimulatory effect, and that the phosphatase MKP1 - an inhibitor of p38 and JNK activity - is downregulated after combined stimulation with IFN-γ and CpG. Our data may represent a novel immunoregulatory role of IFN-γ in B cells after triggering of TLR9, by stimulating IL-10 production.


Subject(s)
B-Lymphocytes/immunology , CpG Islands/genetics , Interferon-gamma/metabolism , Interleukin-10/biosynthesis , JNK Mitogen-Activated Protein Kinases/metabolism , Toll-Like Receptor 9/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/genetics , Cells, Cultured , Dual Specificity Phosphatase 1/biosynthesis , Humans , Interferon-gamma/genetics , Lymphocyte Activation/genetics , MAP Kinase Signaling System/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Signal Transduction/immunology
2.
Virol J ; 14(1): 240, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29258535

ABSTRACT

BACKGROUND: Hemophagocytic lymphohistiocytosis (HLH) is a rare immunological disorder caused by unbridled activation of T cells and macrophages, culminating in a life-threatening cytokine storm. A genetic and acquired subtype are distinguished, termed primary and secondary HLH, respectively. Clinical manifestations of both forms are frequently preceded by a viral infection, predominantly with herpesviruses. The exact role of the viral infection in the development of the hemophagocytic syndrome remains to be further elucidated. METHODS: We utilized a recently developed murine model of cytomegalovirus-associated secondary HLH and dissected the respective contributions of lytic viral replication and immunopathology in its pathogenesis. RESULTS: HLH-like disease only developed in cytomegalovirus-susceptible mouse strains unable to clear the virus, but the severity of symptoms was not correlated to the infectious viral titer. Lytic viral replication and sustained viremia played an essential part in the pathogenesis since abortive viral infection was insufficient to induce a full-blown HLH-like syndrome. Nonetheless, a limited set of symptoms, in particular anemia, thrombocytopenia and elevated levels of soluble CD25, appeared less dependent of the viral replication but rather mediated by the host's immune response, as corroborated by immunosuppressive treatment of infected mice with dexamethasone. CONCLUSION: Both virus-mediated pathology and immunopathology cooperate in the pathogenesis of full-blown virus-associated secondary HLH and are closely entangled. A certain level of viremia appears necessary to elicit the characteristic HLH-like symptoms in the model.


Subject(s)
Disease Models, Animal , Lymphohistiocytosis, Hemophagocytic/physiopathology , Lymphohistiocytosis, Hemophagocytic/virology , Muromegalovirus/physiology , Virus Diseases/physiopathology , Virus Replication/physiology , Animals , Antiviral Agents/pharmacology , Cidofovir , Cytosine/analogs & derivatives , Cytosine/pharmacology , Dexamethasone/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Interleukin-2 Receptor alpha Subunit/drug effects , Interleukin-2 Receptor alpha Subunit/immunology , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Organophosphonates/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/physiology , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/physiology , Virus Replication/drug effects
3.
Front Immunol ; 8: 1102, 2017.
Article in English | MEDLINE | ID: mdl-28936212

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening, hyperinflammatory syndrome, characterized by the uncontrolled activation of macrophages and T cells, eliciting key symptoms such as persistent fever, hepatosplenomegaly, pancytopenia, hemophagocytosis, hyperferritinemia, and coagulopathy. Viral infections are frequently implicated in the onset of active HLH episodes, both in primary, genetic HLH as in the secondary, acquired form. Infections with herpesviruses such as Epstein-Barr virus and cytomegalovirus are the most common. In autoimmune diseases, a link between viral infections and autoreactive immune responses has been recognized for a considerable time. However, the mechanisms by which viruses contribute to HLH pathogenesis remain to be clarified. In this viewpoint, different factors that may come into play are discussed. Viruses, particularly larger DNA viruses such as herpesviruses, are potent modulators of the immune response. By evading immune recognition, interfering with cytokine balances and inhibiting apoptotic pathways, viruses may increase the host's susceptibility to HLH development. In particular cases, a direct connection between the viral infection and inhibition of natural killer cell or T cell cytotoxicity was reported, indicating that viruses may create immunological deficiencies reminiscent of primary HLH.

4.
Arthritis Rheumatol ; 69(1): 213-224, 2017 01.
Article in English | MEDLINE | ID: mdl-27696741

ABSTRACT

OBJECTIVE: Systemic juvenile idiopathic arthritis (JIA) is an immunoinflammatory disease characterized by arthritis and systemic manifestations. The role of natural killer (NK) cells in the pathogenesis of systemic JIA remains unclear. The purpose of this study was to perform a comprehensive analysis of NK cell phenotype and functionality in patients with systemic JIA. METHODS: Transcriptional alterations specific to NK cells were investigated by RNA sequencing of highly purified NK cells from 6 patients with active systemic JIA and 6 age-matched healthy controls. Cytokines (NK cell-stimulating and others) were quantified in plasma samples (n = 18). NK cell phenotype and cytotoxic activity against tumor cells were determined (n = 10), together with their interferon-γ (IFNγ)-producing function (n = 8). RESULTS: NK cells from the systemic JIA patients showed an altered gene expression profile compared to cells from the healthy controls, with enrichment of immunoinflammatory pathways, increased expression of innate genes including TLR4 and S100A9, and decreased expression of immune-regulating genes such as IL10RA and GZMK. In the patients' plasma, interleukin-18 (IL-18) levels were increased, and a decreased ratio of IFNγ to IL-18 was observed. NK cells from the patients exhibited specific alterations in the balance of inhibitory and activating receptors, with decreased killer cell lectin-like receptor G1 and increased NKp44 expression. Although NK cells from the patients showed increased granzyme B expression, consistent with intact cytotoxicity and degranulation against a tumor cell line, decreased granzyme K expression in CD56bright NK cells and defective IL-18-induced IFNγ production and signaling were demonstrated. CONCLUSION: NK cells are active players in the inflammatory environment typical of systemic JIA. Although their cytotoxic function is globally intact, subtle defects in NK-related pathways, such as granzyme K expression and IL-18-driven IFNγ production, may contribute to the immunoinflammatory dysregulation in this disease.


Subject(s)
Arthritis, Juvenile/immunology , Granzymes , Interferon-gamma , Killer Cells, Natural/physiology , Arthritis, Juvenile/genetics , Cells, Cultured , Gene Expression , Granzymes/genetics , Humans , Interferon-gamma/genetics , Phenotype
5.
Br J Haematol ; 174(2): 175-87, 2016 07.
Article in English | MEDLINE | ID: mdl-27292929

ABSTRACT

The cytokine storm syndrome 'haemophagocytic lymphohistiocytosis' (HLH) is an under-recognized hyperinflammatory disorder, causing high morbidity and mortality risk in children and adults. It can be subdivided into a primary, genetic form and a secondary, acquired form that complicates diverse infections, malignancies and autoimmune or autoinflammatory disorders. Both subtypes present with the same spectrum of non-specific symptoms, making accurate diagnosis and rapid treatment initiation challenging. In the last decade, increased awareness and international collaborative efforts fuelled a marked progress in diagnostic protocols and novel treatment strategies for HLH and new diagnostic guidelines are being tailored to specific secondary HLH subtypes. Therapy is gradually shifting its focus from overall immunosuppression towards targeting specific cytokines, cell types or signalling pathways underlying pathophysiology. Nevertheless, continued research efforts remain indispensable to customize therapy to individual patient needs.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/therapy , Diagnosis, Differential , Humans , Lymphohistiocytosis, Hemophagocytic/classification , Lymphohistiocytosis, Hemophagocytic/etiology , Molecular Targeted Therapy
6.
Br J Haematol ; 174(2): 203-17, 2016 07.
Article in English | MEDLINE | ID: mdl-27264204

ABSTRACT

Haemophagocytic lymphohistiocytosis (HLH) comprises a heterogeneous spectrum of hyperinflammatory conditions that are inherited (primary HLH) or acquired in a context of infections, malignancies or autoimmune/autoinflammatory disorders (secondary HLH). Genetic defects in the cytotoxic machinery of natural killer and CD8(+) T cells underlie primary HLH, with residual cytotoxicity determining disease severity. Improved sequencing techniques have expanded the range of causal mutations and have redefined many cases of secondary HLH as primary HLH and vice versa, blurring the distinction between both subtypes. These insights allow HLH to be conceptualized as a threshold disease, in which interplay between various genetic and environmental factors causes progressive inflammation into a critical point, beyond which uncontrolled activation of immune cells and excessive cytokine production give rise to the cardinal symptoms of HLH. Various pathogenic pathways may thus converge to a common end stage of fulminant HLH.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/diagnosis , CD8-Positive T-Lymphocytes/pathology , Diagnosis, Differential , Humans , Inflammation , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/genetics , Mutation , Natural Killer T-Cells/pathology , Sequence Analysis, DNA
7.
J Immunol ; 196(7): 3124-34, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26903481

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening immunological disorder that is characterized by systemic inflammation, widespread organ damage, and hypercytokinemia. Primary HLH is caused by mutations in granule-mediated cytotoxicity, whereas secondary HLH occurs, without a known genetic background, in a context of infections, malignancies, or autoimmune and autoinflammatory disorders. Clinical manifestations of both HLH subtypes are often precipitated by a viral infection, predominantly with Herpesviridae. Exploiting this knowledge, we established an animal model of virus-associated secondary HLH by infecting immunocompetent wild-type mice with the ß-herpesvirus murine CMV. C57BL/6 mice developed a mild inflammatory phenotype, whereas BALB/c mice displayed the clinicopathologic features of HLH, as set forth in the Histiocyte Society diagnostic guidelines: fever, cytopenia, hemophagocytosis, hyperferritinemia, and elevated serum levels of soluble CD25. BALB/c mice also developed lymphadenopathy, liver dysfunction, and decreased NK cell numbers. Lymphoid and myeloid cells were in a hyperactivated state. Nonetheless, depletion of CD8(+) T cells could not inhibit or cure the HLH-like syndrome, highlighting a first dissimilarity from mouse models of primary HLH. Immune cell hyperactivation in BALB/c mice was accompanied by a cytokine storm. Notably, plasma levels of IFN-γ, a key pathogenic cytokine in models of primary HLH, were the highest. Nevertheless, murine CMV-infected IFN-γ-deficient mice still developed the aforementioned HLH-like symptoms. In fact, IFN-γ-deficient mice displayed a more complete spectrum of HLH, including splenomegaly, coagulopathy, and decreased NK cell cytotoxicity, indicating a regulatory role for IFN-γ in the pathogenesis of virus-associated secondary HLH as opposed to its central pathogenic role in primary HLH.


Subject(s)
Herpesviridae Infections/complications , Lymphohistiocytosis, Hemophagocytic/etiology , Muromegalovirus/physiology , Animals , Biomarkers , Cytokines/genetics , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Herpesviridae Infections/virology , Histiocytes/immunology , Histiocytes/metabolism , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Liver/virology , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
8.
PLoS One ; 11(2): e0150075, 2016.
Article in English | MEDLINE | ID: mdl-26914138

ABSTRACT

OBJECTIVES: Indoleamine 2,3-dioxygenase-1 (IDO1) is an immune-modulatory enzyme that catalyzes the degradation of tryptophan (Trp) to kynurenine (Kyn) and is strongly induced by interferon (IFN)-γ. We previously reported highly increased levels of IFN-γ and corresponding IDO activity in patients with hemophagocytic lymphohistiocytosis (HLH), a hyper-inflammatory syndrome. On the other hand, IFN-γ and IDO were low in patients with systemic juvenile idiopathic arthritis (sJIA), an autoinflammatory syndrome. As HLH can occur as a complication of sJIA, the opposing levels of both IFN-γ and IDO are remarkable. In animal models for sJIA and HLH, the role of IFN-γ differs from being protective to pathogenic. In this study, we aimed to unravel the role of IDO1 in the pathogenesis of sJIA and HLH. METHODS: Wild-type and IDO1-knockout (IDO1-KO) mice were used in 3 models of sJIA or HLH: complete Freund's adjuvant (CFA)-injected mice developed an sJIA-like syndrome and secondary HLH (sHLH) was evoked by either repeated injection of unmethylated CpG oligonucleotide or by primary infection with mouse cytomegalovirus (MCMV). An anti-CD3-induced cytokine release syndrome was used as a non-sJIA/HLH control model. RESULTS: No differences were found in clinical, laboratory and hematological features of sJIA/HLH between wild-type and IDO1-KO mice. As IDO modulates the immune response via induction of regulatory T cells and inhibition of T cell proliferation, we investigated both features in a T cell-triggered cytokine release syndrome. Again, no differences were observed in serum cytokine levels, percentages of regulatory T cells, nor of proliferating or apoptotic thymocytes and lymph node cells. CONCLUSIONS: Our data demonstrate that IDO1 deficiency does not affect inflammation in sJIA, sHLH and a T cell-triggered cytokine release model. We hypothesize that other tryptophan-catabolizing enzymes like IDO2 and tryptophan 2,3-dioxygenase (TDO) might compensate for the lack of IDO1.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Juvenile/pathology , Cytokines/blood , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Lymphohistiocytosis, Hemophagocytic/pathology , T-Lymphocytes/immunology , Animals , Apoptosis/immunology , Arthritis, Experimental/immunology , Arthritis, Juvenile/genetics , Cell Proliferation , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Freund's Adjuvant/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphohistiocytosis, Hemophagocytic/genetics , Macrophage Activation Syndrome/genetics , Macrophage Activation Syndrome/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Tryptophan Oxygenase/metabolism
9.
Rheumatology (Oxford) ; 54(8): 1507-17, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25767156

ABSTRACT

OBJECTIVES: To study the role of IFN-γ in the pathogenesis of systemic JIA (sJIA) and haemophagocytic lymphohistiocytosis (HLH) by searching for an IFN-γ profile, and to assess its relationship with other cytokines. METHODS: Patients with inactive (n = 10) and active sJIA (n = 10), HLH [n = 5; of which 3 had sJIA-associated macrophage activation syndrome (MAS)] and healthy controls (n = 16) were enrolled in the study. Cytokines and IFN-γ-induced genes and proteins were determined in plasma, in patient peripheral blood mononuclear cells (PBMCs) and in lymph node biopsies of one patient during both sJIA and MAS episodes. IFN-γ responses were investigated in healthy donor PBMCs, primary fibroblasts and endothelial cells. RESULTS: Plasma IFN-γ, IL-6 and IL-18 were elevated in active sJIA and HLH. Levels of IFN-γ and IFN-γ-induced proteins (IP-10/CXCL-10, IL-18BP and indoleamine 2,3-dioxygenase) in HLH were much higher than levels in active sJIA. Free IL-18 and ratios of IL-18/IFN-γ were higher in active sJIA compared with HLH. HLH PBMCs showed hyporesponsiveness to IFN-γ in vitro when compared with control and sJIA PBMCs. Endothelial cells and fibroblasts expressed IFN-γ-induced proteins in situ in lymph node staining of a MAS patient and in vitro upon stimulation with IFN-γ. CONCLUSION: Patients with active sJIA and HLH/MAS show distinct cytokine profiles, with highly elevated plasma levels of IFN-γ and IFN-γ-induced proteins typically found in HLH/MAS. In addition to PBMCs, histiocytes, endothelial cells and fibroblasts may contribute to an IFN-γ profile in plasma. Increasing levels of IFN-γ compared with IL-18 may raise suspicion about the development of MAS in sJIA.


Subject(s)
Arthritis, Juvenile/metabolism , Cytokines/metabolism , Interferon-gamma/metabolism , Interleukin-18/metabolism , Lymphohistiocytosis, Hemophagocytic/metabolism , Macrophage Activation Syndrome/metabolism , Adolescent , Arthritis, Juvenile/diagnosis , Arthritis, Juvenile/pathology , Biopsy , Case-Control Studies , Child , Child, Preschool , Diagnosis, Differential , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Infant , Interleukin-6/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/pathology , Macrophage Activation Syndrome/diagnosis , Macrophage Activation Syndrome/pathology , Male , Young Adult
10.
Cytokine Growth Factor Rev ; 26(3): 263-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25466631

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) comprises a group of life-threatening immune disorders classified into primary or secondary HLH. The former is caused by mutations in genes involved in granule-mediated cytotoxicity, the latter occurs in a context of infections, malignancies or autoimmune/autoinflammatory disorders. Both are characterized by systemic inflammation, severe cytokine storms and immune-mediated organ damage. Despite recent advances, the pathogenesis of HLH remains incompletely understood. Animal models resembling different subtypes of HLH are therefore of great value to study this disease and to uncover novel treatment strategies. In this review, all known animal models of HLH will be discussed, highlighting findings on cell types, cytokines and signaling pathways involved in disease pathogenesis and extrapolating therapeutic implications for the human situation.


Subject(s)
Cytokines/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , Animals , Disease Models, Animal , Humans , Lymphohistiocytosis, Hemophagocytic/drug therapy
11.
Arthritis Rheumatol ; 66(5): 1340-51, 2014 May.
Article in English | MEDLINE | ID: mdl-24470407

ABSTRACT

OBJECTIVE: Systemic juvenile idiopathic arthritis (JIA) is unique among the rheumatic diseases of childhood, given its distinctive systemic inflammatory character. Inappropriate control of innate immune responses following an initially harmless trigger is thought to account for the excessive inflammatory reaction. The aim of this study was to generate a similar systemic inflammatory syndrome in mice by injecting a relatively innocuous, yet persistent, immune system trigger: Freund's complete adjuvant (CFA), containing heat-killed mycobacteria. METHODS: Given the central role of interferon-γ (IFNγ) in immune regulation, we challenged wild-type (WT) and IFNγ-knockout (KO) BALB/c mice with CFA, and analyzed their clinical symptoms and biologic characteristics. The production of cytokines and the effects of anticytokine antibodies were investigated. RESULTS: In WT mice, CFA injection resulted in splenomegaly, lymphadenopathy, neutrophilia, thrombocytosis, and increased cytokine expression. In the absence of IFNγ, these symptoms were more pronounced and were accompanied by weight loss, arthritis, anemia, hemophagocytosis, abundance of immature blood cells, and increased levels of interleukin-6 (IL-6), all of which are reminiscent of the symptoms of systemic JIA. CFA-challenged IFNγ-KO mice showed increased expression of IL-17 by CD4+ T cells and by innate γ/δ T cells. Inflammatory and hematologic changes were prevented by treatment with anti-IL-12/IL-23p40 and anti-IL-17 antibodies. CONCLUSION: Immune stimulation of IFNγ-KO mice with CFA produces a systemic inflammatory syndrome reflecting the clinical, biologic, and histopathologic picture of systemic JIA. The protective function of IFNγ in preventing anemia and overall systemic inflammation is a striking observation. The finding that both adaptive and innate T cells are important sources of IL-17 may be of relevance in the pathogenesis of systemic JIA.


Subject(s)
Arthritis, Juvenile/chemically induced , Arthritis, Juvenile/physiopathology , Disease Models, Animal , Freund's Adjuvant/adverse effects , Immune System/physiopathology , Interferon-gamma/deficiency , Interferon-gamma/physiology , Adaptive Immunity/physiology , Anemia/metabolism , Anemia/physiopathology , Animals , Arthritis, Juvenile/metabolism , Cytokines/metabolism , Female , Freund's Adjuvant/pharmacology , Immune System/drug effects , Immunity, Innate/physiology , Inflammation/metabolism , Inflammation/physiopathology , Interferon-gamma/genetics , Interleukin-17/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...