Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
Front Immunol ; 10: 3089, 2019.
Article in English | MEDLINE | ID: mdl-32010140

ABSTRACT

Natural killer (NK) cells are innate immune lymphocytes with potent cytolytic and immune-regulatory activities. NK cells are well-known for their ability to kill infected and malignant cells in a fast and non-specific way without prior sensitization. For this purpose, NK cells are equipped with a set of cytotoxic molecules such as perforin and apoptosis-inducing proteins. NK cells also have the capacity to produce large amounts of cytokines and chemokines that synergize with their cytotoxic function and that ensure interaction with other immune cells. A less known feature of NK cells is their capacity to kill non-infected autologous cells, such as immature dendritic cells and activated T cells and monocytes. Via the release of large amounts of TNF-α and IFN-γ, NK cells may contribute to disease pathology. Conversely they may exert a regulatory role through secretion of immuno-regulatory cytokines such as GM-CSF, IL-13, and IL-10. Thus, NK cells may be important target and effector cells in the pathogenesis of autoinflammatory diseases, in particular in those disorders associated with a cytokine storm or in conditions where immune cells are highly activated. Key examples of such diseases are systemic juvenile idiopathic arthritis (sJIA) and its well-associated complication, macrophage activation syndrome (MAS). sJIA is a chronic childhood immune disorder of unknown etiology, characterized by arthritis and systemic inflammation, including a daily spiking fever and evanescent rash. MAS is a potentially fatal complication of autoimmune and autoinflammatory diseases, and most prevalently associated with sJIA. MAS is considered as a subtype of hemophagocytic lymphohistiocytosis (HLH), a systemic hyperinflammatory disorder characterized by defective cytotoxic pathways of cytotoxic T and NK cells. In this review, we describe the established features of NK cells and provide the results of a literature survey on the reported NK cell abnormalities in monogenic and multifactorial autoinflammatory disorders. Finally, we discuss the role of NK cells in the pathogenesis of sJIA and MAS.


Subject(s)
Arthritis, Juvenile/immunology , Killer Cells, Natural/immunology , Macrophage Activation Syndrome/immunology , Arthritis, Juvenile/pathology , Child , Cytokines/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Killer Cells, Natural/pathology , Macrophage Activation Syndrome/pathology
3.
J Allergy Clin Immunol ; 143(1): 359-368, 2019 01.
Article in English | MEDLINE | ID: mdl-30273710

ABSTRACT

BACKGROUND: Postzygotic de novo mutations lead to the phenomenon of gene mosaicism. The 3 main types are called somatic, gonadal, and gonosomal mosaicism, which differ in terms of the body distribution of postzygotic mutations. Mosaicism has been reported occasionally in patients with primary immunodeficiency diseases (PIDs) since the early 1990s, but its real involvement has not been systematically addressed. OBJECTIVE: We sought to investigate the incidence of gene mosaicism in patients with PIDs. METHODS: The amplicon-based deep sequencing method was used in the 3 parts of the study that establish (1) the allele frequency of germline variants (n = 100), (2) the incidence of parental gonosomal mosaicism in families with PIDs with de novo mutations (n = 92), and (3) the incidence of mosaicism in families with PIDs with moderate-to-high suspicion of gene mosaicism (n = 36). Additional investigations evaluated body distribution of postzygotic mutations, their stability over time, and their characteristics. RESULTS: The range of allele frequency (44.1% to 55.6%) was established for germline variants. Those with minor allele frequencies of less than 44.1% were assumed to be postzygotic. Mosaicism was detected in 30 (23.4%) of 128 families with PIDs, with a variable minor allele frequency (0.8% to 40.5%). Parental gonosomal mosaicism was detected in 6 (6.5%) of 92 families with de novo mutations, and a high incidence of mosaicism (63.9%) was detected among families with moderate-to-high suspicion of gene mosaicism. In most analyzed cases mosaicism was found to be both uniformly distributed and stable over time. CONCLUSION: This study represents the largest performed to date to investigate mosaicism in patients with PIDs, revealing that it affects approximately 25% of enrolled families. Our results might have serious consequences regarding treatment and genetic counseling and reinforce the use of next-generation sequencing-based methods in the routine analyses of PIDs.


Subject(s)
Alleles , Gene Frequency , Immunologic Deficiency Syndromes/genetics , Mosaicism , Family , Female , High-Throughput Nucleotide Sequencing , Humans , Immunologic Deficiency Syndromes/immunology , Male
4.
Rheumatology (Oxford) ; 57(7): 1299-1304, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29635517

ABSTRACT

OBJECTIVE: To identify biomarkers of articular and ocular disease activity in patients with Blau syndrome (BS). METHODS: Multiplex plasma protein arrays were performed in five BS patients and eight normal healthy volunteers (NHVs). Plasma S100A12 and S100A8/9 were subsequently measured by ELISA at baseline and 1-year follow-up in all patients from a prospective multicentre cohort study. CRP was measured using Meso Scale Discovery immunoassay. Active joint counts, standardization uveitis nomenclature for anterior uveitis cells and vitreous haze by Nussenblatt scale were the clinical parameters. RESULTS: Multiplex Luminex arrays identified S100A12 as the most significantly elevated protein in five selected BS vs eight NHVs and this was confirmed by ELISA on additional samples from the same five BS patients. In the patient cohort, S100A12 (n = 39) and S100A8/9 (n = 33) were significantly higher compared with NHVs (n = 44 for S100A12, n = 40 for S100A8/9) (P = 0.0000004 and P = 0.0003, respectively). Positive correlations between active joint counts and S100 levels were significant for S100A12 (P = 0.0008) and S100A8/9 (P = 0.015). CRP levels did not correlate with active joint count. Subgroup analysis showed significant association of S100 proteins with active arthritis (S100A12 P = 0.01, S100A8/9 P = 0.008). Active uveitis was not associated with increased S100 levels. CONCLUSION: S100 proteins are biomarkers of articular disease activity in BS and potential outcome measures in future clinical trials. As secreted neutrophil and macrophage products, S100 proteins may reflect the burden of granulomatous tissue in BS.

5.
Am J Ophthalmol ; 187: 158-166, 2018 03.
Article in English | MEDLINE | ID: mdl-28887115

ABSTRACT

PURPOSE: Provide baseline and preliminary follow-up results in a 5-year longitudinal study of Blau syndrome. DESIGN: Multicenter, prospective interventional case series. METHODS: Baseline data from 50 patients from 25 centers worldwide, and follow-up data for patients followed 1, 2, or 3 years at the end of study enrollment. Ophthalmic data were collected at baseline and yearly visits by means of a standardized collection form. RESULTS: Median age at onset of eye disease was 60 months and duration of eye disease at baseline 145 months. At baseline 38 patients (78%) had uveitis, which was bilateral in 37 (97%). Eight patients (21%) had moderate to severe visual impairment. Panuveitis was found in 38 eyes (51%), with characteristic multifocal choroidal infiltrates in 29 eyes (39%). Optic disc pallor in 9 eyes (12%) and peripapillary nodules in 9 eyes (12%) were the commonest signs of optic nerve involvement. Active anterior chamber inflammation was noted in 30 eyes (40%) at baseline and in 16 (34%), 17 (57%), and 11 (61%) eyes at 1, 2, and 3 years, respectively. Panuveitis was associated with longer disease duration. At baseline, 56 eyes (75%) were on topical corticosteroids. Twenty-six patients (68%) received a combination of systemic corticosteroids and immunomodulatory therapy. CONCLUSIONS: Blau uveitis is characterized by progressive panuveitis with multifocal choroiditis, resulting in severe ocular morbidity despite continuous systemic and local immunomodulatory therapy. The frequency and severity of Blau uveitis highlight the need for close ophthalmologic surveillance as well as a search for more effective therapies.


Subject(s)
Arthritis/diagnosis , Synovitis/diagnosis , Uveitis/diagnosis , Adolescent , Adult , Antihypertensive Agents/therapeutic use , Arthritis/drug therapy , Arthritis/physiopathology , Child , Child, Preschool , Choroiditis/diagnosis , Choroiditis/drug therapy , Choroiditis/physiopathology , Cross-Sectional Studies , Disease Progression , Female , Follow-Up Studies , Global Health , Glucocorticoids/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Infant , Male , Middle Aged , Multifocal Choroiditis , Prospective Studies , Sarcoidosis , Synovitis/drug therapy , Synovitis/physiopathology , Uveitis/drug therapy , Uveitis/physiopathology , Visual Acuity/physiology
6.
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
7.
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.

8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Rheumatology (Oxford) ; 54(6): 1008-16, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25416713

ABSTRACT

OBJECTIVE: To report baseline articular, functional and ocular findings of the first international prospective cohort study of Blau syndrome (BS). METHODS: Three-year, multicentre, observational study on articular, functional (HAQ, Childhood HAQ and VAS global and pain), ophthalmological, therapeutic and radiological data in BS patients. RESULTS: Baseline data on the first 31 recruited patients (12 females and 19 males) from 18 centres in 11 countries are presented. Of the 31 patients, 11 carried the p.R334W NOD2 mutation, 9 the p.R334Q and 11 various other NOD2 missense mutations; 20 patients were sporadic and 11 from five BS pedigrees. Median disease duration was 12.8 years (1.1-57). Arthritis, documented in all but one patient, was oligoarticular in 7, polyarticular in 23. The median active joint count was 21. Functional capacity was normal in 41%, mildly impaired in 31% and moderate-severe in 28% of patients. The most frequently involved joints at presentation were wrists, ankles, knees and PIPs. On radiographs, a symmetrical non-erosive arthropathy was shown. Previously unknown dysplastic bony changes were found in two-thirds of patients. Ocular disease was documented in 25 of 31 patients, with vitreous inflammation in 64% and moderate-severe visual loss in 33%. Expanded manifestations (visceral, vascular) beyond the classic clinical triad were seen in 52%. CONCLUSION: BS is associated with severe ocular and articular morbidity. Visceral involvement is common and may be life-threatening. Bone dysplastic changes may show diagnostic value and suggest a previously unknown role of NOD2 in bone morphogenesis. BS is resistant to current drugs, suggesting the need for novel targeted therapies.


Subject(s)
Arthritis , Cranial Nerve Diseases , Eye Diseases , Nod2 Signaling Adaptor Protein/genetics , Skin Diseases , Synovitis , Uveitis , Adolescent , Adult , Arthritis/diagnostic imaging , Arthritis/drug therapy , Arthritis/genetics , Arthritis/physiopathology , Child , Child, Preschool , Cranial Nerve Diseases/diagnostic imaging , Cranial Nerve Diseases/drug therapy , Cranial Nerve Diseases/genetics , Cranial Nerve Diseases/physiopathology , Cross-Sectional Studies , Eye Diseases/drug therapy , Eye Diseases/genetics , Eye Diseases/physiopathology , Female , Humans , Infant , Male , Middle Aged , Mutation, Missense , Prospective Studies , Radiography , Sarcoidosis , Skin Diseases/drug therapy , Skin Diseases/genetics , Skin Diseases/physiopathology , Synovitis/diagnostic imaging , Synovitis/drug therapy , Synovitis/genetics , Synovitis/physiopathology , Treatment Outcome , Uveitis/diagnostic imaging , Uveitis/drug therapy , Uveitis/genetics , Uveitis/physiopathology , Young Adult
15.
Cytokine Growth Factor Rev ; 26(1): 35-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24948570

ABSTRACT

Systemic juvenile idiopathic arthritis (sJIA) is a severe inflammatory childhood disorder, characterized by a specific pattern of systemic features and a typical cytokine profile. Patients are at risk to develop macrophage activation syndrome (MAS), an acute life-threatening condition defined by excessive proliferation and activation of macrophages and T cells. Defects of unknown cause in the natural killer (NK) cell cytotoxic capacity are presumed to underlie the pathogenesis of MAS and have been detected in sJIA patients. Here, we provide an overview of the cytokine profiles in sJIA and related mouse models. We discuss the influence of cytokines on NK cell function, and hypothesize that NK cell dysfunction in sJIA is caused by altered cytokine profiles.


Subject(s)
Arthritis, Juvenile/immunology , Arthritis, Juvenile/physiopathology , Cytokines/metabolism , Killer Cells, Natural/immunology , Animals , Arthritis, Juvenile/complications , Humans , Macrophage Activation Syndrome/etiology , Macrophage Activation Syndrome/physiopathology , Mice , T-Lymphocytes/immunology
16.
Article in English | MEDLINE | ID: mdl-25136265

ABSTRACT

Blau syndrome is a monogenic disease resulting from mutations in the pattern recognition receptor NOD2, and is phenotypically characterized by the triad of granulomatous polyarthritis, dermatitis and uveitis. This paper reviews briefly the classical clinical features of the disease, as well as more recently described extra-triad symptoms. From an ongoing prospective multicenter study, we provide new data on the natural history of Blau syndrome, focusing on functional status and visual outcome. We also present an update of the range of different NOD2 mutations found in Blau syndrome as well as recent data on morphologic and immunohistochemical characteristics of the Blau granuloma. Finally, emerging insights into pathogenic mechanisms including activation of NOD2 signal transduction, and potential biomarkers of disease activity are discussed.


Subject(s)
Cranial Nerve Diseases/genetics , Cranial Nerve Diseases/pathology , Granuloma/genetics , Granuloma/pathology , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Synovitis/genetics , Synovitis/pathology , Uveitis/genetics , Uveitis/pathology , Arthritis/genetics , Arthritis/pathology , Cranial Nerve Diseases/physiopathology , Dermatitis/genetics , Dermatitis/pathology , Granuloma/physiopathology , Hereditary Autoinflammatory Diseases/physiopathology , Humans , Mutation/genetics , Nod2 Signaling Adaptor Protein/genetics , Prospective Studies , Sarcoidosis , Signal Transduction/physiology , Synovitis/physiopathology , Uveitis/physiopathology
18.
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
19.
J Rheumatol ; 39(9): 1888-92, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22859352

ABSTRACT

OBJECTIVE: To put forward a new concept--Blau arteritis, a form of large-vessel vasculitis phenotypically related to Takayasu disease but genetically and clinically part of an expanded phenotype of Blau syndrome. METHODS: We provide a clinical description of a new case and summarize previously published cases of arteritis associated with Blau syndrome. Genetic testing was performed by direct sequencing of exon 4 of the NOD2 gene. RESULTS: The case described and those reviewed from the literature demonstrate the emerging phenotype of Takayasu-like arteritis in patients with Blau syndrome. Although most patients described to date depict an otherwise classic Blau syndrome phenotype, the current case was atypical in that the predominant features were arteritic. A novel substitution, G464W, in a highly conserved position near the nucleotide oligomerization domain of the NOD2 protein is also described. CONCLUSION: Blau arteritis can be observed in the context of both typical and atypical (incomplete) Blau syndrome. The associated mutation in the NOD2 gene raises the question of the potential importance of this gene among patients with "primary" forms of Takayasu arteritis.


Subject(s)
Arteritis/genetics , Cranial Nerve Diseases/genetics , Nod2 Signaling Adaptor Protein/genetics , Synovitis/genetics , Uveitis/genetics , Arthritis , Child , Female , Humans , Mutation , Phenotype , Sarcoidosis , Syndrome
20.
J Allergy Clin Immunol ; 129(4): 1076-84, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22464675

ABSTRACT

BACKGROUND: Blau syndrome (BS) and Crohn disease (CD) are both characterized by granulomatous inflammation and related to nucleotide oligomerization domain 2 (NOD2) mutations. OBJECTIVE: This study aimed to define the morphologic and immunohistochemical characteristics of granulomas in patients with NOD2-related BS and CD. METHODS: Granuloma-containing biopsy specimens from 6 patients with BS and 7 pediatric patients with CD carrying NOD2 mutations or single nucleotide polymorphisms were studied for morphology, cellular composition, and cytokine expression by using hematoxylin and eosin staining and immunohistochemistry. RESULTS: Biopsy specimens from patients with BS typically showed polycyclic granulomas with large lymphocytic coronas, extensive emperipolesis of lymphocytes within multinucleated giant cells (MGCs), MGC death, and fibrinoid necrosis and fibrosis. In contrast, biopsy specimens from patients with CD showed simple granulomas with subtle/absent lymphocytic coronas, sclerosis of the surrounding tissue, and polymorphonuclear cells. Findings found to be similar in all granulomas were as follows: CD68 and HLA-DR expression by epithelioid cells, monocyte-macrophage lineage cells and MGCs, increased lymphocytic HLA-DR expression, increased CD4(+)/CD8(+) T-cell ratio, and CD20(+) B lymphocytes evenly distributed within and around granulomas. In both patient groups prominent IFN-γ expression was found in and around granulomas, and TNF-α and IL-23 receptor expression was moderate. IL-6, IL-17, and TGF-ß expression was prominent in granulomas from patients with BS but sporadic in granulomas from patients with CD. IL-10 expression was absent. CONCLUSION: Granulomas from patients with BS and granulomas from patients with NOD2-associated CD show distinct morphologic features and cytokine expression patterns, suggesting that the T(H)17 axis might be involved in the pathogenesis of BS, whereas T(H)1 is important in both patients with BS and patients with CD.


Subject(s)
Cranial Nerve Diseases/genetics , Cranial Nerve Diseases/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Granuloma/genetics , Granuloma/pathology , Nod2 Signaling Adaptor Protein/genetics , Synovitis/genetics , Synovitis/pathology , Uveitis/genetics , Uveitis/pathology , Adolescent , Arthritis , Child , Child, Preschool , Cranial Nerve Diseases/metabolism , Crohn Disease/immunology , Cytokines/metabolism , Female , Granuloma/immunology , Humans , Immunohistochemistry , Infant , Male , Mutation , Nod2 Signaling Adaptor Protein/immunology , Sarcoidosis , Synovitis/metabolism , Uveitis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...