Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Exp Med ; 220(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37273177

ABSTRACT

Inborn errors of the NF-κB pathways underlie various clinical phenotypes in humans. Heterozygous germline loss-of-expression and loss-of-function mutations in RELA underlie RELA haploinsufficiency, which results in TNF-dependent chronic mucocutaneous ulceration and autoimmune hematological disorders. We here report six patients from five families with additional autoinflammatory and autoimmune manifestations. These patients are heterozygous for RELA mutations, all of which are in the 3' segment of the gene and create a premature stop codon. Truncated and loss-of-function RelA proteins are expressed in the patients' cells and exert a dominant-negative effect. Enhanced expression of TLR7 and MYD88 mRNA in plasmacytoid dendritic cells (pDCs) and non-pDC myeloid cells results in enhanced TLR7-driven secretion of type I/III interferons (IFNs) and interferon-stimulated gene expression in patient-derived leukocytes. Dominant-negative mutations in RELA thus underlie a novel form of type I interferonopathy with systemic autoinflammatory and autoimmune manifestations due to excessive IFN production, probably triggered by otherwise non-pathogenic TLR ligands.


Subject(s)
Autoimmunity , Interferon Type I , Transcription Factor RelA , Humans , Autoimmunity/genetics , Dendritic Cells , Interferon Type I/genetics , Interferon Type I/metabolism , NF-kappa B/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
2.
Chemosphere ; 337: 139331, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37379990

ABSTRACT

The three environmental degradation tests of hydrolysis, indirect photolysis and Zahn-Wellens microbial degradation were conducted according to the OECD and the US EPA guidelines on DEMNUM, a typical linear perfluoropolyether polymer. Low mass degradation products that formed in each test were structurally characterized and indirectly quantified by liquid chromatography mass spectrometry (LC/MS) using a reference compound and an internal standard of similar structure. The degradation of the polymer was assumed to directly correlate with the appearance of lower mass species. The hydrolysis experiment at 50 °C showed the appearance of less than a dozen low mass species with increasing pH but at the negligible total estimated amount of ∼2 ppm relative to polymer. A dozen low mass perfluoro acid entities also appeared following the indirect photolysis experiment in synthetic humic water. Their maximum total amount was at ∼150 ppm relative to polymer. The largest total amount of low mass species formed during the Zahn-Wellens biodegradation test amounted to only ∼80 ppm relative to polymer. The Zahn-Wellens conditions tended to produce larger low mass molecules than the ones formed under photolysis. The results from all three tests indicate that the polymer is stable and non-degradable in the environment.


Subject(s)
Biodegradation, Environmental , Chromatography, Liquid , Mass Spectrometry , Photolysis , Hydrolysis
3.
Front Immunol ; 13: 905960, 2022.
Article in English | MEDLINE | ID: mdl-36211342

ABSTRACT

Purpose: Upregulation of type I interferon (IFN) signaling has been increasingly detected in inflammatory diseases. Recently, upregulation of the IFN signature has been suggested as a potential biomarker of IFN-driven inflammatory diseases. Yet, it remains unclear to what extent type I IFN is involved in the pathogenesis of undifferentiated inflammatory diseases. This study aimed to quantify the type I IFN signature in clinically undiagnosed patients and assess clinical characteristics in those with a high IFN signature. Methods: The type I IFN signature was measured in patients' whole blood cells. Clinical and biological data were collected retrospectively, and an intensive genetic analysis was performed in undiagnosed patients with a high IFN signature. Results: A total of 117 samples from 94 patients with inflammatory diseases, including 37 undiagnosed cases, were analyzed. Increased IFN signaling was observed in 19 undiagnosed patients, with 10 exhibiting clinical features commonly found in type I interferonopathies. Skin manifestations, observed in eight patients, were macroscopically and histologically similar to those found in proteasome-associated autoinflammatory syndrome. Genetic analysis identified novel mutations in the PSMB8 gene of one patient, and rare variants of unknown significance in genes linked to type I IFN signaling in four patients. A JAK inhibitor effectively treated the patient with the PSMB8 mutations. Patients with clinically quiescent idiopathic pulmonary hemosiderosis and A20 haploinsufficiency showed enhanced IFN signaling. Conclusions: Half of the patients examined in this study, with undifferentiated inflammatory diseases, clinically quiescent A20 haploinsufficiency, or idiopathic pulmonary hemosiderosis, had an elevated type I IFN signature.


Subject(s)
Interferon Type I , Janus Kinase Inhibitors , Biomarkers , Humans , Interferon Type I/genetics , Japan , Proteasome Endopeptidase Complex/genetics , Retrospective Studies
4.
Front Immunol ; 13: 917398, 2022.
Article in English | MEDLINE | ID: mdl-35812376

ABSTRACT

Familial Mediterranean fever (FMF) is a hereditary, autoinflammatory disease that causes recurrent fever, arthritis, and serositis. The diagnosis of FMF is based on the presentation of typical clinical symptoms and the Mediterranean fever gene (MEFV) test. However, the challenge lies in diagnosing atypical cases. In this report, we have described a pediatric patient with complex FMF whose diagnosis required trio-whole exome sequencing (WES) and functional validation of a rare MEFV variant. A 3-year-old boy presented with recurrent episodes of elevated liver enzymes and arthralgia. He was diagnosed with autoimmune hepatitis (AIH), and his liver enzymes improved rapidly with steroid treatment. However, he exhibited recurrent arthralgia and severe abdominal attacks. Trio-WES identified compound heterozygous mutations in MEFV (V726A and I692del). Ex vivo functional assays of the patient's monocytes and macrophages, which had been pre-treated with Clostridium difficile toxin A (TcdA) and colchicine, were comparable to those of typical FMF patients, thereby confirming the diagnosis of FMF. Although he was intolerant to colchicine because of liver toxicity, subsequent administration of canakinumab successfully ameliorated his abdominal attacks. However, it was ineffective against liver injury, which recurred after steroid tapering. Therefore, in this case, the pathogenesis of AIH was probably interleukin-1ß (IL-1ß)-independent. In fact, AIH might have been a concurrent disease with FMF, rather than being one of its complications. Nevertheless, further studies are necessary to determine whether FMF-induced inflammasome activation contributes to AIH development. Moreover, we must consider the possibility of mixed phenotypes in such atypical patients who present distinct pathologies simultaneously.


Subject(s)
Familial Mediterranean Fever , Hepatitis, Autoimmune , Arthralgia , Child , Colchicine/therapeutic use , Familial Mediterranean Fever/complications , Familial Mediterranean Fever/diagnosis , Familial Mediterranean Fever/drug therapy , Hepatitis, Autoimmune/complications , Hepatitis, Autoimmune/diagnosis , Hepatitis, Autoimmune/drug therapy , Humans , Male , Mutation , Pyrin/genetics
5.
Front Immunol ; 13: 870535, 2022.
Article in English | MEDLINE | ID: mdl-35603217

ABSTRACT

The concept of autoinflammation, first proposed in 1999, refers to a seemingly unprovoked episode of sterile inflammation manifesting as unexplained fever, skin rashes, and arthralgia. Autoinflammatory diseases are caused mainly by hereditary abnormalities of innate immunity, without the production of autoantibodies or autoreactive T cells. The revolutionary discovery of induced pluripotent stem cells (iPSCs), whereby a patient's somatic cells can be reprogrammed into an embryonic pluripotent state by forced expression of a defined set of transcription factors, has the transformative potential to enable in vitro disease modeling and drug candidate screening, as well as to provide a resource for cell replacement therapy. Recent reports demonstrate that recapitulating a disease phenotype in vitro is feasible for numerous monogenic diseases, including autoinflammatory diseases. In this review, we provide a comprehensive overview of current advances in research into autoinflammatory diseases involving iPSC-derived monocytes/macrophages. This review may aid in the planning of new studies of autoinflammatory diseases.


Subject(s)
Hereditary Autoinflammatory Diseases , Induced Pluripotent Stem Cells , Hereditary Autoinflammatory Diseases/genetics , Humans , Inflammation , Macrophages , Monocytes
6.
J Exp Med ; 219(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35482294

ABSTRACT

Mutations in the C-terminal region of the CDC42 gene cause severe neonatal-onset autoinflammation. Effectiveness of IL-1ß-blocking therapy indicates that the pathology involves abnormal inflammasome activation; however, the mechanism underlying autoinflammation remains to be elucidated. Using induced-pluripotent stem cells established from patients carrying CDC42R186C, we found that patient-derived cells secreted larger amounts of IL-1ß in response to pyrin-activating stimuli. Aberrant palmitoylation and localization of CDC42R186C protein to the Golgi apparatus promoted pyrin inflammasome assembly downstream of pyrin dephosphorylation. Aberrant subcellular localization was the common pathological feature shared by CDC42 C-terminal variants with inflammatory phenotypes, including CDC42*192C*24 that also localizes to the Golgi apparatus. Furthermore, the level of pyrin inflammasome overactivation paralleled that of mutant protein accumulation in the Golgi apparatus, but not that of the mutant GTPase activity. These results reveal an unexpected association between CDC42 subcellular localization and pyrin inflammasome activation that could pave the way for elucidating the mechanism of pyrin inflammasome formation.


Subject(s)
Golgi Apparatus , Inflammasomes , Golgi Apparatus/metabolism , Humans , Inflammasomes/metabolism , Pyrin/genetics , Pyrin/metabolism
8.
Front Immunol ; 13: 825806, 2022.
Article in English | MEDLINE | ID: mdl-35154146

ABSTRACT

Hepatic manifestations of Epstein-Barr virus (EBV) infection are relatively common, mild, and self-limiting. Although fulminant hepatic failure has been reported in a few cases, the contributing factors are unclear. This report discusses a pediatric case of EBV-associated acute liver failure that required urgent liver transplantation; however, liver damage continued to progress post-liver replacement. Monoclonal CD8+ T cells that preferentially infiltrated the native and transplanted liver were positive for EBV-encoded small RNA, suggesting a pathophysiology similar to that of EBV-associated hemophagocytic lymphohistiocytosis and chronic active EBV infection. Therefore, subsequent chemotherapy and hematopoietic cell transplantation was conducted, which led to cure. This is the first case of EBV-associated acute liver failure that relapsed post-liver transplant. As such, it sheds light on an under-recognized clinical entity: liver-restricted hyperinflammation caused by EBV-infected monoclonal CD8+ T cells. This phenomenon needs to be recognized and differentiated from hepatitis/hepatic failure caused by EBV-infected B cells, which has a relatively benign clinical course.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Liver Failure, Acute/virology , Liver/pathology , Lymphohistiocytosis, Hemophagocytic/virology , Child, Preschool , Epstein-Barr Virus Infections/immunology , Hematopoietic Stem Cell Transplantation , Humans , Liver/diagnostic imaging , Liver Failure, Acute/therapy , Liver Transplantation , Lymphohistiocytosis, Hemophagocytic/therapy , Male , Positron Emission Tomography Computed Tomography , RNA, Viral/analysis , Treatment Outcome
9.
Mod Rheumatol Case Rep ; 6(2): 314-318, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35079820

ABSTRACT

Coatomer subunit alpha (COPA) syndrome is an autoinflammatory disease with autoimmune and autoinflammatory manifestations affecting lungs, joints, and kidneys. COPA syndrome is caused by heterozygous loss-of-function mutations in COPA gene, encoding α subunit of coatmer protein complex I (COP-I) coated vesicles. Mutant COPA induces constitutive activation of stimulator of interferon genes, leading to systemic inflammation and elevated type I interferon response. We have previously reported a Japanese family of COPA syndrome with a novel V242G mutation. Two out of four patients required lung transplantation due to intractable interstitial lung disease and respiratory failure. Both of them deceased after lung transplantation, one due to sepsis and the other due to allograft dysfunction possibly caused by the reccurent interstitial lung disease. The literature review indentified unfavorable outcome of the solid organ transplant in COPA syndrome and its related disease, however, precise clinico-pathological description of these cases has been scarce. Here, we report in detail the clinical course of our cases to clarify the pathophysiology of allograft dysfunction in COPA syndrome and propose potential therapeutic approaches to improve post-transplant graft survival.


Subject(s)
Lung Diseases, Interstitial , Lung Transplantation , Allografts , Humans , Lung/pathology , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/etiology , Lung Diseases, Interstitial/surgery , Lung Transplantation/adverse effects , Syndrome
11.
Nat Commun ; 12(1): 6819, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819510

ABSTRACT

Impaired proteasome activity due to genetic variants of certain subunits might lead to proteasome-associated autoinflammatory syndromes (PRAAS). Here we report a de novo heterozygous missense variant of the PSMB9 proteasome subunit gene in two unrelated Japanese infants resulting in amino acid substitution of the glycine (G) by aspartic acid (D) at position 156 of the encoded protein ß1i. In addition to PRAAS-like manifestations, these individuals suffer from pulmonary hypertension and immunodeficiency, which are distinct from typical PRAAS symptoms. The missense variant results in impaired immunoproteasome maturation and activity, yet ubiquitin accumulation is hardly detectable in the patients. A mouse model of the heterozygous human genetic variant (Psmb9G156D/+) recapitulates the proteasome defects and the immunodeficiency phenotype of patients. Structurally, PSMB9 G156D interferes with the ß-ring-ßring interaction of the wild type protein that is necessary for 20S proteasome formation. We propose the term, proteasome-associated autoinflammatory syndrome with immunodeficiency (PRAAS-ID), to indicate a separate category of autoinflammatory diseases, similar to, but distinct from PRAAS, that describes the patients in this study.


Subject(s)
Cysteine Endopeptidases/genetics , Hereditary Autoinflammatory Diseases/genetics , Hypertension, Pulmonary/genetics , Primary Immunodeficiency Diseases/genetics , Proteasome Endopeptidase Complex/metabolism , Animals , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Female , Hereditary Autoinflammatory Diseases/diagnosis , Hereditary Autoinflammatory Diseases/immunology , Hereditary Autoinflammatory Diseases/pathology , Heterozygote , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/immunology , Infant, Newborn , Male , Mice , Mice, Transgenic , Mutation, Missense , Pedigree , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/pathology , Proteasome Endopeptidase Complex/genetics , Syndrome
13.
Pediatr Rheumatol Online J ; 19(1): 77, 2021 May 31.
Article in English | MEDLINE | ID: mdl-34059097

ABSTRACT

BACKGROUND: Cryopyrin-associated periodic syndrome (CAPS) is a life-long, autoinflammatory disease associated with a gain-of-function mutation in the nucleotide-binding domain, leucine-rich repeat family, pyrin domain containing 3 (NLRP3) gene, which result in uncontrolled production of IL-1ß and chronic inflammation. Chronic infantile neurologic cutaneous and articular (CINCA) syndrome/neonatal-Onset multisystem inflammatory disease (NOMID) is the most severe form of CAPS. Although the first symptoms may be presented at birth, there are few reports on the involvement of the placenta and umbilical cord in the disease. Therefore, we present herein a preterm case of CINCA/NOMID syndrome and confirms intrauterine-onset inflammation with conclusive evidence by using fetal and placental histopathological examination. CASE PRESENTATION: The female patient was born at 33weeks of gestation by emergency caesarean section and weighted at 1,514 g. The most common manifestations of CINCA/NOMID syndrome including recurrent fever, urticarial rash, and ventriculomegaly due to aseptic meningitis were presented. She also exhibited atypical symptoms such as severe hepatosplenomegaly with cholestasis. The genetic analysis of NLRP3 revealed a heterozygous c.1698 C > G (p.Phe566Leu) mutation, and she was diagnosed with CINCA/NOMID syndrome. Further, a histopathological examination revealed necrotizing funisitis, mainly inflammation of the umbilical artery, along with focal neutrophilic and lymphocytic villitis. CONCLUSIONS: The necrotizing funisitis, which only involved the artery, was an unusual observation for chorioamnionitis. These evidences suggest that foetal inflammation, probably due to overproduction of IL-1ß, caused tissue damage in utero, and the first symptom of a newborn with CINCA/NOMID.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Chorioamnionitis , Cryopyrin-Associated Periodic Syndromes , Interleukin-1beta/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Placenta/pathology , Umbilical Arteries/pathology , Cesarean Section/methods , Chorioamnionitis/diagnosis , Chorioamnionitis/etiology , Cryopyrin-Associated Periodic Syndromes/diagnosis , Cryopyrin-Associated Periodic Syndromes/genetics , Cryopyrin-Associated Periodic Syndromes/physiopathology , Female , Genetic Carrier Screening , Humans , Immunologic Factors/administration & dosage , Infant, Newborn , Mutation , Necrosis , Obstetric Labor, Premature/etiology , Obstetric Labor, Premature/surgery , Pregnancy , Treatment Outcome
14.
Arthritis Rheumatol ; 73(11): 2105-2115, 2021 11.
Article in English | MEDLINE | ID: mdl-33982886

ABSTRACT

OBJECTIVE: Coatomer subunit alpha (COPA) syndrome, also known as autoinflammatory interstitial lung, joint, and kidney disease, is caused by heterozygous mutations in COPA. We identified a novel COPA variant in 4 patients in one family. We undertook this study to elucidate whether and how the variant causes manifestations of COPA syndrome by studying these 4 patients and by analyzing results from a gene-targeted mouse model. METHODS: We performed whole-exome sequencing in 7 family members and measured the type I interferon (IFN) signature of the peripheral blood cells. We analyzed the effects of COPA variants in in vitro experiments and in Copa mutant mice that were generated. RESULTS: We identified a heterozygous variant of COPA (c.725T>G, p.Val242Gly) in the 4 affected members of the family. The IFN score was high in the members carrying the variant. In vitro analysis revealed that COPA V242G, as well as the previously reported disease-causing variants, augmented stimulator of interferon genes (STING)-induced type I IFN promoter activities. CopaV242G/+ mice manifested interstitial lung disease and STING-dependent elevation of IFN-stimulated gene expression. In CopaV242G/+ dendritic cells, the STING pathway was not constitutively activated but was hyperactivated upon stimulation, leading to increased type I IFN production. CONCLUSION: V242G, a novel COPA variant, was found in 4 patients from one family. In gene-targeted mice with the V242G variant, interstitial lung disease was recapitulated and augmented responses of the STING pathway, leading to an increase in type I IFN production, were demonstrated.


Subject(s)
Coatomer Protein/genetics , Interferon Type I/genetics , Joint Diseases/genetics , Kidney Diseases/genetics , Lung Diseases, Interstitial/genetics , Mutation, Missense , Alleles , DNA Mutational Analysis , Female , Heterozygote , Humans , Joint Diseases/immunology , Kidney Diseases/immunology , Lung Diseases, Interstitial/immunology , Male , Pedigree , Exome Sequencing
15.
J Clin Immunol ; 41(6): 1187-1197, 2021 08.
Article in English | MEDLINE | ID: mdl-33733382

ABSTRACT

PURPOSE: Pathogenic MEFV variants cause pyrin-associated autoinflammatory diseases (PAADs), which include familial Mediterranean fever (FMF), FMF-like disease, and pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND). The diagnosis of PAADs is established by clinical phenotypic and genetic analyses. However, the pathogenicity of most MEFV variants remains controversial, as they have not been functionally evaluated. This study aimed to establish and validate a new functional assay to evaluate the pathogenicity of MEFV variants. METHODS: We transfected THP-1 monocytes with 32 MEFV variants and analyzed their effects on cell death with or without stimulation with Clostridium difficile toxin A (TcdA) or UCN-01. These variants were classified using hierarchical cluster analysis. Macrophages were obtained from three healthy controls and two patients with a novel homozygous MEFVP257L variant, for comparison of IL-1ß secretion using a cell-based assay and a novel THP-1-based assay. RESULTS: Disease-associated MEFV variants induced variable degrees of spontaneous or TcdA/UCN-01-induced cell death in THP-1. Cell death was caspase-1 dependent and was accompanied by ASC speck formation and IL-1ß secretion, indicating that pathogenic MEFV variants induced abnormal pyrin inflammasome activation and subsequent pyroptotic cell deaths in this assay. The MEFV variants (n = 32) exhibiting distinct response signatures were classified into 6 clusters, which showed a good correlation with the clinical phenotypes. Regarding the pathogenicity of MEFVP257L variants, the results were consistent between the cell-based assay and the THP-1-based assay. CONCLUSION: Our assay facilitates a rapid and comprehensive assessment of the pathogenicity of MEFV variants and contributes to a refined definition of PAAD subtypes.


Subject(s)
Flow Cytometry/methods , Genetic Variation/genetics , Pyrin/genetics , Cell Death/genetics , Cell Line , Female , Genetic Predisposition to Disease/genetics , Humans , Inflammasomes/genetics , Male , Middle Aged , Monocytes/metabolism , Phenotype , THP-1 Cells
16.
J Allergy Clin Immunol ; 148(2): 550-562, 2021 08.
Article in English | MEDLINE | ID: mdl-33529688

ABSTRACT

BACKGROUND: Deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive inflammatory disease caused by loss-of-function mutations in both alleles of the ADA2 gene. Most patients with DADA2 exhibit systemic vasculopathy consistent with polyarteritis nodosa, but large phenotypic variability has been reported, and the pathogenesis of DADA2 remains unclear. OBJECTIVES: This study sought to assess the clinical and genetic characteristics of Japanese patients with DADA2 and to gain insight into the pathogenesis of DADA2 by multi-omics analysis. METHODS: Clinical and genetic data were collected from 8 Japanese patients with DADA2 diagnosed between 2016 and 2019. ADA2 variants in this cohort were functionally analyzed by in vitro overexpression analysis. PBMCs from 4 patients with DADA2 were subjected to transcriptome and proteome analyses. Patient samples were collected before and after introduction of anti- TNF-α therapies. Transcriptome data were compared with those of normal controls and patients with other autoinflammatory diseases. RESULTS: Five novel ADA2 variants were identified in these 8 patients and were confirmed pathogenic by in vitro analysis. Anti-TNF-α therapy controlled inflammation in all 8 patients. Transcriptome and proteome analyses showed that upregulation of type II interferon signaling was characteristic of DADA2. Network analysis identified STAT1 as a key regulator and a hub molecule in DADA2 pathogenesis, a finding supported by the hyperactivation of STAT1 in patients' monocytes and B cells after IFN-γ stimulation. CONCLUSIONS: Type II interferon signaling and STAT1 are associated with the pathogenesis of DADA2.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia/immunology , Intercellular Signaling Peptides and Proteins/deficiency , Interferon-gamma/immunology , Leukocytes, Mononuclear/immunology , STAT1 Transcription Factor/immunology , Severe Combined Immunodeficiency/immunology , Adenosine Deaminase/immunology , Adolescent , Adult , Agammaglobulinemia/genetics , Agammaglobulinemia/pathology , Asian People , Child , Child, Preschool , Female , Gene Expression Profiling , Humans , Infant , Intercellular Signaling Peptides and Proteins/immunology , Interferon-gamma/genetics , Japan , Leukocytes, Mononuclear/pathology , Male , Proteomics , STAT1 Transcription Factor/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/pathology
17.
Pediatr Rheumatol Online J ; 19(1): 18, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33602264

ABSTRACT

BACKGROUND: Early-onset sarcoidosis (EOS) and Blau syndrome (BS) are systemic inflammatory granulomatous diseases without visible pulmonary involvement, and are distinguishable from their sporadic and familial forms. The diseases are characterized by a triad of skin rashes, symmetrical polyarthritis, and recurrent uveitis. The most common morbidity is ocular involvement, which is usually refractory to conventional treatment. A gain-of-function mutation in the nucleotide-binding oligomerization domain-containing protein 2 (NOD2) gene has been demonstrated in this disease; however, little is known about the relationship between the activation of NOD2 and the pathophysiology of EOS/BS. Here we describe EOS/BS with a novel mutation in the NOD2 gene, as well as detection of Propionibacterium acnes (P. acnes) in the granulomatous inflammation. CASE PRESENTATION: An 8-year-old Japanese girl presented with refractory bilateral granulomatous panuveitis. Although no joint involvement was evident, she exhibited skin lesions on her legs; a skin biopsy revealed granulomatous dermatitis, and P. acnes was detected within the sarcoid granulomas by immunohistochemistry with P. acnes-specific monoclonal (PAB) antibody. Genetic analyses revealed that the patient had a NOD2 heterozygous D512V mutation that was novel and not present in either of her parents. The mutant NOD2 showed a similar activation pattern to EOS/BS, thus confirming her diagnosis. After starting oral prednisolone treatment, she experienced an anterior vitreous opacity relapse despite gradual prednisolone tapering; oral methotrexate was subsequently administered, and the patient responded positively. CONCLUSIONS: We presented a case of EOS/BS with a novel D512V mutation in the NOD2 gene. In refractory granulomatous panuveitis cases without any joint involvement, EOS/BS should be considered as a differential diagnosis; genetic analyses would lead to a definite diagnosis. Moreover, this is the first report of P. acnes demonstrated in granulomas of EOS/BS. Since intracellular P. acnes activates nuclear factor-kappa B in a NOD2-dependent manner, we hypothesized that the mechanism of granuloma formation in EOS/BS may be the result of NOD2 activity in the presence of the ligand muramyl dipeptide, which is a component of P. acnes. These results indicate that recognition of P. acnes through mutant NOD2 is the etiology in this patient with EOS/BS.


Subject(s)
Arthritis , Dermatitis , Granuloma , Methotrexate/administration & dosage , Nod2 Signaling Adaptor Protein/genetics , Panuveitis , Prednisolone/administration & dosage , Propionibacterium acnes/isolation & purification , Sarcoidosis , Synovitis , Uveitis , Antirheumatic Agents/administration & dosage , Arthritis/diagnosis , Arthritis/drug therapy , Arthritis/genetics , Arthritis/physiopathology , Biopsy/methods , Child , Dermatitis/etiology , Dermatitis/immunology , Dermatitis/microbiology , Dermatitis/pathology , Female , Granuloma/immunology , Granuloma/microbiology , Humans , Immunohistochemistry , Mutation , Panuveitis/diagnosis , Panuveitis/etiology , Sarcoidosis/diagnosis , Sarcoidosis/drug therapy , Sarcoidosis/genetics , Sarcoidosis/physiopathology , Skin/pathology , Synovitis/diagnosis , Synovitis/drug therapy , Synovitis/genetics , Synovitis/physiopathology , Treatment Outcome , Uveitis/diagnosis , Uveitis/drug therapy , Uveitis/genetics , Uveitis/physiopathology
18.
Mod Rheumatol ; 31(2): 493-497, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32011208

ABSTRACT

OBJECTIVES: Haploinsufficiency of A20 (HA20) due to loss-of-function mutations of TNFAIP3 leads to an autoinflammatory disease. These mutations produce a premature termination codon in most cases of HA20. However, exon deletion has not been reported. METHODS: Genomic DNA was extracted from the peripheral blood of the patient clinically suspected of HA20. We examined autoinflammatory disease-causing genes and performed a multiplex ligation-dependent probe amplification (MLPA) assay for copy number analysis. Next, to determine the disconnection point, genomic DNA was amplified with long-range PCR and sequenced. Finally, western blotting was carried out to measure A20 protein expression in mitogen phytohaemagglutinin (PHA)-induced T-cell blasts from the patient and a healthy volunteer. RESULTS: Targeted next-generation sequencing found no pathogenic mutation, but copy number variation (CNV) analysis suggested a heterozygous deletion of exons 2-3. The MLPA assay and long-range PCR confirmed the mutation. Western blotting analysis indicated a marked decrease in expression of A20 protein from the patient compared to a normal control. The results showed that this deletion was a pathogenic mutation. CONCLUSION: This study demonstrates a novel mutation resulting in a deletion of exons 2-3 of TNFAIP3. MLPA analysis is a useful initial screening method for HA20 patients.


Subject(s)
Autoimmune Diseases/genetics , Haploinsufficiency , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Autoimmune Diseases/diagnosis , Exons , Gene Deletion , Heterozygote , Humans , Loss of Function Mutation , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...