Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters

Publication year range
1.
Ann Rheum Dis ; 81(5): 601-613, 2022 05.
Article in English | MEDLINE | ID: mdl-35086813

ABSTRACT

OBJECTIVE: Autoinflammatory type I interferonopathies, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature/proteasome-associated autoinflammatory syndrome (CANDLE/PRAAS), stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI) and Aicardi-Goutières syndrome (AGS) are rare and clinically complex immunodysregulatory diseases. With emerging knowledge of genetic causes and targeted treatments, a Task Force was charged with the development of 'points to consider' to improve diagnosis, treatment and long-term monitoring of patients with these rare diseases. METHODS: Members of a Task Force consisting of rheumatologists, neurologists, an immunologist, geneticists, patient advocates and an allied healthcare professional formulated research questions for a systematic literature review. Then, based on literature, Delphi questionnaires and consensus methodology, 'points to consider' to guide patient management were developed. RESULTS: The Task Force devised consensus and evidence-based guidance of 4 overarching principles and 17 points to consider regarding the diagnosis, treatment and long-term monitoring of patients with the autoinflammatory interferonopathies, CANDLE/PRAAS, SAVI and AGS. CONCLUSION: These points to consider represent state-of-the-art knowledge to guide diagnostic evaluation, treatment and management of patients with CANDLE/PRAAS, SAVI and AGS and aim to standardise and improve care, quality of life and disease outcomes.


Subject(s)
Autoimmune Diseases of the Nervous System , Nervous System Malformations , Rheumatology , Skin Diseases , Erythema Nodosum , Fingers/abnormalities , Humans , Quality of Life
2.
Proc Natl Acad Sci U S A ; 113(6): 1612-7, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26802121

ABSTRACT

Receptor-induced NF-κB activation is controlled by NEMO, the NF-κB essential modulator. Hypomorphic NEMO mutations result in X-linked ectodermal dysplasia with anhidrosis and immunodeficiency, also referred to as NEMO syndrome. Here we describe a distinct group of patients with NEMO C-terminal deletion (ΔCT-NEMO) mutations. Individuals harboring these mutations develop inflammatory skin and intestinal disease in addition to ectodermal dysplasia with anhidrosis and immunodeficiency. Both primary cells from these patients, as well as reconstituted cell lines with this deletion, exhibited increased IκB kinase (IKK) activity and production of proinflammatory cytokines. Unlike previously described loss-of-function mutations, ΔCT-NEMO mutants promoted increased NF-κB activation in response to TNF and Toll-like receptor stimulation. Investigation of the underlying mechanisms revealed impaired interactions with A20, a negative regulator of NF-κB activation, leading to prolonged accumulation of K63-ubiquitinated RIP within the TNFR1 signaling complex. Recruitment of A20 to the C-terminal domain of NEMO represents a novel mechanism limiting NF-κB activation by NEMO, and its absence results in autoinflammatory disease.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Kinase/chemistry , I-kappa B Kinase/metabolism , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Case-Control Studies , Cell Line , Cell Nucleus/metabolism , Cytokines/biosynthesis , Deubiquitinating Enzyme CYLD , Female , Gene Expression Regulation , Humans , I-kappa B Kinase/genetics , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Male , Monocytes/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Pedigree , Phenotype , Polyubiquitin/metabolism , Protein Structure, Tertiary , Protein Transport , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes/metabolism , Toll-Like Receptors/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Suppressor Proteins/metabolism , Ubiquitination
3.
J Immunol ; 185(5): 2665-9, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20693425

ABSTRACT

The signaling and adaptor protein Homer3 plays a role in controlling immune homeostasis and self-reactivity. Homer3 is recruited to the immune synapse (IS) following TCR ligation, although the mechanisms regulating this subcellular localization are unknown. We show that Homer3 specifically associates with a novel ubiquitin-like domain in the IkappaB kinase (IKK) beta subunit of the IKK complex. Homer3 associates with IKKbeta in T cells and colocalizes with the IKK complex at the IS. However, Homer3 is not required for IKK activation, as NF-kappaB signaling is intact in Homer3-deficient T cells. Instead, the IKK complex recruits Homer3 to the IS following TCR engagement, and we present evidence that this association regulates actin dynamics in T cells. These findings identify a novel interaction between two major signaling proteins and reveal an unexpected NF-kappaB-independent function for the IKK complex in regulating the subcellular localization of Homer3.


Subject(s)
Carrier Proteins/metabolism , I-kappa B Proteins/metabolism , Carrier Proteins/physiology , Cell Line , HeLa Cells , Homer Scaffolding Proteins , Humans , I-kappa B Proteins/physiology , Immunological Synapses/enzymology , Immunological Synapses/immunology , Jurkat Cells , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/physiology , Signal Transduction/immunology , Subcellular Fractions/immunology , Subcellular Fractions/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism
4.
J Clin Invest ; 132(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35289316

ABSTRACT

Host defense and inflammation are regulated by the NF-κB essential modulator (NEMO), a scaffolding protein with a broad immune cell and tissue expression profile. Hypomorphic mutations in inhibitor of NF-κB kinase regulatory subunit gamma (IKBKG) encoding NEMO typically present with immunodeficiency. Here, we characterized a pediatric autoinflammatory syndrome in 3 unrelated male patients with distinct X-linked IKBKG germline mutations that led to overexpression of a NEMO protein isoform lacking the domain encoded by exon 5 (NEMO-Δex5). This isoform failed to associate with TANK binding kinase 1 (TBK1), and dermal fibroblasts from affected patients activated NF-κB in response to TNF but not TLR3 or RIG-I-like receptor (RLR) stimulation when isoform levels were high. By contrast, T cells, monocytes, and macrophages that expressed NEMO-Δex5 exhibited increased NF-κB activation and IFN production, and blood cells from these patients expressed a strong IFN and NF-κB transcriptional signature. Immune cells and TNF-stimulated dermal fibroblasts upregulated the inducible IKK protein (IKKi) that was stabilized by NEMO-Δex5, promoting type I IFN induction and antiviral responses. These data revealed how IKBKG mutations that lead to alternative splicing of skipping exon 5 cause a clinical phenotype we have named NEMO deleted exon 5 autoinflammatory syndrome (NDAS), distinct from the immune deficiency syndrome resulting from loss-of-function IKBKG mutations.


Subject(s)
Hereditary Autoinflammatory Diseases , Immunologic Deficiency Syndromes , Alternative Splicing , Child , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Immunologic Deficiency Syndromes/genetics , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype
5.
Arthritis Rheumatol ; 74(5): 735-751, 2022 05.
Article in English | MEDLINE | ID: mdl-35315249

ABSTRACT

OBJECTIVE: Autoinflammatory type I interferonopathies, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature/proteasome-associated autoinflammatory syndrome (CANDLE/PRAAS), stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI), and Aicardi-Goutières syndrome (AGS) are rare and clinically complex immunodysregulatory diseases. With emerging knowledge of genetic causes and targeted treatments, a Task Force was charged with the development of "points to consider" to improve diagnosis, treatment, and long-term monitoring of patients with these rare diseases. METHODS: Members of a Task Force consisting of rheumatologists, neurologists, an immunologist, geneticists, patient advocates, and an allied health care professional formulated research questions for a systematic literature review. Then, based on literature, Delphi questionnaires, and consensus methodology, "points to consider" to guide patient management were developed. RESULTS: The Task Force devised consensus and evidence-based guidance of 4 overarching principles and 17 points to consider regarding the diagnosis, treatment, and long-term monitoring of patients with the autoinflammatory interferonopathies, CANDLE/PRAAS, SAVI, and AGS. CONCLUSION: These points to consider represent state-of-the-art knowledge to guide diagnostic evaluation, treatment, and management of patients with CANDLE/PRAAS, SAVI, and AGS and aim to standardize and improve care, quality of life, and disease outcomes.


Subject(s)
Autoimmune Diseases of the Nervous System , Nervous System Malformations , Rheumatology , Skin Diseases , Autoimmune Diseases of the Nervous System/genetics , Erythema Nodosum , Fingers/abnormalities , Humans , Quality of Life
6.
Ann Allergy Asthma Immunol ; 107(1): 50-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21704885

ABSTRACT

BACKGROUND: Hypomorphic mutations in the nuclear factor-κB (NF-κB) essential modulator (NEMO) gene result in a variable syndrome of somatic and immunologic abnormalities. Clinically relevant genotype-phenotype associations are essential to understanding this complex disease. OBJECTIVE: To study 2 unrelated boys with novel NEMO mutations altering codon 223 for similarity in phenotype in consideration of potential genotype-phenotype associations. METHODS: Clinical and laboratory features, including cell counts, immunoglobulin quantity and quality, natural killer cell cytotoxicity, and Toll-like and tumor necrosis factor receptor signaling, were evaluated. Because both mutations affected NEMO codon 223 and were novel, consideration was given to new potential genotype-phenotype associations. RESULTS: Both patients were diagnosed as having hypohidrotic ectodermal dysplasia and had severe or recurrent infections. One had recurrent sinopulmonary infections and the other necrotizing soft tissue methicillin-resistant Staphylococcus aureus infection and Streptococcus anginosus subdural empyema with bacteremia. NEMO gene sequence demonstrated a 3-nucleotide deletion (c.667_669delGAG) in one patient and a substitution (667G>A) in the other. These findings predict either the deletion of NEMO glutamic acid 223 or it being replaced with lysine, respectively. Both patients had normal serum IgG levels but poor specific antibodies. Natural killer cell cytotoxicity and Toll-like and tumor necrosis factor receptor signaling were also impaired. Serious bacterial infection did not occur in both patients after immunoglobulin replacement therapy. CONCLUSIONS: Two different novel mutations affecting NEMO glutamic acid 223 resulted in clinically relevant similar phenotypes, providing further evidence to support genotype-phenotype correlations in this disease. They suggest NEMO residue 223 is required for ectodermal development and immunity and is apparently dispensable for quantitative IgG production but may be required for specific antibody production.


Subject(s)
I-kappa B Kinase/genetics , Immunoglobulin G/blood , Immunologic Deficiency Syndromes/genetics , Antibody Specificity/immunology , Child, Preschool , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/immunology , Ectodermal Dysplasia 1, Anhidrotic , Humans , I-kappa B Kinase/immunology , Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/immunology , Infant , Killer Cells, Natural/immunology , Male , Mutation , Receptors, Tumor Necrosis Factor/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology
7.
J Clin Immunol ; 30(6): 881-5, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20652730

ABSTRACT

INTRODUCTION: We describe a previously unreported 437 T→G missense mutation producing a V146G substitution in the first coiled-coil (CC1) domain of nuclear factor-κB essential modulator (NEMO) in a 9-month-old boy with ectodermal dysplasia with immunodeficiency who presented with methicillin-resistant Staphylococcus aureus subdural empyema. We performed in vitro experiments to determine if this novel mutation resulted in impaired NF-κB signaling. METHODS: IκBα phosphorylation experiments were performed using a Jurkat T cell line lacking endogenous NEMO expression that was transfected with vectors containing either the wild type or the patient's V146G mutation. The cells were stimulated with TNF-α to activate the NF-κB pathway. Phosphorylated IκBα was detected by immunoblotting with anti-phospho-IκBα antibodies. Peripheral blood mononuclear cells from the patient were stimulated with TNF-α or anti-CD3 and anti-CD28. Impaired IκBα degradation was detected using antibodies against the IκBα protein. RESULTS: While TNF-α stimulation resulted in IκBα phosphorylation in NEMO-deficient Jurkat cells reconstituted with wild-type NEMO, cell transfected with the V146G mutant exhibited a 75% reduction in phospho-IκBα. Peripheral blood mononuclear cells from the patient showed impaired degradation of IκBα after stimulation when compared with normal controls. CONCLUSIONS: The patient's V146G mutation results in impaired NF-κB activation in vitro. The mutation extends the known N-terminal boundary within the CC1 domain that produces an ectodermal dysplasia phenotype, and defines an infectious susceptibility previously unappreciated in ectodermal dysplasia with immunodeficiency (methicillin-resistant S. aureus subdural empyema), broadening the clinical spectrum associated with the disease.


Subject(s)
Drug Resistance, Bacterial , Empyema, Subdural/genetics , I-kappa B Kinase/metabolism , Staphylococcus aureus/immunology , Ectodermal Dysplasia , Empyema, Subdural/drug therapy , Empyema, Subdural/metabolism , Empyema, Subdural/physiopathology , Humans , I-kappa B Kinase/genetics , Infant , Jurkat Cells , Male , Methicillin/therapeutic use , Mutant Proteins/genetics , Mutation, Missense/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Transcriptional Activation/genetics , Transgenes/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
8.
J Exp Med ; 217(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32232430

ABSTRACT

Melorheostosis is a rare sclerosing dysostosis characterized by asymmetric exuberant bone formation. Recently, we reported that somatic mosaicism for MAP2K1-activating mutations causes radiographical "dripping candle wax" melorheostosis. We now report somatic SMAD3 mutations in bone lesions of four unrelated patients with endosteal pattern melorheostosis. In vitro, the SMAD3 mutations stimulated the TGF-ß pathway in osteoblasts, enhanced nuclear translocation and target gene expression, and inhibited proliferation. Osteoblast differentiation and mineralization were stimulated by the SMAD3 mutation, consistent with higher mineralization in affected than in unaffected bone, but differing from MAP2K1 mutation-positive melorheostosis. Conversely, osteoblast differentiation and mineralization were inhibited when osteogenesis of affected osteoblasts was driven in the presence of BMP2. Transcriptome profiling displayed that TGF-ß pathway activation and ossification-related processes were significantly influenced by the SMAD3 mutation. Co-expression clustering illuminated melorheostosis pathophysiology, including alterations in ECM organization, cell growth, and interferon signaling. These data reveal antagonism of TGF-ß/SMAD3 activation by BMP signaling in SMAD3 mutation-positive endosteal melorheostosis, which may guide future therapies.


Subject(s)
Melorheostosis/genetics , Mutation/genetics , Signal Transduction , Smad3 Protein/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation/genetics , Animals , Bone Morphogenetic Protein 2/metabolism , Bone and Bones/pathology , Calcification, Physiologic , Cell Differentiation , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Extracellular Matrix/metabolism , Gain of Function Mutation , Gene Expression Regulation , Humans , MAP Kinase Kinase 1/genetics , Melorheostosis/diagnostic imaging , Melorheostosis/pathology , Mice , Models, Biological , Osteoblasts/metabolism , Osteogenesis , Protein Transport , Transcriptome/genetics
9.
J Clin Invest ; 130(4): 1669-1682, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31874111

ABSTRACT

BACKGROUNDUndifferentiated systemic autoinflammatory diseases (USAIDs) present diagnostic and therapeutic challenges. Chronic interferon (IFN) signaling and cytokine dysregulation may identify diseases with available targeted treatments.METHODSSixty-six consecutively referred USAID patients underwent underwent screening for the presence of an interferon signature using a standardized type-I IFN-response-gene score (IRG-S), cytokine profiling, and genetic evaluation by next-generation sequencing.RESULTSThirty-six USAID patients (55%) had elevated IRG-S. Neutrophilic panniculitis (40% vs. 0%), basal ganglia calcifications (46% vs. 0%), interstitial lung disease (47% vs. 5%), and myositis (60% vs. 10%) were more prevalent in patients with elevated IRG-S. Moderate IRG-S elevation and highly elevated serum IL-18 distinguished 8 patients with pulmonary alveolar proteinosis (PAP) and recurrent macrophage activation syndrome (MAS). Among patients with panniculitis and progressive cytopenias, 2 patients were compound heterozygous for potentially novel LRBA mutations, 4 patients harbored potentially novel splice variants in IKBKG (which encodes NF-κB essential modulator [NEMO]), and 6 patients had de novo frameshift mutations in SAMD9L. Of additional 12 patients with elevated IRG-S and CANDLE-, SAVI- or Aicardi-Goutières syndrome-like (AGS-like) phenotypes, 5 patients carried mutations in either SAMHD1, TREX1, PSMB8, or PSMG2. Two patients had anti-MDA5 autoantibody-positive juvenile dermatomyositis, and 7 could not be classified. Patients with LRBA, IKBKG, and SAMD9L mutations showed a pattern of IRG elevation that suggests prominent NF-κB activation different from the canonical interferonopathies CANDLE, SAVI, and AGS.CONCLUSIONSIn patients with elevated IRG-S, we identified characteristic clinical features and 3 additional autoinflammatory diseases: IL-18-mediated PAP and recurrent MAS (IL-18PAP-MAS), NEMO deleted exon 5-autoinflammatory syndrome (NEMO-NDAS), and SAMD9L-associated autoinflammatory disease (SAMD9L-SAAD). The IRG-S expands the diagnostic armamentarium in evaluating USAIDs and points to different pathways regulating IRG expression.TRIAL REGISTRATIONClinicalTrials.gov NCT02974595.FUNDINGThe Intramural Research Program of the NIH, NIAID, NIAMS, and the Clinical Center.


Subject(s)
Autoimmune Diseases , Interferon Type I , Interleukin-18 , Macrophage Activation Syndrome , Mutation , Panniculitis , Pulmonary Alveolar Proteinosis , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Female , Humans , Interferon Type I/genetics , Interferon Type I/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Macrophage Activation Syndrome/genetics , Macrophage Activation Syndrome/immunology , Male , Panniculitis/genetics , Panniculitis/immunology , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/immunology
10.
J Allergy Clin Immunol ; 122(6): 1169-1177.e16, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18851874

ABSTRACT

BACKGROUND: Human hypomorphic nuclear factor-kappaB essential modulator (NEMO) mutations cause diverse clinical and immunologic phenotypes, but understanding of their scope and mechanistic links to immune function and genotype is incomplete. OBJECTIVE: We created and analyzed a database of hypomorphic NEMO mutations to determine the spectrum of phenotypes and their associated genotypes and sought to establish a standardized NEMO reconstitution system to obtain mechanistic insights. METHODS: Phenotypes of 72 individuals with NEMO mutations were compiled. NEMO L153R and C417R were investigated further in a reconstitution system. TNF-alpha or Toll-like receptor (TLR)-5 signals were evaluated for nuclear factor-kappaB activation, programmed cell death, and A20 gene expression. RESULTS: Thirty-two different mutations were identified; 53% affect the zinc finger domain. Seventy-seven percent were associated with ectodermal dysplasia, 86% with serious pyogenic infection, 39% with mycobacterial infection, 19% with serious viral infection, and 23% with inflammatory diseases. Thirty-six percent of individuals died at a mean age of 6.4 years. CD40, IL-1, TNF-alpha, TLR, and T-cell receptor signals were impaired in 15 of 16 (94%), 6 of 7 (86%), 9 of 11 (82%), 9 of 14 (64%), and 7 of 18 (39%), respectively. Hypomorphism-reconstituted NEMO-deficient cells demonstrated partial restoration of NEMO functions. Although both L153R and C417R impaired TLR and TNF-alpha-induced NF-kappaB activation, L153R also increased TNF-alpha-induced programmed cell death with decreased A20 expression. CONCLUSION: Distinct NEMO hypomorphs define specific disease and genetic characteristics. A reconstitution system can identify attributes of hypomorphisms independent of an individual's genetic background. Apoptosis susceptibility in L153R reconstituted cells defines a specific phenotype of this mutation that likely contributes to the excessive inflammation with which it is clinically associated.


Subject(s)
Databases, Genetic , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Mutation, Missense , Amino Acid Substitution , Apoptosis/genetics , CD40 Antigens/metabolism , DNA-Binding Proteins , Gene Expression Regulation/genetics , Genetic Complementation Test/methods , Genotype , Humans , Intracellular Signaling Peptides and Proteins , Jurkat Cells , NF-kappa B/metabolism , Nuclear Proteins/biosynthesis , Phenotype , Protein Structure, Tertiary/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/genetics , Toll-Like Receptor 5/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/metabolism
11.
J Allergy Clin Immunol ; 121(4): 976-82, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18179816

ABSTRACT

BACKGROUND: Patients with hypomorphic nuclear factor-kappaB essential modulator (NEMO) mutations have extensive phenotypic variability that can include atypical infectious susceptibility. OBJECTIVE: This study may provide important insight into immunologic mechanisms of host defense. METHODS: Immunologic evaluation, including studies of Toll-like receptor (TLR) function, was performed in a 6-month-old boy with normal ectodermal development who was diagnosed with Pneumocystis pneumonia and cytomegalovirus sepsis. RESULTS: Genomic and cDNA sequencing demonstrated a novel NEMO missense mutation, 337G->A, predicted to cause a D113N (aspartic acid to asparagine) substitution in the first coiled-coil region of the NEMO protein. Quantitative serum immunoglobulins, lymphocyte subset numbers, and mitogen-induced lymphocyte proliferation were essentially normal. The PBMC responses to TLR ligands were also surprisingly normal, whereas natural killer cell cytolytic activity, T-cell proliferative responses to specific antigens, and T-cell receptor-induced NF-kappaB activation were diminished. CONCLUSION: Unlike the unique NEMO mutation described here, the most commonly reported mutations are clustered at the 3' end in the tenth exon, which encodes a zinc finger domain. Because specific hypomorphic variants of NEMO are associated with distinctive phenotypes, this particular NEMO mutation highlights a dispensability of the region including amino acid 113 for TLR signaling and ectodysplasin A receptor function. This region is required for certain immunoreceptor functions as demonstrated by his susceptibility to infections as well as natural killer cell and T-cell defects.


Subject(s)
I-kappa B Kinase/genetics , Mutation , Opportunistic Infections/genetics , Opportunistic Infections/metabolism , Pneumocystis carinii , Pneumonia, Pneumocystis/genetics , Pneumonia, Pneumocystis/metabolism , Toll-Like Receptors/physiology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Infant , Killer Cells, Natural/immunology , Male , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Opportunistic Infections/immunology , Pneumonia, Pneumocystis/immunology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Nat Commun ; 9(1): 1390, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29643386

ABSTRACT

Melorheostosis is a sporadic disease of uncertain etiology characterized by asymmetric bone overgrowth and functional impairment. Using whole exome sequencing, we identify somatic mosaic MAP2K1 mutations in affected, but not unaffected, bone of eight unrelated patients with melorheostosis. The activating mutations (Q56P, K57E and K57N) cluster tightly in the MEK1 negative regulatory domain. Affected bone displays a mosaic pattern of increased p-ERK1/2 in osteoblast immunohistochemistry. Osteoblasts cultured from affected bone comprise two populations with distinct p-ERK1/2 levels by flow cytometry, enhanced ERK1/2 activation, and increased cell proliferation. However, these MAP2K1 mutations inhibit BMP2-mediated osteoblast mineralization and differentiation in vitro, underlying the markedly increased osteoid detected in affected bone histology. Mosaicism is also detected in the skin overlying bone lesions in four of five patients tested. Our data show that the MAP2K1 oncogene is important in human bone formation and implicate MEK1 inhibition as a potential treatment avenue for melorheostosis.


Subject(s)
Bone and Bones/metabolism , MAP Kinase Kinase 1/genetics , Melorheostosis/genetics , Mutation , Osteoblasts/metabolism , Osteogenesis/genetics , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone and Bones/pathology , Calcification, Physiologic , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Humans , MAP Kinase Kinase 1/metabolism , Melorheostosis/metabolism , Melorheostosis/pathology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mosaicism , Osteoblasts/pathology , Primary Cell Culture , Signal Transduction , Skin/metabolism , Skin/pathology , Exome Sequencing
14.
Nat Genet ; 48(1): 67-73, 2016 01.
Article in English | MEDLINE | ID: mdl-26642243

ABSTRACT

Systemic autoinflammatory diseases are driven by abnormal activation of innate immunity. Herein we describe a new disease caused by high-penetrance heterozygous germline mutations in TNFAIP3, which encodes the NF-κB regulatory protein A20, in six unrelated families with early-onset systemic inflammation. The disorder resembles Behçet's disease, which is typically considered a polygenic disorder with onset in early adulthood. A20 is a potent inhibitor of the NF-κB signaling pathway. Mutant, truncated A20 proteins are likely to act through haploinsufficiency because they do not exert a dominant-negative effect in overexpression experiments. Patient-derived cells show increased degradation of IκBα and nuclear translocation of the NF-κB p65 subunit together with increased expression of NF-κB-mediated proinflammatory cytokines. A20 restricts NF-κB signals via its deubiquitinase activity. In cells expressing mutant A20 protein, there is defective removal of Lys63-linked ubiquitin from TRAF6, NEMO and RIP1 after stimulation with tumor necrosis factor (TNF). NF-κB-dependent proinflammatory cytokines are potential therapeutic targets for the patients with this disease.


Subject(s)
DNA-Binding Proteins/genetics , Haploinsufficiency/genetics , Hereditary Autoinflammatory Diseases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Nuclear Proteins/genetics , Age of Onset , DNA-Binding Proteins/metabolism , Female , Hereditary Autoinflammatory Diseases/metabolism , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/metabolism , Pedigree , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3
15.
Methods Mol Biol ; 1280: 413-26, 2015.
Article in English | MEDLINE | ID: mdl-25736764

ABSTRACT

Developmental and immune-mediated disease has been linked to genetic mutation of key signaling components involved in NF-κB activation that leads to impaired activation or regulation of the canonical IKK complex. We identify patients with suspected or known defects of the NF-κB signaling pathway through clinical phenotyping and genetic sequencing. To help understand how mutations cause disease, we quantitate the kinetics and dose-response of NF-κB activation signaling events in their cells. Following activation of the canonical IKK complex, phosphorylation of the inhibitor of NF-κB proteins (IκB) leads to their degradation and the subsequent translocation of NF-κB family members from the cell cytoplasm to the nucleus. Here, we provide a method to obtain patient-derived dermal fibroblasts and quantitatively assess the integrity of the signal transduction pathway from receptor activation to nuclear p65 translocation.


Subject(s)
Fibroblasts/metabolism , Skin/cytology , Transcription Factor RelA/metabolism , Active Transport, Cell Nucleus/drug effects , Biopsy , Cell Culture Techniques , Humans , Microscopy, Fluorescence , Tumor Necrosis Factor-alpha/pharmacology
17.
Front Pediatr ; 3: 2, 2015.
Article in English | MEDLINE | ID: mdl-25688341

ABSTRACT

Unbiased genetic diagnosis has increasingly associated seemingly unrelated somatic and immunological phenotypes. We report a male infant who presented within the first year of life with physical growth impairment, feeding difficulties, hyperemesis without diarrhea, and abnormal hair findings suggestive of trichorrhexis nodosa. With advancing age, moderate global developmental delay, susceptibility to frequent viral illnesses, otitis media, and purulent conjunctivitis were identified. Because of the repeated infections, an immunological evaluation was pursued and identified impaired antibody memory responses following pneumococcal vaccine administration. Immunoglobulin replacement therapy and nutritional support were employed as mainstays of therapy. The child is now aged 12 years and still without diarrhea. Whole exome sequencing identified compound heterozygous mutations in the TTC37 gene, a known cause of the trichohepatoenteric syndrome (THES). This case extends the known phenotype of THES and defines a potential subset for inclusion as an immune overlap syndrome.

18.
J Exp Med ; 210(3): 433-43, 2013 Mar 11.
Article in English | MEDLINE | ID: mdl-23440042

ABSTRACT

Primary immunodeficiencies (PIDs) represent exquisite models for studying mechanisms of human host defense. In this study, we report on two unrelated kindreds, with two patients each, who had cryptosporidial infections associated with chronic cholangitis and liver disease. Using exome and candidate gene sequencing, we identified two distinct homozygous loss-of-function mutations in the interleukin-21 receptor gene (IL21R; c.G602T, p.Arg201Leu and c.240_245delCTGCCA, p.C81_H82del). The IL-21R(Arg201Leu) mutation causes aberrant trafficking of the IL-21R to the plasma membrane, abrogates IL-21 ligand binding, and leads to defective phosphorylation of signal transducer and activator of transcription 1 (STAT1), STAT3, and STAT5. We observed impaired IL-21-induced proliferation and immunoglobulin class-switching in B cells, cytokine production in T cells, and NK cell cytotoxicity. Our study indicates that human IL-21R deficiency causes an immunodeficiency and highlights the need for early diagnosis and allogeneic hematopoietic stem cell transplantation in affected children.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Interleukin-21 Receptor alpha Subunit/genetics , Mutation , Child , Child, Preschool , Female , Humans , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/immunology , Interleukin-21 Receptor alpha Subunit/chemistry , Interleukin-21 Receptor alpha Subunit/physiology , Killer Cells, Natural/immunology , Male , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction
19.
Front Immunol ; 2: 61, 2011.
Article in English | MEDLINE | ID: mdl-22566850

ABSTRACT

Ectodermal dysplasias (ED) are uncommon genetic disorders resulting in abnormalities in ectodermally derived structures. Many ED-associated genes have been described, of which ectodysplasin-A (EDA) is one of the more common. The NF-κB essential modulator (NEMO encoded by the IKBKG gene) is unique in that mutations result in severe humoral and cellular immunologic defects in addition to ED. We describe three unrelated kindreds with defects in both EDA and IKBKG resulting from X-chromosome crossover. This demonstrates the importance of thorough immunologic consideration of patients with ED even when an EDA etiology is confirmed, and raises the possibility of a specific phenotype arising from coincident mutations in EDA and IKBKG.

SELECTION OF CITATIONS
SEARCH DETAIL