Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Article in English | MEDLINE | ID: mdl-38185418

ABSTRACT

BACKGROUND: Dedicator of cytokinesis 8 (DOCK8)-deficient patients have severe eczema, elevated IgE, and eosinophilia, features of atopic dermatitis (AD). OBJECTIVE: We sought to understand the mechanisms of eczema in DOCK8 deficiency. METHODS: Skin biopsy samples were characterized by histology, immunofluorescence microscopy, and gene expression. Skin barrier function was measured by transepidermal water loss. Allergic skin inflammation was elicited in mice by epicutaneous sensitization with ovalbumin (OVA) or cutaneous application of Staphylococcus aureus. RESULTS: Skin lesions of DOCK8-deficient patients exhibited type 2 inflammation, and the patients' skin was colonized by Saureus, as in AD. Unlike in AD, DOCK8-deficient patients had a reduced FOXP3:CD4 ratio in their skin lesions, and their skin barrier function was intrinsically intact. Dock8-/- mice exhibited reduced numbers of cutaneous T regulatory (Treg) cells and a normal skin barrier. Dock8-/- and mice with an inducible Dock8 deletion in Treg cells exhibited increased allergic skin inflammation after epicutaneous sensitization with OVA. DOCK8 was shown to be important for Treg cell stability at sites of allergic inflammation and for the generation, survival, and suppressive activity of inducible Treg cells. Adoptive transfer of wild-type, but not DOCK8-deficient, OVA-specific, inducible Treg cells suppressed allergic inflammation in OVA-sensitized skin of Dock8-/- mice. These mice developed severe allergic skin inflammation and elevated serum IgE levels after topical exposure to Saureus. Both were attenuated after adoptive transfer of WT but not DOCK8-deficient Treg cells. CONCLUSION: Treg cell dysfunction increases susceptibility to allergic skin inflammation in DOCK8 deficiency and synergizes with cutaneous exposure to Saureus to drive eczema in DOCK8 deficiency.

2.
J Allergy Clin Immunol ; 151(5): 1296-1306.e7, 2023 05.
Article in English | MEDLINE | ID: mdl-36690254

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is characterized by TH2-dominated skin inflammation and systemic response to cutaneously encountered antigens. The TH2 cytokines IL-4 and IL-13 play a critical role in the pathogenesis of AD. The Q576->R576 polymorphism in the IL-4 receptor alpha (IL-4Rα) chain common to IL-4 and IL-13 receptors alters IL-4 signaling and is associated with asthma severity. OBJECTIVE: We sought to investigate whether the IL-4Rα R576 polymorphism is associated with AD severity and exaggerates allergic skin inflammation in mice. METHODS: Nighttime itching interfering with sleep, Rajka-Langeland, and Eczema Area and Severity Index scores were used to assess AD severity. Allergic skin inflammation following epicutaneous sensitization of mice 1 or 2 IL-4Rα R576 alleles (QR and RR) and IL-4Rα Q576 (QQ) controls was assessed by flow cytometric analysis of cells and quantitative RT-PCR analysis of cytokines in skin. RESULTS: The frequency of nighttime itching in 190 asthmatic inner-city children with AD, as well as Rajka-Langeland and Eczema Area and Severity Index scores in 1116 White patients with AD enrolled in the Atopic Dermatitis Research Network, was higher in subjects with the IL-4Rα R576 polymorphism compared with those without, with statistical significance for the Rajka-Langeland score. Following epicutaneous sensitization of mice with ovalbumin or house dust mite, skin infiltration by CD4+ cells and eosinophils, cutaneous expression of Il4 and Il13, transepidermal water loss, antigen-specific IgE antibody levels, and IL-13 secretion by antigen-stimulated splenocytes were significantly higher in RR and QR mice compared with QQ controls. Bone marrow radiation chimeras demonstrated that both hematopoietic cells and stromal cells contribute to the mutants' exaggerated allergic skin inflammation. CONCLUSIONS: The IL-4Rα R576 polymorphism predisposes to more severe AD and increases allergic skin inflammation in mice.


Subject(s)
Dermatitis, Atopic , Eczema , Mice , Animals , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Th2 Cells , Skin/metabolism , Cytokines/metabolism , Inflammation/metabolism , Pruritus/metabolism , Eczema/metabolism
3.
Sci Immunol ; 6(63): eabf6723, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34533979

ABSTRACT

Inhibitor of nuclear factor kappa B kinase alpha (IKKα) is critical for p100/NF-κB2 phosphorylation and processing into p52 and activation of the noncanonical NF-κB pathway. A patient with recurrent infections, skeletal abnormalities, absent secondary lymphoid structures, reduced B cell numbers, hypogammaglobulinemia, and lymphocytic infiltration of intestine and liver was found to have a homozygous p.Y580C mutation in the helix-loop-helix domain of IKKα. The mutation preserves IKKα kinase activity but abolishes the interaction of IKKα with its activator NF-κB­inducing kinase and impairs lymphotoxin-ß­driven p100/NF-κB2 processing and VCAM1 expression. Homozygous IKKαY580C/Y580C mutant mice phenocopy the patient findings; lack marginal zone B cells, germinal centers, and antigen-specific T cell response to cutaneous immunization; have impaired Il17a expression; and are susceptible to cutaneous Staphylococcus aureus infection. In addition, these mice demonstrate a severe reduction in medullary thymic epithelial cells, impaired thymocyte negative selection, a restricted TCRVß repertoire, a selective expansion of potentially autoreactive T cell clones, a decreased frequency of regulatory T cells, and infiltration of liver, pancreas, and lung by activated T cells coinciding with organ damage. Hence, this study identifies IKKα deficiency as a previously undescribed cause of primary immunodeficiency with associated autoimmunity.


Subject(s)
Autoimmunity/immunology , I-kappa B Kinase/immunology , Mutation, Missense/genetics , Animals , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense/immunology
4.
Nature ; 591(7851): 633-638, 2021 03.
Article in English | MEDLINE | ID: mdl-33627867

ABSTRACT

The gut microbiota of preterm infants develops predictably1-7, with pioneer species colonizing the gut after birth, followed by an ordered succession of microorganisms. The gut microbiota is vital to the health of preterm infants8,9, but the forces that shape these predictable dynamics of microbiome assembly are unknown. The environment, the host and interactions between microorganisms all potentially shape the dynamics of the microbiota, but in such a complex ecosystem, identifying the specific role of any individual factor is challenging10-14. Here we use multi-kingdom absolute abundance quantification, ecological modelling and experimental validation to address this challenge. We quantify the absolute dynamics of bacteria, fungi and archaea in a longitudinal cohort of 178 preterm infants. We uncover microbial blooms and extinctions, and show that there is an inverse correlation between bacterial and fungal loads in the infant gut. We infer computationally and demonstrate experimentally in vitro and in vivo that predictable assembly dynamics may be driven by directed, context-dependent interactions between specific microorganisms. Mirroring the dynamics of macroscopic ecosystems15-17, a late-arriving member of the microbiome, Klebsiella, exploits the pioneer microorganism, Staphylococcus, to gain a foothold within the gut. Notably, we find that interactions between different kingdoms can influence assembly, with a single fungal species-Candida albicans-inhibiting multiple dominant genera of gut bacteria. Our work reveals the centrality of simple microbe-microbe interactions in shaping host-associated microbiota, which is critical both for our understanding of microbiota ecology and for targeted microbiota interventions.


Subject(s)
Biodiversity , Gastrointestinal Microbiome , Infant, Premature , Bacterial Load , Diet , Female , Gestational Age , Humans , Infant , Infant, Newborn , Male , Microbial Interactions , Reproducibility of Results
5.
J Clin Invest ; 131(3)2021 02 01.
Article in English | MEDLINE | ID: mdl-33529166

ABSTRACT

The coat protein I (COPI) complex mediates retrograde trafficking from the Golgi to the endoplasmic reticulum (ER). Five siblings with persistent bacterial and viral infections and defective humoral and cellular immunity had a homozygous p.K652E mutation in the γ1 subunit of COPI (γ1-COP). The mutation disrupts COPI binding to the KDEL receptor and impairs the retrieval of KDEL-bearing chaperones from the Golgi to the ER. Homozygous Copg1K652E mice had increased ER stress in activated T and B cells, poor antibody responses, and normal numbers of T cells that proliferated normally, but underwent increased apoptosis upon activation. Exposure of the mutants to pet store mice caused weight loss, lymphopenia, and defective T cell proliferation that recapitulated the findings in the patients. The ER stress-relieving agent tauroursodeoxycholic acid corrected the immune defects of the mutants and reversed the phenotype they acquired following exposure to pet store mice. This study establishes the role of γ1-COP in the ER retrieval of KDEL-bearing chaperones and thereby the importance of ER homeostasis in adaptive immunity.


Subject(s)
Apoptosis/immunology , B-Lymphocytes/immunology , Endoplasmic Reticulum Stress/immunology , Lymphocyte Activation , Mutation, Missense , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology , Amino Acid Substitution , Animals , Apoptosis/genetics , Coatomer Protein/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum Stress/genetics , Golgi Apparatus/genetics , Golgi Apparatus/immunology , Humans , Mice , Mice, Mutant Strains , Receptors, Peptide/genetics , Receptors, Peptide/immunology , Severe Combined Immunodeficiency/genetics
6.
J Allergy Clin Immunol ; 147(2): 723-726, 2021 02.
Article in English | MEDLINE | ID: mdl-32888943

ABSTRACT

BACKGROUND: Next-generation sequencing has become a first-line tool for the diagnosis of primary immunodeficiency. However, patient access remains limited because of restricted insurance coverage and a lack of guidelines addressing the use of targeted panels versus whole-exome sequencing (WES). OBJECTIVES: We sought to compare targeted next-generation sequencing with WES in a global population of patients with primary immunodeficiency. METHODS: This was a longitudinal study of 878 patients with likely primary immunodeficiency sequenced between 2010 and 2020. Most patients (n = 780) were first sequenced using a 264 gene panel. This was followed by WES in selected cases if a candidate gene was not found. A subset of patients (n = 98) were selected for a WES-only pipeline if the history was atypical for genes within the targeted panel. RESULTS: Disease-causing variants were identified in 498 of the 878 probands (56%), encompassing 152 distinct monogenic disorders. Sixteen patients had disorders that were novel at the time of sequencing (1.8%). Diagnostic yield in patients sequenced by targeted panel was 56% (433 of 780 patients), with subsequent WES leading to an additional 18 diagnoses (overall diagnostic yield 58%, 451 of 780 patients). The WES-only approach had a diagnostic yield of 45% (45 of 98 patients), reflecting that these cases had less common clinical and laboratory phenotypes. Cost analysis, based on current commercial WES and targeted panel prices, demonstrated savings ranging from $300 to $950 with a WES-only approach, depending on diagnostic yield. CONCLUSIONS: Advantages of WES over targeted next-generation sequencing include simplified workflow, reduced overall cost, and the potential for identification of novel diseases.


Subject(s)
Exome Sequencing/methods , High-Throughput Nucleotide Sequencing/methods , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Infant , Longitudinal Studies , Male , Middle Aged , Sequence Analysis, DNA , Young Adult
7.
J Allergy Clin Immunol ; 145(6): 1664-1672.e10, 2020 06.
Article in English | MEDLINE | ID: mdl-31945408

ABSTRACT

BACKGROUND: Deficiency of adenosine deaminase 2 (DADA2) is a syndrome with pleiotropic manifestations including vasculitis and hematologic compromise. A systematic definition of the relationship between adenosine deaminase 2 (ADA2) mutations and clinical phenotype remains unavailable. OBJECTIVE: We sought to test whether the impact of ADA2 mutations on enzyme function correlates with clinical presentation. METHODS: Patients with DADA2 with severe hematologic manifestations were compared with vasculitis-predominant patients. Enzymatic activity was assessed using expression constructs reflecting all 53 missense, nonsense, insertion, and deletion genotypes from 152 patients across the DADA2 spectrum. RESULTS: We identified patients with DADA2 presenting with pure red cell aplasia (n = 5) or bone marrow failure (BMF, n = 10) syndrome. Most patients did not exhibit features of vasculitis. Recurrent infection, hepatosplenomegaly, and gingivitis were common in patients with BMF, of whom half died from infection. Unlike patients with DADA2 with vasculitis, patients with pure red cell aplasia and BMF proved largely refractory to TNF inhibitors. ADA2 variants associated with vasculitis predominantly reflected missense mutations with at least 3% residual enzymatic activity. In contrast, pure red cell aplasia and BMF were associated with missense mutations with minimal residual enzyme activity, nonsense variants, and insertions/deletions resulting in complete loss of function. CONCLUSIONS: Functional interrogation of ADA2 mutations reveals an association of subtotal function loss with vasculitis, typically responsive to TNF blockade, whereas more extensive loss is observed in hematologic disease, which may be refractory to treatment. These findings establish a genotype-phenotype spectrum in DADA2.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Bone Marrow Failure Disorders/genetics , Child , Child, Preschool , Female , Genotype , Humans , Infant , Male , Mutation/genetics , Phenotype , Red-Cell Aplasia, Pure/genetics , Vasculitis/genetics
8.
J Allergy Clin Immunol ; 146(1): 192-202, 2020 07.
Article in English | MEDLINE | ID: mdl-31862378

ABSTRACT

BACKGROUND: The gene AK2 encodes the phosphotransferase adenylate kinase 2 (AK2). Human variants in AK2 cause reticular dysgenesis, a severe combined immunodeficiency with agranulocytosis, lymphopenia, and sensorineural deafness that requires hematopoietic stem cell transplantation for survival. OBJECTIVE: We investigated the mechanisms underlying recurrent sinopulmonary infections and hypogammaglobulinemia in 15 patients, ranging from 3 to 34 years of age, from 9 kindreds. Only 2 patients, both of whom had mildly impaired T-cell proliferation, each had a single clinically significant opportunistic infection. METHODS: Patient cells were studied with next-generation DNA sequencing, tandem mass spectrometry, and assays of lymphocyte and mitochondrial function. RESULTS: We identified 2 different homozygous variants in AK2. AK2G100S and AK2A182D permit residual protein expression, enzymatic activity, and normal numbers of neutrophils and lymphocytes. All but 1 patient had intact hearing. The patients' B cells had severely impaired proliferation and in vitro immunoglobulin secretion. With activation, the patients' B cells exhibited defective mitochondrial respiration and impaired regulation of mitochondrial membrane potential and quality. Although activated T cells from the patients with opportunistic infections demonstrated impaired mitochondrial function, the mitochondrial quality in T cells was preserved. Consistent with the capacity of activated T cells to utilize nonmitochondrial metabolism, these findings revealed a less strict cellular dependence of T-cell function on AK2 activity. Chemical inhibition of ATP synthesis in control T and B cells similarly demonstrated the greater dependency of B cells on mitochondrial function. CONCLUSIONS: Our patients demonstrate the in vivo sequelae of the cell-specific requirements for the functions of AK2 and mitochondria, particularly in B-cell activation and antibody production.


Subject(s)
Adenylate Kinase/genetics , B-Lymphocytes/immunology , Homozygote , Lymphocyte Activation/genetics , Mutation, Missense , Severe Combined Immunodeficiency/genetics , Adenylate Kinase/immunology , Adult , Amino Acid Substitution , Child , Child, Preschool , Female , Humans , Male , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology
10.
Turk J Pediatr ; 61(3): 413-417, 2019.
Article in English | MEDLINE | ID: mdl-31916720

ABSTRACT

Çakan M, Aktay-Ayaz N, Karadag SG, Tahir-Turanli E, Stafstrom K, Bainter W, Geha RS, Chou J. Atypical phenotype of an old disease or typical phenotype of a new disease: deficiency of adenosine deaminase 2. Turk J Pediatr 2019; 61: 413-417. Deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive autoinflammatory disorder caused by mutations in CECR1 (cat eye syndrome chromosome region, canditate 1) gene, which encodes the enzyme adenosine deaminase 2 necessary for endothelial cell survival and function. The diversity of the clinical phenotypes associated with DADA2 include polyarteritis nodosa-like vasculitic features, early-onset stroke, mild to severe immunodeficiency and cytopenias. The diagnosis of the disease may be difficult due to complex clinical phenotype. Herein, we present a case of DADA2 presenting with vasculitis, amarousis fugax, gastrointestinal bleeding and silent lacunar infarct successfully treated with etanercept.


Subject(s)
Adenosine Deaminase/deficiency , Agammaglobulinemia/blood , Brain Infarction/etiology , Intercellular Signaling Peptides and Proteins/deficiency , Polyarteritis Nodosa/etiology , Severe Combined Immunodeficiency/blood , Adenosine Deaminase/blood , Adenosine Deaminase/genetics , Adolescent , Brain Infarction/diagnosis , Brain Stem/pathology , DNA/genetics , DNA Mutational Analysis , Humans , Intercellular Signaling Peptides and Proteins/blood , Intercellular Signaling Peptides and Proteins/genetics , Magnetic Resonance Imaging , Male , Mutation , Phenotype , Polyarteritis Nodosa/diagnosis
12.
Clin Immunol ; 191: 21-26, 2018 06.
Article in English | MEDLINE | ID: mdl-29307770

ABSTRACT

Pattern recognition receptors (PRRs), receptors of the innate immune system, are important in interaction with pathogens. Caspase Recruitment Domain-containing protein 9 (CARD9), a member of PRRs, is an intracellular adaptor protein important in fungal defense. CARD9 deficiency causes a rare primary immunodeficiency (PID) characterized by superficial and deep fungal infections. We report a 17year-old female with a homozygous nonsense mutation in CARD9, who presented with severe cerebral fungal infection of the central nervous system. She was also found to have an heterozygous NLRP12 mutation, which may have had add-on effect on the severity of the infection.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Central Nervous System Fungal Infections/etiology , Codon, Nonsense , Mycoses/etiology , Adolescent , CARD Signaling Adaptor Proteins/deficiency , Central Nervous System Fungal Infections/genetics , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mycoses/genetics
13.
J Allergy Clin Immunol ; 141(4): 1450-1458, 2018 04.
Article in English | MEDLINE | ID: mdl-28916186

ABSTRACT

BACKGROUND: Combined immunodeficiencies (CIDs) are diseases of defective adaptive immunity with diverse clinical phenotypes. Although CIDs are more prevalent in the Middle East than Western countries, the resources for genetic diagnosis are limited. OBJECTIVES: This study aims to characterize the categories of patients with CIDs in Iran clinically and genetically. METHODS: Clinical and laboratory data were obtained from 696 patients with CIDs. Patients were subdivided into those with syndromic (344 patients) and nonsyndromic (352 patients) CIDs. Targeted DNA sequencing was performed on 243 (34.9%) patients. RESULTS: The overall diagnostic yield of the 243 sequenced patients was 77.8% (189 patients). The clinical diagnosis of hyper-IgE syndrome (P < .001), onset of disease at greater than 5 years (P = .02), and absence of multiple affected family members (P = .04) were significantly more frequent in the patients without a genetic diagnosis. An autosomal recessive disease was found in 62.9% of patients, reflecting the high rate of consanguinity in this cohort. Mutations impairing VDJ recombination and DNA repair were the most common underlying causes of CIDs. However, in patients with syndromic CIDs, autosomal recessive mutations in ataxia-telangiectasia mutated (ATM), autosomal dominant mutations in signal transducer and activator of transcription 3 (STAT3), and microdeletions in 22q11.21 were the most commonly affected genomic loci. Patients with syndromic CIDs had a significantly lower 5-year survival rate rather than those with nonsyndromic CIDs. CONCLUSIONS: This study provides proof of principle for the application of targeted next-generation sequencing panels in countries with limited diagnostic resources. The effect of genetic diagnosis on clinical care requires continued improvements in therapeutic resources for these patients.


Subject(s)
Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Adolescent , Child , Child, Preschool , Consanguinity , Female , Genes, Recessive/genetics , Genes, Recessive/immunology , Genetic Predisposition to Disease/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Immunologic Deficiency Syndromes/mortality , Infant , Iran , Job Syndrome/genetics , Job Syndrome/immunology , Job Syndrome/mortality , Male , Mutation/genetics , Mutation/immunology , Phenotype , Retrospective Studies , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Sequence Analysis, DNA/methods , Survival Rate
14.
Front Immunol ; 9: 3146, 2018.
Article in English | MEDLINE | ID: mdl-30697212

ABSTRACT

Objective: To present the genetic causes of patients with primary immune deficiencies (PIDs) in Kuwait between 2004 and 2017. Methods: The data was obtained from the Kuwait National Primary Immunodeficiency Disorders Registry. Genomic DNA from patients with clinical and immunological features of PID was sequenced using Sanger sequencing (SS), next generation sequencing (NGS) of targeted genes, whole exome sequencing (WES), and/or whole genome sequencing (WGS). Functional assays were utilized to assess the biologic effect of identified variants. Fluorescence in situ hybridization (FISH) for 22q11.2 deletion and genomic hybridizations arrays were performed when thymic defects were suspected. Results: A total of 264 patients were registered during the study period with predominance of patients with immunodeficiencies affecting cellular and humoral immunity (35.2%), followed by combined immunodeficiencies with associated syndromic features (24%). Parental consanguinity and family history suggestive of PID were reported in 213 (81%) and 145 patients (55%), respectively. Genetic testing of 206 patients resulted in a diagnostic yield of 70%. Mutations were identified in 46 different genes and more than 90% of the reported genetic defects were transmitted by in an autosomal recessive pattern. The majority of the mutations were missense mutations (57%) followed by deletions and frame shift mutations. Five novel disease-causing genes were discovered. Conclusions: Genetic testing should be an integral part in the management of primary immunodeficiency patients. This will help the delivery of precision medicine and facilitate proper genetic counseling. Studying inbred populations using sophisticated diagnostic methods can allow better understanding of the genetics of primary immunodeficiency disorders.


Subject(s)
Consanguinity , Genetic Association Studies , Genetic Predisposition to Disease , Immunologic Deficiency Syndromes/epidemiology , Immunologic Deficiency Syndromes/genetics , Alleles , Biomarkers , Cohort Studies , Female , Genetic Association Studies/methods , Genetics, Population , Humans , Immunologic Deficiency Syndromes/diagnosis , In Situ Hybridization, Fluorescence , Kuwait/epidemiology , Male , Mutation , Pedigree , Polymorphism, Single Nucleotide , Exome Sequencing
15.
J Clin Invest ; 127(12): 4415-4420, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29106381

ABSTRACT

Primary immunodeficiencies are often monogenic disorders characterized by vulnerability to specific infectious pathogens. Here, we performed whole-exome sequencing of a patient with disseminated Mycobacterium abscessus, Streptococcus viridians bacteremia, and cytomegalovirus (CMV) viremia and identified mutations in 2 genes that regulate distinct IFN pathways. The patient had a homozygous frameshift deletion in IFNGR2, which encodes the signal transducing chain of the IFN-γ receptor, that resulted in minimal protein expression and abolished downstream signaling. The patient also harbored a homozygous deletion in IFNAR1 (IFNAR1*557Gluext*46), which encodes the IFN-α receptor signaling subunit. The IFNAR1*557Gluext*46 resulted in replacement of the stop codon with 46 additional codons at the C-terminus. The level of IFNAR1*557Gluext*46 mutant protein expressed in patient fibroblasts was comparable to levels of WT IFNAR1 in control fibroblasts. IFN-α-induced signaling was impaired in the patient fibroblasts, as evidenced by decreased STAT1/STAT2 phosphorylation, nuclear translocation of STAT1, and expression of IFN-α-stimulated genes critical for CMV immunity. Pretreatment with IFN-α failed to suppress CMV protein expression in patient fibroblasts, whereas expression of WT IFNAR1 restored IFN-α-mediated suppression of CMV. This study identifies a human IFNAR1 mutation and describes a digenic immunodeficiency specific to type I and type II IFNs.


Subject(s)
Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Mutation , Receptor, Interferon alpha-beta , Receptors, Interferon , Bacteremia/genetics , Bacteremia/immunology , Bacteremia/microbiology , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Female , Fibroblasts/immunology , Fibroblasts/microbiology , Fibroblasts/virology , Genetic Diseases, Inborn/microbiology , Genetic Diseases, Inborn/virology , Humans , Immunologic Deficiency Syndromes/microbiology , Immunologic Deficiency Syndromes/virology , Male , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium abscessus/immunology , Phosphorylation/genetics , Phosphorylation/immunology , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptors, Interferon/genetics , Receptors, Interferon/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/immunology , Streptococcal Infections/genetics , Streptococcal Infections/immunology , Viremia/genetics , Viremia/immunology , Viremia/virology , Viridans Streptococci/immunology
16.
Clin Immunol ; 183: 198-200, 2017 10.
Article in English | MEDLINE | ID: mdl-28917720

ABSTRACT

JAK3 is a tyrosine kinase essential for signaling downstream of the common gamma chain subunit shared by multiple cytokine receptors. JAK3 deficiency results in T-B+NK- severe combined immune deficiency (SCID). We report a patient with SCID due to a novel mutation in the JAK3 JH4 domain. The function of the JH4 domain remains unknown. This is the first report of a missense mutation in the JAK3 JH4 domain, thereby demonstrating the importance of the JH4 domain of JAK3 in host immunity.


Subject(s)
Janus Kinase 3/metabolism , Osteomyelitis/genetics , Severe Combined Immunodeficiency/genetics , Spine/pathology , Fatal Outcome , Female , Humans , Infant , Janus Kinase 3/genetics , Mutation , Osteomyelitis/complications , Protein Domains , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/pathology
17.
J Exp Med ; 214(7): 1937-1947, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28600438

ABSTRACT

The treatment of chronic mucocutaneous ulceration is challenging, and only some patients respond selectively to inhibitors of tumor necrosis factor-α (TNF). TNF activates opposing pathways leading to caspase-8-mediated apoptosis as well as nuclear factor κB (NF-κB)-dependent cell survival. We investigated the etiology of autosomal-dominant, mucocutaneous ulceration in a family whose proband was dependent on anti-TNF therapy for sustained remission. A heterozygous mutation in RELA, encoding the NF-κB subunit RelA, segregated with the disease phenotype and resulted in RelA haploinsufficiency. The patients' fibroblasts exhibited increased apoptosis in response to TNF, impaired NF-κB activation, and defective expression of NF-κB-dependent antiapoptotic genes. Rela+/- mice have similarly impaired NF-κB activation, develop cutaneous ulceration from TNF exposure, and exhibit severe dextran sodium sulfate-induced colitis, ameliorated by TNF inhibition. These findings demonstrate an essential contribution of biallelic RELA expression in protecting stromal cells from TNF-mediated cell death, thus delineating the mechanisms driving the effectiveness of TNF inhibition in this disease.


Subject(s)
Genes, Dominant/genetics , Haploinsufficiency , Oral Ulcer/genetics , Skin Ulcer/genetics , Transcription Factor RelA/genetics , Animals , Base Sequence , Cells, Cultured , Chronic Disease , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate , Exome/genetics , Female , Fibroblasts/metabolism , Gene Expression Profiling/methods , Humans , Male , Mice, Knockout , NF-kappa B/metabolism , Pedigree , Sequence Analysis, DNA/methods , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
J Allergy Clin Immunol ; 140(6): 1651-1659.e1, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28192143

ABSTRACT

BACKGROUND: Leucine-rich repeat containing 8A (LRRC8A) is an ubiquitously expressed transmembrane protein with 17 leucine-rich repeats (LRRs) at its C-terminal end and is an essential component of the volume-regulated anion channel (VRAC), which controls cellular volume. A heterozygous mutation in LRRC8A that truncates the 2 terminal LRRs was reported in a patient with agammaglobulinemia and absent B cells and was demonstrated to exert a dominant negative effect on T- and B-cell development in mice. Lrrc8a-/- mice have severely defective T-cell development and function. It is not known whether the T- and B-cell defects caused by LRRC8A deficiency are caused by loss of VRAC activity. OBJECTIVE: We sought to determine whether VRAC activity is required for normal T-cell development and function. METHODS: VRAC activity was examined by using patch-clamp analysis. Flow cytometry was used to examine T-cell development. T-cell proliferation, cytokine secretion, and antibody titers were measured by using standard techniques. RESULTS: We demonstrate that the spontaneous mouse mutant ébouriffé (ebo/ebo) harbors a homozygous 2-bp frameshift mutation in Lrrc8a that truncates the 15 terminal LRRs of LRRC8A. The Lrrc8aebo mutation does not affect protein expression but drastically diminishes VRAC activity in T cells. ebo/ebo mice share features with Lrrc8a-/- mice that include curly hair, infertility, reduced longevity, and kidney abnormalities. However, in contrast to Lrrc8a-/- mice, ebo/ebo mice have normal T-cell development and function and intact antibody response to T-dependent antigen. CONCLUSION: LRRC8A-dependent VRAC activity is dispensable for T-cell development and function.


Subject(s)
Agammaglobulinemia/genetics , Anion Transport Proteins/metabolism , B-Lymphocytes/physiology , Membrane Proteins/metabolism , T-Lymphocytes/physiology , Animals , Antibodies/blood , Cell Differentiation , Cell Proliferation , Cell Size , Cells, Cultured , Humans , Ion Transport/genetics , Lymphocyte Activation , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Sequence Deletion/genetics
19.
Clin Immunol ; 171: 65-66, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27538774

ABSTRACT

Whole exome sequencing is increasingly used in the diagnosis of primary immunodeficiencies due to the overlapping and atypical presentations of these disorders. We report two patients who presented with recurrent infections and early onset colitis. They were investigated by whole exome sequencing due to suspicion of primary immunodeficiency and found to have mutations in pyrin known to cause familial Mediterranean fever.


Subject(s)
Colitis/genetics , Familial Mediterranean Fever/genetics , Immunologic Deficiency Syndromes/genetics , Pyrin/genetics , Child, Preschool , Colitis/diagnosis , Consanguinity , Diagnosis, Differential , Familial Mediterranean Fever/diagnosis , Female , Humans , Immunologic Deficiency Syndromes/diagnosis , Infant , Male , Mutation
20.
Clin Immunol ; 166-167: 100-2, 2016 05.
Article in English | MEDLINE | ID: mdl-27063589

ABSTRACT

ORAI1 is the pore-forming subunit of the calcium release-activated calcium channel responsible for calcium influx into cells triggered by endoplasmic reticulum store depletion. We report here a patient with severe combined immunodeficiency and absent store-operated calcium entry due to a novel mutation in ORAI1 that results in the expression of a C-terminally truncated protein that abolishes ORAI1 binding to STIM1.


Subject(s)
Mutation , Neoplasm Proteins/genetics , ORAI1 Protein/genetics , Severe Combined Immunodeficiency/genetics , Stromal Interaction Molecule 1/genetics , Base Sequence , Exome/genetics , Fatal Outcome , Female , HEK293 Cells , Homozygote , Humans , Infant , Neoplasm Proteins/metabolism , ORAI1 Protein/metabolism , Protein Binding/genetics , Sequence Analysis, DNA , Sequence Deletion , Severe Combined Immunodeficiency/metabolism , Stromal Interaction Molecule 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...