Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Publication year range
1.
Cell ; 187(2): 390-408.e23, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38157855

ABSTRACT

We describe a human lung disease caused by autosomal recessive, complete deficiency of the monocyte chemokine receptor C-C motif chemokine receptor 2 (CCR2). Nine children from five independent kindreds have pulmonary alveolar proteinosis (PAP), progressive polycystic lung disease, and recurrent infections, including bacillus Calmette Guérin (BCG) disease. The CCR2 variants are homozygous in six patients and compound heterozygous in three, and all are loss-of-expression and loss-of-function. They abolish CCR2-agonist chemokine C-C motif ligand 2 (CCL-2)-stimulated Ca2+ signaling in and migration of monocytic cells. All patients have high blood CCL-2 levels, providing a diagnostic test for screening children with unexplained lung or mycobacterial disease. Blood myeloid and lymphoid subsets and interferon (IFN)-γ- and granulocyte-macrophage colony-stimulating factor (GM-CSF)-mediated immunity are unaffected. CCR2-deficient monocytes and alveolar macrophage-like cells have normal gene expression profiles and functions. By contrast, alveolar macrophage counts are about half. Human complete CCR2 deficiency is a genetic etiology of PAP, polycystic lung disease, and recurrent infections caused by impaired CCL2-dependent monocyte migration to the lungs and infected tissues.


Subject(s)
Pulmonary Alveolar Proteinosis , Receptors, CCR2 , Child , Humans , Lung/metabolism , Macrophages, Alveolar/metabolism , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/diagnosis , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Reinfection/metabolism
2.
Cell ; 184(4): 1017-1031.e14, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33548172

ABSTRACT

Antibodies mediate natural and vaccine-induced immunity against viral and bacterial pathogens, whereas fungi represent a widespread kingdom of pathogenic species for which neither vaccine nor neutralizing antibody therapies are clinically available. Here, using a multi-kingdom antibody profiling (multiKAP) approach, we explore the human antibody repertoires against gut commensal fungi (mycobiota). We identify species preferentially targeted by systemic antibodies in humans, with Candida albicans being the major inducer of antifungal immunoglobulin G (IgG). Fungal colonization of the gut induces germinal center (GC)-dependent B cell expansion in extraintestinal lymphoid tissues and generates systemic antibodies that confer protection against disseminated C. albicans or C. auris infection. Antifungal IgG production depends on the innate immunity regulator CARD9 and CARD9+CX3CR1+ macrophages. In individuals with invasive candidiasis, loss-of-function mutations in CARD9 are associated with impaired antifungal IgG responses. These results reveal an important role of gut commensal fungi in shaping the human antibody repertoire through CARD9-dependent induction of host-protective antifungal IgG.


Subject(s)
Antibodies, Fungal/immunology , CARD Signaling Adaptor Proteins/metabolism , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Immunity , Immunoglobulin G/immunology , Mycobiome/immunology , Animals , B-Lymphocytes/immunology , Candida albicans/immunology , Candidiasis/immunology , Candidiasis/microbiology , Feces/microbiology , Germinal Center/immunology , Humans , Mice, Inbred C57BL , Phagocytes/metabolism , Polymorphism, Single Nucleotide/genetics , Protein Binding , Signal Transduction
4.
Nature ; 628(8008): 620-629, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509369

ABSTRACT

Epstein-Barr virus (EBV) infection can engender severe B cell lymphoproliferative diseases1,2. The primary infection is often asymptomatic or causes infectious mononucleosis (IM), a self-limiting lymphoproliferative disorder3. Selective vulnerability to EBV has been reported in association with inherited mutations impairing T cell immunity to EBV4. Here we report biallelic loss-of-function variants in IL27RA that underlie an acute and severe primary EBV infection with a nevertheless favourable outcome requiring a minimal treatment. One mutant allele (rs201107107) was enriched in the Finnish population (minor allele frequency = 0.0068) and carried a high risk of severe infectious mononucleosis when homozygous. IL27RA encodes the IL-27 receptor alpha subunit5,6. In the absence of IL-27RA, phosphorylation of STAT1 and STAT3 by IL-27 is abolished in T cells. In in vitro studies, IL-27 exerts a synergistic effect on T-cell-receptor-dependent T cell proliferation7 that is deficient in cells from the patients, leading to impaired expansion of potent anti-EBV effector cytotoxic CD8+ T cells. IL-27 is produced by EBV-infected B lymphocytes and an IL-27RA-IL-27 autocrine loop is required for the maintenance of EBV-transformed B cells. This potentially explains the eventual favourable outcome of the EBV-induced viral disease in patients with IL-27RA deficiency. Furthermore, we identified neutralizing anti-IL-27 autoantibodies in most individuals who developed sporadic infectious mononucleosis and chronic EBV infection. These results demonstrate the critical role of IL-27RA-IL-27 in immunity to EBV, but also the hijacking of this defence by EBV to promote the expansion of infected transformed B cells.


Subject(s)
Epstein-Barr Virus Infections , Interleukin-27 , Receptors, Interleukin , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Young Adult , Alleles , B-Lymphocytes/pathology , B-Lymphocytes/virology , CD8-Positive T-Lymphocytes/pathology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/therapy , Finland , Gene Frequency , Herpesvirus 4, Human , Homozygote , Infectious Mononucleosis/complications , Infectious Mononucleosis/genetics , Infectious Mononucleosis/therapy , Interleukin-27/immunology , Interleukin-27/metabolism , Loss of Function Mutation , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Treatment Outcome
5.
Nature ; 623(7988): 803-813, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37938781

ABSTRACT

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Subject(s)
Autoantibodies , Genetic Predisposition to Disease , Interferon Type I , NF-kappa B , Humans , Autoantibodies/immunology , COVID-19/genetics , COVID-19/immunology , Gain of Function Mutation , Heterozygote , I-kappa B Proteins/deficiency , I-kappa B Proteins/genetics , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Loss of Function Mutation , NF-kappa B/deficiency , NF-kappa B/genetics , NF-kappa B p52 Subunit/deficiency , NF-kappa B p52 Subunit/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , Thymus Gland/abnormalities , Thymus Gland/immunology , Thymus Gland/pathology , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , AIRE Protein , NF-kappaB-Inducing Kinase
6.
Proc Natl Acad Sci U S A ; 119(21): e2200413119, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35576468

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection fatality rate (IFR) doubles with every 5 y of age from childhood onward. Circulating autoantibodies neutralizing IFN-α, IFN-ω, and/or IFN-ß are found in ∼20% of deceased patients across age groups, and in ∼1% of individuals aged <70 y and in >4% of those >70 y old in the general population. With a sample of 1,261 unvaccinated deceased patients and 34,159 individuals of the general population sampled before the pandemic, we estimated both IFR and relative risk of death (RRD) across age groups for individuals carrying autoantibodies neutralizing type I IFNs, relative to noncarriers. The RRD associated with any combination of autoantibodies was higher in subjects under 70 y old. For autoantibodies neutralizing IFN-α2 or IFN-ω, the RRDs were 17.0 (95% CI: 11.7 to 24.7) and 5.8 (4.5 to 7.4) for individuals <70 y and ≥70 y old, respectively, whereas, for autoantibodies neutralizing both molecules, the RRDs were 188.3 (44.8 to 774.4) and 7.2 (5.0 to 10.3), respectively. In contrast, IFRs increased with age, ranging from 0.17% (0.12 to 0.31) for individuals <40 y old to 26.7% (20.3 to 35.2) for those ≥80 y old for autoantibodies neutralizing IFN-α2 or IFN-ω, and from 0.84% (0.31 to 8.28) to 40.5% (27.82 to 61.20) for autoantibodies neutralizing both. Autoantibodies against type I IFNs increase IFRs, and are associated with high RRDs, especially when neutralizing both IFN-α2 and IFN-ω. Remarkably, IFRs increase with age, whereas RRDs decrease with age. Autoimmunity to type I IFNs is a strong and common predictor of COVID-19 death.


Subject(s)
Antibodies, Neutralizing , Autoantibodies , Autoimmunity , COVID-19 , Interferon Type I , SARS-CoV-2 , Adult , Age Factors , Aged , Aged, 80 and over , Antibodies, Neutralizing/blood , Autoantibodies/blood , COVID-19/immunology , COVID-19/mortality , Female , Humans , Interferon Type I/immunology , Male , Middle Aged , Risk
7.
J Clin Immunol ; 44(7): 163, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008214

ABSTRACT

BACKGROUND: Cryptococcosis is a life-threatening disease caused by Cryptococcus neoformans or C. gattii. Neutralizing autoantibodies (auto-Abs) against granulocyte-macrophage colony-stimulating factor (GM-CSF) in otherwise healthy adults with cryptococcal meningitis have been described since 2013. We searched for neutralizing auto-Abs in sera collected from Colombian patients with non-HIV-associated cryptococcosis in a retrospective national cohort from 1997 to 2016. METHODS: We reviewed clinical and laboratory records and assessed the presence of neutralizing auto-Abs against GM-CSF in 30 HIV negative adults with cryptococcosis (13 caused by C. gattii and 17 caused by C. neoformans). RESULTS: We detected neutralizing auto-Abs against GM-CSF in the sera of 10 out of 13 (77%) patients infected with C. gattii and one out of 17 (6%) patients infected with C. neoformans. CONCLUSIONS: We report eleven Colombian patients diagnosed with cryptococcosis who had auto-Abs that neutralize GM-CSF. Among these patients, ten were infected with C. gattii and only one with C. neoformans.


Subject(s)
Antibodies, Neutralizing , Autoantibodies , Cryptococcosis , Cryptococcus gattii , Cryptococcus neoformans , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Autoantibodies/blood , Autoantibodies/immunology , Male , Colombia , Female , Adult , Cryptococcus gattii/immunology , Middle Aged , Cryptococcus neoformans/immunology , Cryptococcosis/immunology , Cryptococcosis/diagnosis , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Retrospective Studies , HIV Seronegativity/immunology , Young Adult , Aged
8.
J Clin Immunol ; 44(3): 62, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38363432

ABSTRACT

PURPOSE: Inborn errors of IFN-γ immunity underlie Mendelian susceptibility to mycobacterial disease (MSMD). Twenty-two genes with products involved in the production of, or response to, IFN-γ and variants of which underlie MSMD have been identified. However, pathogenic variants of IFNG encoding a defective IFN-γ have been described in only two siblings, who both underwent hematopoietic stem cell transplantation (HCST). METHODS: We characterized a new patient with MSMD by genetic, immunological, and clinical means. Therapeutic decisions were taken on the basis of these findings. RESULTS: The patient was born to consanguineous Turkish parents and developed bacillus Calmette-Guérin (BCG) disease following vaccination at birth. Whole-exome sequencing revealed a homozygous private IFNG variant (c.224 T > C, p.F75S). Upon overexpression in recipient cells or constitutive expression in the patient's cells, the mutant IFN-γ was produced within the cells but was not correctly folded or secreted. The patient was treated for 6 months with two or three antimycobacterial drugs only and then for 30 months with subcutaneous recombinant IFN-γ1b plus two antimycobacterial drugs. Treatment with IFN-γ1b finally normalized all biological parameters. The patient presented no recurrence of mycobacterial disease or other related infectious diseases. The treatment was well tolerated, without the production of detectable autoantibodies against IFN-γ. CONCLUSION: We describe a patient with a new form of autosomal recessive IFN-γ deficiency, with intracellular, but not extracellular IFN-γ. IFN-γ1b treatment appears to have been beneficial in this patient, with no recurrence of mycobacterial infection over a period of more than 30 months. This targeted treatment provides an alternative to HCST in patients with complete IFN-γ deficiency or at least an option to better control mycobacterial infection prior to HCST.


Subject(s)
Mycobacterium Infections , Mycobacterium bovis , Infant, Newborn , Humans , Genetic Predisposition to Disease , Interferon-gamma , Mycobacterium Infections/genetics , Homozygote
9.
J Clin Immunol ; 43(5): 921-932, 2023 07.
Article in English | MEDLINE | ID: mdl-36821021

ABSTRACT

BACKGROUND: Cryptococcosis is a potentially life-threatening fungal disease caused by encapsulated yeasts of the genus Cryptococcus, mostly C. neoformans or C. gattii. Cryptococcal meningitis is the most frequent clinical manifestation in humans. Neutralizing autoantibodies (auto-Abs) against granulocyte-macrophage colony-stimulating factor (GM-CSF) have recently been discovered in otherwise healthy adult patients with cryptococcal meningitis, mostly caused by C. gattii. We hypothesized that three Colombian patients with cryptococcal meningitis caused by C. neoformans in two of them would carry high plasma levels of neutralizing auto-Abs against GM-CSF. METHODS: We reviewed medical and laboratory records, performed immunological evaluations, and tested for anti-cytokine auto-Abs three previously healthy HIV-negative adults with disseminated cryptococcosis. RESULTS: Peripheral blood leukocyte subset levels and serum immunoglobulin concentrations were within the normal ranges. We detected high levels of neutralizing auto-Abs against GM-CSF in the plasma of all three patients. CONCLUSIONS: We report three Colombian patients with disseminated cryptococcosis associated with neutralizing auto-Abs against GM-CSF. Further studies should evaluate the genetic contribution to anti-GM-CSF autoantibody production and the role of the GM-CSF signaling pathway in the immune response to Cryptococcus spp.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Meningitis, Cryptococcal , Adult , Humans , Granulocyte-Macrophage Colony-Stimulating Factor , Meningitis, Cryptococcal/diagnosis , Autoantibodies , Colombia , Cryptococcosis/diagnosis
10.
J Clin Immunol ; 43(6): 1093-1103, 2023 08.
Article in English | MEDLINE | ID: mdl-37209324

ABSTRACT

Autoantibodies (auto-Abs) neutralizing type I interferons (IFNs) are found in the blood of at least 15% of unvaccinated patients with life-threatening COVID-19 pneumonia. We report here the presence of auto-Abs neutralizing type I IFNs in the bronchoalveolar lavage (BAL) of 54 of the 415 unvaccinated patients (13%) with life-threatening COVID-19 pneumonia tested. The 54 individuals with neutralizing auto-Abs in the BAL included 45 (11%) with auto-Abs against IFN-α2, 37 (9%) with auto-Abs against IFN-ω, 54 (13%) with auto-Abs against IFN-α2 and/or ω, and five (1%) with auto-Abs against IFN-ß, including three (0.7%) with auto-Abs neutralizing IFN-α2, IFN-ω, and IFN-ß, and two (0.5%) with auto-Abs neutralizing IFN-α2 and IFN-ß. Auto-Abs against IFN-α2 also neutralize the other 12 subtypes of IFN-α. Paired plasma samples were available for 95 patients. All seven patients with paired samples who had detectable auto-Abs in BAL also had detectable auto-Abs in plasma, and one patient had auto-Abs detectable only in blood. Auto-Abs neutralizing type I IFNs are, therefore, present in the alveolar space of at least 10% of patients with life-threatening COVID-19 pneumonia. These findings suggest that these auto-Abs impair type I IFN immunity in the lower respiratory tract, thereby contributing to hypoxemic COVID-19 pneumonia.


Subject(s)
COVID-19 , Interferon Type I , Humans , Autoantibodies , Interferon-alpha , Bronchoalveolar Lavage
11.
J Clin Immunol ; 42(2): 336-349, 2022 02.
Article in English | MEDLINE | ID: mdl-34791587

ABSTRACT

BACKGROUND: CARD9 deficiency is an autosomal recessive primary immunodeficiency underlying increased susceptibility to fungal infection primarily presenting as invasive CNS Candida and/or cutaneous/invasive dermatophyte infections. More recently, a rare heterozygous dominant negative CARD9 variant c.1434 + 1G > C was reported to be protective from inflammatory bowel disease. OBJECTIVE: We studied two siblings carrying homozygous CARD9 variants (c.1434 + 1G > C) and born to heterozygous asymptomatic parents. One sibling was asymptomatic and the other presented with candida esophagitis, upper respiratory infections, hypogammaglobulinemia, and low class-switched memory B cells. METHODS AND RESULTS: The CARD9 c.1434 + 1G > C variant generated two mutant transcripts confirmed by mRNA and protein expression: an out-of-frame c.1358-1434 deletion/ ~ 55 kDa protein (CARD9Δex.11) and an in-frame c.1417-1434 deletion/ ~ 61 kDa protein (CARD9Δ18 nt.). Neither transcript was able to form a complete/functional CBM complex, which includes TRIM62. Based on the index patient's CVID-like phenotype, CARD9 expression was tested and detected in lymphocytes and monocytes from humans and mice. The functional impact of different CARD9 mutations and gene dosage conditions was evaluated in heterozygous and homozygous c.1434 + 1 G > C members of the index family, and in WT (two WT alleles), haploinsufficiency (one WT, one null allele), and null (two null alleles) individuals. CARD9 gene dosage impacted lymphocyte and monocyte functions including cytokine generation, MAPK activation, T-helper commitment, transcription, plasmablast differentiation, and immunoglobulin production in a differential manner. CONCLUSIONS: CARD9 exon 11 integrity is critical to CBM complex function. CARD9 is expressed and affects particular T and B cell functions in a gene dosage-dependent manner, which in turn may contribute to the phenotype of CARD9 deficiency.


Subject(s)
Candidiasis, Chronic Mucocutaneous , Alleles , Animals , CARD Signaling Adaptor Proteins/genetics , Gene Dosage , Homozygote , Humans , Mice , Phenotype
12.
Med Mycol ; 59(2): 180-188, 2021 Feb 04.
Article in English | MEDLINE | ID: mdl-32526033

ABSTRACT

Candidiasis is characterized by susceptibility to recurrent or persistent infections caused by Candida spp., typically Candida albicans, of cutaneous and mucosal surfaces. In this report, function and frequency of Th17 cells as well as genetics of patients susceptible to mucocutaneous candidiasis were studied. For patients, T-cell proliferation tests in response to Candida antigen, Th17 cell proportions, and STAT1 phosphorylation were evaluated through flow cytometry. Expression of IL17A, IL17F and IL22 genes were measured by real-time quantitative PCR. At the same time, whole exome sequencing was performed for all patients. We identified two heterozygous substitutions, one: c.821G > A (p. R274Q) was found in a multiplex family with three individuals affected, the second one: c.812A > C (p. Q271P) was found in a sporadic case. Both mutations are located in the coiled-coil domain (CCD) of STAT1. The frequency of Th17 cells, IL17A, IL17F, and IL22 gene expression in patients' peripheral blood mononuclear cells (PBMCs), and T-cell proliferation to Candida antigens were significantly reduced in the patients as compared to healthy controls. An increased STAT1 phosphorylation was observed in patients' PBMCs upon interferon (IFN)-γ stimulation as compared to healthy controls. We report two different but neighboring heterozygous mutations, located in exon 10 of the STAT1 gene, in four Iranian patients with CMC, one of whom also had hypothyroidism. These mutations were associated with impaired T cell proliferation to Candida antigen, low Th17 cell proportions, and increased STAT1 phosphorylation upon IFN-γ. We suggest that interfering with STAT1 phosphorylation might be a promising way for potential therapeutic measurements for such patients.


Subject(s)
Candidiasis, Chronic Mucocutaneous/genetics , Candidiasis, Chronic Mucocutaneous/microbiology , Family , Gain of Function Mutation , STAT1 Transcription Factor/genetics , Th17 Cells/immunology , Adolescent , Cell Proliferation , Child, Preschool , Female , Flow Cytometry , Gene Expression , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Iran , Male , Phosphorylation , Th17 Cells/pathology , Young Adult
13.
J Clin Immunol ; 40(2): 359-366, 2020 02.
Article in English | MEDLINE | ID: mdl-31940125

ABSTRACT

PURPOSE: Caspase-associated recruitment domain-9 (CARD9) deficiency is an inborn error of immunity that typically predisposes otherwise healthy patients to single fungal infections and the occurrence of multiple invasive fungal infections is rare. It has been described as the first known condition that predisposes to extrapulmonary Aspergillus infection with preserved lungs. We present a patient that expands the clinical variability of CARD9 deficiency. MATERIALS AND METHODS: Genetic analysis was performed by Sanger sequencing. Neutrophils and mononuclear phagocyte response to fungal stimulation were evaluated through luminol-enhanced chemiluminescence and whole blood production of the proinflammatory mediator interleukin (IL)-6, respectively. RESULTS: We report a 56-year-old Argentinean woman, whose invasive Exophiala spinifera infection at the age of 32 years was unexplained and reported in year 2004. At the age of 49 years, she presented with chronic pulmonary disease due to Aspergillus nomius. After partial improvement following treatment with caspofungin and posaconazole, right pulmonary bilobectomy was performed. Despite administration of multiple courses of antifungals, sustained clinical remission could not be achieved. We recently found that the patient's blood showed an impaired production of IL-6 when stimulated with zymosan. We also found that she is homozygous for a previously reported CARD9 loss-of-function mutation (Q289*). CONCLUSIONS: This is the first report of a patient with inherited CARD9 deficiency and chronic invasive pulmonary aspergillosis (IPA) due to A. nomius. Inherited CARD9 deficiency should be considered in otherwise healthy children and adults with one or more invasive fungal diseases.


Subject(s)
Aspergillus/physiology , CARD Signaling Adaptor Proteins/genetics , Candidiasis, Chronic Mucocutaneous/diagnosis , Exophiala/physiology , Mutation/genetics , Phaeohyphomycosis/diagnosis , Pulmonary Aspergillosis/diagnosis , Candidiasis, Chronic Mucocutaneous/genetics , Cells, Cultured , Female , Humans , Interleukin-6/metabolism , Middle Aged , Pedigree , Phaeohyphomycosis/genetics , Pneumonectomy , Pulmonary Aspergillosis/genetics
14.
J Clin Immunol ; 40(6): 807-819, 2020 08.
Article in English | MEDLINE | ID: mdl-32572726

ABSTRACT

Down syndrome (DS) is characterized by the occurrence of three copies of human chromosome 21 (HSA21). HSA21 contains a cluster of four interferon receptor (IFN-R) genes: IFNAR1, IFNAR2, IFNGR2, and IL10RB. DS patients often develop mucocutaneous infections and autoimmune diseases, mimicking patients with heterozygous gain-of-function (GOF) STAT1 mutations, which enhance cellular responses to three types of interferon (IFN). A gene dosage effect at these four loci may contribute to the infectious and autoimmune manifestations observed in individuals with DS. We report high levels of IFN-αR1, IFN-αR2, and IFN-γR2 expression on the surface of monocytes and EBV-transformed-B (EBV-B) cells from studying 45 DS patients. Total and phosphorylated STAT1 (STAT1 and pSTAT1) levels were constitutively high in unstimulated and IFN-α- and IFN-γ-stimulated monocytes from DS patients but lower than those in patients with GOF STAT1 mutations. Following stimulation with IFN-α or -γ, but not with IL-6 or IL-21, pSTAT1 and IFN-γ activation factor (GAF) DNA-binding activities were significantly higher in the EBV-B cells of DS patients than in controls. These responses resemble the dysregulated responses observed in patients with STAT1 GOF mutations. Concentrations of plasma type I IFNs were high in 12% of the DS patients tested (1.8% in the healthy controls). Levels of type I IFNs, IFN-Rs, and STAT1 were similar in DS patients with and without recurrent skin infections. We performed a genome-wide transcriptomic analysis based on principal component analysis and interferon modules on circulating monocytes. We found that DS monocytes had levels of both IFN-α- and IFN-γ-inducible ISGs intermediate to those of monocytes from healthy controls and from patients with GOF STAT1 mutations. Unlike patients with GOF STAT1 mutations, patients with DS had normal circulating Th17 counts and a high proportion of terminally differentiated CD8+ T cells with low levels of STAT1 expression. We conclude a mild interferonopathy in Down syndrome leads to an incomplete penetrance at both cellular and clinical level, which is not correlate with recurrent skin bacterial or fungal infections. The constitutive upregulation of type I and type II IFN-R, at least in monocytes of DS patients, may contribute to the autoimmune diseases observed in these individuals.


Subject(s)
Down Syndrome/genetics , Down Syndrome/metabolism , Gene Dosage , Interferon Type I/metabolism , Receptors, Interferon/genetics , Adolescent , Adult , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , Child , Child, Preschool , Chromosome Mapping , Cytokines/metabolism , Disease Susceptibility , Down Syndrome/immunology , Female , Gene Expression Profiling , Genetic Loci , Genetic Predisposition to Disease , Humans , Interferon Type I/genetics , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Receptors, Interferon/metabolism , STAT1 Transcription Factor/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome , Young Adult
15.
Immunol Invest ; 49(3): 299-306, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31588815

ABSTRACT

Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED) syndrome is a rare monogenic autosomal recessive disorder caused by biallelic mutations in the AIRE (autoimmune regulator) gene. Patients with APECED present with heterogeneous endocrine and non-endocrine manifestations. In this study, we report an Iranian patient who presented with Addison disease, chronic mucocutaneous candidiasis, alopecia totalis, keratopathy and asplenia treated as an isolated endocrinopathy for 25 years. In the adulthood, the diagnosis of APECED was made by genetic analysis which demonstrated homozygous nonsense p.R257* (c.769C>T) mutation of AIRE. APECED has been shown to be frequent in some ethnicities including Iranian Jews. Therefore, we reviewed 39 Iranian APECED patients published in the literature. We found that most of the Iranian patients were of Jewish ethnic background and presented hypoparathyroidism, adrenal insufficiency, and candidiasis as the main clinical manifestation.


Subject(s)
Polyendocrinopathies, Autoimmune/diagnosis , Polyendocrinopathies, Autoimmune/genetics , Transcription Factors/genetics , Adult , Autoantibodies/blood , Cytokines/immunology , Homozygote , Humans , Male , Mutation , Pedigree , Polyendocrinopathies, Autoimmune/pathology , Polyendocrinopathies, Autoimmune/therapy , AIRE Protein
16.
Proc Natl Acad Sci U S A ; 113(51): E8277-E8285, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930337

ABSTRACT

Chronic mucocutaneous candidiasis (CMC) is defined as recurrent or persistent infection of the skin, nails, and/or mucosae with commensal Candida species. The first genetic etiology of isolated CMC-autosomal recessive (AR) IL-17 receptor A (IL-17RA) deficiency-was reported in 2011, in a single patient. We report here 21 patients with complete AR IL-17RA deficiency, including this first patient. Each patient is homozygous for 1 of 12 different IL-17RA alleles, 8 of which create a premature stop codon upstream from the transmembrane domain and have been predicted and/or shown to prevent expression of the receptor on the surface of circulating leukocytes and dermal fibroblasts. Three other mutant alleles create a premature stop codon downstream from the transmembrane domain, one of which encodes a surface-expressed receptor. Finally, the only known missense allele (p.D387N) also encodes a surface-expressed receptor. All of the alleles tested abolish cellular responses to IL-17A and -17F homodimers and heterodimers in fibroblasts and to IL-17E/IL-25 in leukocytes. The patients are currently aged from 2 to 35 y and originate from 12 unrelated kindreds. All had their first CMC episode by 6 mo of age. Fourteen patients presented various forms of staphylococcal skin disease. Eight were also prone to various bacterial infections of the respiratory tract. Human IL-17RA is, thus, essential for mucocutaneous immunity to Candida and Staphylococcus, but otherwise largely redundant. A diagnosis of AR IL-17RA deficiency should be considered in children or adults with CMC, cutaneous staphylococcal disease, or both, even if IL-17RA is detected on the cell surface.


Subject(s)
Bacterial Infections/immunology , Candidiasis/immunology , Mycoses/immunology , Receptors, Interleukin-17/deficiency , Receptors, Interleukin-17/genetics , Alleles , Candida , Cell Membrane , Child , Child, Preschool , Family Health , Female , Fibroblasts/metabolism , Genes, Recessive , Genome-Wide Association Study , HEK293 Cells , Homozygote , Humans , Immunophenotyping , Infant , Infant, Newborn , Interleukin-17/metabolism , Male , Mutation , Open Reading Frames , Pedigree , Receptors, Interleukin-17/metabolism , Skin/microbiology , T-Lymphocytes/cytology
17.
J Clin Immunol ; 38(5): 617-627, 2018 07.
Article in English | MEDLINE | ID: mdl-29995221

ABSTRACT

PURPOSE: Inborn errors of IFN-γ immunity underlie Mendelian susceptibility to mycobacterial disease (MSMD). Autosomal recessive complete IL-12Rß1 deficiency is the most frequent genetic etiology of MSMD. Only two of the 84 known mutations are copy number variations (CNVs), identified in two of the 213 IL-12Rß1-deficient patients and two of the 164 kindreds reported. These two CNVs are large deletions found in the heterozygous or homozygous state. We searched for novel families with IL-12Rß1 deficiency due to CNVs. METHODS: We studied six MSMD patients from five unrelated kindreds displaying adverse reactions to BCG vaccination. Three of the patients also presented systemic salmonellosis, two had mucocutaneous candidiasis, and one had disseminated histoplasmosis. We searched for CNVs and other variations by IL12RB1-targeted next-generation sequencing (NGS). RESULTS: We identified six new IL-12Rß1-deficient patients with a complete loss of IL-12Rß1 expression on phytohemagglutinin-activated T cells and/or EBV-transformed B cells. The cells of these patients did not respond to IL-12 and IL-23. Five different CNVs encompassing IL12RB1 (four deletions and one duplication) were identified in these patients by NGS coverage analysis, either in the homozygous state (n = 1) or in trans (n = 4) with a single-nucleotide variation (n = 3) or a small indel (n = 1). Seven of the nine mutations are novel. Interestingly, four of the five CNVs were predicted to be driven by nearby Alu elements, as well as the two previously reported large deletions. The IL12RB1 locus is actually enriched in Alu elements (44.7%), when compared with the rest of the genome (10.5%). CONCLUSION: The IL12RB1 locus is Alu-enriched and therefore prone to rearrangements at various positions. CNVs should be considered in the genetic diagnosis of IL-12Rß1 deficiency.


Subject(s)
Alu Elements , DNA Copy Number Variations , Genetic Association Studies , Genetic Predisposition to Disease , Interleukin-12 Receptor beta 1 Subunit/deficiency , Alleles , Base Sequence , Chromosome Mapping , Female , Gene Expression , Humans , Interferon-gamma , Male , Mutation , Mycobacterium Infections/diagnosis , Mycobacterium Infections/etiology , Mycobacterium Infections/metabolism , Pedigree , Phenotype
18.
Pediatr Blood Cancer ; 64(6)2017 06.
Article in English | MEDLINE | ID: mdl-27873456

ABSTRACT

Mutations of the IL12B and IL12RB1 genes underlie the development of IL-12 p40 and IL-12Rß1 deficiencies, respectively, both of which cause predisposition to infection with weakly virulent mycobacteria and Salmonella. Infections with other intramacrophagic organisms have only been rarely observed. We identified two patients with visceral leishmaniasis who had autosomal recessive IL-12 p40 and IL-12Rß1 deficiencies, respectively. This finding demonstrates the importance of IFN-γ immunity in the control of leishmaniasis. We also searched the literature for similar reports in patients with these and other primary immunodeficiencies.


Subject(s)
Genetic Diseases, Inborn , Immunologic Deficiency Syndromes , Interleukin-12 Subunit p40/deficiency , Leishmaniasis, Visceral , Receptors, Interleukin-12/deficiency , Adolescent , Child , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/pathology , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Leishmaniasis, Visceral/genetics , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/pathology , Male
19.
J Allergy Clin Immunol ; 138(1): 241-248.e3, 2016 07.
Article in English | MEDLINE | ID: mdl-26936803

ABSTRACT

BACKGROUND: Chronic granulomatous disease (CGD) is a rare primary immunodeficiency caused by inborn errors of the phagocyte nicotinamide adenine dinucleotide phosphate oxidase complex. From the first year of life onward, most affected patients display multiple, severe, and recurrent infections caused by bacteria and fungi. Mycobacterial infections have also been reported in some patients. OBJECTIVE: Our objective was to assess the effect of mycobacterial disease in patients with CGD. METHODS: We analyzed retrospectively the clinical features of mycobacterial disease in 71 patients with CGD. Tuberculosis and BCG disease were diagnosed on the basis of microbiological, pathological, and/or clinical criteria. RESULTS: Thirty-one (44%) patients had tuberculosis, and 53 (75%) presented with adverse effects of BCG vaccination; 13 (18%) had both tuberculosis and BCG infections. None of these patients displayed clinical disease caused by environmental mycobacteria, Mycobacterium leprae, or Mycobacterium ulcerans. Most patients (76%) also had other pyogenic and fungal infections, but 24% presented solely with mycobacterial disease. Most patients presented a single localized episode of mycobacterial disease (37%), but recurrence (18%), disseminated disease (27%), and even death (18%) were also observed. One common feature in these patients was an early age at presentation for BCG disease. Mycobacterial disease was the first clinical manifestation of CGD in 60% of these patients. CONCLUSION: Mycobacterial disease is relatively common in patients with CGD living in countries in which tuberculosis is endemic, BCG vaccine is mandatory, or both. Adverse reactions to BCG and severe forms of tuberculosis should lead to a suspicion of CGD. BCG vaccine is contraindicated in patients with CGD.


Subject(s)
Granulomatous Disease, Chronic/complications , Mycobacterium Infections/diagnosis , Mycobacterium Infections/etiology , BCG Vaccine/administration & dosage , Bacterial Infections/diagnosis , Bacterial Infections/epidemiology , Bacterial Infections/etiology , Bacterial Infections/mortality , Child , Child, Preschool , Female , Granulomatous Disease, Chronic/epidemiology , Granulomatous Disease, Chronic/mortality , Granulomatous Disease, Chronic/therapy , Humans , Infant , Male , Mycobacterium Infections/epidemiology , Mycobacterium Infections/mortality , Mycoses/diagnosis , Mycoses/epidemiology , Mycoses/etiology , Mycoses/mortality , Patient Outcome Assessment , Retrospective Studies , Tuberculosis/diagnosis , Tuberculosis/etiology
20.
Am J Hum Genet ; 92(3): 407-14, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23415668

ABSTRACT

Only a small fraction of individuals infected with Mycobacterium tuberculosis develop clinical tuberculosis (TB) in their lifetime. Genetic epidemiological evidence suggests a genetic determinism of pulmonary TB (PTB), but the molecular basis of genetic predisposition to PTB remains largely unknown. We used a positional-cloning approach to carry out ultrafine linkage-disequilibrium mapping of a previously identified susceptibility locus in chromosomal region 8q12-13 by genotyping 3,216 SNPs in a family-based Moroccan sample including 286 offspring with PTB. We observed 44 PTB-associated SNPs (p < 0.01), which were genotyped in an independent set of 317 cases and 650 controls from Morocco. A single signal, consisting of two correlated SNPs close to TOX, rs1568952 and rs2726600 (combined p = 1.1 × 10(-5) and 9.2 × 10(-5), respectively), was replicated. Stronger evidence of association was found in individuals who developed PTB before the age of 25 years (combined p for rs1568952 = 4.4 × 10(-8); odds ratio of PTB for AA versus AG/GG = 3.09 [1.99-4.78]). The association with rs2726600 (p = 0.04) was subsequently replicated in PTB-affected subjects under 25 years in a study of 243 nuclear families from Madagascar. Stronger evidence of replication in Madagascar was obtained for additional SNPs in strong linkage disequilibrium with the two initial SNPs (p = 0.003 for rs2726597), further confirming the signal. We thus identified around rs1568952 and rs2726600 a cluster of SNPs strongly associated with early-onset PTB in Morocco and Madagascar. SNP rs2726600 is located in a transcription-factor binding site in the 3' region of TOX, and further functional explorations will focus on CD4 T lymphocytes.


Subject(s)
Chromosomes, Human, Pair 8 , Genetic Linkage , High Mobility Group Proteins/genetics , Tuberculosis, Pulmonary/genetics , Adult , Age Factors , Alleles , Case-Control Studies , Female , Genetic Loci , Genetic Predisposition to Disease , Genotype , Humans , Linkage Disequilibrium , Madagascar , Male , Morocco , Mycobacterium tuberculosis/isolation & purification , Polymorphism, Single Nucleotide , Tuberculosis, Pulmonary/microbiology , White People
SELECTION OF CITATIONS
SEARCH DETAIL