Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Publication year range
1.
Cell ; 183(7): 1826-1847.e31, 2020 12 23.
Article in English | MEDLINE | ID: mdl-33296702

ABSTRACT

Inborn errors of human interferon gamma (IFN-γ) immunity underlie mycobacterial disease. We report a patient with mycobacterial disease due to inherited deficiency of the transcription factor T-bet. The patient has extremely low counts of circulating Mycobacterium-reactive natural killer (NK), invariant NKT (iNKT), mucosal-associated invariant T (MAIT), and Vδ2+ γδ T lymphocytes, and of Mycobacterium-non reactive classic TH1 lymphocytes, with the residual populations of these cells also producing abnormally small amounts of IFN-γ. Other lymphocyte subsets develop normally but produce low levels of IFN-γ, with the exception of CD8+ αß T and non-classic CD4+ αß TH1∗ lymphocytes, which produce IFN-γ normally in response to mycobacterial antigens. Human T-bet deficiency thus underlies mycobacterial disease by preventing the development of innate (NK) and innate-like adaptive lymphocytes (iNKT, MAIT, and Vδ2+ γδ T cells) and IFN-γ production by them, with mycobacterium-specific, IFN-γ-producing, purely adaptive CD8+ αß T, and CD4+ αß TH1∗ cells unable to compensate for this deficit.


Subject(s)
Adaptive Immunity , Immunity, Innate , Interferon-gamma/immunology , Mycobacterium/immunology , T-Box Domain Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cell Lineage , Child, Preschool , Chromatin/metabolism , CpG Islands/genetics , DNA Methylation/genetics , Dendritic Cells/metabolism , Epigenesis, Genetic , Female , Homozygote , Humans , INDEL Mutation/genetics , Infant , Interferon-gamma/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Loss of Function Mutation/genetics , Male , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Mycobacterium Infections/microbiology , Pedigree , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Lymphocytes, Helper-Inducer/immunology , Transcriptome/genetics
2.
Nat Immunol ; 19(9): 973-985, 2018 09.
Article in English | MEDLINE | ID: mdl-30127434

ABSTRACT

Human inborn errors of IFN-γ immunity underlie mycobacterial diseases. We describe patients with Mycobacterium bovis (BCG) disease who are homozygous for loss-of-function mutations of SPPL2A. This gene encodes a transmembrane protease that degrades the N-terminal fragment (NTF) of CD74 (HLA invariant chain) in antigen-presenting cells. The CD74 NTF therefore accumulates in the HLA class II+ myeloid and lymphoid cells of SPPL2a-deficient patients. This toxic fragment selectively depletes IL-12- and IL-23-producing CD1c+ conventional dendritic cells (cDC2s) and their circulating progenitors. Moreover, SPPL2a-deficient memory TH1* cells selectively fail to produce IFN-γ when stimulated with mycobacterial antigens in vitro. Finally, Sppl2a-/- mice lack cDC2s, have CD4+ T cells that produce small amounts of IFN-γ after BCG infection, and are highly susceptible to infection with BCG or Mycobacterium tuberculosis. These findings suggest that inherited SPPL2a deficiency in humans underlies mycobacterial disease by decreasing the numbers of cDC2s and impairing IFN-γ production by mycobacterium-specific memory TH1* cells.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Dendritic Cells/immunology , Membrane Proteins/metabolism , Mycobacterium Infections/immunology , Mycobacterium bovis/physiology , Mycobacterium tuberculosis/physiology , Th1 Cells/immunology , Tuberculosis/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Cells, Cultured , HLA Antigens/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immunity , Immunologic Memory , Infant , Interferon-gamma/metabolism , Lymphadenopathy , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Mycobacterium Infections/genetics , Vaccination
3.
Rev Med Liege ; 79(3): 175-180, 2024 Mar.
Article in French | MEDLINE | ID: mdl-38487912

ABSTRACT

It is estimated that in highly medicalised countries, median life expectancy for most newborns with cystic fibrosis now exceeds 70 years, approaching that of the general population. However, socio-economic disparities between countries continue to have a devastating impact on the prognosis of patients in Eastern Europe, Africa, India and South America. In Morocco, very limited genetic data suggest that the prevalence of this disease is at least of the same order as in Belgium. But as it is not really recognised by the national health system, patients are denied access even to symptomatic treatment. As a result, their outcome is tragic, similar to what it was 60 years ago in the most medicalised countries. A pilot project for a first paediatric reference centre in Casablanca is currently being set up. If properly resourced, this project can only be a success and should be the first step on the road towards cystic fibrosis care in this country. In a very humble way, several Belgian stakeholders are trying to support this project.


Dans les pays les plus médicalisés, l'espérance de vie médiane de la plupart des nouveau-nés atteints de mucoviscidose excède aujourd'hui 70 ans et se rapproche de celle de la population générale. Ailleurs, en Europe de l'Est comme en Afrique, en Inde ou en Amérique du Sud, les disparités socio-économiques des pays continuent à impacter très durement le pronostic des patients. Au Maroc, des données génétiques très fragmentaires suggèrent que la prévalence de la mucoviscidose est au moins du même ordre qu'en Belgique. Mais la maladie n'y est pas réellement reconnue par le système de santé, de telle sorte que même le traitement symptomatique reste inaccessible aux patients et leur pronostic est tragique, similaire à ce qu'il était il y a 60 ans dans les pays les plus médicalisés. À Casablanca, le projet pilote d'un premier Centre pédiatrique de Référence est en train de se mettre en place. S'il bénéficie d'un support adéquat, ce projet ne peut être qu'un succès et doit constituer un tout premier pas sur le chemin vers une prise en charge des patients dans ce pays. Très modestement, plusieurs intervenants belges tentent d'y apporter leur soutien.


Subject(s)
Cystic Fibrosis , Child , Humans , Infant, Newborn , Cystic Fibrosis/epidemiology , Cystic Fibrosis/therapy , Pilot Projects , Belgium/epidemiology
4.
J Clin Immunol ; 42(7): 1508-1520, 2022 10.
Article in English | MEDLINE | ID: mdl-36198931

ABSTRACT

The International Union of Immunological Societies (IUIS) expert committee (EC) on Inborn Errors of Immunity (IEI) reports here the 2022 updated phenotypic classification, which accompanies and complements the most-recent genotypic classification. This phenotypic classification is aimed for clinicians at the bedside and focuses on clinical features and laboratory phenotypes of specific IEI. In this classification, 485 IEI underlying phenotypes as diverse as infection, malignancy, allergy, auto-immunity and auto-inflammation are described, including 55 novel monogenic defects and 1 autoimmune phenocopy. Therefore, all 485 diseases of the genetic classification are presented in this paper in the form of colored tables with essential clinical or immunological phenotype entries.


Subject(s)
Hypersensitivity , Immunologic Deficiency Syndromes , Neoplasms , Humans , Immunologic Deficiency Syndromes/genetics , Phenotype , Genotype
5.
J Clin Immunol ; 42(7): 1473-1507, 2022 10.
Article in English | MEDLINE | ID: mdl-35748970

ABSTRACT

We report the updated classification of inborn errors of immunity, compiled by the International Union of Immunological Societies Expert Committee. This report documents the key clinical and laboratory features of 55 novel monogenic gene defects, and 1 phenocopy due to autoantibodies, that have either been discovered since the previous update (published January 2020) or were characterized earlier but have since been confirmed or expanded in subsequent studies. While variants in additional genes associated with immune diseases have been reported in the literature, this update includes only those that the committee assessed that reached the necessary threshold to represent novel inborn errors of immunity. There are now a total of 485 inborn errors of immunity. These advances in discovering the genetic causes of human immune diseases continue to significantly further our understanding of molecular, cellular, and immunological mechanisms of disease pathogenesis, thereby simultaneously enhancing immunological knowledge and improving patient diagnosis and management. This report is designed to serve as a resource for immunologists and geneticists pursuing the molecular diagnosis of individuals with heritable immunological disorders and for the scientific dissection of cellular and molecular mechanisms underlying monogenic and related human immune diseases.


Subject(s)
Immune System Diseases , Immunologic Deficiency Syndromes , Humans , Immunologic Deficiency Syndromes/diagnosis , Phenotype , Research Report
6.
J Clin Immunol ; 41(3): 666-679, 2021 04.
Article in English | MEDLINE | ID: mdl-33598806

ABSTRACT

The most recent updated classification of inborn errors of immunity/primary immunodeficiencies, compiled by the International Union of Immunological Societies Expert Committee, was published in January 2020. Within days of completing this report, it was already out of date, evidenced by the frequent publication of genetic variants proposed to cause novel inborn errors of immunity. As the next formal report from the IUIS Expert Committee will not be published until 2022, we felt it important to provide the community with a brief update of recent contributions to the field of inborn errors of immunity. Herein, we highlight studies that have identified 26 additional monogenic gene defects that reach the threshold to represent novel causes of immune defects.


Subject(s)
Genetic Predisposition to Disease , Primary Immunodeficiency Diseases/epidemiology , Primary Immunodeficiency Diseases/genetics , Alleles , COVID-19/complications , COVID-19/epidemiology , COVID-19/virology , Diagnosis, Differential , Disease Management , Genetic Association Studies/methods , Genotype , Humans , Immunity/genetics , Inheritance Patterns , Phenotype , Primary Immunodeficiency Diseases/diagnosis , Public Health Surveillance , Risk Factors
7.
J Clin Immunol ; 41(6): 1339-1351, 2021 08.
Article in English | MEDLINE | ID: mdl-34052995

ABSTRACT

BACKGROUND: Inborn errors of immunity (IEIs) are a heterogeneous group of genetic defects of immunity, which cause high rates of morbidity and mortality mainly among children due to infectious and non-infectious complications. The IEI burden has been critically underestimated in countries from middle- and low-income regions and the majority of patients with IEI in these regions lack a molecular diagnosis. METHODS: We analyzed the clinical, immunologic, and genetic data of IEI patients from 22 countries in the Middle East and North Africa (MENA) region. The data was collected from national registries and diverse databases such as the Asian Pacific Society for Immunodeficiencies (APSID) registry, African Society for Immunodeficiencies (ASID) registry, Jeffrey Modell Foundation (JMF) registry, J Project centers, and International Consortium on Immune Deficiency (ICID) centers. RESULTS: We identified 17,120 patients with IEI, among which females represented 39.4%. Parental consanguinity was present in 60.5% of cases and 27.3% of the patients were from families with a confirmed previous family history of IEI. The median age of patients at the onset of disease was 36 months and the median delay in diagnosis was 41 months. The rate of registered IEI patients ranges between 0.02 and 7.58 per 100,000 population, and the lowest rates were in countries with the highest rates of disability-adjusted life years (DALY) and death rates for children. Predominantly antibody deficiencies were the most frequent IEI entities diagnosed in 41.2% of the cohort. Among 5871 patients genetically evaluated, the diagnostic yield was 83% with the majority (65.2%) having autosomal recessive defects. The mortality rate was the highest in patients with non-syndromic combined immunodeficiency (51.7%, median age: 3.5 years) and particularly in patients with mutations in specific genes associated with this phenotype (RFXANK, RAG1, and IL2RG). CONCLUSIONS: This comprehensive registry highlights the importance of a detailed investigation of IEI patients in the MENA region. The high yield of genetic diagnosis of IEI in this region has important implications for prevention, prognosis, treatment, and resource allocation.


Subject(s)
Genetic Diseases, Inborn/epidemiology , Primary Immunodeficiency Diseases/epidemiology , Adolescent , Adult , Africa, Northern/epidemiology , Aged , Child , Consensus , Disability-Adjusted Life Years , Female , Humans , Male , Middle Aged , Middle East/epidemiology , Registries , Young Adult
8.
J Clin Immunol ; 40(1): 96-104, 2020 01.
Article in English | MEDLINE | ID: mdl-31696364

ABSTRACT

Autosomal recessive agammaglobulinemia (ARA) is a primary immunodeficiency characterized by absent peripheral B cells, severe hypogammaglobulinemia, and absent BTK gene mutations. In ARA, mutations occur in genes encoding the pre-B cell receptor (pre-BCR) or downstream signaling proteins. In this work, we used candidate gene and whole-exome sequencing to investigate the molecular basis of ARA in 6 patients from 4 consanguineous North-African families. Sanger sequencing of candidate genes encoding the pre-BCR components (ΙGΗΜ, CD79A, CD79B, IGLL1, and VPREB1) was initially performed and determined the genetic defect in five patients. Two novel mutations in IGHM (p.Val378Alafs*1 and p.Ile184Serfs*21) were identified in three patients from two unrelated kindred and a novel nonsense mutation was identified in CD79A (p.Trp66*) in two siblings from a third kindred. Whole-exome sequencing (WES) was performed on the sixth patient who harbored a homozygous stop mutation at position 407 in the RAG2 gene (p.Glu407*). We concluded that conventional gene sequencing, especially when multiple genes are involved in the defect as is the case in ARA, is costly and time-consuming, resulting in delayed diagnosis that contributes to increased morbidity and mortality. In addition, it fails to identify the involvement of novel and unsuspected gene defects when the phenotype of the patients is atypical. WES has the potential to provide a rapid and more accurate genetic diagnosis in ARA, which is crucial for the treatment of the patients.


Subject(s)
Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Child, Preschool , Codon, Nonsense/genetics , Consanguinity , Exome/genetics , Female , Homozygote , Humans , Infant , Infant, Newborn , Male , North America , Pedigree , Phenotype , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA/methods , Exome Sequencing/methods
9.
J Clin Immunol ; 40(1): 24-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31953710

ABSTRACT

We report the updated classification of Inborn Errors of Immunity/Primary Immunodeficiencies, compiled by the International Union of Immunological Societies Expert Committee. This report documents the key clinical and laboratory features of 430 inborn errors of immunity, including 64 gene defects that have either been discovered in the past 2 years since the previous update (published January 2018) or were characterized earlier but have since been confirmed or expanded upon in subsequent studies. The application of next-generation sequencing continues to expedite the rapid identification of novel gene defects, rare or common; broaden the immunological and clinical phenotypes of conditions arising from known gene defects and even known variants; and implement gene-specific therapies. These advances are contributing to greater understanding of the molecular, cellular, and immunological mechanisms of disease, thereby enhancing immunological knowledge while improving the management of patients and their families. This report serves as a valuable resource for the molecular diagnosis of individuals with heritable immunological disorders and also for the scientific dissection of cellular and molecular mechanisms underlying inborn errors of immunity and related human diseases.

10.
J Clin Immunol ; 40(1): 66-81, 2020 01.
Article in English | MEDLINE | ID: mdl-32048120

ABSTRACT

Since 2013, the International Union of Immunological Societies (IUIS) expert committee (EC) on Inborn Errors of Immunity (IEI) has published an updated phenotypic classification of IEI, which accompanies and complements their genotypic classification into ten tables. This phenotypic classification is user-friendly and serves as a resource for clinicians at the bedside. There are now 430 single-gene IEI underlying phenotypes as diverse as infection, malignancy, allergy, autoimmunity, and autoinflammation. We herein report the 2019 phenotypic classification, including the 65 new conditions. The diagnostic algorithms are based on clinical and laboratory phenotypes for each of the ten broad categories of IEI.


Subject(s)
Immunity/genetics , Immunologic Deficiency Syndromes/genetics , Autoimmunity/genetics , Genotype , Hereditary Autoinflammatory Diseases/genetics , Humans , Hypersensitivity , Phenotype
11.
12.
Blood ; 132(22): 2362-2374, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30254128

ABSTRACT

ARPC1B is a key factor for the assembly and maintenance of the ARP2/3 complex that is involved in actin branching from an existing filament. Germline biallelic mutations in ARPC1B have been recently described in 6 patients with clinical features of combined immunodeficiency (CID), whose neutrophils and platelets but not T lymphocytes were studied. We hypothesized that ARPC1B deficiency may also lead to cytoskeleton and functional defects in T cells. We have identified biallelic mutations in ARPC1B in 6 unrelated patients with early onset disease characterized by severe infections, autoimmune manifestations, and thrombocytopenia. Immunological features included T-cell lymphopenia, low numbers of naïve T cells, and hyper-immunoglobulin E. Alteration in ARPC1B protein structure led to absent/low expression by flow cytometry and confocal microscopy. This molecular defect was associated with the inability of patient-derived T cells to extend an actin-rich lamellipodia upon T-cell receptor (TCR) stimulation and to assemble an immunological synapse. ARPC1B-deficient T cells additionally displayed impaired TCR-mediated proliferation and SDF1-α-directed migration. Gene transfer of ARPC1B in patients' T cells using a lentiviral vector restored both ARPC1B expression and T-cell proliferation in vitro. In 2 of the patients, in vivo somatic reversion restored ARPC1B expression in a fraction of lymphocytes and was associated with a skewed TCR repertoire. In 1 revertant patient, memory CD8+ T cells expressing normal levels of ARPC1B displayed improved T-cell migration. Inherited ARPC1B deficiency therefore alters T-cell cytoskeletal dynamics and functions, contributing to the clinical features of CID.


Subject(s)
Actin-Related Protein 2-3 Complex/genetics , Germ-Line Mutation , Immunologic Deficiency Syndromes/genetics , T-Lymphocytes/pathology , Actin-Related Protein 2-3 Complex/chemistry , Female , Homozygote , Humans , Immunologic Deficiency Syndromes/pathology , Male , Models, Molecular , Pedigree , Protein Conformation , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/metabolism
13.
J Allergy Clin Immunol ; 143(6): 2215-2226.e7, 2019 06.
Article in English | MEDLINE | ID: mdl-30578871

ABSTRACT

BACKGROUND: Inflammatory activation of CD8+ T cells can, when left unchecked, drive severe immunopathology. Hyperstimulation of CD8+ T cells through a broad set of triggering signals can precipitate hemophagocytic lymphohistiocytosis (HLH), a life-threatening systemic inflammatory disorder. OBJECTIVE: The mechanism linking CD8+ T-cell hyperactivation to pathology is controversial, with excessive production of IFN-γ and, more recently, excessive consumption of IL-2, which are proposed as competing hypotheses. We formally tested the proximal mechanistic events of each pathway in a mouse model of HLH. METHODS: In addition to reporting a complete autosomal recessive IFN-γ receptor 1-deficient patient with multiple aspects of HLH pathology, we used the mouse model of perforin (Prf1)KO mice infected with lymphocytic choriomeningitis virus to genetically eliminate either IFN-γ production or CD25 expression and assess the immunologic, hematologic, and physiologic disease measurement. RESULTS: We found a striking dichotomy between the mechanistic basis of the hematologic and inflammatory components of CD8+ T cell-mediated pathology. The hematologic features of HLH were completely dependent on IFN-γ production, with complete correction after loss of IFN-γ production without any role for CD8+ T cell-mediated IL-2 consumption. By contrast, the mechanistic contribution of the immunologic features was reversed, with no role for IFN-γ production but substantial correction after reduction of IL-2 consumption by hyperactivated CD8+ T cells. These results were complemented by the characterization of an IFN-γ receptor 1-deficient patients with HLH-like disease, in whom multiple aspects of HLH pathology were observed in the absence of IFN-γ signaling. CONCLUSION: These results synthesize the competing mechanistic models of HLH pathology into a dichotomous pathogenesis driven through discrete pathways. A holistic model provides a new paradigm for understanding HLH and, more broadly, the consequences of CD8+ T-cell hyperactivation, thereby paving the way for clinical intervention based on the features of HLH in individual patients.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Interferon-gamma/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/physiology , Lymphohistiocytosis, Hemophagocytic/immunology , Receptors, Interferon/deficiency , Animals , Consanguinity , Disease Models, Animal , Female , Humans , Infant , Lymphocyte Activation , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/genetics , Mice , Mice, Knockout , Models, Immunological , Morocco , Perforin/genetics , Signal Transduction , Interferon gamma Receptor
14.
Proc Natl Acad Sci U S A ; 113(24): 6713-8, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27247391

ABSTRACT

Principal component analysis (PCA), homozygosity rate estimations, and linkage studies in humans are classically conducted through genome-wide single-nucleotide variant arrays (GWSA). We compared whole-exome sequencing (WES) and GWSA for this purpose. We analyzed 110 subjects originating from different regions of the world, including North Africa and the Middle East, which are poorly covered by public databases and have high consanguinity rates. We tested and applied a number of quality control (QC) filters. Compared with GWSA, we found that WES provided an accurate prediction of population substructure using variants with a minor allele frequency > 2% (correlation = 0.89 with the PCA coordinates obtained by GWSA). WES also yielded highly reliable estimates of homozygosity rates using runs of homozygosity with a 1,000-kb window (correlation = 0.94 with the estimates provided by GWSA). Finally, homozygosity mapping analyses in 15 families including a single offspring with high homozygosity rates showed that WES provided 51% less genome-wide linkage information than GWSA overall but 97% more information for the coding regions. At the genome-wide scale, 76.3% of linked regions were found by both GWSA and WES, 17.7% were found by GWSA only, and 6.0% were found by WES only. For coding regions, the corresponding percentages were 83.5%, 7.4%, and 9.1%, respectively. With appropriate QC filters, WES can be used for PCA and adjustment for population substructure, estimating homozygosity rates in individuals, and powerful linkage analyses, particularly in coding regions.


Subject(s)
Consanguinity , Genetic Linkage , Genome-Wide Association Study , Homozygote , Female , Humans , Male , Middle East , North America
15.
Immunol Rev ; 264(1): 103-20, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25703555

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis (M.tb) and a few related mycobacteria, is a devastating disease, killing more than a million individuals per year worldwide. However, its pathogenesis remains largely elusive, as only a small proportion of infected individuals develop clinical disease either during primary infection or during reactivation from latency or secondary infection. Subacute, hematogenous, and extrapulmonary disease tends to be more frequent in infants, children, and teenagers than in adults. Life-threatening primary TB of childhood can result from known acquired or inherited immunodeficiencies, although the vast majority of cases remain unexplained. We review here the conditions conferring a predisposition to childhood clinical diseases caused by mycobacteria, including not only M.tb but also weakly virulent mycobacteria, such as BCG vaccines and environmental mycobacteria. Infections with weakly virulent mycobacteria are much rarer than TB, but the inherited and acquired immunodeficiencies underlying these infections are much better known. Their study has also provided genetic and immunological insights into childhood TB, as illustrated by the discovery of single-gene inborn errors of IFN-γ immunity underlying severe cases of TB. Novel findings are expected from ongoing and future human genetic studies of childhood TB in countries that combine a high proportion of consanguineous marriages, a high incidence of TB, and an excellent clinical care, such as Iran, Morocco, and Turkey.


Subject(s)
Disease Susceptibility/immunology , Genetic Predisposition to Disease , Immunocompromised Host , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/etiology , Mycobacterium tuberculosis/immunology , Tuberculosis/etiology , Age Factors , Child , Genes, Dominant , Genes, Recessive , Humans , Immunologic Deficiency Syndromes/diagnosis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
16.
J Clin Immunol ; 38(1): 129-143, 2018 01.
Article in English | MEDLINE | ID: mdl-29226301

ABSTRACT

Since the 1990s, the International Union of Immunological Societies (IUIS) PID expert committee (EC), now called Inborn Errors of Immunity Committee, has published every other year a classification of the inborn errors of immunity. This complete catalog serves as a reference for immunologists and researchers worldwide. However, it was unadapted for clinicians at the bedside. For those, the IUIS PID EC is now publishing a phenotypical classification since 2013, which proved to be more user-friendly. There are now 320 single-gene inborn errors of immunity underlying phenotypes as diverse as infection, malignancy, allergy, auto-immunity, and auto-inflammation. We herein propose the revised 2017 phenotypic classification, based on the accompanying 2017 IUIS Inborn Errors of Immunity Committee classification.


Subject(s)
Allergy and Immunology , Immunity/genetics , Immunologic Deficiency Syndromes/immunology , Humans , Immunologic Deficiency Syndromes/genetics , International Cooperation , Phenotype
17.
J Clin Immunol ; 38(1): 96-128, 2018 01.
Article in English | MEDLINE | ID: mdl-29226302

ABSTRACT

Beginning in 1970, a committee was constituted under the auspices of the World Health Organization (WHO) to catalog primary immunodeficiencies. Twenty years later, the International Union of Immunological Societies (IUIS) took the remit of this committee. The current report details the categorization and listing of 354 (as of February 2017) inborn errors of immunity. The growth and increasing complexity of the field have been impressive, encompassing an increasing variety of conditions, and the classification described here will serve as a critical reference for immunologists and researchers worldwide.


Subject(s)
Allergy and Immunology , Immunity/genetics , Immunologic Deficiency Syndromes , Humans , Research Report , Societies, Scientific , World Health Organization
18.
Blood ; 127(25): 3154-64, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27114460

ABSTRACT

Since their discovery in patients with autosomal dominant (AD) chronic mucocutaneous candidiasis (CMC) in 2011, heterozygous STAT1 gain-of-function (GOF) mutations have increasingly been identified worldwide. The clinical spectrum associated with them needed to be delineated. We enrolled 274 patients from 167 kindreds originating from 40 countries from 5 continents. Demographic data, clinical features, immunological parameters, treatment, and outcome were recorded. The median age of the 274 patients was 22 years (range, 1-71 years); 98% of them had CMC, with a median age at onset of 1 year (range, 0-24 years). Patients often displayed bacterial (74%) infections, mostly because of Staphylococcus aureus (36%), including the respiratory tract and the skin in 47% and 28% of patients, respectively, and viral (38%) infections, mostly because of Herpesviridae (83%) and affecting the skin in 32% of patients. Invasive fungal infections (10%), mostly caused by Candida spp. (29%), and mycobacterial disease (6%) caused by Mycobacterium tuberculosis, environmental mycobacteria, or Bacille Calmette-Guérin vaccines were less common. Many patients had autoimmune manifestations (37%), including hypothyroidism (22%), type 1 diabetes (4%), blood cytopenia (4%), and systemic lupus erythematosus (2%). Invasive infections (25%), cerebral aneurysms (6%), and cancers (6%) were the strongest predictors of poor outcome. CMC persisted in 39% of the 202 patients receiving prolonged antifungal treatment. Circulating interleukin-17A-producing T-cell count was low for most (82%) but not all of the patients tested. STAT1 GOF mutations underlie AD CMC, as well as an unexpectedly wide range of other clinical features, including not only a variety of infectious and autoimmune diseases, but also cerebral aneurysms and carcinomas that confer a poor prognosis.


Subject(s)
Candidiasis, Chronic Mucocutaneous/genetics , Genetic Association Studies , Mutation , STAT1 Transcription Factor/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Female , Genetic Predisposition to Disease , Heterozygote , Humans , Infant , Male , Middle Aged , Phenotype , Young Adult
19.
J Allergy Clin Immunol ; 140(1): 232-241, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28011069

ABSTRACT

BACKGROUND: Germline heterozygous mutations in human signal transducer and activator of transcription 1 (STAT1) can cause loss of function (LOF), as in patients with Mendelian susceptibility to mycobacterial diseases, or gain of function (GOF), as in patients with chronic mucocutaneous candidiasis. LOF and GOF mutations are equally rare and can affect the same domains of STAT1, especially the coiled-coil domain (CCD) and DNA-binding domain (DBD). Moreover, 6% of patients with chronic mucocutaneous candidiasis with a GOF STAT1 mutation have mycobacterial disease, obscuring the functional significance of the identified STAT1 mutations. Current computational approaches, such as combined annotation-dependent depletion, do not distinguish LOF and GOF variants. OBJECTIVE: We estimated variations in the CCD/DBD of STAT1. METHODS: We mutagenized 342 individual wild-type amino acids in the CCD/DBD (45.6% of full-length STAT1) to alanine and tested the mutants for STAT1 transcriptional activity. RESULTS: Of these 342 mutants, 201 were neutral, 30 were LOF, and 111 were GOF mutations in a luciferase assay. This assay system correctly estimated all previously reported LOF mutations (100%) and slightly fewer GOF mutations (78.1%) in the CCD/DBD of STAT1. We found that GOF alanine mutants occurred at the interface of the antiparallel STAT1 dimer, suggesting that they destabilize this dimer. This assay also precisely predicted the effect of 2 hypomorphic and dominant negative mutations, E157K and G250E, in the CCD of STAT1 that we found in 2 unrelated patients with Mendelian susceptibility to mycobacterial diseases. CONCLUSION: The systematic alanine-scanning assay is a useful tool to estimate the GOF or LOF status and the effect of heterozygous missense mutations in STAT1 identified in patients with severe infectious diseases, including mycobacterial and fungal diseases.


Subject(s)
Alanine/genetics , Mycobacterium Infections/genetics , STAT1 Transcription Factor/genetics , Biological Assay , Female , Genetic Predisposition to Disease , Humans , Male , Mutagenesis , Mutation , Protein Domains
20.
J Allergy Clin Immunol ; 136(4): 993-1006.e1, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26162572

ABSTRACT

BACKGROUND: Follicular helper T (TFH) cells underpin T cell-dependent humoral immunity and the success of most vaccines. TFH cells also contribute to human immune disorders, such as autoimmunity, immunodeficiency, and malignancy. Understanding the molecular requirements for the generation and function of TFH cells will provide strategies for targeting these cells to modulate their behavior in the setting of these immunologic abnormalities. OBJECTIVE: We sought to determine the signaling pathways and cellular interactions required for the development and function of TFH cells in human subjects. METHODS: Human primary immunodeficiencies (PIDs) resulting from monogenic mutations provide a unique opportunity to assess the requirement for particular molecules in regulating human lymphocyte function. Circulating follicular helper T (cTFH) cell subsets, memory B cells, and serum immunoglobulin levels were quantified and functionally assessed in healthy control subjects, as well as in patients with PIDs resulting from mutations in STAT3, STAT1, TYK2, IL21, IL21R, IL10R, IFNGR1/2, IL12RB1, CD40LG, NEMO, ICOS, or BTK. RESULTS: Loss-of-function (LOF) mutations in STAT3, IL10R, CD40LG, NEMO, ICOS, or BTK reduced cTFH cell frequencies. STAT3 and IL21/R LOF and STAT1 gain-of-function mutations skewed cTFH cell differentiation toward a phenotype characterized by overexpression of IFN-γ and programmed death 1. IFN-γ inhibited cTFH cell function in vitro and in vivo, as corroborated by hypergammaglobulinemia in patients with IFNGR1/2, STAT1, and IL12RB1 LOF mutations. CONCLUSION: Specific mutations affect the quantity and quality of cTFH cells, highlighting the need to assess TFH cells in patients by using multiple criteria, including phenotype and function. Furthermore, IFN-γ functions in vivo to restrain TFH cell-induced B-cell differentiation. These findings shed new light on TFH cell biology and the integrated signaling pathways required for their generation, maintenance, and effector function and explain the compromised humoral immunity seen in patients with some PIDs.


Subject(s)
Immunologic Deficiency Syndromes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Agammaglobulinaemia Tyrosine Kinase , B-Lymphocytes/immunology , CD40 Ligand/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Humans , I-kappa B Kinase/genetics , Immunity, Humoral/genetics , Immunologic Deficiency Syndromes/genetics , Immunologic Memory , Inducible T-Cell Co-Stimulator Protein/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lymphocyte Activation , Mutation/genetics , Protein-Tyrosine Kinases/genetics , Receptors, Cytokine/genetics , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL