126 results on '"Casanova Jean-Laurent"'
Search Results
2. Biallelic human SHARPIN loss of function induces autoinflammation and immunodeficiency.
- Author
-
Oda H, Manthiram K, Chavan PP, Rieser E, Veli Ö, Kaya Ö, Rauch C, Nakabo S, Kuehn HS, Swart M, Wang Y, Çelik NI, Molitor A, Ziaee V, Movahedi N, Shahrooei M, Parvaneh N, Alipour-Olyei N, Carapito R, Xu Q, Preite S, Beck DB, Chae JJ, Nehrebecky M, Ombrello AK, Hoffmann P, Romeo T, Deuitch NT, Matthíasardóttir B, Mullikin J, Komarow H, Stoddard J, Niemela J, Dobbs K, Sweeney CL, Anderton H, Lawlor KE, Yoshitomi H, Yang D, Boehm M, Davis J, Mudd P, Randazzo D, Tsai WL, Gadina M, Kaplan MJ, Toguchida J, Mayer CT, Rosenzweig SD, Notarangelo LD, Iwai K, Silke J, Schwartzberg PL, Boisson B, Casanova JL, Bahram S, Rao AP, Peltzer N, Walczak H, Lalaoui N, Aksentijevich I, and Kastner DL
- Subjects
- Humans, Female, Male, NF-kappa B metabolism, Ubiquitin-Protein Ligases genetics, Inflammation immunology, Inflammation genetics, B-Lymphocytes immunology, Loss of Function Mutation, Fibroblasts metabolism, Fibroblasts immunology, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins metabolism, Animals, Mice, Alleles, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Nerve Tissue Proteins, Ubiquitins
- Abstract
The linear ubiquitin assembly complex (LUBAC) consists of HOIP, HOIL-1 and SHARPIN and is essential for proper immune responses. Individuals with HOIP and HOIL-1 deficiencies present with severe immunodeficiency, autoinflammation and glycogen storage disease. In mice, the loss of Sharpin leads to severe dermatitis due to excessive keratinocyte cell death. Here, we report two individuals with SHARPIN deficiency who manifest autoinflammatory symptoms but unexpectedly no dermatological problems. Fibroblasts and B cells from these individuals showed attenuated canonical NF-κB responses and a propensity for cell death mediated by TNF superfamily members. Both SHARPIN-deficient and HOIP-deficient individuals showed a substantial reduction of secondary lymphoid germinal center B cell development. Treatment of one SHARPIN-deficient individual with anti-TNF therapies led to complete clinical and transcriptomic resolution of autoinflammation. These findings underscore the critical function of the LUBAC as a gatekeeper for cell death-mediated immune dysregulation in humans., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
3. Hyperimmunoglobulinemia E and hereditary immune deficiencies.
- Author
-
Fadil I, Ailal F, Beziat V, Casanova JL, Boisson B, and Bousfiha AA
- Subjects
- Humans, Immunoglobulin E, Hypergammaglobulinemia, Hypersensitivity, Immunologic Deficiency Syndromes
- Abstract
The detection of a high serum immunoglobulin E (IgE) level is first suggestive of allergy, atopy or parasitosis. However, some very high values can be a sign of more severe diseases. We propose a diagnostic strategy based on clinical and biological data to identify the various hereditary immune diseases that also present with abnormally high serum IgE levels.
- Published
- 2023
4. Inborn Errors of Immunity in Algerian Children and Adults: A Single-Center Experience Over a Period of 13 Years (2008-2021).
- Author
-
Belaid B, Lamara Mahammed L, Drali O, Oussaid AM, Touri NS, Melzi S, Dehimi A, Berkani LM, Merah F, Larab Z, Allam I, Khemici O, Kirane SY, Boutaba M, Belbouab R, Bekkakcha H, Guedouar A, Chelali A, Baamara B, Noui D, Baaziz H, Rezak R, Azzouz SM, Aichaoui M, Moktefi A, Benhatchi RM, Oussalah M, Benaissa N, Laredj A, Bouchetara A, Adria A, Habireche B, Tounsi N, Dahmoun F, Touati R, Boucenna H, Bouferoua F, Sekfali L, Bouhafs N, Aboura R, Kherra S, Inouri Y, Dib S, Medouri N, Khelfaoui N, Redjedal A, Zelaci A, Yahiaoui S, Medjadj S, Touhami TK, Kadi A, Amireche F, Frada I, Houasnia S, Benarab K, Boubidi C, Ferhani Y, Benalioua H, Sokhal S, Benamar N, Aggoune S, Hadji K, Bellouti A, Rahmoune H, Boutrid N, Okka K, Ammour A, Saadoune H, Amroun M, Belhadj H, Ghanem A, Abbaz H, Boudrioua S, Zebiche B, Ayad A, Hamadache Z, Ouaras N, Achour N, Bouchair N, Boudiaf H, Bekkat-Berkani D, Maouche H, Bouzrar Z, Aissat L, Ibsaine O, Bioud B, Kedji L, Dahlouk D, Bensmina M, Radoui A, Bessahraoui M, Bensaadi N, Mekki A, Zeroual Z, Chan KW, Leung D, Tebaibia A, Ayoub S, Mekideche D, Gharnaout M, Casanova JL, Puel A, Lau YL, Cherif N, Ladj S, Smati L, Boukari R, Benhalla N, and Djidjik R
- Subjects
- Adult, Algeria epidemiology, Child, Female, Humans, Male, Retrospective Studies, Immunologic Deficiency Syndromes genetics, Primary Immunodeficiency Diseases genetics, Severe Combined Immunodeficiency
- Abstract
Background: Inborn errors of immunity (IEI) predispose patients to various infectious and non-infectious complications. Thanks to the development and expanding use of flow cytometry and increased awareness, the diagnostic rate of IEI has markedly increased in Algeria the last decade., Aim: This study aimed to describe a large cohort of Algerian patients with probable IEI and to determine their clinical characteristics and outcomes., Methods: We collected and analyzed retrospectively the demographic data, clinical manifestations, immunologic, genetic data, and outcome of Algerian IEI patients - diagnosed in the department of medical immunology of Beni Messous university hospital center, Algiers, from 2008 to 2021., Results: Eight hundred and seven patients with IEI (482 males and 325 females) were enrolled, 9.7% of whom were adults. Consanguinity was reported in 50.3% of the cases and a positive family history in 32.34%. The medium age at disease onset was 8 months and at diagnosis was 36 months. The median delay in diagnosis was 16 months. Combined immunodeficiencies were the most frequent (33.8%), followed by antibody deficiencies (24.5%) and well-defined syndromes with immunodeficiency (24%). Among 287 patients tested for genetic disorders, 129 patients carried pathogenic mutations; 102 having biallelic variants mostly in a homozygous state (autosomal recessive disorders). The highest mortality rate was observed in patients with combined immunodeficiency (70.1%), especially in patients with severe combined immunodeficiency (SCID), Omenn syndrome, or Major Histocompatibility Complex (MHC) class II deficiency., Conclusion: The spectrum of IEI in Algeria is similar to that seen in most countries of the Middle East and North Africa (MENA) region, notably regarding the frequency of autosomal recessive and/or combined immunodeficiencies., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Belaid, Lamara Mahammed, Drali, Oussaid, Touri, Melzi, Dehimi, Berkani, Merah, Larab, Allam, Khemici, Kirane, Boutaba, Belbouab, Bekkakcha, Guedouar, Chelali, Baamara, Noui, Baaziz, Rezak, Azzouz, Aichaoui, Moktefi, Benhatchi, Oussalah, Benaissa, Laredj, Bouchetara, Adria, Habireche, Tounsi, Dahmoun, Touati, Boucenna, Bouferoua, Sekfali, Bouhafs, Aboura, Kherra, Inouri, Dib, Medouri, Khelfaoui, Redjedal, Zelaci, Yahiaoui, Medjadj, Touhami, Kadi, Amireche, Frada, Houasnia, Benarab, Boubidi, Ferhani, Benalioua, Sokhal, Benamar, Aggoune, Hadji, Bellouti, Rahmoune, Boutrid, Okka, Ammour, Saadoune, Amroun, Belhadj, Ghanem, Abbaz, Boudrioua, Zebiche, Ayad, Hamadache, Ouaras, Achour, Bouchair, Boudiaf, Bekkat-Berkani, Maouche, Bouzrar, Aissat, Ibsaine, Bioud, Kedji, Dahlouk, Bensmina, Radoui, Bessahraoui, Bensaadi, Mekki, Zeroual, Chan, Leung, Tebaibia, Ayoub, Mekideche, Gharnaout, Casanova, Puel, Lau, Cherif, Ladj, Smati, Boukari, Benhalla and Djidjik.)
- Published
- 2022
- Full Text
- View/download PDF
5. From Dysgammaglobulinemia to Autosomal-Dominant Activation-Induced Cytidine Deaminase Deficiency: Unraveling an Inherited Immunodeficiency after 50 Years.
- Author
-
Fadlallah J, Chentout L, Boisson B, Pouliet A, Masson C, Morin F, Durandy A, Casanova JL, Oksenhendler E, and Kracker S
- Subjects
- Cytidine Deaminase metabolism, DNA Mutational Analysis, Dysgammaglobulinemia complications, Dysgammaglobulinemia metabolism, Female, Follow-Up Studies, Humans, Middle Aged, Cytidine Deaminase genetics, Dysgammaglobulinemia genetics, Forecasting, Immunologic Deficiency Syndromes complications, Mutation
- Abstract
The genetic investigation of a family presenting with a dominant form of hyper IgM syndrome published in 1963 and 1975 revealed a R190X nonsense mutation in activation-induced cytidine deaminase. This report illustrates the progress made over 6 decades in the characterization of primary immunodeficiencies, from immunochemistry to whole-exome sequencing., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
6. Autoimmune Lymphoproliferative Syndrome Presenting with Invasive Streptococcus pneumoniae Infection.
- Author
-
Oksenhendler E, Spaan AN, Neven B, Stolzenberg MC, Fusaro M, Casanova JL, Rieux-Laucat F, Boisson B, and Magérus A
- Subjects
- Autoimmune Lymphoproliferative Syndrome genetics, Female, Humans, Immunoglobulin M metabolism, Immunologic Deficiency Syndromes genetics, Immunologic Memory genetics, Infant, Male, Pedigree, Pneumococcal Infections genetics, Autoimmune Lymphoproliferative Syndrome diagnosis, B-Lymphocytes physiology, Immunologic Deficiency Syndromes diagnosis, Mutation genetics, Pneumococcal Infections diagnosis, Streptococcus pneumoniae physiology, fas Receptor genetics
- Published
- 2020
- Full Text
- View/download PDF
7. Human BCL10 Deficiency due to Homozygosity for a Rare Allele.
- Author
-
Van Den Rym A, Taur P, Martinez-Barricarte R, Lorenzo L, Puel A, Gonzalez-Navarro P, Pandrowala A, Gowri V, Safa A, Toledano V, Cubillos-Zapata C, López-Collazo E, Vela M, Pérez-Martínez A, Sánchez-Ramón S, Recio MJ, Casanova JL, Desai MM, and Perez de Diego R
- Subjects
- Cells, Cultured, Chromosome Disorders, Homozygote, Humans, Immunologic Memory, Infant, Lectins, C-Type metabolism, Male, Respiratory Tract Infections, Toll-Like Receptors metabolism, B-Cell CLL-Lymphoma 10 Protein genetics, B-Lymphocytes immunology, Immunologic Deficiency Syndromes genetics, Mutation genetics, T-Lymphocytes immunology
- Abstract
In 2014, a child with broad combined immunodeficiency (CID) who was homozygous for a private BCL10 allele was reported to have complete inherited human BCL10 deficiency. In the present study, we report a new BCL10 mutation in another child with CID who was homozygous for a BCL10 variant (R88X), previously reported as a rare allele in heterozygosis (minor allele frequency, 0.000003986). The mutant allele was a loss-of-expression and loss-of-function allele. As with the previously reported patient, this patient had complete BCL10 deficiency. The clinical phenotype shared features, such as respiratory infections, but differed from that of the previous patient that he did not develop significant gastroenteritis episodes or chronic colitis. Cellular and immunological phenotypes were similar to those of the previous patient. TLR4, TLR2/6, and Dectin-1 responses were found to depend on BCL10 in fibroblasts, and final maturation of T cell and B cell maturation into memory cells was affected. Autosomal-recessive BCL10 deficiency should therefore be considered in children with CID.
- Published
- 2020
- Full Text
- View/download PDF
8. Human inborn errors of immunity to herpes viruses.
- Author
-
Jouanguy E, Béziat V, Mogensen TH, Casanova JL, Tangye SG, and Zhang SY
- Subjects
- Genetic Diseases, Inborn diagnosis, Genetic Diseases, Inborn virology, Herpesviridae isolation & purification, Herpesviridae Infections transmission, Herpesviridae Infections virology, Humans, Immunologic Deficiency Syndromes diagnosis, Immunologic Deficiency Syndromes virology, Genetic Diseases, Inborn immunology, Herpesviridae immunology, Herpesviridae Infections immunology, Immunity, Innate immunology, Immunologic Deficiency Syndromes immunology
- Abstract
Infections with any of the nine human herpes viruses (HHV) can be asymptomatic or life-threatening. The study of patients with severe diseases caused by HHVs, in the absence of overt acquired immunodeficiency, has led to the discovery or diagnosis of various inborn errors of immunity. The related inborn errors of adaptive immunity disrupt α/β T-cell rather than B-cell immunity. Affected patients typically develop HHV infections in the context of other infectious diseases. However, this is not always the case, as illustrated by inborn errors of SAP-dependent T-cell immunity to EBV-infected B cells. The related inborn errors of innate immunity disrupt leukocytes other than T and B cells, non-hematopoietic cells, or both. Patients typically develop only a single type of infection due to HHV, although, again, this is not always the case, as illustrated by inborn errors of TLR3 immunity resulting in HSV1 encephalitis in some patients and influenza pneumonitis in others. Most severe HHV infections in otherwise healthy patients remains unexplained. The forward human genetic dissection of isolated and syndromic HHV-driven illnesses will establish the molecular and cellular basis of protective immunity to HHVs, paving the way for novel diagnosis and management strategies., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. Human Inborn Errors of Immunity: 2019 Update of the IUIS Phenotypical Classification.
- Author
-
Bousfiha A, Jeddane L, Picard C, Al-Herz W, Ailal F, Chatila T, Cunningham-Rundles C, Etzioni A, Franco JL, Holland SM, Klein C, Morio T, Ochs HD, Oksenhendler E, Puck J, Torgerson TR, Casanova JL, Sullivan KE, and Tangye SG
- Subjects
- Autoimmunity genetics, Genotype, Hereditary Autoinflammatory Diseases genetics, Humans, Hypersensitivity, Phenotype, Immunity genetics, Immunologic Deficiency Syndromes genetics
- 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.
- Published
- 2020
- Full Text
- View/download PDF
10. A combined immunodeficiency with severe infections, inflammation, and allergy caused by ARPC1B deficiency.
- Author
-
Volpi S, Cicalese MP, Tuijnenburg P, Tool ATJ, Cuadrado E, Abu-Halaweh M, Ahanchian H, Alzyoud R, Akdemir ZC, Barzaghi F, Blank A, Boisson B, Bottino C, Brigida I, Caorsi R, Casanova JL, Chiesa S, Chinn IK, Dückers G, Enders A, Erichsen HC, Forbes LR, Gambin T, Gattorno M, Karimiani EG, Giliani S, Gold MS, Jacobsen EM, Jansen MH, King JR, Laxer RM, Lupski JR, Mace E, Marcenaro S, Maroofian R, Meijer AB, Niehues T, Notarangelo LD, Orange J, Pannicke U, Pearson C, Picco P, Quinn PJ, Schulz A, Seeborg F, Stray-Pedersen A, Tawamie H, van Leeuwen EMM, Aiuti A, Yeung R, Schwarz K, and Kuijpers TW
- Subjects
- Actin-Related Protein 2-3 Complex genetics, Adolescent, Child, Child, Preschool, Female, Humans, Hypersensitivity genetics, Hypersensitivity immunology, Immunologic Deficiency Syndromes immunology, Infant, Infections genetics, Infections immunology, Inflammation genetics, Inflammation immunology, Male, Mutation, Actin-Related Protein 2-3 Complex deficiency, Immunologic Deficiency Syndromes genetics
- Published
- 2019
- Full Text
- View/download PDF
11. Molecular, Immunological, and Clinical Features of 16 Iranian Patients with Mendelian Susceptibility to Mycobacterial Disease.
- Author
-
Sarrafzadeh SA, Nourizadeh M, Mahloojirad M, Fazlollahi MR, Shokouhi Shoormasti R, Badalzadeh M, Deswarte C, Casanova JL, Pourpak Z, Bustamante J, and Moin M
- Subjects
- Adolescent, Cells, Cultured, Child, Child, Preschool, Cytokines metabolism, Female, Humans, Immunity genetics, Infant, Interferon-gamma genetics, Interferon-gamma metabolism, Iran, Male, Pedigree, BCG Vaccine immunology, Immunologic Deficiency Syndromes diagnosis, Mycobacterium Infections, Nontuberculous genetics, Receptors, Interleukin-12 genetics, Sequence Deletion genetics
- Abstract
Purpose: Mendelian susceptibility to mycobacterial disease (MSMD) is a rare primary immunodeficiency, triggered by non-tuberculous mycobacteria or Bacillus Calmette-Guérin (BCG) vaccines and characterized by severe diseases. All known genetic etiologies are inborn errors of IFN-γ-mediated immunity. Here, we report the molecular, cellular, and clinical features of patients from 15 Iranian families with disseminated disease without vaccination (2 patients) or following live BCG vaccination (14 patients)., Methods: We used whole blood samples from 16 patients and 12 age-matched healthy controls. To measure IL-12 and IFN-γ, samples were activated by BCG plus recombinant human IFN-γ or recombinant human IL-12. Immunological assessments and genetic analysis were also done for the patients., Results: Eight patients affected as a result of parental first-cousin marriages. Seven patients originated from multiplex kindred with positive history of death because of tuberculosis or finding the MSMD-related gene mutations. Two patients died due to mycobacterial disease at the ages of 8 months and 3.7 years. The remaining patients were alive at the last follow-up and were aged between 2 and 13 years. Patients suffered from infections including chronic mucocutaneous candidiasis (n = 10), salmonellosis (n = 2), and Leishmania (responsible for visceral form) (n = 2). Thirteen patients presented with autosomal recessive (AR) IL-12Rβ1 deficiency, meaning their cells produced low levels of IFN-γ. Bi-allelic IL12RB1 mutations were detected in nine of patients. Three patients with AR IL-12p40 deficiency (bi-allelic IL12B mutations) produced low levels of both IL-12 and IFN-γ. Overall, we found five mutations in the IL12RB1 gene and three mutations in the IL12B gene. Except one mutation in exon 5 (c.510C>A) of IL12B, all others were previously reported to be loss-of-function mutations., Conclusions: We found low levels of IFN-γ production and failure to respond to IL12 in 13 Iranian MSMD patients. Due to complicated clinical manifestations in affected children, early cellular and molecular diagnostics is crucial in susceptible patients.
- Published
- 2019
- Full Text
- View/download PDF
12. Biallelic mutations in DNA ligase 1 underlie a spectrum of immune deficiencies.
- Author
-
Maffucci P, Chavez J, Jurkiw TJ, O'Brien PJ, Abbott JK, Reynolds PR, Worth A, Notarangelo LD, Felgentreff K, Cortes P, Boisson B, Radigan L, Cobat A, Dinakar C, Ehlayel M, Ben-Omran T, Gelfand EW, Casanova JL, and Cunningham-Rundles C
- Subjects
- HEK293 Cells, Humans, Alleles, DNA Ligase ATP genetics, DNA Ligase ATP immunology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Mutation
- Abstract
We report the molecular, cellular, and clinical features of 5 patients from 3 kindreds with biallelic mutations in the autosomal LIG1 gene encoding DNA ligase 1. The patients exhibited hypogammaglobulinemia, lymphopenia, increased proportions of circulating γδT cells, and erythrocyte macrocytosis. Clinical severity ranged from a mild antibody deficiency to a combined immunodeficiency requiring hematopoietic stem cell transplantation. Using engineered LIG1-deficient cell lines, we demonstrated chemical and radiation defects associated with the mutant alleles, which variably impaired the DNA repair pathway. We further showed that these LIG1 mutant alleles are amorphic or hypomorphic, and exhibited variably decreased enzymatic activities, which lead to premature release of unligated adenylated DNA. The variability of the LIG1 genotypes in the patients was consistent with that of their immunological and clinical phenotypes. These data suggest that different forms of autosomal recessive, partial DNA ligase 1 deficiency underlie an immunodeficiency of variable severity.
- Published
- 2018
- Full Text
- View/download PDF
13. T-cell defects in patients with ARPC1B germline mutations account for combined immunodeficiency.
- Author
-
Brigida I, Zoccolillo M, Cicalese MP, Pfajfer L, Barzaghi F, Scala S, Oleaga-Quintas C, Álvarez-Álvarez JA, Sereni L, Giannelli S, Sartirana C, Dionisio F, Pavesi L, Benavides-Nieto M, Basso-Ricci L, Capasso P, Mazzi B, Rosain J, Marcus N, Lee YN, Somech R, Degano M, Raiola G, Caorsi R, Picco P, Moncada Velez M, Khourieh J, Arias AA, Bousfiha A, Issekutz T, Issekutz A, Boisson B, Dobbs K, Villa A, Lombardo A, Neven B, Moshous D, Casanova JL, Franco JL, Notarangelo LD, Scielzo C, Volpi S, Dupré L, Bustamante J, Gattorno M, and Aiuti A
- Subjects
- 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, Actin-Related Protein 2-3 Complex genetics, Germ-Line Mutation, Immunologic Deficiency Syndromes genetics, T-Lymphocytes pathology
- 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., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
14. Mechanism of dysfunction of human variants of the IRAK4 kinase and a role for its kinase activity in interleukin-1 receptor signaling.
- Author
-
De S, Karim F, Kiessu E, Cushing L, Lin LL, Ghandil P, Hoarau C, Casanova JL, Puel A, and Rao VR
- Subjects
- Cell Line, Crystallography, X-Ray, Humans, Immunity, Innate genetics, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes pathology, Interleukin-1 chemistry, Interleukin-1 Receptor-Associated Kinases deficiency, Mutation, Myeloid Differentiation Factor 88 chemistry, NF-kappa B genetics, Polymorphism, Single Nucleotide genetics, Receptors, Interleukin-1 chemistry, Receptors, Interleukin-1 genetics, Signal Transduction, Immunologic Deficiency Syndromes genetics, Interleukin-1 genetics, Interleukin-1 Receptor-Associated Kinases chemistry, Interleukin-1 Receptor-Associated Kinases genetics, Myeloid Differentiation Factor 88 genetics
- Abstract
Interleukin-1 receptor (IL1R)-associated kinase 4 (IRAK4) is a central regulator of innate immune signaling, controlling IL1R and Toll-like receptor (TLR)-mediated responses and containing both scaffolding and kinase activities. Humans deficient in IRAK4 activity have autosomal recessive primary immune deficiency (PID). Here, we characterized the molecular mechanism of dysfunction of two IRAK4 PID variants, G298D and the compound variant R12C (R12C/R391H/T458I). Using these variants and the kinase-inactive D329A variant to delineate the contributions of IRAK4's scaffolding and kinase activities to IL1R signaling, we found that the G298D variant is kinase-inactive and expressed at extremely low levels, acting functionally as a null mutation. The R12C compound variant possessed WT kinase activity, but could not interact with myeloid differentiation primary response 88 (MyD88) and IRAK1, causing impairment of IL-1-induced signaling and cytokine production. Quantitation of IL-1 signaling in IRAK4-deficient cells complemented with either WT or the R12C or D329A variant indicated that the loss of MyD88 interaction had a greater impact on IL-1-induced signaling and cytokine expression than the loss of IRAK4 kinase activity. Importantly, kinase-inactive IRAK4 exhibited a greater association with MyD88 and a weaker association with IRAK1 in IRAK4-deficient cells expressing kinase-inactive IRAK4 and in primary cells treated with a selective IRAK4 inhibitor. Loss of IRAK4 kinase activity only partially inhibited IL-1-induced cytokine and NF-κB signaling. Therefore, the IRAK4-MyD88 scaffolding function is essential for IL-1 signaling, but IRAK4 kinase activity can control IL-1 signal strength by modulating the association of IRAK4, MyD88, and IRAK1., (© 2018 De et al.)
- Published
- 2018
- Full Text
- View/download PDF
15. Incomplete penetrance for isolated congenital asplenia in humans with mutations in translated and untranslated RPSA exons.
- Author
-
Bolze A, Boisson B, Bosch B, Antipenko A, Bouaziz M, Sackstein P, Chaker-Margot M, Barlogis V, Briggs T, Colino E, Elmore AC, Fischer A, Genel F, Hewlett A, Jedidi M, Kelecic J, Krüger R, Ku CL, Kumararatne D, Lefevre-Utile A, Loughlin S, Mahlaoui N, Markus S, Garcia JM, Nizon M, Oleastro M, Pac M, Picard C, Pollard AJ, Rodriguez-Gallego C, Thomas C, Von Bernuth H, Worth A, Meyts I, Risolino M, Selleri L, Puel A, Klinge S, Abel L, and Casanova JL
- Subjects
- 5' Untranslated Regions, Female, Founder Effect, Heterozygote, Humans, Immunologic Deficiency Syndromes metabolism, Male, Primary Immunodeficiency Diseases, Receptors, Laminin biosynthesis, Ribosomal Proteins biosynthesis, Spleen metabolism, Exons, Immunologic Deficiency Syndromes genetics, Mutation, Penetrance, Protein Biosynthesis genetics, RNA Splicing genetics, Receptors, Laminin genetics, Ribosomal Proteins genetics, Spleen abnormalities
- Abstract
Isolated congenital asplenia (ICA) is the only known human developmental defect exclusively affecting a lymphoid organ. In 2013, we showed that private deleterious mutations in the protein-coding region of RPSA , encoding ribosomal protein SA, caused ICA by haploinsufficiency with complete penetrance. We reported seven heterozygous protein-coding mutations in 8 of the 23 kindreds studied, including 6 of the 8 multiplex kindreds. We have since enrolled 33 new kindreds, 5 of which are multiplex. We describe here 11 new heterozygous ICA-causing RPSA protein-coding mutations, and the first two mutations in the 5'-UTR of this gene, which disrupt mRNA splicing. Overall, 40 of the 73 ICA patients (55%) and 23 of the 56 kindreds (41%) carry mutations located in translated or untranslated exons of RPSA. Eleven of the 43 kindreds affected by sporadic disease (26%) carry RPSA mutations, whereas 12 of the 13 multiplex kindreds (92%) carry RPSA mutations. We also report that 6 of 18 (33%) protein-coding mutations and the two (100%) 5'-UTR mutations display incomplete penetrance. Three mutations were identified in two independent kindreds, due to a hotspot or a founder effect. Finally, RPSA ICA-causing mutations were demonstrated to be de novo in 7 of the 23 probands. Mutations in RPSA exons can affect the translated or untranslated regions and can underlie ICA with complete or incomplete penetrance., Competing Interests: Conflict of interest statement: A.B. works at Helix.
- Published
- 2018
- Full Text
- View/download PDF
16. Laboratory evaluation of the IFN-γ circuit for the molecular diagnosis of Mendelian susceptibility to mycobacterial disease.
- Author
-
Esteve-Solé A, Sologuren I, Martínez-Saavedra MT, Deyà-Martínez À, Oleaga-Quintas C, Martinez-Barricarte R, Martin-Nalda A, Juan M, Casanova JL, Rodriguez-Gallego C, Alsina L, and Bustamante J
- Subjects
- BCG Vaccine, Humans, Mycobacterium, Genetic Predisposition to Disease genetics, Immunologic Deficiency Syndromes diagnosis, Immunologic Deficiency Syndromes genetics, Interferon-gamma genetics, Molecular Diagnostic Techniques, Mycobacterium Infections genetics
- Abstract
The integrity of the interferon (IFN)-γ circuit is necessary to mount an effective immune response to intra-macrophagic pathogens, especially Mycobacteria. Inherited monogenic defects in this circuit that disrupt the production of, or response to, IFN-γ underlie a primary immunodeficiency known as Mendelian susceptibility to mycobacterial disease (MSMD). Otherwise healthy patients display a selective susceptibility to clinical disease caused by poorly virulent mycobacteria such as BCG (bacille Calmette-Guérin) vaccines and environmental mycobacteria, and more rarely by other intra-macrophagic pathogens, particularly Salmonella and M. tuberculosis. There is high genetic and allelic heterogeneity, with 19 genetic etiologies due to mutations in 10 genes that account for only about half of the patients reported. An efficient laboratory diagnostic approach to suspected MSMD patients is important, because it enables the establishment of specific therapeutic measures that will improve the patient's prognosis and quality of life. Moreover, it is essential to offer genetic counseling to affected families. Herein, we review the various genetic and immunological diagnostic approaches that can be used in concert to reach a molecular and cellular diagnosis in patients with MSMD.
- Published
- 2018
- Full Text
- View/download PDF
17. Human genetics of infectious diseases: Unique insights into immunological redundancy.
- Author
-
Casanova JL and Abel L
- Subjects
- Asymptomatic Diseases, Biological Evolution, Communicable Diseases immunology, Genetic Diseases, X-Linked immunology, Genetic Predisposition to Disease, Host-Pathogen Interactions, Humans, Immunologic Deficiency Syndromes immunology, Phenotype, Viral Tropism, Communicable Diseases genetics, Genetic Diseases, X-Linked genetics, Genotype, Immunity genetics, Immunologic Deficiency Syndromes genetics
- Abstract
For almost any given human-tropic virus, bacterium, fungus, or parasite, the clinical outcome of primary infection is enormously variable, ranging from asymptomatic to lethal infection. This variability has long been thought to be largely determined by the germline genetics of the human host, and this is increasingly being demonstrated to be the case. The number and diversity of known inborn errors of immunity is continually increasing, and we focus here on autosomal and X-linked recessive traits underlying complete deficiencies of the encoded protein. Schematically, four types of infectious phenotype have been observed in individuals with such deficiencies, each providing information about the redundancy of the corresponding human gene, in terms of host defense in natural conditions. The lack of a protein can confer vulnerability to a broad range of microbes in most, if not all patients, through the disruption of a key immunological component. In such cases, the gene concerned is of low redundancy. However, the lack of a protein may also confer vulnerability to a narrow range of microbes, sometimes a single pathogen, and not necessarily in all patients. In such cases, the gene concerned is highly redundant. Conversely, the deficiency may be apparently neutral, conferring no detectable predisposition to infection in any individual. In such cases, the gene concerned is completely redundant. Finally, the lack of a protein may, paradoxically, be advantageous to the host, conferring resistance to one or more infections. In such cases, the gene is considered to display beneficial redundancy. These findings reflect the current state of evolution of humans and microbes, and should not be considered predictive of redundancy, or of a lack of redundancy, in the distant future. Nevertheless, these observations are of potential interest to present-day biologists testing immunological hypotheses experimentally and physicians managing patients with immunological or infectious conditions., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
18. Mechanisms of genotype-phenotype correlation in autosomal dominant anhidrotic ectodermal dysplasia with immune deficiency.
- Author
-
Petersheim D, Massaad MJ, Lee S, Scarselli A, Cancrini C, Moriya K, Sasahara Y, Lankester AC, Dorsey M, Di Giovanni D, Bezrodnik L, Ohnishi H, Nishikomori R, Tanita K, Kanegane H, Morio T, Gelfand EW, Jain A, Secord E, Picard C, Casanova JL, Albert MH, Torgerson TR, and Geha RS
- Subjects
- Carrier Proteins genetics, Carrier Proteins immunology, Chemokine CCL20 genetics, Chemokine CCL20 immunology, Ectodermal Dysplasia pathology, Genetic Diseases, X-Linked pathology, HEK293 Cells, Humans, Immunologic Deficiency Syndromes pathology, Intercellular Adhesion Molecule-1 genetics, Intercellular Adhesion Molecule-1 immunology, Interleukin-6 genetics, Interleukin-6 immunology, NF-KappaB Inhibitor alpha genetics, NF-KappaB Inhibitor alpha immunology, NF-kappa B p52 Subunit genetics, NF-kappa B p52 Subunit immunology, Point Mutation, Primary Immunodeficiency Diseases, Proto-Oncogene Mas, Ectodermal Dysplasia genetics, Ectodermal Dysplasia immunology, Genetic Diseases, X-Linked genetics, Genetic Diseases, X-Linked immunology, Genotype, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Signal Transduction genetics, Signal Transduction immunology
- Abstract
Background: Autosomal dominant anhidrotic ectodermal dysplasia with immune deficiency (AD EDA-ID) is caused by heterozygous point mutations at or close to serine 32 and serine 36 or N-terminal truncations in IκBα that impair its phosphorylation and degradation and thus activation of the canonical nuclear factor κ light chain enhancer of activated B cells (NF-κB) pathway. The outcome of hematopoietic stem cell transplantation is poor in patients with AD EDA-ID despite achievement of chimerism. Mice heterozygous for the serine 32I mutation in IκBα have impaired noncanonical NF-κB activity and defective lymphorganogenesis., Objective: We sought to establish genotype-phenotype correlation in patients with AD EDA-ID., Methods: A disease severity scoring system was devised. Stability of IκBα mutants was examined in transfected cells. Immunologic, biochemical, and gene expression analyses were performed to evaluate canonical and noncanonical NF-κB signaling in skin-derived fibroblasts., Results: Disease severity was greater in patients with IκBα point mutations than in those with truncation mutations. IκBα point mutants were expressed at significantly higher levels in transfectants compared with truncation mutants. Canonical NF-κB-dependent IL-6 secretion and upregulation of the NF-κB subunit 2/p100 and RELB proto-oncogene, NF-κB subunit (RelB) components of the noncanonical NF-κB pathway were diminished significantly more in patients with point mutations compared with those with truncations. Noncanonical NF-κB-driven generation of the transcriptionally active p100 cleavage product p52 and upregulation of CCL20, intercellular adhesion molecule 1 (ICAM1), and vascular cell adhesion molecule 1 (VCAM1), which are important for lymphorganogenesis, were diminished significantly more in LPS plus α-lymphotoxin β receptor-stimulated fibroblasts from patients with point mutations compared with those with truncations., Conclusions: IκBα point mutants accumulate at higher levels compared with truncation mutants and are associated with more severe disease and greater impairment of canonical and noncanonical NF-κB activity in patients with AD EDA-ID., (Copyright © 2017 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
19. International Union of Immunological Societies: 2017 Primary Immunodeficiency Diseases Committee Report on Inborn Errors of Immunity.
- Author
-
Picard C, Bobby Gaspar H, Al-Herz W, Bousfiha A, Casanova JL, Chatila T, Crow YJ, Cunningham-Rundles C, Etzioni A, Franco JL, Holland SM, Klein C, Morio T, Ochs HD, Oksenhendler E, Puck J, Tang MLK, Tangye SG, Torgerson TR, and Sullivan KE
- Subjects
- Humans, Research Report, Societies, Scientific, World Health Organization, Allergy and Immunology, Immunity genetics, Immunologic Deficiency Syndromes
- 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.
- Published
- 2018
- Full Text
- View/download PDF
20. The 2017 IUIS Phenotypic Classification for Primary Immunodeficiencies.
- Author
-
Bousfiha A, Jeddane L, Picard C, Ailal F, Bobby Gaspar H, Al-Herz W, Chatila T, Crow YJ, Cunningham-Rundles C, Etzioni A, Franco JL, Holland SM, Klein C, Morio T, Ochs HD, Oksenhendler E, Puck J, Tang MLK, Tangye SG, Torgerson TR, Casanova JL, and Sullivan KE
- Subjects
- Humans, Immunologic Deficiency Syndromes genetics, International Cooperation, Phenotype, Allergy and Immunology, Immunity genetics, Immunologic Deficiency Syndromes immunology
- 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.
- Published
- 2018
- Full Text
- View/download PDF
21. Pathogenesis of infections in HIV-infected individuals: insights from primary immunodeficiencies.
- Author
-
Zhang Q, Frange P, Blanche S, and Casanova JL
- Subjects
- Animals, CD4-Positive T-Lymphocytes virology, Candidiasis complications, HIV Infections complications, Humans, Immunologic Deficiency Syndromes complications, Infections complications, Signal Transduction, CD4-Positive T-Lymphocytes immunology, Candidiasis immunology, HIV Infections immunology, HIV-1 immunology, Immunologic Deficiency Syndromes immunology, Infections immunology, Th17 Cells immunology
- Abstract
Following infection with almost any given microorganism other than an emerging pathogen, only a minority of individuals develop life-threatening clinical disease, implying that these individuals have some form of immunodeficiency. A growing number of inherited and acquired immunodeficiencies have been deciphered over the last 50 years. HIV infection is probably the best-known acquired immunodeficiency. It emerged about 40 years ago and precipitates various severe infections, the occurrence of which is associated with a fall in circulating CD4
+ T cells. However, despite the strength of this correlation, infection rates differ between patients with similar levels and durations of CD4+ T lymphopenia in the presence or absence of antiretroviral treatment. Moreover, a few infections seem to be less dependent on total CD4+ T-cell levels. The fine detail of the mechanisms underlying these infections is unknown. We discuss here how studies of the human genetics and immunology of some of these infections in patients with primary immunodeficiencies (PIDs) have provided unique insights into their molecular and cellular basis. Defects of specific CD4+ Th-cell subsets account for some of these infections, as best exemplified by Th1* for mycobacteriosis and Th17 for candidiasis. PIDs are individually rare, but collectively much more common than initially thought, with new disorders being discovered at an ever-increasing pace and a global prevalence worldwide approaching that of HIV infection. Studies of known and new PIDs should make it possible to dissect the pathogenesis of most human infections at an unprecedented level of molecular and cellular precision. The predictive, preventive, and therapeutic implications of studies of immunity to infection in PIDs may extend to HIV-infected patients and patients with infectious diseases in other settings., (Copyright © 2017 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
22. Human IκBα Gain of Function: a Severe and Syndromic Immunodeficiency.
- Author
-
Boisson B, Puel A, Picard C, and Casanova JL
- Subjects
- Alleles, Animals, Enzyme Activation, Gene Expression Regulation, Genetic Association Studies, Genetic Predisposition to Disease, Germ-Line Mutation, Hematopoietic Stem Cell Transplantation, Heterozygote, Humans, Immunity genetics, Immunologic Deficiency Syndromes diagnosis, Immunologic Deficiency Syndromes therapy, Organ Specificity genetics, Phenotype, Phosphorylation, Proteolysis, Severity of Illness Index, Treatment Outcome, Gain of Function Mutation, Immunologic Deficiency Syndromes etiology, Immunologic Deficiency Syndromes metabolism, NF-KappaB Inhibitor alpha genetics, NF-KappaB Inhibitor alpha metabolism
- Abstract
Germline heterozygous gain-of-function (GOF) mutations of NFKBIA, encoding IκBα, cause an autosomal dominant (AD) form of anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID). Fourteen unrelated patients have been reported since the identification of the first case in 2003. All mutations enhanced the inhibitory activity of IκBα, by preventing its phosphorylation on serine 32 or 36 and its subsequent degradation. The mutation certainly or probably occurred de novo in 13 patients, whereas it was inherited from a parent with somatic mosaicism in one patient. Eleven mutations, belonging to two groups, were identified: (i) missense mutations affecting S32, S36, or neighboring residues (8 mutations, 11 patients) and (ii) nonsense mutations upstream from S32 associated with the reinitiation of translation downstream from S36 (3 mutations, 3 patients). Thirteen patients had developmental features of EDA, the severity and nature of which differed between cases. All patient cells tested displayed impaired NF-κB-mediated responses to the stimulation of various surface receptors involved in cell-intrinsic (fibroblasts), innate (monocytes), and adaptive (B and T cells) immunity, including TLRs, IL-1Rs, TNFRs, TCR, and BCR. All patients had profound B-cell deficiency. Specific immunological features, found in some, but not all patients, included a lack of peripheral lymph nodes, lymphocytosis, dysfunctional α/β T cells, and a lack of circulating γ/δ T cells. The patients had various pyogenic, mycobacterial, fungal, and viral severe infections. Patients with a missense mutation tended to display more severe phenotypes, probably due to higher levels of GOF proteins. In the absence of hematopoietic stem cell transplantation (HSCT), this condition cause death before the age of 1 year (one child). Two survivors have been on prophylaxis (at 9 and 22 years). Six children died after HSCT. Five survived, four of whom have been on prophylaxis (3 to 21 years post HSCT), whereas one has been well with no prophylaxis. Heterozygous GOF mutations in IκBα underlie a severe and syndromic immunodeficiency, the interindividual variability of which might partly be ascribed to the dichotomy of missense and nonsense mutations, and the hematopoietic component of which can be rescued by HSCT.
- Published
- 2017
- Full Text
- View/download PDF
23. Visceral leishmaniasis in two patients with IL-12p40 and IL-12Rβ1 deficiencies.
- Author
-
Parvaneh N, Barlogis V, Alborzi A, Deswarte C, Boisson-Dupuis S, Migaud M, Farnaria C, Markle J, Parvaneh L, Casanova JL, and Bustamante J
- Subjects
- Adolescent, Child, Female, Humans, Male, Genetic Diseases, Inborn genetics, Genetic Diseases, Inborn immunology, Genetic Diseases, Inborn pathology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes pathology, Interleukin-12 Subunit p40 deficiency, Leishmaniasis, Visceral genetics, Leishmaniasis, Visceral immunology, Leishmaniasis, Visceral pathology, Receptors, Interleukin-12 deficiency
- Abstract
Mutations of the IL12B and IL12RB1 genes underlie the development of IL-12 p40 and IL-12Rβ1 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-12Rβ1 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., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
24. Inherited GINS1 deficiency underlies growth retardation along with neutropenia and NK cell deficiency.
- Author
-
Cottineau J, Kottemann MC, Lach FP, Kang YH, Vély F, Deenick EK, Lazarov T, Gineau L, Wang Y, Farina A, Chansel M, Lorenzo L, Piperoglou C, Ma CS, Nitschke P, Belkadi A, Itan Y, Boisson B, Jabot-Hanin F, Picard C, Bustamante J, Eidenschenk C, Boucherit S, Aladjidi N, Lacombe D, Barat P, Qasim W, Hurst JA, Pollard AJ, Uhlig HH, Fieschi C, Michon J, Bermudez VP, Abel L, de Villartay JP, Geissmann F, Tangye SG, Hurwitz J, Vivier E, Casanova JL, Smogorzewska A, and Jouanguy E
- Subjects
- Animals, DNA-Binding Proteins immunology, Female, Fetal Growth Retardation genetics, Fetal Growth Retardation immunology, Humans, Infant, Male, Mice, DNA-Binding Proteins deficiency, Genetic Diseases, Inborn genetics, Genetic Diseases, Inborn immunology, Growth Disorders genetics, Growth Disorders immunology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Killer Cells, Natural, Neutropenia genetics, Neutropenia immunology
- Abstract
Inborn errors of DNA repair or replication underlie a variety of clinical phenotypes. We studied 5 patients from 4 kindreds, all of whom displayed intrauterine growth retardation, chronic neutropenia, and NK cell deficiency. Four of the 5 patients also had postnatal growth retardation. The association of neutropenia and NK cell deficiency, which is unusual among primary immunodeficiencies and bone marrow failures, was due to a blockade in the bone marrow and was mildly symptomatic. We discovered compound heterozygous rare mutations in Go-Ichi-Ni-San (GINS) complex subunit 1 (GINS1, also known as PSF1) in the 5 patients. The GINS complex is essential for eukaryotic DNA replication, and homozygous null mutations of GINS component-encoding genes are embryonic lethal in mice. The patients' fibroblasts displayed impaired GINS complex assembly, basal replication stress, impaired checkpoint signaling, defective cell cycle control, and genomic instability, which was rescued by WT GINS1. The residual levels of GINS1 activity reached 3% to 16% in patients' cells, depending on their GINS1 genotype, and correlated with the severity of growth retardation and the in vitro cellular phenotype. The levels of GINS1 activity did not influence the immunological phenotype, which was uniform. Autosomal recessive, partial GINS1 deficiency impairs DNA replication and underlies intra-uterine (and postnatal) growth retardation, chronic neutropenia, and NK cell deficiency.
- Published
- 2017
- Full Text
- View/download PDF
25. Dedicator of cytokinesis 8-deficient CD4 + T cells are biased to a T H 2 effector fate at the expense of T H 1 and T H 17 cells.
- Author
-
Tangye SG, Pillay B, Randall KL, Avery DT, Phan TG, Gray P, Ziegler JB, Smart JM, Peake J, Arkwright PD, Hambleton S, Orange J, Goodnow CC, Uzel G, Casanova JL, Lugo Reyes SO, Freeman AF, Su HC, and Ma CS
- Subjects
- Adolescent, Adult, Allergens immunology, Child, Child, Preschool, Cytokines immunology, Female, Guanine Nucleotide Exchange Factors immunology, Humans, Immunoglobulin E blood, Leukocytes, Mononuclear immunology, Male, Young Adult, Guanine Nucleotide Exchange Factors deficiency, Immunologic Deficiency Syndromes immunology, T-Lymphocytes immunology
- Abstract
Background: Dedicator of cytokinesis 8 (DOCK8) deficiency is a combined immunodeficiency caused by autosomal recessive loss-of-function mutations in DOCK8. This disorder is characterized by recurrent cutaneous infections, increased serum IgE levels, and severe atopic disease, including food-induced anaphylaxis. However, the contribution of defects in CD4
+ T cells to disease pathogenesis in these patients has not been thoroughly investigated., Objective: We sought to investigate the phenotype and function of DOCK8-deficient CD4+ T cells to determine (1) intrinsic and extrinsic CD4+ T-cell defects and (2) how defects account for the clinical features of DOCK8 deficiency., Methods: We performed in-depth analysis of the CD4+ T-cell compartment of DOCK8-deficient patients. We enumerated subsets of CD4+ T helper cells and assessed cytokine production and transcription factor expression. Finally, we determined the levels of IgE specific for staple foods and house dust mite allergens in DOCK8-deficient patients and healthy control subjects., Results: DOCK8-deficient memory CD4+ T cells were biased toward a TH 2 type, and this was at the expense of TH 1 and TH 17 cells. In vitro polarization of DOCK8-deficient naive CD4+ T cells revealed the TH 2 bias and TH 17 defect to be T-cell intrinsic. Examination of allergen-specific IgE revealed plasma IgE from DOCK8-deficient patients is directed against staple food antigens but not house dust mites., Conclusion: Investigations into the DOCK8-deficient CD4+ T cells provided an explanation for some of the clinical features of this disorder: the TH 2 bias is likely to contribute to atopic disease, whereas defects in TH 1 and TH 17 cells compromise antiviral and antifungal immunity, respectively, explaining the infectious susceptibility of DOCK8-deficient patients., (Copyright © 2016 American Academy of Allergy, Asthma & Immunology. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
26. Clinical spectrum and features of activated phosphoinositide 3-kinase δ syndrome: A large patient cohort study.
- Author
-
Coulter TI, Chandra A, Bacon CM, Babar J, Curtis J, Screaton N, Goodlad JR, Farmer G, Steele CL, Leahy TR, Doffinger R, Baxendale H, Bernatoniene J, Edgar JD, Longhurst HJ, Ehl S, Speckmann C, Grimbacher B, Sediva A, Milota T, Faust SN, Williams AP, Hayman G, Kucuk ZY, Hague R, French P, Brooker R, Forsyth P, Herriot R, Cancrini C, Palma P, Ariganello P, Conlon N, Feighery C, Gavin PJ, Jones A, Imai K, Ibrahim MA, Markelj G, Abinun M, Rieux-Laucat F, Latour S, Pellier I, Fischer A, Touzot F, Casanova JL, Durandy A, Burns SO, Savic S, Kumararatne DS, Moshous D, Kracker S, Vanhaesebroeck B, Okkenhaug K, Picard C, Nejentsev S, Condliffe AM, and Cant AJ
- Subjects
- Adolescent, Adult, Animals, Antibiotic Prophylaxis, Child, Child, Preschool, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, Cohort Studies, Enzyme Inhibitors therapeutic use, Female, Hematopoietic Stem Cell Transplantation, Herpesviridae Infections genetics, Herpesviridae Infections mortality, Herpesviridae Infections therapy, Humans, Immunoglobulins, Intravenous therapeutic use, Immunologic Deficiency Syndromes mortality, Immunologic Deficiency Syndromes therapy, Infant, International Cooperation, Lymphoproliferative Disorders mortality, Lymphoproliferative Disorders therapy, Male, Mice, Middle Aged, Recurrence, Respiratory Tract Infections mortality, Respiratory Tract Infections therapy, Surveys and Questionnaires, Survival Analysis, Young Adult, Class I Phosphatidylinositol 3-Kinases genetics, Immunologic Deficiency Syndromes genetics, Lymphoproliferative Disorders genetics, Mutation genetics, Respiratory Tract Infections genetics
- Abstract
Background: Activated phosphoinositide 3-kinase δ syndrome (APDS) is a recently described combined immunodeficiency resulting from gain-of-function mutations in PIK3CD, the gene encoding the catalytic subunit of phosphoinositide 3-kinase δ (PI3Kδ)., Objective: We sought to review the clinical, immunologic, histopathologic, and radiologic features of APDS in a large genetically defined international cohort., Methods: We applied a clinical questionnaire and performed review of medical notes, radiology, histopathology, and laboratory investigations of 53 patients with APDS., Results: Recurrent sinopulmonary infections (98%) and nonneoplastic lymphoproliferation (75%) were common, often from childhood. Other significant complications included herpesvirus infections (49%), autoinflammatory disease (34%), and lymphoma (13%). Unexpectedly, neurodevelopmental delay occurred in 19% of the cohort, suggesting a role for PI3Kδ in the central nervous system; consistent with this, PI3Kδ is broadly expressed in the developing murine central nervous system. Thoracic imaging revealed high rates of mosaic attenuation (90%) and bronchiectasis (60%). Increased IgM levels (78%), IgG deficiency (43%), and CD4 lymphopenia (84%) were significant immunologic features. No immunologic marker reliably predicted clinical severity, which ranged from asymptomatic to death in early childhood. The majority of patients received immunoglobulin replacement and antibiotic prophylaxis, and 5 patients underwent hematopoietic stem cell transplantation. Five patients died from complications of APDS., Conclusion: APDS is a combined immunodeficiency with multiple clinical manifestations, many with incomplete penetrance and others with variable expressivity. The severity of complications in some patients supports consideration of hematopoietic stem cell transplantation for severe childhood disease. Clinical trials of selective PI3Kδ inhibitors offer new prospects for APDS treatment., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
27. Combined immunodeficiency and Epstein-Barr virus-induced B cell malignancy in humans with inherited CD70 deficiency.
- Author
-
Abolhassani H, Edwards ES, Ikinciogullari A, Jing H, Borte S, Buggert M, Du L, Matsuda-Lennikov M, Romano R, Caridha R, Bade S, Zhang Y, Frederiksen J, Fang M, Bal SK, Haskologlu S, Dogu F, Tacyildiz N, Matthews HF, McElwee JJ, Gostick E, Price DA, Palendira U, Aghamohammadi A, Boisson B, Rezaei N, Karlsson AC, Lenardo MJ, Casanova JL, Hammarström L, Tangye SG, Su HC, and Pan-Hammarström Q
- Subjects
- Adolescent, Adult, CD27 Ligand genetics, CD8-Positive T-Lymphocytes immunology, Child, Cytotoxicity, Immunologic, Female, Herpesvirus 4, Human immunology, Humans, Immunologic Memory, Male, Mutation, Tumor Necrosis Factor Receptor Superfamily, Member 7 physiology, B-Lymphocytes immunology, CD27 Ligand deficiency, Epstein-Barr Virus Infections complications, Hodgkin Disease etiology, Immunologic Deficiency Syndromes complications
- Abstract
In this study, we describe four patients from two unrelated families of different ethnicities with a primary immunodeficiency, predominantly manifesting as susceptibility to Epstein-Barr virus (EBV)-related diseases. Three patients presented with EBV-associated Hodgkin's lymphoma and hypogammaglobulinemia; one also had severe varicella infection. The fourth had viral encephalitis during infancy. Homozygous frameshift or in-frame deletions in CD70 in these patients abolished either CD70 surface expression or binding to its cognate receptor CD27. Blood lymphocyte numbers were normal, but the proportions of memory B cells and EBV-specific effector memory CD8
+ T cells were reduced. Furthermore, although T cell proliferation was normal, in vitro-generated EBV-specific cytotoxic T cell activity was reduced because of CD70 deficiency. This reflected impaired activation by, rather than effects during killing of, EBV-transformed B cells. Notably, expression of 2B4 and NKG2D, receptors implicated in controlling EBV infection, on memory CD8+ T cells from CD70-deficient individuals was reduced, consistent with their impaired killing of EBV-infected cells. Thus, autosomal recessive CD70 deficiency is a novel cause of combined immunodeficiency and EBV-associated diseases, reminiscent of inherited CD27 deficiency. Overall, human CD70-CD27 interactions therefore play a nonredundant role in T and B cell-mediated immunity, especially for protection against EBV and humoral immunity., (© 2017 Abolhassani et al.)- Published
- 2017
- Full Text
- View/download PDF
28. Exome and genome sequencing for inborn errors of immunity.
- Author
-
Meyts I, Bosch B, Bolze A, Boisson B, Itan Y, Belkadi A, Pedergnana V, Moens L, Picard C, Cobat A, Bossuyt X, Abel L, and Casanova JL
- Subjects
- Genetic Variation, High-Throughput Nucleotide Sequencing, Humans, Exome genetics, Genetic Diseases, Inborn, Genome, Human genetics, Immunologic Deficiency Syndromes genetics
- Abstract
The advent of next-generation sequencing (NGS) in 2010 has transformed medicine, particularly the growing field of inborn errors of immunity. NGS has facilitated the discovery of novel disease-causing genes and the genetic diagnosis of patients with monogenic inborn errors of immunity. Whole-exome sequencing (WES) is presently the most cost-effective approach for research and diagnostics, although whole-genome sequencing offers several advantages. The scientific or diagnostic challenge consists in selecting 1 or 2 candidate variants among thousands of NGS calls. Variant- and gene-level computational methods, as well as immunologic hypotheses, can help narrow down this genome-wide search. The key to success is a well-informed genetic hypothesis on 3 key aspects: mode of inheritance, clinical penetrance, and genetic heterogeneity of the condition. This determines the search strategy and selection criteria for candidate alleles. Subsequent functional validation of the disease-causing effect of the candidate variant is critical. Even the most up-to-date dry lab cannot clinch this validation without a seasoned wet lab. The multifariousness of variations entails an experimental rigor even greater than traditional Sanger sequencing-based approaches in order not to assign a condition to an irrelevant variant. Finding the needle in the haystack takes patience, prudence, and discernment., (Copyright © 2016 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
29. Chronic and Invasive Fungal Infections in a Family with CARD9 Deficiency.
- Author
-
Alves de Medeiros AK, Lodewick E, Bogaert DJ, Haerynck F, Van Daele S, Lambrecht B, Bosma S, Vanderdonckt L, Lortholary O, Migaud M, Casanova JL, Puel A, Lanternier F, Lambert J, Brochez L, and Dullaers M
- Subjects
- CARD Signaling Adaptor Proteins deficiency, CARD Signaling Adaptor Proteins immunology, Candida growth & development, Candida pathogenicity, Candidiasis, Chronic Mucocutaneous immunology, Candidiasis, Chronic Mucocutaneous pathology, Child, Consanguinity, Female, Gene Expression, Genes, Recessive, Genetic Predisposition to Disease, Granulocyte-Macrophage Colony-Stimulating Factor genetics, Granulocyte-Macrophage Colony-Stimulating Factor immunology, Homozygote, Humans, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes pathology, Interleukin-17 genetics, Interleukin-17 immunology, Interleukin-6 genetics, Interleukin-6 immunology, Interleukins genetics, Interleukins immunology, Invasive Fungal Infections immunology, Invasive Fungal Infections pathology, Male, Middle Aged, Mutation, Pedigree, T-Lymphocytes, Tinea immunology, Tinea pathology, Trichophyton growth & development, Trichophyton pathogenicity, Turkey, Interleukin-22, CARD Signaling Adaptor Proteins genetics, Candidiasis, Chronic Mucocutaneous genetics, Immunologic Deficiency Syndromes genetics, Invasive Fungal Infections genetics, Tinea genetics
- Abstract
Chronic mucocutaneous or invasive fungal infections are generally the result of primary or secondary immune dysfunction. Patients with autosomal recessive CARD9 mutations are also predisposed to recurrent mucocutaneous and invasive fungal infections with Candida spp., dermatophytes (e.g., Trichophyton spp.) and phaeohyphomycetes (Exophiala spp., Phialophora verrucosa). We study a consanguineous family of Turkish origin in which three members present with distinct clinical phenotypes of chronic mucocutaneous and invasive fungal infections, ranging from chronic mucocutaneous candidiasis (CMC) in one patient, treatment-resistant cutaneous dermatophytosis and deep dermatophytosis in a second patient, to CMC with Candida encephalitis and endocrinopathy in a third patient. Two patients consented to genetic testing and were found to have a previously reported homozygous R70W CARD9 mutation. Circulating IL-17 and IL-22 producing T cells were decreased as was IL-6 and granulocyte/macrophage colony-stimulating factor (GM-CSF) secretion upon stimulation with Candida albicans. Patients with recurrent fungal infections in the absence of known immunodeficiencies should be analyzed for CARD9 gene mutations as the cause of fungal infection predisposition.
- Published
- 2016
- Full Text
- View/download PDF
30. Actin polymerisation after FCγR stimulation of human fibroblasts is BCL10 independent.
- Author
-
Garcia-Gomez S, Alvarez Doforno R, Martinez-Barricarte R, Torres JM, Ferreira Cerdan A, Davila M, Hernández-Jiménez E, Toledano V, Cubillos-Zapata C, Vallejo-Cremades MT, López-Collazo E, Fernández Arquero M, Sánchez-Ramón S, Casanova JL, and Pérez de Diego R
- Subjects
- Adaptor Proteins, Signal Transducing immunology, B-Cell CLL-Lymphoma 10 Protein, CARD Signaling Adaptor Proteins immunology, Case-Control Studies, Caspases immunology, Fibroblasts immunology, Humans, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes metabolism, Microscopy, Fluorescence, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, NF-kappa B immunology, Neoplasm Proteins immunology, Signal Transduction immunology, Actins metabolism, Adaptor Proteins, Signal Transducing genetics, Fibroblasts metabolism, Immunologic Deficiency Syndromes genetics, Polymerization, Receptors, IgG immunology
- Abstract
B-cell lymphoma 10 (BCL10) is not essential for actin polymerisation after FcγR stimulation in human fibroblasts., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
31. Severe infectious diseases of childhood as monogenic inborn errors of immunity.
- Author
-
Casanova JL
- Subjects
- Adolescent, Candidiasis, Chronic Mucocutaneous genetics, Candidiasis, Chronic Mucocutaneous immunology, Child, Complement System Proteins genetics, Encephalitis, Herpes Simplex genetics, Encephalitis, Herpes Simplex immunology, Epidermodysplasia Verruciformis genetics, Epidermodysplasia Verruciformis immunology, Genetic Predisposition to Disease, Humans, Influenza, Human genetics, Influenza, Human immunology, Interferon-gamma genetics, Interferon-gamma immunology, Lymphoproliferative Disorders genetics, Lymphoproliferative Disorders immunology, Malaria genetics, Malaria immunology, Models, Genetic, Models, Immunological, Mycobacterium Infections genetics, Mycobacterium Infections immunology, Neisseria immunology, Neisseria pathogenicity, Pneumococcal Infections genetics, Pneumococcal Infections immunology, Tinea genetics, Tinea immunology, Young Adult, Genetic Diseases, Inborn genetics, Genetic Diseases, Inborn immunology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Infections genetics, Infections immunology
- Abstract
This paper reviews the developments that have occurred in the field of human genetics of infectious diseases from the second half of the 20th century onward. In particular, it stresses and explains the importance of the recently described monogenic inborn errors of immunity underlying resistance or susceptibility to specific infections. The monogenic component of the genetic theory provides a plausible explanation for the occurrence of severe infectious diseases during primary infection. Over the last 20 y, increasing numbers of life-threatening infectious diseases striking otherwise healthy children, adolescents, and even young adults have been attributed to single-gene inborn errors of immunity. These studies were inspired by seminal but neglected findings in plant and animal infections. Infectious diseases typically manifest as sporadic traits because human genotypes often display incomplete penetrance (most genetically predisposed individuals remain healthy) and variable expressivity (different infections can be allelic at the same locus). Infectious diseases of childhood, once thought to be archetypal environmental diseases, actually may be among the most genetically determined conditions of mankind. This nascent and testable notion has interesting medical and biological implications.
- Published
- 2015
- Full Text
- View/download PDF
32. Primary Immunodeficiency Diseases: an Update on the Classification from the International Union of Immunological Societies Expert Committee for Primary Immunodeficiency 2015.
- Author
-
Picard C, Al-Herz W, Bousfiha A, Casanova JL, Chatila T, Conley ME, Cunningham-Rundles C, Etzioni A, Holland SM, Klein C, Nonoyama S, Ochs HD, Oksenhendler E, Puck JM, Sullivan KE, Tang ML, Franco JL, and Gaspar HB
- Subjects
- Autoimmune Diseases genetics, Carcinogenesis genetics, Carcinogenesis immunology, Consensus Development Conferences as Topic, Evidence-Based Medicine, Expert Testimony, Genetic Predisposition to Disease, Humans, Immunity genetics, Immunologic Deficiency Syndromes classification, Infections genetics, International Cooperation, Mutation genetics, Autoimmune Diseases immunology, Immunologic Deficiency Syndromes immunology, Infections immunology
- Abstract
We report the updated classification of primary immunodeficiencies compiled by the Primary Immunodeficiency Expert Committee (PID EC) of the International Union of Immunological Societies (IUIS). In the two years since the previous version, 34 new gene defects are reported in this updated version. For each disorder, the key clinical and laboratory features are provided. In this new version we continue to see the increasing overlap between immunodeficiency, as manifested by infection and/or malignancy, and immune dysregulation, as manifested by auto-inflammation, auto-immunity, and/or allergy. There is also an increased number of genetic defects that lead to susceptibility to specific organisms which reflects the finely tuned nature of immune defense systems. This classification is the most up to date catalogue of all known and published primary immunodeficiencies and acts as a current reference of the knowledge of these conditions and is an important aid for the genetic and molecular diagnosis of patients with these rare diseases.
- Published
- 2015
- Full Text
- View/download PDF
33. The 2015 IUIS Phenotypic Classification for Primary Immunodeficiencies.
- Author
-
Bousfiha A, Jeddane L, Al-Herz W, Ailal F, Casanova JL, Chatila T, Conley ME, Cunningham-Rundles C, Etzioni A, Franco JL, Gaspar HB, Holland SM, Klein C, Nonoyama S, Ochs HD, Oksenhendler E, Picard C, Puck JM, Sullivan KE, and Tang ML
- Subjects
- Autoimmunity, Expert Testimony, Humans, Immunity genetics, Immunologic Deficiency Syndromes classification, Phenotype, Hypersensitivity immunology, Immunologic Deficiency Syndromes immunology, Infections immunology, Inflammation immunology, Neoplasms immunology
- Abstract
There are now nearly 300 single-gene inborn errors of immunity underlying phenotypes as diverse as infection, malignancy, allergy, auto-immunity, and auto-inflammation. For each of these five categories, a growing variety of phenotypes are ascribed to Primary Immunodeficiency Diseases (PID), making PIDs a rapidly expanding field of medicine. The International Union of Immunological Societies (IUIS) PID expert committee (EC) has published every other year a classification of these disorders into tables, defined by shared pathogenesis and/or clinical consequences. In 2013, the IUIS committee also proposed a more user-friendly, phenotypic classification, based on the selection of key phenotypes at the bedside. We herein propose the revised figures, based on the accompanying 2015 IUIS PID EC classification.
- Published
- 2015
- Full Text
- View/download PDF
34. Genetic errors of the human caspase recruitment domain-B-cell lymphoma 10-mucosa-associated lymphoid tissue lymphoma-translocation gene 1 (CBM) complex: Molecular, immunologic, and clinical heterogeneity.
- Author
-
Pérez de Diego R, Sánchez-Ramón S, López-Collazo E, Martínez-Barricarte R, Cubillos-Zapata C, Ferreira Cerdán A, Casanova JL, and Puel A
- Subjects
- Adaptive Immunity, Adaptor Proteins, Signal Transducing metabolism, Animals, B-Cell CLL-Lymphoma 10 Protein, CARD Signaling Adaptor Proteins metabolism, Caspases metabolism, Genetic Predisposition to Disease, Guanylate Cyclase genetics, Humans, Immunity, Innate, Mice, Mice, Knockout, Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein, Mutation genetics, Neoplasm Proteins metabolism, Polymorphism, Genetic, Adaptor Proteins, Signal Transducing genetics, CARD Signaling Adaptor Proteins genetics, Caspases genetics, Immunologic Deficiency Syndromes genetics, Mycoses genetics, Neoplasm Proteins genetics
- Abstract
Three members of the caspase recruitment domain (CARD) family of adaptors (CARD9, CARD10, and CARD11) are known to form heterotrimers with B-cell lymphoma 10 (BCL10) and mucosa-associated lymphoid tissue lymphoma-translocation gene 1 (MALT1). These 3 CARD-BCL10-MALT1 (CBM) complexes activate nuclear factor κB in both the innate and adaptive arms of immunity. Human inherited defects of the 3 components of the CBM complex, including the 2 adaptors CARD9 and CARD11 and the 2 core components BCL10 and MALT1, have recently been reported. Biallelic loss-of-function mutant alleles underlie several different immunologic and clinical phenotypes, which can be assigned to 2 distinct categories. Isolated invasive fungal infections of unclear cellular basis are associated with CARD9 deficiency, whereas a broad range of clinical manifestations, including those characteristic of T- and B-lymphocyte defects, are associated with CARD11, MALT1, and BCL10 deficiencies. Interestingly, human subjects with these mutations have some features in common with the corresponding knockout mice, but other features are different between human subjects and mice. Moreover, germline and somatic gain-of-function mutations of MALT1, BCL10, and CARD11 have also been found in patients with other lymphoproliferative disorders. This broad range of germline and somatic CBM lesions, including loss-of-function and gain-of-function mutations, highlights the contribution of each of the components of the CBM complex to human immunity., (Copyright © 2015 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Monogenic mutations differentially affect the quantity and quality of T follicular helper cells in patients with human primary immunodeficiencies.
- Author
-
Ma CS, Wong N, Rao G, Avery DT, Torpy J, Hambridge T, Bustamante J, Okada S, Stoddard JL, Deenick EK, Pelham SJ, Payne K, Boisson-Dupuis S, Puel A, Kobayashi M, Arkwright PD, Kilic SS, El Baghdadi J, Nonoyama S, Minegishi Y, Mahdaviani SA, Mansouri D, Bousfiha A, Blincoe AK, French MA, Hsu P, Campbell DE, Stormon MO, Wong M, Adelstein S, Smart JM, Fulcher DA, Cook MC, Phan TG, Stepensky P, Boztug K, Kansu A, İkincioğullari A, Baumann U, Beier R, Roscioli T, Ziegler JB, Gray P, Picard C, Grimbacher B, Warnatz K, Holland SM, Casanova JL, Uzel G, and Tangye SG
- Subjects
- 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, Immunologic Deficiency Syndromes immunology, T-Lymphocyte Subsets immunology, T-Lymphocytes, Helper-Inducer immunology
- 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., Competing Interests: The authors declare no conflicts of interest, (Copyright © 2015 American Academy of Allergy, Asthma & Immunology. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. A homozygous mutation of RTEL1 in a child presenting with an apparently isolated natural killer cell deficiency.
- Author
-
Hanna S, Béziat V, Jouanguy E, Casanova JL, and Etzioni A
- Subjects
- Female, Humans, Infant, DNA Helicases genetics, Genetic Diseases, Inborn genetics, Homozygote, Immunologic Deficiency Syndromes genetics, Killer Cells, Natural, Mutation
- Published
- 2015
- Full Text
- View/download PDF
37. Editorial for JoCI.
- Author
-
Bonagura VR and Casanova JL
- Subjects
- Humans, International Cooperation, Publishing trends, Education, Medical, Continuing, Immunologic Deficiency Syndromes therapy
- Published
- 2015
- Full Text
- View/download PDF
38. Inherited DOCK2 Deficiency in Patients with Early-Onset Invasive Infections.
- Author
-
Dobbs K, Domínguez Conde C, Zhang SY, Parolini S, Audry M, Chou J, Haapaniemi E, Keles S, Bilic I, Okada S, Massaad MJ, Rounioja S, Alwahadneh AM, Serwas NK, Capuder K, Çiftçi E, Felgentreff K, Ohsumi TK, Pedergnana V, Boisson B, Haskoloğlu Ş, Ensari A, Schuster M, Moretta A, Itan Y, Patrizi O, Rozenberg F, Lebon P, Saarela J, Knip M, Petrovski S, Goldstein DB, Parrott RE, Savas B, Schambach A, Tabellini G, Bock C, Chatila TA, Comeau AM, Geha RS, Abel L, Buckley RH, İkincioğulları A, Al-Herz W, Helminen M, Doğu F, Casanova JL, Boztuğ K, and Notarangelo LD
- Subjects
- B-Lymphocytes immunology, B-Lymphocytes metabolism, Child, Preschool, Fatal Outcome, Female, GTPase-Activating Proteins, Genes, Recessive, Genetic Diseases, Inborn therapy, Guanine Nucleotide Exchange Factors deficiency, Hematopoietic Stem Cell Transplantation, Humans, Immunologic Deficiency Syndromes therapy, Infant, Killer Cells, Natural immunology, Male, Pedigree, T-Lymphocytes metabolism, rac1 GTP-Binding Protein metabolism, Genetic Diseases, Inborn genetics, Guanine Nucleotide Exchange Factors genetics, Immunologic Deficiency Syndromes genetics, Mutation, T-Lymphocytes immunology
- Abstract
Background Combined immunodeficiencies are marked by inborn errors of T-cell immunity in which the T cells that are present are quantitatively or functionally deficient. Impaired humoral immunity is also common. Patients have severe infections, autoimmunity, or both. The specific molecular, cellular, and clinical features of many types of combined immunodeficiencies remain unknown. Methods We performed genetic and cellular immunologic studies involving five unrelated children with early-onset invasive bacterial and viral infections, lymphopenia, and defective T-cell, B-cell, and natural killer (NK)-cell responses. Two patients died early in childhood; after allogeneic hematopoietic stem-cell transplantation, the other three had normalization of T-cell function and clinical improvement. Results We identified biallelic mutations in the dedicator of cytokinesis 2 gene (DOCK2) in these five patients. RAC1 activation was impaired in the T cells. Chemokine-induced migration and actin polymerization were defective in the T cells, B cells, and NK cells. NK-cell degranulation was also affected. Interferon-α and interferon-λ production by peripheral-blood mononuclear cells was diminished after viral infection. Moreover, in DOCK2-deficient fibroblasts, viral replication was increased and virus-induced cell death was enhanced; these conditions were normalized by treatment with interferon alfa-2b or after expression of wild-type DOCK2. Conclusions Autosomal recessive DOCK2 deficiency is a new mendelian disorder with pleiotropic defects of hematopoietic and nonhematopoietic immunity. Children with clinical features of combined immunodeficiencies, especially with early-onset, invasive infections, may have this condition. (Supported by the National Institutes of Health and others.).
- Published
- 2015
- Full Text
- View/download PDF
39. Deficiency of interleukin-1 receptor-associated kinase 4 presenting as fatal Pseudomonas aeruginosa bacteremia in two siblings.
- Author
-
Stergiopoulou T, Walsh TJ, Seghaye MC, Netea MG, Casanova JL, Moutschen M, and Picard C
- Subjects
- Fatal Outcome, Humans, Infant, Interleukin-1 Receptor-Associated Kinases, Male, Primary Immunodeficiency Diseases, Pseudomonas Infections diagnosis, Pseudomonas Infections metabolism, Bacteremia, Immunologic Deficiency Syndromes, Pseudomonas Infections genetics, Pseudomonas Infections microbiology, Pseudomonas aeruginosa, Siblings
- Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK-4) deficiency is a primary immunodeficiency of innate immunity. This is the case of a previous healthy toddler and his sibling, who both died of fulminant sepsis due to Pseudomonas aeruginosa. Subsequent genetic analysis demonstrated IRAK-4 deficiency with compound heterozygous splice mutations. Fulminant fatal P. aeruginosa sepsis may be the first manifestation of IRAK-4 deficiency.
- Published
- 2015
- Full Text
- View/download PDF
40. Inherited and acquired immunodeficiencies underlying tuberculosis in childhood.
- Author
-
Boisson-Dupuis S, Bustamante J, El-Baghdadi J, Camcioglu Y, Parvaneh N, El Azbaoui S, Agader A, Hassani A, El Hafidi N, Mrani NA, Jouhadi Z, Ailal F, Najib J, Reisli I, Zamani A, Yosunkaya S, Gulle-Girit S, Yildiran A, Cipe FE, Torun SH, Metin A, Atikan BY, Hatipoglu N, Aydogmus C, Kilic SS, Dogu F, Karaca N, Aksu G, Kutukculer N, Keser-Emiroglu M, Somer A, Tanir G, Aytekin C, Adimi P, Mahdaviani SA, Mamishi S, Bousfiha A, Sanal O, Mansouri D, Casanova JL, and Abel L
- Subjects
- Age Factors, Child, Genes, Dominant, Genes, Recessive, Humans, Immunologic Deficiency Syndromes diagnosis, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Disease Susceptibility immunology, Genetic Predisposition to Disease, Immunocompromised Host, Immunologic Deficiency Syndromes complications, Immunologic Deficiency Syndromes etiology, Mycobacterium tuberculosis immunology, Tuberculosis etiology
- 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., (© 2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
41. Immunological loss-of-function due to genetic gain-of-function in humans: autosomal dominance of the third kind.
- Author
-
Boisson B, Quartier P, and Casanova JL
- Subjects
- Animals, Autoimmunity genetics, Autoimmunity immunology, Genes, Dominant, Humans, Hypersensitivity genetics, Hypersensitivity immunology, Hypersensitivity metabolism, Immunologic Deficiency Syndromes metabolism, Infections genetics, Infections immunology, Infections metabolism, Inflammation genetics, Inflammation immunology, Inflammation metabolism, Genetic Association Studies, Genetic Predisposition to Disease, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology
- Abstract
All the human primary immunodeficiencies (PIDs) recognized as such in the 1950s were Mendelian traits and, whether autosomal or X-linked, displayed recessive inheritance. The first autosomal dominant (AD) PID, hereditary angioedema, was recognized in 1963. However, since the first identification of autosomal recessive (AR), X-linked recessive (XR) and AD PID-causing genes in 1985 (ADA; severe combined immunodeficiency), 1986 (CYBB, chronic granulomatous disease) and 1989 (SERPING1; hereditary angioedema), respectively, the number of genetically defined AD PIDs has increased more rapidly than that of any other type of PID. AD PIDs now account for 61 of the 260 known conditions (23%). All known AR PIDs are caused by alleles with some loss-of-function (LOF). A single XR PID is caused by gain-of-function (GOF) mutations (WASP-related neutropenia, 2001). In contrast, only 44 of 61 AD defects are caused by LOF alleles, which exert dominance by haploinsufficiency or negative dominance. Since 2003, up to 17 AD disorders of the third kind, due to GOF alleles, have been described. Remarkably, six of the 17 genes concerned also harbor monoallelic (STAT3), biallelic (C3, CFB, CARD11, PIK3R1) or both monoallelic and biallelic (STAT1) LOF alleles in patients with other clinical phenotypes. Most heterozygous GOF alleles result in auto-inflammation, auto-immunity, or both, with a wide range of immunological and clinical forms. Some also underlie infections and, fewer, allergies, by impairing or enhancing immunity to non-self. Malignancies are also rare. The enormous diversity of immunological and clinical phenotypes is thought provoking and mirrors the diversity and pleiotropy of the underlying genotypes. These experiments of nature provide a unique insight into the quantitative regulation of human immunity., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
42. Phenotypic complementation of genetic immunodeficiency by chronic herpesvirus infection.
- Author
-
MacDuff DA, Reese TA, Kimmey JM, Weiss LA, Song C, Zhang X, Kambal A, Duan E, Carrero JA, Boisson B, Laplantine E, Israel A, Picard C, Colonna M, Edelson BT, Sibley LD, Stallings CL, Casanova JL, Iwai K, and Virgin HW
- Subjects
- Acute Disease, Animals, Bone Marrow Cells cytology, Caspase 1 metabolism, Cell Compartmentation, Chronic Disease, Citrobacter physiology, Cytokines biosynthesis, Genetic Complementation Test, Herpesviridae Infections virology, Humans, Immunity, Innate, Inflammation pathology, Inflammation Mediators metabolism, Interleukin-6 metabolism, Listeria monocytogenes physiology, Listeriosis immunology, Listeriosis microbiology, Listeriosis pathology, Macrophages metabolism, Mice, Mice, Knockout, Mycobacterium tuberculosis physiology, Phenotype, Rhadinovirus physiology, Toxoplasma, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Herpesviridae physiology, Herpesviridae Infections immunology, Herpesviridae Infections pathology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes virology
- Abstract
Variation in the presentation of hereditary immunodeficiencies may be explained by genetic or environmental factors. Patients with mutations in HOIL1 (RBCK1) present with amylopectinosis-associated myopathy with or without hyper-inflammation and immunodeficiency. We report that barrier-raised HOIL-1-deficient mice exhibit amylopectin-like deposits in the myocardium but show minimal signs of hyper-inflammation. However, they show immunodeficiency upon acute infection with Listeria monocytogenes, Toxoplasma gondii or Citrobacter rodentium. Increased susceptibility to Listeria was due to HOIL-1 function in hematopoietic cells and macrophages in production of protective cytokines. In contrast, HOIL-1-deficient mice showed enhanced control of chronic Mycobacterium tuberculosis or murine γ-herpesvirus 68 (MHV68), and these infections conferred a hyper-inflammatory phenotype. Surprisingly, chronic infection with MHV68 complemented the immunodeficiency of HOIL-1, IL-6, Caspase-1 and Caspase-1;Caspase-11-deficient mice following Listeria infection. Thus chronic herpesvirus infection generates signs of auto-inflammation and complements genetic immunodeficiency in mutant mice, highlighting the importance of accounting for the virome in genotype-phenotype studies.
- Published
- 2015
- Full Text
- View/download PDF
43. IRAK-4 and MyD88 deficiencies impair IgM responses against T-independent bacterial antigens.
- Author
-
Maglione PJ, Simchoni N, Black S, Radigan L, Overbey JR, Bagiella E, Bussel JB, Bossuyt X, Casanova JL, Meyts I, Cerutti A, Picard C, and Cunningham-Rundles C
- Subjects
- Adolescent, Adult, Animals, Antibodies, Bacterial genetics, B-Lymphocytes pathology, Bacterial Infections genetics, Bacterial Infections pathology, Cells, Cultured, Child, Child, Preschool, Female, Humans, Immunoglobulin G immunology, Immunoglobulin M genetics, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes pathology, Infant, Interleukin-1 Receptor-Associated Kinases genetics, Interleukin-1 Receptor-Associated Kinases immunology, Male, Middle Aged, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 immunology, Primary Immunodeficiency Diseases, Toll-Like Receptor 7 genetics, Toll-Like Receptor 7 immunology, Toll-Like Receptor 9 genetics, Toll-Like Receptor 9 immunology, Antibodies, Bacterial immunology, Antigens, Bacterial immunology, B-Lymphocytes immunology, Bacterial Infections immunology, Immunoglobulin M immunology, Immunologic Deficiency Syndromes immunology, Polysaccharides, Bacterial immunology
- Abstract
IRAK-4 and MyD88 deficiencies impair interleukin 1 receptor and Toll-like receptor (TLR) signaling and lead to heightened susceptibility to invasive bacterial infections. Individuals with these primary immunodeficiencies have fewer immunoglobulin M (IgM)(+)IgD(+)CD27(+) B cells, a population that resembles murine splenic marginal zone B cells that mount T-independent antibody responses against bacterial antigens. However, the significance of this B-cell subset in humans is poorly understood. Using both a 610 carbohydrate array and enzyme-linked immunosorbent assay, we found that patients with IRAK-4 and MyD88 deficiencies have reduced serum IgM, but not IgG antibody, recognizing T-independent bacterial antigens. Moreover, the quantity of specific IgM correlated with IgM(+)IgD(+)CD27(+) B-cell frequencies. As with mouse marginal zone B cells, human IgM(+)CD27(+) B cells activated by TLR7 or TLR9 agonists produced phosphorylcholine-specific IgM. Further linking splenic IgM(+)IgD(+)CD27(+) B cells with production of T-independent IgM, serum from splenectomized subjects, who also have few IgM(+)IgD(+)CD27(+) B cells, had reduced antibacterial IgM. IRAK-4 and MyD88 deficiencies impaired TLR-induced proliferation of this B-cell subset, suggesting a means by which loss of this activation pathway leads to reduced cell numbers. Thus, by bolstering the IgM(+)IgD(+)CD27(+) B-cell subset, IRAK-4 and MyD88 promote optimal T-independent IgM antibody responses against bacteria in humans., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
44. Inherited BCL10 deficiency impairs hematopoietic and nonhematopoietic immunity.
- Author
-
Torres JM, Martinez-Barricarte R, García-Gómez S, Mazariegos MS, Itan Y, Boisson B, Rholvarez R, Jiménez-Reinoso A, del Pino L, Rodríguez-Pena R, Ferreira A, Hernández-Jiménez E, Toledano V, Cubillos-Zapata C, Díaz-Almirón M, López-Collazo E, Unzueta-Roch JL, Sánchez-Ramón S, Regueiro JR, López-Granados E, Casanova JL, and Pérez de Diego R
- Subjects
- Animals, B-Cell CLL-Lymphoma 10 Protein, B-Lymphocytes immunology, B-Lymphocytes pathology, Child, Preschool, Disease Models, Animal, Fibroblasts immunology, Fibroblasts pathology, Humans, Mice, Myeloid Cells immunology, Myeloid Cells pathology, NF-kappa B genetics, NF-kappa B immunology, T-Lymphocytes immunology, T-Lymphocytes pathology, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing immunology, Gene Expression Regulation genetics, Gene Expression Regulation immunology, Genetic Diseases, Inborn genetics, Genetic Diseases, Inborn immunology, Genetic Diseases, Inborn pathology, Hematopoiesis genetics, Hematopoiesis immunology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes pathology
- Abstract
Heterotrimers composed of B cell CLL/lymphoma 10 (BCL10), mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1), and caspase recruitment domain-containing (CARD) family adaptors play a role in NF-κB activation and have been shown to be involved in both the innate and the adaptive arms of immunity in murine models. Moreover, individuals with inherited defects of MALT1, CARD9, and CARD11 present with immunological and clinical phenotypes. Here, we characterized a case of autosomal-recessive, complete BCL10 deficiency in a child with a broad immunodeficiency, including defects of both hematopoietic and nonhematopoietic immunity. The patient died at 3 years of age and was homozygous for a loss-of-expression, loss-of-function BCL10 mutation. The effect of BCL10 deficiency was dependent on the signaling pathway, and, for some pathways, the cell type affected. Despite the noted similarities to BCL10 deficiency in mice, including a deficient adaptive immune response, human BCL10 deficiency in this patient resulted in a number of specific features within cell populations. Treatment of the patient's myeloid cells with a variety of pathogen-associated molecular pattern molecules (PAMPs) elicited a normal response; however, NF-κB-mediated fibroblast functions were dramatically impaired. The results of this study indicate that inherited BCL10 deficiency should be considered in patients with combined immunodeficiency with B cell, T cell, and fibroblast defects.
- Published
- 2014
- Full Text
- View/download PDF
45. A narrow repertoire of transcriptional modules responsive to pyogenic bacteria is impaired in patients carrying loss-of-function mutations in MYD88 or IRAK4.
- Author
-
Alsina L, Israelsson E, Altman MC, Dang KK, Ghandil P, Israel L, von Bernuth H, Baldwin N, Qin H, Jin Z, Banchereau R, Anguiano E, Ionan A, Abel L, Puel A, Picard C, Pascual V, Casanova JL, and Chaussabel D
- Subjects
- Adolescent, Child, Child, Preschool, Cluster Analysis, Female, Gene Expression Profiling, Humans, Immunity, Innate genetics, Immunity, Innate immunology, Infant, Interleukin-1 Receptor-Associated Kinases immunology, Male, Oligonucleotide Array Sequence Analysis, Primary Immunodeficiency Diseases, Transcriptome, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Interleukin-1 Receptor-Associated Kinases genetics, Mutation, Myeloid Differentiation Factor 88 genetics
- Abstract
Loss of function of the kinase IRAK4 or the adaptor MyD88 in humans interrupts a pathway critical for pathogen sensing and ignition of inflammation. However, patients with loss-of-function mutations in the genes encoding these factors are, unexpectedly, susceptible to only a limited range of pathogens. We employed a systems approach to investigate transcriptome responses following in vitro exposure of patients' blood to agonists of Toll-like receptors (TLRs) and receptors for interleukin 1 (IL-1Rs) and to whole pathogens. Responses to purified agonists were globally abolished, but variable residual responses were present following exposure to whole pathogens. Further delineation of the latter responses identified a narrow repertoire of transcriptional programs affected by loss of MyD88 function or IRAK4 function. Our work introduces the use of a systems approach for the global assessment of innate immune responses and the characterization of human primary immunodeficiencies.
- Published
- 2014
- Full Text
- View/download PDF
46. Mycobacterium simiae infection in two unrelated patients with different forms of inherited IFN-γR2 deficiency.
- Author
-
Martínez-Barricarte R, Megged O, Stepensky P, Casimir P, Moncada-Velez M, Averbuch D, Assous MV, Abuzaitoun O, Kong XF, Pedergnana V, Deswarte C, Migaud M, Rose-John S, Itan Y, Boisson B, Belkadi A, Conti F, Abel L, Vogt G, Boisson-Dupuis S, Casanova JL, and Bustamante J
- Subjects
- Child, Preschool, Fatal Outcome, Female, Genetic Predisposition to Disease, Humans, Immunologic Deficiency Syndromes immunology, Male, Mycobacterium Infections immunology, Mycobacterium Infections mortality, Immunologic Deficiency Syndromes complications, Immunologic Deficiency Syndromes genetics, Mycobacterium Infections complications, Mycobacterium Infections pathology, Receptors, Interferon deficiency, Receptors, Interferon genetics
- Abstract
Interferon-γ receptor 2 (IFN-γR2) deficiency is a rare primary immunodeficiency characterized by predisposition to infections with weakly virulent mycobacteria, such as environmental mycobacteria and BCG vaccines. We describe here two children with IFN-γR2 deficiency, from unrelated, consanguineous kindreds of Arab and Israeli descent. The first patient was a boy who died at the age of 4.5 years, from recurrent, disseminated disease caused by Mycobacterium simiae. His IFN-γR2 defect was autosomal recessive and complete. The second patient was a girl with multiple disseminated mycobacterial infections, including infection with M. simiae. She died at the age of 5 years, a short time after the transplantation of umbilical cord blood cells from an unrelated donor. Her IFN-γR2 defect was autosomal recessive and partial. Autosomal recessive IFN-γR2 deficiency is life-threatening, even in its partial form, and genetic diagnosis and familial counseling are therefore particularly important for this condition. These two cases are the first of IFN-γR2 deficiency associated with M. simiae infection to be described.
- Published
- 2014
- Full Text
- View/download PDF
47. Guidelines for genetic studies in single patients: lessons from primary immunodeficiencies.
- Author
-
Casanova JL, Conley ME, Seligman SJ, Abel L, and Notarangelo LD
- Subjects
- Genetic Association Studies, Genetic Testing, Guidelines as Topic, Humans, Immunologic Deficiency Syndromes diagnosis, Genetic Research legislation & jurisprudence, Immunologic Deficiency Syndromes genetics
- Abstract
Can genetic and clinical findings made in a single patient be considered sufficient to establish a causal relationship between genotype and phenotype? We report that up to 49 of the 232 monogenic etiologies (21%) of human primary immunodeficiencies (PIDs) were initially reported in single patients. The ability to incriminate single-gene inborn errors in immunodeficient patients results from the relative ease in validating the disease-causing role of the genotype by in-depth mechanistic studies demonstrating the structural and functional consequences of the mutations using blood samples. The candidate genotype can be causally connected to a clinical phenotype using cellular (leukocytes) or molecular (plasma) substrates. The recent advent of next generation sequencing (NGS), with whole exome and whole genome sequencing, induced pluripotent stem cell (iPSC) technology, and gene editing technologies-including in particular the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology-offer new and exciting possibilities for the genetic exploration of single patients not only in hematology and immunology but also in other fields. We propose three criteria for deciding if the clinical and experimental data suffice to establish a causal relationship based on only one case. The patient's candidate genotype must not occur in individuals without the clinical phenotype. Experimental studies must indicate that the genetic variant impairs, destroys, or alters the expression or function of the gene product (or two genetic variants for compound heterozygosity). The causal relationship between the candidate genotype and the clinical phenotype must be confirmed via a relevant cellular phenotype, or by default via a relevant animal phenotype. When supported by satisfaction of rigorous criteria, the report of single patient-based discovery of Mendelian disorders should be encouraged, as it can provide the first step in the understanding of a group of human diseases, thereby revealing crucial pathways underlying physiological and pathological processes., (© 2014 Casanova et al.)
- Published
- 2014
- Full Text
- View/download PDF
48. Inborn errors of metabolism underlying primary immunodeficiencies.
- Author
-
Parvaneh N, Quartier P, Rostami P, Casanova JL, and de Lonlay P
- Subjects
- Energy Metabolism genetics, Genetic Testing, Humans, Immunity genetics, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes therapy, Infant, Newborn, Metabolism, Inborn Errors genetics, Metabolism, Inborn Errors therapy, Molecular Targeted Therapy, Protein Processing, Post-Translational genetics, Immunologic Deficiency Syndromes diagnosis, Metabolism, Inborn Errors diagnosis, Mitochondria metabolism
- Abstract
A number of inborn errors of metabolism (IEM) have been shown to result in predominantly immunologic phenotypes, manifesting in part as inborn errors of immunity. These phenotypes are mostly caused by defects that affect the (i) quality or quantity of essential structural building blocks (e.g., nucleic acids, and amino acids), (ii) cellular energy economy (e.g., glucose metabolism), (iii) post-translational protein modification (e.g., glycosylation) or (iv) mitochondrial function. Presenting as multisystemic defects, they also affect innate or adaptive immunity, or both, and display various types of immune dysregulation. Specific and potentially curative therapies are available for some of these diseases, whereas targeted treatments capable of inducing clinical remission are available for others. We will herein review the pathogenesis, diagnosis, and treatment of primary immunodeficiencies (PIDs) due to underlying metabolic disorders.
- Published
- 2014
- Full Text
- View/download PDF
49. Functional characterization of the human dendritic cell immunodeficiency associated with the IRF8(K108E) mutation.
- Author
-
Salem S, Langlais D, Lefebvre F, Bourque G, Bigley V, Haniffa M, Casanova JL, Burk D, Berghuis A, Butler KM, Leahy TR, Hambleton S, and Gros P
- Subjects
- Amino Acid Substitution, Antigen Presentation genetics, Antigen Presentation immunology, Clonal Anergy genetics, Clonal Anergy immunology, Cord Blood Stem Cell Transplantation, Female, HEK293 Cells, Homozygote, Humans, Immunologic Deficiency Syndromes therapy, Infant, Interferon Regulatory Factors metabolism, Lymphohistiocytosis, Hemophagocytic genetics, Lymphohistiocytosis, Hemophagocytic immunology, Lymphohistiocytosis, Hemophagocytic therapy, Mutant Proteins genetics, Mutant Proteins immunology, Mutant Proteins metabolism, Protein Processing, Post-Translational, Protein Stability, RNA genetics, T-Lymphocyte Subsets immunology, Dendritic Cells immunology, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Interferon Regulatory Factors deficiency, Interferon Regulatory Factors genetics, Mutation, Missense
- Abstract
We have previously reported on a unique patient in whom homozygosity for a mutation at IRF8 (IRF8(K108E)) causes a severe immunodeficiency. Laboratory evaluation revealed a highly unusual myeloid compartment, remarkable for the complete absence of CD141 and CD161 monocytes, absence of CD11c1 conventional dendritic cells (DCs) and CD11c1/CD1231 plasmacytoid DCs, and striking granulocytic hyperplasia. The patient initially presented with severe disseminated mycobacterial and mucocutaneous fungal infections and was ultimately cured by cord blood transplant. Sequencing RNA from the IRF8(K108E) patient's primary blood cells prior to transplant shows not only depletion of IRF8-bound and IRF8-regulated transcriptional targets, in keeping with the distorted composition of the myeloid compartment, but also a paucity of transcripts associated with activated CD41 and CD81 T lymphocytes. This suggests that T cells reared in the absence of a functional antigen-presenting compartment in IRF8(K108E) are anergic. Biochemical characterization of the IRF8(K108E) mutant in vitro shows that loss of the positively charged side chain at K108 causes loss of nuclear localization and loss of transcriptional activity, which is concomitant with decreased protein stability, increased ubiquitination, increased small ubiquitin-like modification, and enhanced proteasomal degradation. These findings provide functional insight into the molecular basis of immunodeficiency associated with loss of IRF8.
- Published
- 2014
- Full Text
- View/download PDF
50. Invasive pneumococcal disease in children can reveal a primary immunodeficiency.
- Author
-
Gaschignard J, Levy C, Chrabieh M, Boisson B, Bost-Bru C, Dauger S, Dubos F, Durand P, Gaudelus J, Gendrel D, Gras Le Guen C, Grimprel E, Guyon G, Jeudy C, Jeziorski E, Leclerc F, Léger PL, Lesage F, Lorrot M, Pellier I, Pinquier D, de Pontual L, Sachs P, Thomas C, Tissières P, Valla FV, Desprez P, Frémeaux-Bacchi V, Varon E, Bossuyt X, Cohen R, Abel L, Casanova JL, Puel A, and Picard C
- Subjects
- Adolescent, Child, Child, Preschool, Disease Susceptibility, Female, France, Humans, Infant, Male, Prospective Studies, Immunologic Deficiency Syndromes complications, Pneumococcal Infections epidemiology, Pneumococcal Infections immunology
- Abstract
Background: About 10% of pediatric patients with invasive pneumococcal disease (IPD) die from the disease. Some primary immunodeficiencies (PIDs) are known to confer predisposition to IPD. However, a systematic search for these PIDs has never been carried out in children presenting with IPD., Methods: We prospectively identified pediatric cases of IPD requiring hospitalization between 2005 and 2011 in 28 pediatric wards throughout France. IPD was defined as a positive pneumococcal culture, polymerase chain reaction result, and/or soluble antigen detection at a normally sterile site. The immunological assessment included abdominal ultrasound, whole-blood counts and smears, determinations of plasma immunoglobulin and complement levels, and the evaluation of proinflammatory cytokines., Results: We included 163 children with IPD (male-to-female ratio, 1.3; median age, 13 months). Seventeen children had recurrent IPD. Meningitis was the most frequent type of infection (87%); other infections included pleuropneumonitis, isolated bloodstream infection, osteomyelitis, endocarditis, and mastoiditis. One patient with recurrent meningitis had a congenital cerebrospinal fluid fistula. The results of immunological explorations were abnormal in 26 children (16%), and a PID was identified in 17 patients (10%), including 1 case of MyD88 deficiency, 3 of complement fraction C2 or C3 deficiencies, 1 of isolated congenital asplenia, and 2 of Bruton disease (X-linked agammaglobulinemia). The proportion of PIDs was much higher in children aged >2 years than in younger children (26% vs 3%; P < .001)., Conclusions: Children with IPD should undergo immunological investigations, particularly those aged >2 years, as PIDs may be discovered in up to 26% of cases., (© The Author 2014. Published by Oxford University Press on behalf of the Infectious Diseases Society of America. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.