1
|
Reid W, Romberg N. Inborn Errors of Immunity and Cytokine Storm Syndromes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:185-207. [PMID: 39117816 DOI: 10.1007/978-3-031-59815-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Inborn errors of immunity (IEI) are a diverse and growing category of more than 430 chronic disorders that share susceptibilities to infections. Whether the result of a genetic lesion that causes defective granule-dependent cytotoxicity, excessive lymphoproliferation, or an overwhelming infection represents a unique antigenic challenge, IEIs can display a proclivity for cytokine storm syndrome (CSS) development. This chapter provides an overview of CSS pathophysiology as it relates to IEIs. For each IEI, the immunologic defect and how it promotes or discourages CSS phenomena are reviewed. The IEI-associated molecular defects in pathways that are postulated to be critical to CSS physiology (i.e., toll-like receptors, T regulatory cells, the IL-12/IFNγ axis, IL-6) and, whenever possible, review strategies for treating CSS in IEI patients with molecularly directed therapies are highlighted.
Collapse
Affiliation(s)
- Whitney Reid
- Department of Pediatrics, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Neil Romberg
- Department of Pediatrics, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Worley MJ. Salmonella Bloodstream Infections. Trop Med Infect Dis 2023; 8:487. [PMID: 37999606 PMCID: PMC10675298 DOI: 10.3390/tropicalmed8110487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Salmonella is a major foodborne pathogen of both animals and humans. This bacterium is responsible for considerable morbidity and mortality world-wide. Different serovars of this genus cause diseases ranging from self-limiting gastroenteritis to a potentially fatal systemic disease known as enteric fever. Gastrointestinal infections with Salmonella are usually self-limiting and rarely require medical intervention. Bloodstream infections, on the other hand, are often fatal even with hospitalization. This review describes the routes and underlying mechanisms of the extraintestinal dissemination of Salmonella and the chronic infections that sometimes result. It includes information on the pathogenicity islands and individual virulence factors involved in systemic dissemination as well as a discussion of the host factors that mediate susceptibility. Also, the major outbreaks of invasive Salmonella disease in the tropics are described.
Collapse
Affiliation(s)
- Micah J Worley
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
3
|
Ricci S, Abu-Rumeileh S, Campagna N, Barbati F, Stagi S, Canessa C, Lodi L, Palterer B, Maggi L, Matucci A, Vultaggio A, Annunziato F, Azzari C. Case Report: A child with NFKB1 haploinsufficiency explaining the linkage between immunodeficiency and short stature. Front Immunol 2023; 14:1224603. [PMID: 37600787 PMCID: PMC10434558 DOI: 10.3389/fimmu.2023.1224603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
We report the case of a patient with common variable immunodeficiency (CVID) presenting with short stature and treated with recombinant human growth hormone (rhGH). Whole exome sequencing revealed a novel single-nucleotide duplication in the NFKB1 gene (c.904dup, p.Ser302fs), leading to a frameshift and thus causing NFKB1 haploinsufficiency. The variant was considered pathogenic and was later found in the patient's mother, also affected by CVID. This is the first reported case of a patient with CVID due to NFKB1 mutation presenting with short stature. We analyzed the interconnection between NFKB1 and GH - IGF-1 pathways and we hypothesized a common ground for both CVID and short stature in our patient.
Collapse
Affiliation(s)
- S. Ricci
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Division, Section of Pediatrics, Meyer Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Florence, Italy
| | - S. Abu-Rumeileh
- Department of Health Sciences, University of Florence, Florence, Italy
| | - N. Campagna
- Department of Health Sciences, University of Florence, Florence, Italy
| | - F. Barbati
- Department of Health Sciences, University of Florence, Florence, Italy
| | - S. Stagi
- Department of Health Sciences, University of Florence, Florence, Italy
- Endocrinology Division, Section of Pediatrics, Meyer Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Florence, Italy
| | - C. Canessa
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Division, Section of Pediatrics, Meyer Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Florence, Italy
| | - L. Lodi
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Division, Section of Pediatrics, Meyer Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Florence, Italy
| | - B. Palterer
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - L. Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - A. Matucci
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | - A. Vultaggio
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Immunoallergology Unit, Careggi University Hospital, Florence, Italy
| | - F. Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, Careggi University Hospital, Florence, Italy
| | - C. Azzari
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Division, Section of Pediatrics, Meyer Children’s Hospital Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Florence, Italy
| |
Collapse
|
4
|
Sharfe N, Dalal I, Naghdi Z, Lefaudeux D, Vong L, Dadi H, Navarro H, Tasher D, Ovadia A, Zangen T, Ater D, Ngan B, Hoffmann A, Roifman CM. NFκB pathway dysregulation due to reduced RelB expression leads to severe autoimmune disorders and declining immunity. J Autoimmun 2023; 137:102946. [PMID: 36402602 DOI: 10.1016/j.jaut.2022.102946] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Genetic aberrations in the NFκB pathway lead to primary immunodeficiencies with various degrees of severity. We previously demonstrated that complete ablation of the RelB transcription factor, a key component of the alternative pathway, results in an early manifested combined immunodeficiency requiring stem cell transplantation. OBJECTIVE To study the molecular basis of a progressive severe autoimmunity and immunodeficiency in three patients. METHODS Whole exome sequencing was performed to identify the genetic defect. Molecular and cellular techniques were utilized to assess the variant impact on NFκB signaling, canonical and alternative pathway crosstalk, as well as the resultant effects on immune function. RESULTS Patients presented with multiple autoimmune progressive severe manifestations encompassing the liver, gut, lung, and skin, becoming debilitating in the second decade of life. This was accompanied by a deterioration of the immune system, demonstrating an age-related decline in naïve T cells and responses to mitogens, accompanied by a gradual loss of all circulating CD19+ cells. Whole exome sequencing identified a novel homozygous c. C1091T (P364L) transition in RELB. The P364L RelB protein was unstable, with extremely low expression, but retained some function and could be transiently and partially upregulated following Toll-like receptor stimulation. Stimulation of P364L patient fibroblasts resulted in a marked rise in a cluster of pro-inflammatory hyper-expressed transcripts consistent with the removal of RelB inhibitory effect on RelA function. This is likely the main driver of autoimmune manifestations in these patients. CONCLUSION Incomplete loss of RelB provided a unique opportunity to gain insights into NFκB's pathway interactions as well as the pathogenesis of autoimmunity. The P364L RelB mutation leads to gradual decline in immune function with progression of severe debilitating autoimmunity.
Collapse
Affiliation(s)
- Nigel Sharfe
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Ilan Dalal
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Zahra Naghdi
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Diane Lefaudeux
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Linda Vong
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Harjit Dadi
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada
| | - Hector Navarro
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Diana Tasher
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Ovadia
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tzili Zangen
- Pediatric Department, E. Wolfson Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Ater
- Pediatric Pulmonology Unit, Assuta Medical Center, Tel Aviv, Israel
| | - Bo Ngan
- Department of Laboratory Medicine and Pathobiology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Chaim M Roifman
- The Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Division of Immunology/Allergy, Department of Pediatrics, Hospital for Sick Children, and the University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Of Mycelium and Men: Inherent Human Susceptibility to Fungal Diseases. Pathogens 2023; 12:pathogens12030456. [PMID: 36986378 PMCID: PMC10058615 DOI: 10.3390/pathogens12030456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
In medical mycology, the main context of disease is iatrogenic-based disease. However, historically, and occasionally, even today, fungal diseases affect humans with no obvious risk factors, sometimes in a spectacular fashion. The field of “inborn errors of immunity” (IEI) has deduced at least some of these previously enigmatic cases; accordingly, the discovery of single-gene disorders with penetrant clinical effects and their immunologic dissection have provided a framework with which to understand some of the key pathways mediating human susceptibility to mycoses. By extension, they have also enabled the identification of naturally occurring auto-antibodies to cytokines that phenocopy such susceptibility. This review provides a comprehensive update of IEI and autoantibodies that inherently predispose humans to various fungal diseases.
Collapse
|
6
|
A Novel De Novo NFKBIA Missense Mutation Associated to Ectodermal Dysplasia with Dysgammaglobulinemia. Genes (Basel) 2022; 13:genes13101900. [PMID: 36292785 PMCID: PMC9602067 DOI: 10.3390/genes13101900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/26/2022] Open
Abstract
Background: Inborn errors of immunity (IEIs) are comprised of heterogeneous groups of genetic disorders affecting immune function. In this report, a 17-month-old Malay patient suspected of having Hyper IgM syndrome, a type of IEIs, was described. However, the diagnosis of Hyper IgM syndrome was excluded by the normal functional studies and the mild features of ectodermal dysplasia observed from a further clinical phenotype inspection. Methods: Whole-exome sequencing (WES) was performed to unravel the causative mutation in this patient. Results: The variant analysis demonstrated a novel missense mutation in NFKBIA (NM_020529:c.94A > T,NP_065390:p.Ser32Cys) and was predicted as damaging by in silico prediction tools. The NFKBIA gene encodes for IκBα, a member of nuclear factor kappa B (NF-κB) inhibitors, playing an important role in regulating NF-κB activity. The mutation occurred at the six degrons (Asp31-Ser36) in IκBα which were evolutionarily conserved across several species. Prediction analysis suggested that the substitution of Ser32Cys may cause a loss of the phosphorylation site at residue 32 and a gain of the sumoylation site at residue 38, resulting in the alteration of post-translational modifications of IκBα required for NF-κB activation. Conclusion: Our analysis hints that the post-translational modification in the NFKBIA Ser32Cys mutant would alter the signaling pathway of NF-κB. Our findings support the usefulness of WES in diagnosing IEIs and suggest the role of post-translational modification of IκBα.
Collapse
|
7
|
Gold NB, Harrison SM, Rowe JH, Gold J, Furutani E, Biffi A, Duncan CN, Shimamura A, Lehmann LE, Green RC. Low frequency of treatable pediatric disease alleles in gnomAD: An opportunity for future genomic screening of newborns. HGG ADVANCES 2022; 3:100059. [PMID: 35047849 PMCID: PMC8756496 DOI: 10.1016/j.xhgg.2021.100059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/20/2021] [Indexed: 01/18/2023] Open
Abstract
Hematopoietic stem cell transplant (HSCT) can prevent progression of several genetic disorders. Although a subset of these disorders are identified on newborn screening panels, others are not identified until irreversible symptoms develop. Genetic testing is an efficient methodology to ascertain pre-symptomatic children, but the penetrance of risk-associated variants in the general population is not well understood. We developed a list of 127 genes associated with disorders treatable with HSCT. We identified likely pathogenic or pathogenic (LP/P) and loss-of-function (LoF) variants in these genes in the Genome Aggregation Database (gnomAD), a dataset containing exome and genome sequencing data from 141,456 healthy adults. Within gnomAD, we identified 59 individuals with a LP/P or LoF variant in 15 genes. Genes were associated with bone marrow failure syndromes, bleeding disorders, primary immunodeficiencies, osteopetrosis, metabolic disorders, and epidermolysis bullosa. In conclusion, few ostensibly healthy adults had genotypes associated with pediatric disorders treatable with HSCTs. Given that most of these disorders do not have biomarkers that could be cheaply and universally assessed on a standard newborn screen, our data suggest that genetic testing may be a complementary approach to traditional newborn screening methodology that has the potential to improve mortality and is not expected to lead to a high burden of false-positive results.
Collapse
Affiliation(s)
- Nina B. Gold
- Massachusetts General Hospital for Children, Division of Medical Genetics and Metabolism, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Jared H. Rowe
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Jessica Gold
- Department of Pediatrics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elissa Furutani
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
| | - Alessandra Biffi
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Christine N. Duncan
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Akiko Shimamura
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Leslie E. Lehmann
- Harvard Medical School, Boston, MA, USA
- Boston Children’s Hospital, Division of Hematology and Oncology, Boston, MA, USA
- Dana-Farber Cancer Institute Division of Pediatric Oncology, Boston, MA, USA
| | - Robert C. Green
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital, Boston, MA, USA
- Ariadne Labs, Boston, MA, USA
| |
Collapse
|
8
|
Wen W, Wang L, Deng M, Li Y, Tang X, Mao H, Zhao X. A heterozygous N-terminal truncation mutation of NFKBIA results in an impaired NF-κB dependent inflammatory response. Genes Dis 2022; 9:176-186. [PMID: 35005117 PMCID: PMC8720704 DOI: 10.1016/j.gendis.2021.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/21/2021] [Accepted: 03/24/2021] [Indexed: 11/19/2022] Open
Abstract
Germline heterozygous gain-of-function (GOF) mutation of NFKBIA, encoding IκBα, would affect the activation of NF-κB pathway and cause an autosomal dominant (AD) form of anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID). Here we reported a Chinese patient with a heterozygous N-terminal truncation mutation of NFKBIA/IκBα. She presented recurrent fever, infectious pneumonia and chronic diarrhea with EDA-ID. Impaired NF-κB translocation and IL1R and TLR4 pathway activation were revealed in this patient. The findings suggested that the truncation mutation of IκBα caused medium impaired of activation of NF-κB but the early death. Furthermore, we reviewed all the reported patients with NFKBIA mutation to learn more about this disease.
Collapse
Affiliation(s)
- Wen Wen
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
- Pediatric Research Institute, Chongqing, 400014 PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014 PR China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China
- Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
| | - Li Wang
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Pediatric Research Institute, Chongqing, 400014 PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014 PR China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China
- Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
| | - Mengyue Deng
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
- Pediatric Research Institute, Chongqing, 400014 PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014 PR China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China
- Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
| | - Yue Li
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
- Pediatric Research Institute, Chongqing, 400014 PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014 PR China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China
- Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
| | - Xuemei Tang
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Pediatric Research Institute, Chongqing, 400014 PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014 PR China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China
- Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
| | - Huawei Mao
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Pediatric Research Institute, Chongqing, 400014 PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014 PR China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China
- Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
| | - Xiaodong Zhao
- Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University, Chongqing, 400014 PR China
- Pediatric Research Institute, Chongqing, 400014 PR China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, PR China
- National Clinical Research Center for Child Health and Disorders, Chongqing, 400014 PR China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, PR China
- Children's Hospital of Chongqing Medical University, Chongqing 400014, PR China
| |
Collapse
|
9
|
Aluri J, Cooper MA, Schuettpelz LG. Toll-Like Receptor Signaling in the Establishment and Function of the Immune System. Cells 2021; 10:cells10061374. [PMID: 34199501 PMCID: PMC8228919 DOI: 10.3390/cells10061374] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors that play a central role in the development and function of the immune system. TLR signaling promotes the earliest emergence of hematopoietic cells during development, and thereafter influences the fate and function of both primitive and effector immune cell types. Aberrant TLR signaling is associated with hematopoietic and immune system dysfunction, and both loss- and gain-of- function variants in TLR signaling-associated genes have been linked to specific infection susceptibilities and immune defects. Herein, we will review the role of TLR signaling in immune system development and the growing number of heritable defects in TLR signaling that lead to inborn errors of immunity.
Collapse
|
10
|
Abstract
Acute bacterial gastroenteritis is among the most common infections worldwide, with millions of infections annually in the United States. Much of the illness is foodborne, occurring as both sporadic cases and large multistate outbreaks. Pathogen evolution through genetic exchange of virulence traits and antibiotic resistance determinants poses challenges for empiric therapy. Culture-independent diagnostic tests in clinical laboratories afford rapid diagnosis and expanded identification of pathogens. However, cultures remain important to generate sensitivity data and strain archiving for outbreak investigations. Most infections are self-limited, permitting judicious selection of antibiotic use in more severe forms of illness.
Collapse
Affiliation(s)
- James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine, Campus Box 8051, 660 South Euclid Avenue, Saint Louis, MO 63110, USA; Infectious Disease Section, Medicine Service, Veterans Affairs Saint Louis Health Care System, 915 North Grand Boulevard, Saint Louis, MO 63106, USA.
| | - F Matthew Kuhlmann
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine, Campus Box 8051, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine, Campus Box 8051, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| |
Collapse
|
11
|
Sogkas G, Atschekzei F, Adriawan IR, Dubrowinskaja N, Witte T, Schmidt RE. Cellular and molecular mechanisms breaking immune tolerance in inborn errors of immunity. Cell Mol Immunol 2021; 18:1122-1140. [PMID: 33795850 PMCID: PMC8015752 DOI: 10.1038/s41423-020-00626-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/11/2020] [Indexed: 02/01/2023] Open
Abstract
In addition to susceptibility to infections, conventional primary immunodeficiency disorders (PIDs) and inborn errors of immunity (IEI) can cause immune dysregulation, manifesting as lymphoproliferative and/or autoimmune disease. Autoimmunity can be the prominent phenotype of PIDs and commonly includes cytopenias and rheumatological diseases, such as arthritis, systemic lupus erythematosus (SLE), and Sjogren's syndrome (SjS). Recent advances in understanding the genetic basis of systemic autoimmune diseases and PIDs suggest an at least partially shared genetic background and therefore common pathogenic mechanisms. Here, we explore the interconnected pathogenic pathways of autoimmunity and primary immunodeficiency, highlighting the mechanisms breaking the different layers of immune tolerance to self-antigens in selected IEI.
Collapse
Affiliation(s)
- Georgios Sogkas
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany.
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany.
| | - Faranaz Atschekzei
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Ignatius Ryan Adriawan
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Natalia Dubrowinskaja
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| | - Reinhold Ernst Schmidt
- Department of Rheumatology and Immunology, Hannover Medical School, Hanover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hanover, Germany
| |
Collapse
|
12
|
Tan EE, Hopkins RA, Lim CK, Jamuar SS, Ong C, Thoon KC, Koh MJ, Shin EM, Lian DW, Weerasooriya M, Lee CZ, Soetedjo AAP, Lim CS, Au VB, Chua E, Lee HY, Jones LA, James SS, Kaliaperumal N, Kwok J, Tan ES, Thomas B, Wu LX, Ho L, Fairhurst AM, Ginhoux F, Teo AK, Zhang YL, Ong KH, Yu W, Venkatesh B, Tergaonkar V, Reversade B, Chin KC, Tan AM, Liew WK, Connolly JE. Dominant-negative NFKBIA mutation promotes IL-1β production causing hepatic disease with severe immunodeficiency. J Clin Invest 2021; 130:5817-5832. [PMID: 32750042 DOI: 10.1172/jci98882] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Although IKK-β has previously been shown as a negative regulator of IL-1β secretion in mice, this role has not been proven in humans. Genetic studies of NF-κB signaling in humans with inherited diseases of the immune system have not demonstrated the relevance of the NF-κB pathway in suppressing IL-1β expression. Here, we report an infant with a clinical pathology comprising neutrophil-mediated autoinflammation and recurrent bacterial infections. Whole-exome sequencing revealed a de novo heterozygous missense mutation of NFKBIA, resulting in a L34P IκBα variant that severely repressed NF-κB activation and downstream cytokine production. Paradoxically, IL-1β secretion was elevated in the patient's stimulated leukocytes, in her induced pluripotent stem cell-derived macrophages, and in murine bone marrow-derived macrophages containing the L34P mutation. The patient's hypersecretion of IL-1β correlated with activated neutrophilia and liver fibrosis with neutrophil accumulation. Hematopoietic stem cell transplantation reversed neutrophilia, restored a resting state in neutrophils, and normalized IL-1β release from stimulated leukocytes. Additional therapeutic blockade of IL-1 ameliorated liver damage, while decreasing neutrophil activation and associated IL-1β secretion. Our studies reveal a previously unrecognized role of human IκBα as an essential regulator of canonical NF-κB signaling in the prevention of neutrophil-dependent autoinflammatory diseases. These findings also highlight the therapeutic potential of IL-1 inhibitors in treating complications arising from systemic NF-κB inhibition.
Collapse
Affiliation(s)
- Enrica Ek Tan
- Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Richard A Hopkins
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Chrissie K Lim
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Saumya S Jamuar
- Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Christina Ong
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Koh C Thoon
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Mark Ja Koh
- Duke-NUS Medical School, Singapore.,Dermatology Service, KK Women's and Children's Hospital, Singapore
| | - Eun Mong Shin
- Institute of Molecular and Cell Biology, A*STAR, Singapore.,Cancer Science Institute of Singapore, Singapore.,National University of Singapore, Singapore
| | - Derrick Wq Lian
- Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore.,Duke-NUS Medical School, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Madhushanee Weerasooriya
- Department of Microbiology and Immunology and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | | | | | - Veonice B Au
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Edmond Chua
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Hui Yin Lee
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Leigh Ann Jones
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Sharmy S James
- Department of Microbiology and Immunology and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Nivashini Kaliaperumal
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Jeffery Kwok
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Ee Shien Tan
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Biju Thomas
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - Lynn Xue Wu
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Lena Ho
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | | | - Adrian Kk Teo
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Yong Liang Zhang
- Department of Microbiology and Immunology and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Kok Huar Ong
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Weimiao Yu
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | - Vinay Tergaonkar
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology, A*STAR, Singapore.,Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia.,Faculty of Health Sciences, University of Macau, Macau, China
| | - Bruno Reversade
- Institute of Molecular and Cell Biology, A*STAR, Singapore.,Department of Medical Genetics, School of Medicine, Koç University, Istanbul, Turkey.,Department of Paediatrics, National University of Singapore, Singapore.,Institute of Medical Biology, A*STAR, Singapore
| | - Keh Chuang Chin
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore.,Department of Physiology and
| | | | - Woei Kang Liew
- Duke-NUS Medical School, Singapore.,Department of Paediatrics and
| | - John E Connolly
- Program in Translational Immunology, Institute of Molecular and Cell Biology, A*STAR, Singapore.,Department of Paediatrics and.,Department of Microbiology and Immunity, National University of Singapore, Singapore.,Institute of Biomedical Studies, Baylor University Medical Center, Waco, Texas, USA
| |
Collapse
|
13
|
De Luca F. Regulatory role of NF-κB in growth plate chondrogenesis and its functional interaction with Growth Hormone. Mol Cell Endocrinol 2020; 514:110916. [PMID: 32569858 DOI: 10.1016/j.mce.2020.110916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 11/26/2022]
Abstract
Nuclear Factor kappa B (NF-kB) is a family of transcription factors that participates in the regulation of cell proliferation, migration, and apoptosis. Impaired NF-kB activity appears to be involved in the pathophysiology of inflammatory states, autoimmune diseases, and cancer. Genetic manipulation in mice leading to impaired NF-kB function is associated with abnormal limb development and delayed bone growth. We have previously shown in rodent cultured chondrocytes and cultured metatarsal bones that NF-kB promotes longitudinal bone growth and growth plate chondrocyte function. These NF-kB growth-promoting effects appear to be facilitated by Growth Hormone (GH) and Insulin-like Growth factor-1 (IGF-1). These stimulatory effects of GH and IGF-1 on NF-kB activity are supported by observational evidence in humans; a number of individuals carrying mutations that alter NF-kB function exhibit growth failure and GH insensitivity.
Collapse
Affiliation(s)
- Francesco De Luca
- Division of Endocrinology and Diabetes, Children's Mercy Kansas City, University of Missouri Kansas City-School of Medicine, Kansas City, MO, 64111, USA.
| |
Collapse
|
14
|
Li Y, Yang Q, Shi ZH, Zhou M, Yan L, Li H, Xie YH, Wang SW. The Anti-Inflammatory Effect of Feiyangchangweiyan Capsule and Its Main Components on Pelvic Inflammatory Disease in Rats via the Regulation of the NF- κB and BAX/BCL-2 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:9585727. [PMID: 31312226 PMCID: PMC6595388 DOI: 10.1155/2019/9585727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 04/23/2019] [Indexed: 11/18/2022]
Abstract
Although gastroenteritis and pelvic inflammatory disease (PID) occur in the gastrointestinal tract and pelvis, respectively, they display similar pathogeneses. The incidence of inflammation in these conditions is usually associated with dysbacteriosis, and, at times, they are caused by the same pathogenic bacteria, Escherichia coli and Streptococcus aureus. Feiyangchangweiyan capsule (FYC) is a traditional Chinese patent medicine that is widely used to treat bacterial dysentery and acute and chronic gastroenteritis. However, whether it has an effect on PID is unclear. The aim of this study was to investigate the anti-inflammatory effect of FYC and its main components, gallic acid (GA), ellagic acid (EA), and syringin (SY), on a pathogen-induced PID model and illustrate their potential mechanism of action. Female specific pathogen-free SD rats (n = 1110) were randomly divided into control, PID, FYC, GA, EA, SY, GA + EA, GA + SY, EA + SY, GA + EA + SY, and Fuke Qianjin capsule (FKC) positive groups. Histological examination and enzyme-linked immunosorbent assay (ELISA) were carried out as well as western blot analysis to detect the expression of NF-κB, BAX, BCL-2, and JNK. In this study, FYC and its main components dramatically suppressed the infiltration of inflammatory cells, reduced the production of IL-1β, TNF-α, and MCP-1, and elevated the IL-10 level to varying degrees. We also found that FYC and its main components inhibited the expression of BAX induced by infection and increased the expression of Bcl-2. FYC, GA, EA, and SY could also block the activation of the NF-κB pathway. Finally, we found that the phosphorylation of JNK could be decreased by FYC, GA, and SY. FYC and its main components exhibit anti-inflammatory effect on a pathogen-induced PID model by regulating the NF-κB and apoptosis signaling pathways.
Collapse
Affiliation(s)
- Yao Li
- The College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Qian Yang
- Department of Natural Medicine, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Zhi-hui Shi
- Shaanxi Junbisha Pharmaceutical Limited Company, Xianyang, 712000, China
| | - Min Zhou
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - Li Yan
- Department of Natural Medicine, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hua Li
- Department of Natural Medicine, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yan-hua Xie
- The College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Si-wang Wang
- The College of Life Sciences, Northwest University, Xi'an, 710069, China
- Department of Natural Medicine, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
15
|
Scott O, Roifman CM. NF-κB pathway and the Goldilocks principle: Lessons from human disorders of immunity and inflammation. J Allergy Clin Immunol 2019; 143:1688-1701. [PMID: 30940520 DOI: 10.1016/j.jaci.2019.03.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 01/12/2023]
Abstract
Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) signaling pathways play a key role in various cell processes related to host immunity. The last few years have seen an explosion of disorders associated with NF-κB components from core members of the canonical and noncanonical cascades to adaptor protein and ubiquitination-related enzymes. Disease phenotypes have extended beyond susceptibility to infections and include autoimmunity, lymphoproliferation, atopy, and inflammation. Concurrently, studies are unveiling a tightly regulated system marked by extensive cross-talk between the canonical and noncanonical pathways, as well as among the NF-κB and other signaling pathways. As the rate of discovery in the realm of NF-κB defects accelerates, this review presents a timely summary of major known defects causing human disease, as well as diagnostic, therapeutic, and research challenges and opportunities.
Collapse
Affiliation(s)
- Ori Scott
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada
| | - Chaim M Roifman
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children.
| |
Collapse
|
16
|
Hart M, Walch-Rückheim B, Friedmann KS, Rheinheimer S, Tänzer T, Glombitza B, Sester M, Lenhof HP, Hoth M, Schwarz EC, Keller A, Meese E. miR-34a: a new player in the regulation of T cell function by modulation of NF-κB signaling. Cell Death Dis 2019; 10:46. [PMID: 30718475 PMCID: PMC6362007 DOI: 10.1038/s41419-018-1295-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 12/13/2018] [Accepted: 12/18/2018] [Indexed: 12/15/2022]
Abstract
NF-κB functions as modulator of T cell receptor-mediated signaling and transcriptional regulator of miR-34a. Our in silico analysis revealed that miR-34a impacts the NF-κB signalosome with miR-34a binding sites in 14 key members of the NF-κB signaling pathway. Functional analysis identified five target genes of miR-34a including PLCG1, CD3E, PIK3CB, TAB2, and NFΚBIA. Overexpression of miR-34a in CD4+ and CD8+ T cells led to a significant decrease of NFΚBIA as the most downstream cytoplasmic NF-κB member, a reduced cell surface abundance of TCRA and CD3E, and to a reduction of T cell killing capacity. Inhibition of miR-34a caused an increase of NFΚBIA, TCRA, and CD3E. Notably, activation of CD4+ and CD8+ T cells entrails a gradual increase of miR-34a. Our results lend further support to a model with miR-34a as a central NF-κB regulator in T cells.
Collapse
Affiliation(s)
- Martin Hart
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany.
| | - Barbara Walch-Rückheim
- Institute of Virology and Center of Human and Molecular Biology, Saarland University Medical School, 66421, Homburg, Germany
| | - Kim S Friedmann
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | | | - Tanja Tänzer
- Institute of Virology and Center of Human and Molecular Biology, Saarland University Medical School, 66421, Homburg, Germany
| | - Birgit Glombitza
- Institute of Virology and Center of Human and Molecular Biology, Saarland University Medical School, 66421, Homburg, Germany
| | - Martina Sester
- Department of Transplant and Infection Immunology, Saarland University, 66421, Homburg, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Markus Hoth
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | | | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany
| |
Collapse
|
17
|
Yazdani R, Fekrvand S, Shahkarami S, Azizi G, Moazzami B, Abolhassani H, Aghamohammadi A. The hyper IgM syndromes: Epidemiology, pathogenesis, clinical manifestations, diagnosis and management. Clin Immunol 2018; 198:19-30. [PMID: 30439505 DOI: 10.1016/j.clim.2018.11.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 11/11/2018] [Indexed: 12/17/2022]
Abstract
Hyper Immunoglobulin M syndrome (HIGM) is a rare primary immunodeficiency disorder characterized by low or absent levels of serum IgG, IgA, IgE and normal or increased levels of serum IgM. Various X-linked and autosomal recessive/dominant mutations have been reported as the underlying cause of the disease. Based on the underlying genetic defect, the affected patients present a variety of clinical manifestations including pulmonary and gastrointestinal complications, autoimmune disorders, hematologic abnormalities, lymphoproliferation and malignancies which could be controlled by multiple relevant therapeutic approaches. Herein, the epidemiology, pathogenesis, clinical manifestations, diagnosis, management, prognosis and treatment in patients with HIGM syndrome have been reviewed.
Collapse
Affiliation(s)
- Reza Yazdani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Sepideh Shahkarami
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Bobak Moazzami
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
18
|
Cuvelier GDE, Rubin TS, Junker A, Sinha R, Rosenberg AM, Wall DA, Schroeder ML. Clinical presentation, immunologic features, and hematopoietic stem cell transplant outcomes for IKBKB immune deficiency. Clin Immunol 2018; 205:138-147. [PMID: 30391351 PMCID: PMC7106064 DOI: 10.1016/j.clim.2018.10.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/10/2018] [Accepted: 10/30/2018] [Indexed: 11/24/2022]
Abstract
IKBKB immune deficiency is a rare but life-threatening primary immunodeficiency disorder, involving activation defects in adaptive and innate immunity. We present sixteen cases of a homozygous IKBKB mutation (c.1292dupG) in infants characterized by early-onset bacterial, viral, fungal and Mycobacterial infections. In most cases, T- and B-cells were quantitatively normal, but phenotypically naïve, with severe hypogammaglobulinemia. T-cell receptor excision circles were normal, meaning newborn screening by TREC analysis would miss IKBKB cases. Although IKBKB immune deficiency does not meet traditional laboratory based definitions for SCID, this combined immune deficiency appears to be at least as profound. Urgent HSCT, performed in eight patients, remains the only known curative therapy, although only three patients are survivors. Ongoing infections after transplant remain a concern, and may be due to combinations of poor social determinants of health, secondary graft failure, and failure of HSCT to replace non-hematopoietic cells important in immune function and dependent upon IKK/NF-κB pathways.
Collapse
Affiliation(s)
- Geoffrey D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, Division of Pediatric Hematology-Oncology-BMT, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Tamar S Rubin
- Winnipeg Children's Hospital, Division of Pediatric Allergy and Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Anne Junker
- British Columbia Children's Hospital, Division of Clinical Immunology and Allergy, University of British Columbia, Vancouver, BC, Canada
| | - Roona Sinha
- Royal University Hospital, Division of Pediatric Hematology-Oncology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Alan M Rosenberg
- Royal University Hospital, Department of Pediatrics, University of Saskatchewan, Saskatoon, SK, Canada
| | - Donna A Wall
- Hospital for Sick Children, Pediatric Blood and Marrow Transplant, University of Toronto, Toronto, ON, Canada
| | - Marlis L Schroeder
- Manitoba Blood and Marrow Transplant Program, CancerCare Manitoba, Division of Pediatric Hematology-Oncology-BMT, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
19
|
Teku GN, Vihinen M. Simulation of the Dynamics of Primary Immunodeficiencies in B Cells. Front Immunol 2018; 9:1785. [PMID: 30116248 PMCID: PMC6082931 DOI: 10.3389/fimmu.2018.01785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022] Open
Abstract
Primary immunodeficiencies (PIDs) are a group of over 300 hereditary, heterogeneous, and mainly rare disorders that affect the immune system. Various aspects of immune system and PID proteins and genes have been investigated and facilitate systems biological studies of effects of PIDs on B cell physiology and response. We reconstructed a B cell network model based on data for the core B cell receptor activation and response processes and performed semi-quantitative dynamic simulations for normal and B cell PID failure modes. The results for several knockout simulations correspond to previously reported molecular studies and reveal novel mechanisms for PIDs. The simulations for CD21, CD40, LYN, MS4A1, ORAI1, PLCG2, PTPRC, and STIM1 indicated profound changes to major transcription factor signaling and to the network. Significant effects were observed also in the BCL10, BLNK, BTK, loss-of-function CARD11, IKKB, MALT1, and NEMO, simulations whereas only minor effects were detected for PIDs that are caused by constitutively active proteins (PI3K, gain-of-function CARD11, KRAS, and NFKBIA). This study revealed the underlying dynamics of PID diseases, confirms previous observations, and identifies novel candidates for PID diagnostics and therapy.
Collapse
Affiliation(s)
| | - Mauno Vihinen
- Department of Experimental Medical Science, BMC B13, Lund University, Lund, Sweden
| |
Collapse
|
20
|
Villafuerte B, Natera-de-Benito D, González A, Mori MA, Palomares M, Nevado J, García-Miñaur S, Lapunzina P, González-Granado LI, Allende LM, Moreno JC. The Brain-Lung-Thyroid syndrome (BLTS): A novel deletion in chromosome 14q13.2-q21.1 expands the phenotype to humoral immunodeficiency. Eur J Med Genet 2018; 61:393-398. [DOI: 10.1016/j.ejmg.2018.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/13/2017] [Accepted: 02/17/2018] [Indexed: 12/11/2022]
|
21
|
Zhu F, Hu Y. Integrity of IKK/NF-κB Shields Thymic Stroma That Suppresses Susceptibility to Autoimmunity, Fungal Infection, and Carcinogenesis. Bioessays 2018. [PMID: 29522649 DOI: 10.1002/bies.201700131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A pathogenic connection between autoreactive T cells, fungal infection, and carcinogenesis has been demonstrated in studies of human autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) as well as in a mouse model in which kinase-dead Ikkα knock-in mice develop impaired central tolerance, autoreactive T cell-mediated autoimmunity, chronic fungal infection, and esophageal squamous cell carcinoma, which recapitulates APECED. IκB kinase α (IKKα) is one subunit of the IKK complex required for NF-κB activation. IKK/NF-κB is essential for central tolerance establishment by regulating the development of medullary thymic epithelial cells (mTECs) that facilitate the deletion of autoreactive T cells in the thymus. In this review, we extensively discuss the pathogenic roles of inborn errors in the IKK/NF-κB loci in the phenotypically related diseases APECED, immune deficiency syndrome, and severe combined immunodeficiency; differentiate how IKK/NF-κB components, through mTEC (stroma), T cells/leukocytes, or epithelial cells, contribute to the pathogenesis of infectious diseases, autoimmunity, and cancer; and highlight the medical significance of IKK/NF-κB in these diseases.
Collapse
Affiliation(s)
- Feng Zhu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21701, Maryland, USA
| | - Yinling Hu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, 21701, Maryland, USA
| |
Collapse
|
22
|
Chew CL, Conos SA, Unal B, Tergaonkar V. Noncoding RNAs: Master Regulators of Inflammatory Signaling. Trends Mol Med 2017; 24:66-84. [PMID: 29246760 DOI: 10.1016/j.molmed.2017.11.003] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/15/2022]
Abstract
Inflammatory signaling underlies many diseases, from arthritis to cancer. Our understanding of inflammation has thus far been limited to the world of proteins, because we are only just beginning to understand the role that noncoding RNAs (ncRNA) might play. It is now clear that ncRNA do not constitute transcriptional 'noise' but instead harbor physiological functions in controlling signaling pathways. In this review, we cover the newly discovered mechanisms and functions of ncRNAs in the regulation of inflammatory signaling. We also describe advances in experimental techniques allowing this field of research to take root. These findings have opened new avenues for putative therapeutic intervention in inflammatory diseases, which may be seen translated into clinical outcomes in the future.
Collapse
Affiliation(s)
- Chen Li Chew
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore; These authors contributed equally
| | - Stephanie Ana Conos
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore; These authors contributed equally
| | - Bilal Unal
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore
| | - Vinay Tergaonkar
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore; Cancer Science Institute of Singapore, Singapore 117599, Singapore; Centre for Cancer Biology (University of South Australia and SA Pathology), Adelaide, SA 5000, Australia.
| |
Collapse
|
23
|
Cirillo F, Lazzeroni P, Sartori C, Street ME. Inflammatory Diseases and Growth: Effects on the GH-IGF Axis and on Growth Plate. Int J Mol Sci 2017; 18:E1878. [PMID: 28858208 PMCID: PMC5618527 DOI: 10.3390/ijms18091878] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 08/23/2017] [Accepted: 08/29/2017] [Indexed: 02/08/2023] Open
Abstract
This review briefly describes the most common chronic inflammatory diseases in childhood, such as cystic fibrosis (CF), inflammatory bowel diseases (IBDs), juvenile idiopathic arthritis (JIA), and intrauterine growth restriction (IUGR) that can be considered, as such, for the changes reported in the placenta and cord blood of these subjects. Changes in growth hormone (GH) secretion, GH resistance, and changes in the insulin-like growth factor (IGF) system are described mainly in relationship with the increase in nuclear factor-κB (NF-κB) and pro-inflammatory cytokines. Changes in the growth plate are also reported as well as a potential role for microRNAs (miRNAs) and thus epigenetic changes in chronic inflammation. Many mechanisms leading to growth failure are currently known; however, it is clear that further research in the field is still warranted.
Collapse
Affiliation(s)
- Francesca Cirillo
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| | - Pietro Lazzeroni
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| | - Chiara Sartori
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| | - Maria Elisabeth Street
- Division of Paediatric Endocrinology and Diabetology, Department of Obstetrics, Gynaecology and Paediatrics, Azienda AUSL-IRCCS, Viale Risorgimento, 80, 42123 Reggio Emilia, Italy.
| |
Collapse
|
24
|
Hematopoietic stem cell transplantation in 29 patients hemizygous for hypomorphic IKBKG/NEMO mutations. Blood 2017; 130:1456-1467. [PMID: 28679735 DOI: 10.1182/blood-2017-03-771600] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 06/29/2017] [Indexed: 12/18/2022] Open
Abstract
X-linked recessive ectodermal dysplasia with immunodeficiency is a rare primary immunodeficiency caused by hypomorphic mutations of the IKBKG gene encoding the nuclear factor κB essential modulator (NEMO) protein. This condition displays enormous allelic, immunological, and clinical heterogeneity, and therapeutic decisions are difficult because NEMO operates in both hematopoietic and nonhematopoietic cells. Hematopoietic stem cell transplantation (HSCT) is potentially life-saving, but the small number of case reports available suggests it has been reserved for only the most severe cases. Here, we report the health status before HSCT, transplantation outcome, and clinical follow-up for a series of 29 patients from unrelated kindreds from 11 countries. Between them, these patients carry 23 different hypomorphic IKBKG mutations. HSCT was performed from HLA-identical related donors (n = 7), HLA-matched unrelated donors (n = 12), HLA-mismatched unrelated donors (n = 8), and HLA-haploidentical related donors (n = 2). Engraftment was documented in 24 patients, and graft-versus-host disease in 13 patients. Up to 7 patients died 0.2 to 12 months after HSCT. The global survival rate after HSCT among NEMO-deficient children was 74% at a median follow-up after HSCT of 57 months (range, 4-108 months). Preexisting mycobacterial infection and colitis were associated with poor HSCT outcome. The underlying mutation does not appear to have any influence, as patients with the same mutation had different outcomes. Transplantation did not appear to cure colitis, possibly as a result of cell-intrinsic disorders of the epithelial barrier. Overall, HSCT can cure most clinical features of patients with a variety of IKBKG mutations.
Collapse
|
25
|
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, Geha RS. Mechanisms of genotype-phenotype correlation in autosomal dominant anhidrotic ectodermal dysplasia with immune deficiency. J Allergy Clin Immunol 2017. [PMID: 28629746 DOI: 10.1016/j.jaci.2017.05.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
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.
Collapse
Affiliation(s)
- Daniel Petersheim
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Michel J Massaad
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Saetbyul Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Alessia Scarselli
- Division of Immunology and Infectious Diseases, Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, and University of Rome Tor Vergata, Rome, Italy
| | - Caterina Cancrini
- Division of Immunology and Infectious Diseases, Department of Pediatrics, Bambino Gesù Children's Hospital, Rome, and University of Rome Tor Vergata, Rome, Italy
| | | | - Yoji Sasahara
- Department of Pediatrics, Tohoku University, Tohoku, Japan
| | - Arjan C Lankester
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Morna Dorsey
- Division of Allergy, Immunology, and Bone Marrow Transplantation, University of California, San Francisco, Calif
| | - Daniela Di Giovanni
- Immunology Service, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - Liliana Bezrodnik
- Immunology Service, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | | | | | - Kay Tanita
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hirokazu Kanegane
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Erwin W Gelfand
- Immunodeficiency Diagnosis and Treatment Program, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Ashish Jain
- Merck Research Laboratories Boston, Boston, Mass
| | - Elizabeth Secord
- Division of Allergy, Asthma, and Immunology, Children's Hospital of Michigan, Detroit, Mich
| | - Capucine Picard
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital, Paris Descartes University, Paris, France
| | - Jean-Laurent Casanova
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital, Paris Descartes University, Paris, France; St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Michael H Albert
- Department of Pediatric Hematology and Oncology, Dr von Hauner University Children's Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Troy R Torgerson
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Wash
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
26
|
Boisson B, Puel A, Picard C, Casanova JL. Human IκBα Gain of Function: a Severe and Syndromic Immunodeficiency. J Clin Immunol 2017; 37:397-412. [PMID: 28597146 DOI: 10.1007/s10875-017-0400-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/01/2017] [Indexed: 02/05/2023]
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.
Collapse
Affiliation(s)
- Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, USA. .,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Necker Hospital for Sick Children, Paris, France. .,Imagine Institute, Paris Descartes University, Paris, France.
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, Paris Descartes University, Paris, France
| | - Capucine Picard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, Paris Descartes University, Paris, France.,Pediatric Hematology-Immunology and Rheumatology Unit, AP-HP, Necker Hospital for Sick Children, Paris, France.,Study Center for Immunodeficiencies, AP-HP, Necker Hospital for Sick Children, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR1163, Necker Hospital for Sick Children, Paris, France.,Imagine Institute, Paris Descartes University, Paris, France.,Pediatric Hematology-Immunology and Rheumatology Unit, AP-HP, Necker Hospital for Sick Children, Paris, France.,Howard Hughes Medical Institute, New York, NY, USA
| |
Collapse
|
27
|
Teku GN, Vihinen M. Simulation of the dynamics of primary immunodeficiencies in CD4+ T-cells. PLoS One 2017; 12:e0176500. [PMID: 28448599 PMCID: PMC5407609 DOI: 10.1371/journal.pone.0176500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/11/2017] [Indexed: 01/05/2023] Open
Abstract
Primary immunodeficiencies (PIDs) form a large and heterogeneous group of mainly rare disorders that affect the immune system. T-cell deficiencies account for about one-tenth of PIDs, most of them being monogenic. Apart from genetic and clinical information, lots of other data are available for PID proteins and genes, including functions and interactions. Thus, it is possible to perform systems biology studies on the effects of PIDs on T-cell physiology and response. To achieve this, we reconstructed a T-cell network model based on literature mining and TPPIN, a previously published core T-cell network, and performed semi-quantitative dynamic network simulations on both normal and T-cell PID failure modes. The results for several loss-of-function PID simulations correspond to results of previously reported molecular studies. The simulations for TCR PTPRC, LCK, ZAP70 and ITK indicate profound changes to numerous proteins in the network. Significant effects were observed also in the BCL10, CARD11, MALT1, NEMO, IKKB and MAP3K14 simulations. No major effects were observed for PIDs that are caused by constitutively active proteins. The T-cell model facilitates the understanding of the underlying dynamics of PID disease processes. The approach confirms previous knowledge about T-cell signaling network and indicates several new important proteins that may be of interest when developing novel diagnosis and therapies to treat immunological defects.
Collapse
Affiliation(s)
- Gabriel N. Teku
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Mauno Vihinen
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
28
|
Staples E, Morillo-Gutierrez B, Davies J, Petersheim D, Massaad M, Slatter M, Dimou D, Doffinger R, Hackett S, Kumararatne D, Hadfield J, Eldridge MD, Geha RS, Abinun M, Thaventhiran JED. Disseminated Mycobacterium malmoense and Salmonella Infections Associated with a Novel Variant in NFKBIA. J Clin Immunol 2017; 37:415-418. [PMID: 28417298 PMCID: PMC5489571 DOI: 10.1007/s10875-017-0390-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/27/2017] [Indexed: 01/20/2023]
Affiliation(s)
- Emily Staples
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Beatriz Morillo-Gutierrez
- Department of Paediatric Immunology, Great North Children's Hospital, Newcastle upon Tyne, NE1 4LP, UK
| | - Jessica Davies
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - Daniel Petersheim
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michel Massaad
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mary Slatter
- Department of Paediatric Immunology, Great North Children's Hospital, Newcastle upon Tyne, NE1 4LP, UK
| | - Dimitra Dimou
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Rainer Doffinger
- Department of Clinical Immunology, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Scott Hackett
- Paediatric Immunology Department, Birmingham Heartlands Hospital, Birmingham, B9 5SS, UK
| | | | - James Hadfield
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - Matthew D Eldridge
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mario Abinun
- Department of Paediatric Immunology, Great North Children's Hospital, Newcastle upon Tyne, NE1 4LP, UK
| | - James E D Thaventhiran
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK. .,Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
29
|
Puel A, Kun Yang, Ku CL, von Bernuth H, Bustamante J, Santos OF, Lawrence T, Chang HH, Al-Mousa H, Picard C, Casanova JL. Heritable defects of the human TLR signalling pathways. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519050110040601] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Recently, three human primary immunodeficiencies associated with impaired TLR signalling were described. Anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID), either X-linked recessive or autosomal dominant, is caused by hypomorphic mutations in NEMO or hypermorphic mutation in IKBA, respectively, both involved in nuclear factor-κB (NF-κB) activation. These patients present with abnormal development of ectoderm-derived structures and suffer from a broad spectrum of infectious diseases. In vitro studies of the patients' cells showed an impaired, but not abolished, NF-κB activation in response to a large set of stimuli, including TLR agonists. More recently, patients with autosomal recessive amorphic mutations in IRAK4 have been reported, presenting no developmental defect and a more restricted spectrum of infectious diseases, mostly caused by pyogenic encapsulated bacteria, principally, but not exclusively Gram-positive. In vitro studies carried out with these patients' cells showed a specific impairment of the Toll—interleukin-1 receptor (TIR)—interleukin-1 receptor associated kinase (IRAK) signalling pathway. NF-κB- and mitogen activated protein kinase (MAPK) pathways are impaired in response to all TIR agonists tested. These data, therefore, suggest that TLRs play a critical role in host defence against pyogenic bacteria, but may be dispensable or redundant for immunity to most other infectious agents in humans.
Collapse
Affiliation(s)
- Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France,
| | - Kun Yang
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France, French-Chinese Laboratory of Genetics and Life Sciences, Rui-Jin Hospital, Shanghai University, Shanghai, China
| | - Cheng-Lung Ku
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France
| | - Horst von Bernuth
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France
| | - Orchidée Filipe Santos
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France
| | - Tatiana Lawrence
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France
| | - Huey-Hsuan Chang
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France
| | - Hamoud Al-Mousa
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France, Pediatric Hematology-Immunology Unit, Necker Hospital, Paris, France
| | - Capucine Picard
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France, Pediatric Hematology-Immunology Unit, Necker Hospital, Paris, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, University of Paris-INSERM U550, Necker Medical School, Paris, France, Pediatric Hematology-Immunology Unit, Necker Hospital, Paris, France
| |
Collapse
|
30
|
Gain-of-function mutations and immunodeficiency: at a loss for proper tuning of lymphocyte signaling. Curr Opin Allergy Clin Immunol 2016; 15:533-8. [PMID: 26406182 DOI: 10.1097/aci.0000000000000217] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To present recent advances in the discovery and characterization of new immunodeficiency disorders linked to gain-of-function (GOF) mutations in immune signaling molecules. (Figure is included in full-text article.) RECENT FINDINGS In the past 2 years, extensive cellular and molecular studies have illuminated the root causes of pathogenesis for several new monogenic primary immunodeficiency disorders (PIDs) linked to GOF mutations in signaling molecules. Here we discuss on two disorders (BENTA and APDS/PASLI) featuring shared clinical presentation (e.g. lymphoproliferation, selective antibody deficiencies, recurrent sinopulmonary infections). These findings highlight an emerging theme: both loss-of-function and gain-of-function mutations in key molecules can disrupt finely tuned immunoreceptor signaling modalities, resulting in the dysregulation of lymphocyte differentiation and impaired adaptive immunity. SUMMARY Continued research on the molecular pathogenesis of PIDs defined by hyperactive signaling molecules will better distinguish these and related disorders, and pinpoint tailored therapeutic interventions for 'retuning' the immune response in these patients.
Collapse
|
31
|
Wit JM, de Luca F. Atypical defects resulting in growth hormone insensitivity. Growth Horm IGF Res 2016; 28:57-61. [PMID: 26670721 DOI: 10.1016/j.ghir.2015.11.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/27/2015] [Accepted: 11/28/2015] [Indexed: 12/13/2022]
Abstract
Besides four well-documented genetic causes of GH insensitivity (GHI) (GHR, STAT5B, IGF1, IGFALS defects), several other congenital and acquired conditions are associated with GHI. With respect to its anabolic actions, GH induces transcription of IGF1, IGFBP3 and IGFALS through a complex regulatory cascade including GH binding to its receptor (GHR), activation of JAK2 and phosphorylation of STAT5b, which then trafficks to the nucleus. GH also activates the MAPK and PI3K pathways. The synthesis of GHR can be reduced by estrogen deficiency or corticosteroid excess, and is possibly decreased in African pygmies. An increased degradation of GHRs because of overexpression of cytokine-inducible SH2-containing protein (CIS) was suggested for some children with idiopathic short stature. Effects on several downstream components of GH signaling were observed for FGF21, cytokines, sepsis, fever and chronic renal failure. In Noonan syndrome and other "rasopathies" the activation of the RAS-RAF-MAPK-ERK pathway leads to inhibition of the JAK/STAT pathway. In contrast, fibroblasts from tall patients with Sotos syndrome showed a downregulation of this axis. Experimental and clinical evidence suggests that the NF-κB pathway plays a role in GH signaling. In a patient with an IκBα mutation presenting with short stature, GHI, severe immune deficiency and other features, NF-κB nuclear transportation and STAT5 and PI3K expression and activity were reduced. A patient with a mosaic de novo duplication of 17q21-25 presented with several congenital anomalies, GHI and mild immunodeficiency. Studies in blood lymphocytes showed disturbed signaling of the CD28 pathway, involving NF-κB and related proteins. Functional studies on skin fibroblasts revealed that NF-κB activation, PI3K activity and STAT5 phosphorylation in response to GH were suppressed, while the sensitivity to GH in terms of MAPK phosphorylation was increased. The expression of one of the duplicated genes, PRKCA, was significantly higher than in control cells, which might be the cause of this clinical syndrome.
Collapse
Affiliation(s)
- Jan M Wit
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Francesco de Luca
- Section of Endocrinology and Diabetes, St. Christopher's Hospital for Children, Drexel University, College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
32
|
Gentile M, De Mattia D, Pansini A, Schettini F, Buonadonna AL, Capozza M, Ficarella R, Laforgia N. 14q13 distal microdeletion encompassingNKX2-1andPAX9: Patient report and refinement of the associated phenotype. Am J Med Genet A 2016; 170:1884-8. [DOI: 10.1002/ajmg.a.37691] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/15/2016] [Indexed: 02/03/2023]
Affiliation(s)
- Mattia Gentile
- Department of Medical Genetics; Hospital Di Venere - ASL BARI; Bari Italy
| | - Delia De Mattia
- Neonatology and NICU; Department of Biochemical Sciences and Human Oncology; University of Bari; Bari Italy
| | - Angela Pansini
- Department of Medical Genetics; Hospital Di Venere - ASL BARI; Bari Italy
| | - Federico Schettini
- Neonatology and NICU; Department of Biochemical Sciences and Human Oncology; University of Bari; Bari Italy
| | | | - Manuela Capozza
- Neonatology and NICU; Department of Biochemical Sciences and Human Oncology; University of Bari; Bari Italy
| | - Romina Ficarella
- Department of Medical Genetics; Hospital Di Venere - ASL BARI; Bari Italy
| | - Nicola Laforgia
- Neonatology and NICU; Department of Biochemical Sciences and Human Oncology; University of Bari; Bari Italy
| |
Collapse
|
33
|
Pedersen GK, Ádori M, Stark JM, Khoenkhoen S, Arnold C, Beutler B, Karlsson Hedestam GB. Heterozygous Mutation in IκBNS Leads to Reduced Levels of Natural IgM Antibodies and Impaired Responses to T-Independent Type 2 Antigens. Front Immunol 2016; 7:65. [PMID: 26973645 PMCID: PMC4771772 DOI: 10.3389/fimmu.2016.00065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/12/2016] [Indexed: 11/24/2022] Open
Abstract
Mice deficient in central components of classical NF-κB signaling have low levels of circulating natural IgM antibodies and fail to respond to immunization with T-independent type 2 (TI-2) antigens. A plausible explanation for these defects is the severely reduced numbers of B-1 and marginal zone B (MZB) cells in such mice. By using an ethyl-N-nitrosourea mutagenesis screen, we identified a role for the atypical IκB protein IκBNS in humoral immunity. IκBNS-deficient mice lack B-1 cells and have severely reduced numbers of MZB cells, and thus resemble several other strains with defects in classical NF-κB signaling. We analyzed mice heterozygous for the identified IκBNS mutation and demonstrate that these mice have an intermediary phenotype in terms of levels of circulating IgM antibodies and responses to TI-2 antigens. However, in contrast to mice that are homozygous for the IκBNS mutation, the heterozygous mice had normal frequencies of B-1 and MZB cells. These results suggest that there is a requirement for IκBNS expression from two functional alleles for maintaining normal levels of circulating natural IgM antibodies and responses to TI-2 antigens.
Collapse
Affiliation(s)
- Gabriel K Pedersen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Monika Ádori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Julian M Stark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Sharesta Khoenkhoen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Carrie Arnold
- Department of Genetics, The Scripps Research Institute , La Jolla, CA , USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center , Dallas, TX , USA
| | | |
Collapse
|
34
|
Severe Mycobacterial Diseases in a Patient with GOF IκBα Mutation Without EDA. J Clin Immunol 2015; 36:12-5. [PMID: 26691317 DOI: 10.1007/s10875-015-0223-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 12/09/2015] [Indexed: 10/22/2022]
|
35
|
Genetic Variation in NFKBIE Is Associated With Increased Risk of Pneumococcal Meningitis in Children. EBioMedicine 2015; 3:93-99. [PMID: 26870821 PMCID: PMC4739413 DOI: 10.1016/j.ebiom.2015.11.048] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 11/29/2022] Open
Abstract
Background Streptococcus pneumoniae and Neisseria meningitidis are frequent pathogens in life-threatening infections. Genetic variation in the immune system may predispose to these infections. Nuclear factor-κB is a key component of the TLR-pathway, controlled by inhibitors, encoded by the genes NFKBIA, NFKBIE and NFKBIZ. We aimed to replicate previous findings of genetic variation associated with invasive pneumococcal disease (IPD), and to assess whether similar associations could be found in invasive meningococcal disease (IMD). Methods Cases with IPD and IMD and controls were identified by linking Danish national registries. DNA was obtained from the Danish Neonatal Screening Biobank. The association between SNPs and susceptibility to IPD and IMD, mortality and pneumococcal serotypes was investigated. Results 372 children with pneumococcal meningitis, 907 with pneumococcal bacteremia and 1273 controls were included. We included 406 cases with meningococcal meningitis, 272 with meningococcal bacteremia, and 672 controls. The NFKBIE SNP was associated with increased risk of pneumococcal meningitis (aOR 1.68; 95% CI: 1.20–2.36), but not bacteremia (aOR 1.08; 95% CI: 0.86–1.35). The remaining SNPs were not associated with susceptibility to invasive disease. None of the SNPs were associated with risk of IMD or mortality. Conclusions A NFKBIE polymorphism was associated with increased risk of pneumococcal meningitis. A polymorphism in the NFKBIE gene was associated with an increased risk of pneumococcal meningitis in children. This single nucleotide polymorphism (SNP) was not associated with bacteremia. None of the studied SNPs were associated with risk or severity of invasive meningococcal disease.
Collapse
Key Words
- CI, confidence intervals
- CRS, Danish Civil Registration System
- CSF, cerebrospinal fluid
- DNPR, Danish National Patient Registry
- DNSB, Danish Neonatal Screening Biobank
- HWE, Hardy–Weinberg equilibrium
- IMD, invasive meningococcal disease
- IPD, Invasive pneumococcal disease
- IQR, interquartile range
- Invasive Meningococcal Disease
- Invasive Pneumococcal Disease
- LD, linkage disequilibrium
- NF, nuclear factor-κB
- Nuclear Factor-κB
- OR, odds ratio
- Pneumoccoccal Serotypes
- RSV, respiratory syncytial virus
- SNPs, single nucleotide polymorphisms
- SSI, Statens Serum Institut
- WGA, whole-genome-amplification
- aOR, adjusted odds ratio
Collapse
|
36
|
Sharfe N, Merico D, Karanxha A, Macdonald C, Dadi H, Ngan B, Herbrick JA, Roifman CM. The effects of RelB deficiency on lymphocyte development and function. J Autoimmun 2015; 65:90-100. [DOI: 10.1016/j.jaut.2015.09.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/03/2015] [Accepted: 09/05/2015] [Indexed: 10/23/2022]
|
37
|
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, Abel L. Inherited and acquired immunodeficiencies underlying tuberculosis in childhood. Immunol Rev 2015; 264:103-20. [PMID: 25703555 DOI: 10.1111/imr.12272] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Stéphanie Boisson-Dupuis
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale, INSERM-U1163, Paris, France; Paris Descartes University, Imagine Institute, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Gilchrist JJ, MacLennan CA, Hill AVS. Genetic susceptibility to invasive Salmonella disease. Nat Rev Immunol 2015; 15:452-63. [PMID: 26109132 DOI: 10.1038/nri3858] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Invasive Salmonella disease, in the form of enteric fever and invasive non-typhoidal Salmonella (iNTS) disease, causes substantial morbidity and mortality in children and adults in the developing world. The study of genetic variations in humans and mice that influence susceptibility of the host to Salmonella infection provides important insights into immunity to Salmonella. In this Review, we discuss data that have helped to elucidate the host genetic determinants of human enteric fever and iNTS disease, alongside data from the mouse model of Salmonella infection. Considered together, these studies provide a detailed picture of the immunobiology of human invasive Salmonella disease.
Collapse
Affiliation(s)
- James J Gilchrist
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, UK
| | - Calman A MacLennan
- 1] Jenner Institute, Nuffield Department of Medicine, Old Road Campus Research Building, Roosevelt Drive, University of Oxford, Oxford, OX3 7DQ, UK. [2] Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Adrian V S Hill
- 1] Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, UK. [2] Jenner Institute, Nuffield Department of Medicine, Old Road Campus Research Building, Roosevelt Drive, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
39
|
RelB deficiency causes combined immunodeficiency. LYMPHOSIGN JOURNAL-THE JOURNAL OF INHERITED IMMUNE DISORDERS 2015. [DOI: 10.14785/lpsn-2015-0005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Combined immunodeficiency (CID) presents in infancy with severe microbial infections due to either the depletion or dysfunction of lymphocytes. Several mutated genes have been implicated in causing this condition. These encoded proteins are involved in gene recombination, signal transduction from receptors to transcription factors, or they are critical for lymphocyte development. There remain 20%–30% of patients with similar phenotypes but with no known genetic aberration. The objective of this study was to define the molecular basis of CID in a group of patients. Genotyping was performed using linkage panel chips, and the results were analyzed for parametric linkage. Whole genome sequencing was also performed. In vitro mitogen stimulation, flow cytometry, real time PCR, Western blotting, and cytokine ELISA were used to assess immunological status and signal transduction pathways. We identified a homozygous mutation in the gene for the NFκB transcription factor RelB in 3 patients who suffered repeated infection despite the presence of circulating T and B cells. This mutation introduces a premature stop, resulting in an ablation of RelB expression. Evaluation of patient immune systems revealed reduced response to mitogens and an inability to maintain an adequate antibody response to immunizations. Lack of RelB expression results in a clinical presentation of CID. Statement of novelty: We describe RelB deficiency for the first time.
Collapse
|
40
|
Senegas A, Gautheron J, Maurin AGD, Courtois G. IKK-related genetic diseases: probing NF-κB functions in humans and other matters. Cell Mol Life Sci 2015; 72:1275-87. [PMID: 25432706 PMCID: PMC11113297 DOI: 10.1007/s00018-014-1793-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 11/04/2014] [Accepted: 11/20/2014] [Indexed: 12/28/2022]
Abstract
The transcription factor NF-κB plays a key role in numerous physiological processes such as inflammation, immunity, cell proliferation or control of cell death. Its activation is tightly controlled by a kinase complex, IκB kinase (IKK), composed of three core proteins: IKK1/IKKα, IKK2/IKKβ and NEMO/IKKγ. The first two are structurally related kinases whereas the third one is a regulatory subunit exhibiting affinity for upstream activators modified by polyubiquitin chains. Over the years, several inherited diseases caused by mutations of each of the three subunits of IKK have been identified in humans together with diseases caused by mutations of several of its substrates. They are associated with very specific and complex phenotypes involving a broad range of abnormalities such as impaired innate and acquired immune response, perturbed skin development and defects of the central nervous system. Here, we summarize the diverse clinical, cellular and molecular manifestations of IKK-related genetic diseases and show that studying patient-related mutations affecting the IKK subunits and some of their substrates offers the opportunity to understand the various functions of NF-κB in humans, complementing studies performed with mouse models. This analysis also provides glimpses about putative functions of IKK subunits that may be NF-κB-independent.
Collapse
Affiliation(s)
- Anna Senegas
- INSERM U1038, iRTSV, CEA Grenoble, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| | - Jérémie Gautheron
- Department of Gastroenterology, University Hospital RWTH Aachen, Aachen, Germany
| | - Alice Gentil Dit Maurin
- INSERM U1038, iRTSV, CEA Grenoble, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| | - Gilles Courtois
- INSERM U1038, iRTSV, CEA Grenoble, Grenoble, France
- Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
41
|
Rare mendelian primary immunodeficiency diseases associated with impaired NF-κB signaling. Genes Immun 2015; 16:239-46. [PMID: 25764117 PMCID: PMC4457537 DOI: 10.1038/gene.2015.3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/07/2015] [Accepted: 01/09/2015] [Indexed: 12/14/2022]
Abstract
Mendelian Primary Immunodeficiency Diseases (MPIDs) are rare disorders affecting distinct constituents of the innate and adaptive immune system. Although they are genetically heterogeneous a substantial group of MPIDs is due to mutations in genes affecting the NF-κB transcription pathway, essential for cell proliferation, cell survival, and involved in innate immunity and in inflammation. Many of these genes encode for crucial regulatory components of NF-κB pathway and their mutations are associated with immunological and developmental signs somehow overlapping in patients with MPIDs. At present nine different MPIDs listed in the OMIM are caused by mutations in at least nine different genes strictly involved in the NF-κB pathway that result in defects in immune responses. We will report here on the distinct function of each causative gene, on the impaired NF-κB step and more in general on the molecular mechanisms underlining the pathogenesis of the disease. Overall, the MPIDs affecting NF-κB signalosome require a careful integrated diagnosis and appropriate genetic tests to be molecularly identified. Their discovery at an ever-increasing rate will help to establish common therapeutic strategy for a subclass of immunodeficient patients.
Collapse
|
42
|
Boisson B, Quartier P, Casanova JL. Immunological loss-of-function due to genetic gain-of-function in humans: autosomal dominance of the third kind. Curr Opin Immunol 2015; 32:90-105. [PMID: 25645939 PMCID: PMC4364384 DOI: 10.1016/j.coi.2015.01.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 01/06/2015] [Accepted: 01/12/2015] [Indexed: 12/29/2022]
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.
Collapse
Affiliation(s)
- Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Pierre Quartier
- Paris Descartes University, Imagine Institute, Paris 75015, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Paris 75015, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
- Paris Descartes University, Imagine Institute, Paris 75015, France
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Paris 75015, France
- Howard Hughes Medical Institute, New York, NY 10065, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris 75015, France
| |
Collapse
|
43
|
Mooster JL, Le Bras S, Massaad MJ, Jabara H, Yoon J, Galand C, Heesters BA, Burton OT, Mattoo H, Manis J, Geha RS. Defective lymphoid organogenesis underlies the immune deficiency caused by a heterozygous S32I mutation in IκBα. ACTA ACUST UNITED AC 2015; 212:185-202. [PMID: 25601653 PMCID: PMC4322042 DOI: 10.1084/jem.20140979] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Mooster et al. created a knock-in mouse harboring a mutation (S32I) in IκBα that has been identified in a patient with ectodermal dysplasia with immunodeficiency. The mice are characterized by defective architectural cell function; they lack lymph nodes, Peyer’s patches, splenic marginal zones, and follicular DCs and fail to develop germinal centers. These features have not been previously recognized in patients. Patients with ectodermal dysplasia with immunodeficiency (ED-ID) caused by mutations in the inhibitor of NF-κB α (IκBα) are susceptible to severe recurrent infections, despite normal T and B cell numbers and intact in vitro lymphocyte function. Moreover, the outcome of hematopoietic stem cell transplantation (HSCT) in these patients is poor despite good engraftment. Mice heterozygous for the IκBα S32I mutation found in patients exhibited typical features of ED-ID. Strikingly, the mice lacked lymph nodes, Peyer’s patches, splenic marginal zones, and follicular dendritic cells and failed to develop contact hypersensitivity (CHS) or form germinal centers (GCs), all features not previously recognized in patients and typical of defective noncanonical NF-κB signaling. Lymphotoxin β receptor (LTβR)–driven induction of chemokines and adhesion molecules mediated by both canonical and noncanonical NF-κB pathways was impaired, and levels of p100 were markedly diminished in the mutant. IκBα mutant→Rag2−/−, but not WT→IκBα mutant, bone marrow chimeras formed proper lymphoid organs and developed CHS and GCs. Defective architectural cell function explains the immunodeficiency and poor outcome of HSCT in patients with IκBα deficiency and suggests that correction of this niche is critical for reconstituting their immune function.
Collapse
Affiliation(s)
- Jana L Mooster
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Severine Le Bras
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Michel J Massaad
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Haifa Jabara
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Juhan Yoon
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Claire Galand
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Balthasar A Heesters
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, Netherlands
| | - Oliver T Burton
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Hamid Mattoo
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114
| | - John Manis
- Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| | - Raif S Geha
- Division of Allergy and Immunology and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115 Department of Pediatrics, Division of Transfusion Medicine, and Department of Pathology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
44
|
Uhlig HH, Schwerd T, Koletzko S, Shah N, Kammermeier J, Elkadri A, Ouahed J, Wilson DC, Travis SP, Turner D, Klein C, Snapper SB, Muise AM. The diagnostic approach to monogenic very early onset inflammatory bowel disease. Gastroenterology 2014; 147:990-1007.e3. [PMID: 25058236 PMCID: PMC5376484 DOI: 10.1053/j.gastro.2014.07.023] [Citation(s) in RCA: 440] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 07/13/2014] [Accepted: 07/15/2014] [Indexed: 02/07/2023]
Abstract
Patients with a diverse spectrum of rare genetic disorders can present with inflammatory bowel disease (monogenic IBD). Patients with these disorders often develop symptoms during infancy or early childhood, along with endoscopic or histological features of Crohn's disease, ulcerative colitis, or IBD unclassified. Defects in interleukin-10 signaling have a Mendelian inheritance pattern with complete penetrance of intestinal inflammation. Several genetic defects that disturb intestinal epithelial barrier function or affect innate and adaptive immune function have incomplete penetrance of the IBD-like phenotype. Several of these monogenic conditions do not respond to conventional therapy and are associated with high morbidity and mortality. Due to the broad spectrum of these extremely rare diseases, a correct diagnosis is frequently a challenge and often delayed. In many cases, these diseases cannot be categorized based on standard histological and immunologic features of IBD. Genetic analysis is required to identify the cause of the disorder and offer the patient appropriate treatment options, which include medical therapy, surgery, or allogeneic hematopoietic stem cell transplantation. In addition, diagnosis based on genetic analysis can lead to genetic counseling for family members of patients. We describe key intestinal, extraintestinal, and laboratory features of 50 genetic variants associated with IBD-like intestinal inflammation. In addition, we provide approaches for identifying patients likely to have these disorders. We also discuss classic approaches to identify these variants in patients, starting with phenotypic and functional assessments that lead to analysis of candidate genes. As a complementary approach, we discuss parallel genetic screening using next-generation sequencing followed by functional confirmation of genetic defects.
Collapse
Affiliation(s)
- Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, England; Department of Pediatrics, University of Oxford, Oxford, England.
| | - Tobias Schwerd
- Translational Gastroenterology Unit, University of Oxford, Oxford, England
| | - Sibylle Koletzko
- Dr von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Neil Shah
- Great Ormond Street Hospital London, London, England; Catholic University, Leuven, Belgium
| | | | - Abdul Elkadri
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jodie Ouahed
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts; Division of Gastroenterology and Hepatology, Brigham & Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - David C Wilson
- Child Life and Health, University of Edinburgh, Edinburgh, Scotland; Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Royal Hospital for Sick Children, Edinburgh, Scotland
| | - Simon P Travis
- Translational Gastroenterology Unit, University of Oxford, Oxford, England
| | - Dan Turner
- Pediatric Gastroenterology Unit, Shaare Zedek Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Christoph Klein
- Dr von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Scott B Snapper
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts; Division of Gastroenterology and Hepatology, Brigham & Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Aleixo M Muise
- SickKids Inflammatory Bowel Disease Center and Cell Biology Program, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
45
|
Epidermal RelA Specifically Restricts Contact Allergen–Induced Inflammation and Apoptosis in Skin. J Invest Dermatol 2014; 134:2541-2550. [DOI: 10.1038/jid.2014.193] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/18/2014] [Accepted: 04/04/2014] [Indexed: 11/08/2022]
|
46
|
Platt C, Geha RS, Chou J. Gene hunting in the genomic era: approaches to diagnostic dilemmas in patients with primary immunodeficiencies. J Allergy Clin Immunol 2014; 134:262-8. [PMID: 24100122 PMCID: PMC3976463 DOI: 10.1016/j.jaci.2013.08.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/25/2013] [Accepted: 08/26/2013] [Indexed: 12/22/2022]
Abstract
There are more than 180 different genetic causes of primary immunodeficiencies identified to date. Approaches for identifying causative mutations can be broadly classified into 3 strategies: (1) educated guesses based on known signaling pathways essential for immune cell development and function, (2) similarity of clinical phenotypes to mouse models, and (3) unbiased genetic approaches. Next-generation DNA sequencing permits efficient sequencing of whole genomes or exomes but also requires strategies for filtering vast amounts of data. Recent studies have identified ways to solve difficult cases, such as diseases with autosomal dominant inheritance, incomplete penetrance, or mutations in noncoding regions. This review focuses on recently identified primary immunodeficiencies to illustrate the strategies, technologies, and potential pitfalls in finding novel causes of these diseases.
Collapse
Affiliation(s)
- Craig Platt
- Division of Immunology and the Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Raif S Geha
- Division of Immunology and the Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Janet Chou
- Division of Immunology and the Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
47
|
Routes J, Abinun M, Al-Herz W, Bustamante J, Condino-Neto A, De La Morena MT, Etzioni A, Gambineri E, Haddad E, Kobrynski L, Le Deist F, Nonoyama S, Oliveira JB, Perez E, Picard C, Rezaei N, Sleasman J, Sullivan KE, Torgerson T. ICON: the early diagnosis of congenital immunodeficiencies. J Clin Immunol 2014; 34:398-424. [PMID: 24619621 DOI: 10.1007/s10875-014-0003-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/17/2014] [Indexed: 01/27/2023]
Abstract
Primary immunodeficiencies are intrinsic defects in the immune system that result in a predisposition to infection and are frequently accompanied by a propensity to autoimmunity and/or immunedysregulation. Primary immunodeficiencies can be divided into innate immunodeficiencies, phagocytic deficiencies, complement deficiencies, disorders of T cells and B cells (combined immunodeficiencies), antibody deficiencies and immunodeficiencies associated with syndromes. Diseases of immune dysregulation and autoinflammatory disorder are many times also included although the immunodeficiency in these disorders are often secondary to the autoimmunity or immune dysregulation and/or secondary immunosuppression used to control these disorders. Congenital primary immunodeficiencies typically manifest early in life although delayed onset are increasingly recognized. The early diagnosis of congenital immunodeficiencies is essential for optimal management and improved outcomes. In this International Consensus (ICON) document, we provide the salient features of the most common congenital immunodeficiencies.
Collapse
Affiliation(s)
- John Routes
- Department of Pediatrics, Medical College of Wisconsin, and Children's Research Institute, Milwaukee, WI, 53226-4874, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Immunodeficiencies with nonfunctional T cells comprise a heterogeneous group of conditions characterized by altered function of T lymphocytes in spite of largely preserved T cell development. Some of these forms are due to hypomorphic mutations in genes causing severe combined immunodeficiency. More recently, advances in human genome sequencing have facilitated the identification of novel genetic defects that do not affect T cell development, but alter T cell function and homeostasis. Along with increased susceptibility to infections, these conditions are characterized by autoimmunity and higher risk of malignancies. The study of these diseases, and of corresponding animal models, has provided fundamental insights on the mechanisms that govern immune homeostasis.
Collapse
|
49
|
Pannicke U, Baumann B, Fuchs S, Henneke P, Rensing-Ehl A, Rizzi M, Janda A, Hese K, Schlesier M, Holzmann K, Borte S, Laux C, Rump EM, Rosenberg A, Zelinski T, Schrezenmeier H, Wirth T, Ehl S, Schroeder ML, Schwarz K. Deficiency of innate and acquired immunity caused by an IKBKB mutation. N Engl J Med 2013; 369:2504-14. [PMID: 24369075 DOI: 10.1056/nejmoa1309199] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Severe combined immunodeficiency (SCID) comprises a heterogeneous group of heritable deficiencies of humoral and cell-mediated immunity. Many patients with SCID have lymphocyte-activation defects that remain uncharacterized. METHODS We performed genetic studies in four patients, from four families of Northern Cree ancestry, who had clinical characteristics of SCID, including early onset of severe viral, bacterial, and fungal infections despite normal B-cell and T-cell counts. Genomewide homozygosity mapping was used to identify a candidate region, which was found on chromosome 8; all genes within this interval were sequenced. Immune-cell populations, signal transduction on activation, and effector functions were studied. RESULTS The patients had hypogammaglobulinemia or agammaglobulinemia, and their peripheral-blood B cells and T cells were almost exclusively of naive phenotype. Regulatory T cells and γδ T cells were absent. All patients carried a homozygous duplication--c.1292dupG in exon 13 of IKBKB, which encodes IκB kinase 2 (IKK2, also known as IKKβ)--leading to loss of expression of IKK2, a component of the IKK-nuclear factor κB (NF-κB) pathway. Immune cells from the patients had impaired responses to stimulation through T-cell receptors, B-cell receptors, toll-like receptors, inflammatory cytokine receptors, and mitogens. CONCLUSIONS A form of human SCID is characterized by normal lymphocyte development despite a loss of IKK2 function. IKK2 deficiency results in an impaired response to activation stimuli in a variety of immune cells, leading to clinically relevant impairment of adaptive and innate immunity. Although Ikk2 deficiency is lethal in mouse embryos, our observations suggest a more restricted, unique role of IKK2-NF-κB signaling in humans. (Funded by the German Federal Ministry of Education and Research and others.).
Collapse
Affiliation(s)
- Ulrich Pannicke
- From the Institute for Transfusion Medicine, University Hospital Ulm (U.P., C.L., H.S., K.S.), the Institute of Physiological Chemistry (B.B., T.W.), the Center for Biomedical Research, Genomics Core Facility (K. Holzmann), University of Ulm, and the Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg-Hessen (E.-M.R., H.S., K.S.), Ulm; the Center of Chronic Immunodeficiency, University Medical Center Freiburg (S.F., P.H., A.R.-E., K. Hese, M.R., A.J., M.S., S.E.), the Faculty of Biology, University of Freiburg (S.F.), the Center for Pediatrics and Adolescent Medicine (P.H., S.E.), and the Department of Rheumatology and Clinical Immunology (M.S.), University Hospital Freiburg, Freiburg; and the Translational Center for Regenerative Medicine, University of Leipzig, Leipzig (S.B.) - all in Germany; the Division of Clinical Immunology and Transfusion Medicine, Department of Laboratory Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm (S.B.); and the Department of Pediatrics, University of Saskatchewan, Saskatoon (A.R.), and the Departments of Biochemistry and Medical Genetics (T.Z.) and Pediatrics and Child Health (T.Z., M.L.S.), University of Manitoba, Winnipeg - both in Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Yoshioka T, Nishikomori R, Hara J, Okada K, Hashii Y, Okafuji I, Nodomi S, Kawai T, Izawa K, Ohnishi H, Yasumi T, Nakahata T, Heike T. Autosomal dominant anhidrotic ectodermal dysplasia with immunodeficiency caused by a novel NFKBIA mutation, p.Ser36Tyr, presents with mild ectodermal dysplasia and non-infectious systemic inflammation. J Clin Immunol 2013; 33:1165-74. [PMID: 23864385 DOI: 10.1007/s10875-013-9924-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/02/2013] [Indexed: 12/23/2022]
Abstract
PURPOSE Anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID) is characterized by hypohidrosis, dental abnormalities, sparse hair, and immunodeficiency. Autosomal dominant (AD)-EDA-ID, caused by a heterozygous mutation within NFKBIA, is very rare and its clinical features remain largely unknown. This study describes a patient with AD-EDA-ID harboring a novel NFKBIA mutation who presented with mild EDA and non-infectious systemic inflammation. METHODS The clinical presentation of an AD-EDA-ID patient was described and immunological, genetic, and biochemical analyses were performed, with a focus on nuclear factor kappa B (NF-κB) activation. RESULTS The patient presented with symptoms of mild EDA-ID, namely sparse hair and hypohidrosis, although a skin biopsy confirmed the presence of sweat glands. There were no dental abnormalities. The patient also suffered from non-infectious inflammation, which responded to systemic corticosteroid therapy; however, the patient remained ill. Immunological analyses revealed reduced Toll-like receptor/IL-1 (TLR/IL-1) and tumor necrosis factor (TNF) receptor family responses to various stimuli. Genetic analysis identified a de novo heterozygous missense mutation, p.Ser36Tyr, in NFKBIA, resulting in defective NFKBIA degradation and impaired NF-κB activation. The patient was diagnosed with AD-EDA-ID and underwent hematopoietic stem cell transplantation. Engraftment was successful, with few signs of acute graft versus host disease. However, the patient suffered hemolytic anemia and thrombocytopenia, and died from a brain hemorrhage due to intractable thrombocytopenia. CONCLUSION AD-EDA-ID patients can present with mild ectodermal dysplasia and non-infectious inflammation, rather than with recurrent infections. Also, hematopoietic stem cell transplantation for AD-EDA-ID is still a clinical challenge.
Collapse
Affiliation(s)
- Takakazu Yoshioka
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|