Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genes Immun ; 23(2): 66-72, 2022 04.
Article in English | MEDLINE | ID: mdl-35264785

ABSTRACT

XMEN (X-linked immunodeficiency with magnesium defect) is caused by loss-of-function mutations in MAGT1 which is encoded on the X chromosome. The disorder is characterised by CD4 lymphopenia, severe chronic viral infections and defective T-lymphocyte activation. XMEN patients are susceptible to Epstein-Barr virus infections and persistently low levels of intracellular Mg2+. Here we describe a patient that presented with multiple recurrent infections and a subsequent diffuse B-cell lymphoma. Molecular genetic analysis by exome sequencing identified a novel hemizygous MAGT1 nonsense mutation c.1005T>A (NM_032121.5) p.(Cys335*), confirming a diagnosis of XMEN deficiency. Follow-up immunophenotyping was performed by antibody staining and flow cytometry; proliferation was determined by 3H-thymidine uptake after activation by PHA and anti-CD3. Cytotoxic natural killer cell activity was assessed with K562 target cells using the NKTESTTM assay. While lymphocyte populations were superficially intact, B cells were largely naive with a reduced memory cell compartment. Translated NKG2D was absent on both NK and T cells in the proband, and normally expressed in the carrier mother. In vitro NK cell activity was intact in both the proband and his mother. This report adds to the growing number of identified XMEN cases, raising awareness of a, still rare, X-linked immunodeficiency.


Subject(s)
Cation Transport Proteins , Epstein-Barr Virus Infections , Neoplasms , X-Linked Combined Immunodeficiency Diseases , Cation Transport Proteins/genetics , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human , Humans , Mutation , Neoplasms/genetics , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics
2.
J Clin Immunol ; 40(3): 531-533, 2020 04.
Article in English | MEDLINE | ID: mdl-31970544

ABSTRACT

The hyper IgM syndromes are a group of rare primary immunodeficiency disorders. Currently 6 classes of HIGM are described. X-linked HIGM is also called the type 1 HIGM is the commonest variant in which children present in early infancy with features of combined immunodeficiency. Tuberculosis is a very rare presentation as a presenting symptom in HIGM. Here, we describe a child with XHIGM with recurrent tuberculosis.


Subject(s)
Hyper-IgM Immunodeficiency Syndrome, Type 1/diagnosis , Tuberculosis, Pulmonary/diagnosis , X-Linked Combined Immunodeficiency Diseases/diagnosis , Child, Preschool , Humans , Image-Guided Biopsy , Infant , Male , Recurrence , Respiratory Insufficiency , Skin Tests , Tomography, X-Ray Computed
3.
J Clin Immunol ; 40(7): 1001-1009, 2020 10.
Article in English | MEDLINE | ID: mdl-32681206

ABSTRACT

We report the case of a patient with X-linked severe combined immunodeficiency (X-SCID) who survived for over 20 years without hematopoietic stem cell transplantation (HSCT) because of a somatic reversion mutation. An important feature of this rare case included the strategy to validate the pathogenicity of a variant of the IL2RG gene when the T and B cell lineages comprised only revertant cells. We studied the X-inactivation of sorted T cells from the mother to show that the pathogenic variant was indeed the cause of his SCID. One interesting feature was a progressive loss of B cells over 20 years. CyTOF (cytometry time of flight) analysis of bone marrow offered a potential explanation of the B cell failure, with expansions of progenitor populations that suggest a developmental block. Another interesting feature was that the patient bore extensive granulomatous disease and skin cancers that contained T cells, despite severe T cell lymphopenia in the blood. Finally, the patient had a few hundred T cells on presentation but his TCRs comprised a very limited repertoire, supporting the important conclusion that repertoire size trumps numbers of T cells.


Subject(s)
B-Lymphocytes/immunology , Disease Susceptibility , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/etiology , B-Lymphocytes/metabolism , Biomarkers , Biopsy , Child, Preschool , Cytokines/metabolism , Disease Susceptibility/immunology , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Immunophenotyping , Infant , Lymphocyte Count , Male , Phenotype , Skin/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Exome Sequencing , X Chromosome Inactivation
4.
J Clin Immunol ; 40(3): 503-514, 2020 04.
Article in English | MEDLINE | ID: mdl-32072341

ABSTRACT

Hypomorphic IL2RG mutations may lead to milder phenotypes than X-SCID, named variably as atypical X-SCID or X-CID. We report an 11-year-old boy with a novel c. 172C>T;p.(Pro58Ser) mutation in IL2RG, presenting with atypical X-SCID phenotype. We also review the growing number of hypomorphic IL2RG mutations causing atypical X-SCID. We studied the patient's clinical phenotype, B, T, NK, and dendritic cell phenotypes, IL2RG and CD25 cell surface expression, and IL-2 target gene expression, STAT tyrosine phosphorylation, PBMC proliferation, and blast formation in response to IL-2 stimulation, as well as protein-protein interactions of the mutated IL2RG by BioID proximity labeling. The patient suffered from recurrent upper and lower respiratory tract infections, bronchiectasis, and reactive arthritis. His total lymphocyte counts have remained normal despite skewed T and B cells subpopulations, with very low numbers of plasmacytoid dendritic cells. Surface expression of IL2RG was reduced on his lymphocytes. This led to impaired STAT tyrosine phosphorylation in response to IL-2 and IL-21, reduced expression of IL-2 target genes in patient CD4+ T cells, and reduced cell proliferation in response to IL-2 stimulation. BioID proximity labeling showed aberrant interactions between mutated IL2RG and ER/Golgi proteins causing mislocalization of the mutated IL2RG to the ER/Golgi interface. In conclusion, IL2RG p.(Pro58Ser) causes X-CID. Failure of IL2RG plasma membrane targeting may lead to atypical X-SCID. We further identified another carrier of this mutation from newborn SCID screening, lost to closer scrutiny.


Subject(s)
Dendritic Cells/immunology , Interleukin Receptor Common gamma Subunit/genetics , Lymphocytes/physiology , Multiprotein Complexes/metabolism , Mutation/genetics , Receptors, Interleukin-2/metabolism , X-Linked Combined Immunodeficiency Diseases/diagnosis , Cells, Cultured , Child , Gene Expression Regulation , Hemizygote , Humans , Male , Multiprotein Complexes/genetics , Pedigree , Receptors, Interleukin-2/genetics , STAT5 Transcription Factor/metabolism , X-Linked Combined Immunodeficiency Diseases/genetics
5.
Clin Exp Immunol ; 200(1): 61-72, 2020 04.
Article in English | MEDLINE | ID: mdl-31799703

ABSTRACT

In addition to their detection in typical X-linked severe combined immunodeficiency, hypomorphic mutations in the interleukin (IL)-2 receptor common gamma chain gene (IL2RG) have been described in patients with atypical clinical and immunological phenotypes. In this leaky clinical phenotype the diagnosis is often delayed, limiting prompt therapy in these patients. Here, we report the biochemical and functional characterization of a nonsense mutation in exon 8 (p.R328X) of IL2RG in two siblings: a 4-year-old boy with lethal Epstein-Barr virus-related lymphoma and his asymptomatic 8-month-old brother with a Tlow B+ natural killer (NK)+ immunophenotype, dysgammaglobulinemia, abnormal lymphocyte proliferation and reduced levels of T cell receptor excision circles. After confirming normal IL-2RG expression (CD132) on T lymphocytes, signal transducer and activator of transcription-1 (STAT-5) phosphorylation was examined to evaluate the functionality of the common gamma chain (γc ), which showed partially preserved function. Co-immunoprecipitation experiments were performed to assess the interaction capacity of the R328X mutant with Janus kinase (JAK)3, concluding that R328X impairs JAK3 binding to γc . Here, we describe how the R328X mutation in IL-2RG may allow partial phosphorylation of STAT-5 through a JAK3-independent pathway. We identified a region of three amino acids in the γc intracellular domain that may be critical for receptor stabilization and allow this alternative signaling. Identification of the functional consequences of pathogenic IL2RG variants at the cellular level is important to enable clearer understanding of partial defects leading to leaky phenotypes.


Subject(s)
Codon, Nonsense , Interleukin Receptor Common gamma Subunit/genetics , STAT5 Transcription Factor/metabolism , T-Lymphocytes/metabolism , X-Linked Combined Immunodeficiency Diseases/genetics , Animals , COS Cells , Child, Preschool , Chlorocebus aethiops , DNA Mutational Analysis/methods , Female , Humans , Infant , Male , Phenotype , Phosphorylation , Siblings , X-Linked Combined Immunodeficiency Diseases/diagnosis
6.
Immunol Invest ; 47(3): 221-228, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29388853

ABSTRACT

BACKGROUND: Primary immunodeficiency diseases are a group of genetic disorders that lead to increased propensity to a variety of infections, sometimes with fatal outcomes. METHOD: In this study, whole-exome sequencing (WES) was used to identify mutations in two patients suspected of having primary immunodeficiency. Sanger sequencing was used to confirm the results in the patients and their family. RESULT: One patient was diagnosed as X-linked severe combined immunodeficiency (X-SCID) and another patient as X-linked chronic granulomatous disease (X-CGD) by WES. Sequencing analysis of IL2RG gene revealed a novel mutation (c.794T>A, p.I265N) and CYBB gene revealed a missense mutation (c.935T>A, p.M312K). DISCUSSION AND CONCLUSION: This study identifies one novel mutation in the IL2RG gene and another, previously described mutation in the CYBB genes. It is the first report establishing a diagnosis of X-SCID and X-CGD using WES in Chinese patients.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Interleukin Receptor Common gamma Subunit/genetics , Mutation , NADPH Oxidase 2/genetics , Alleles , Amino Acid Substitution , Asian People/genetics , Biomarkers , China , DNA Mutational Analysis , Genotype , Granulomatous Disease, Chronic/diagnosis , Granulomatous Disease, Chronic/genetics , Humans , Immunophenotyping , Infant , Interleukin Receptor Common gamma Subunit/chemistry , Male , Models, Molecular , NADPH Oxidase 2/chemistry , Pedigree , Phenotype , Structure-Activity Relationship , Whole Genome Sequencing , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics
7.
J Pediatr Hematol Oncol ; 40(6): e348-e349, 2018 08.
Article in English | MEDLINE | ID: mdl-29620683

ABSTRACT

Severe combined immunodeficiency is an inherited disease with profoundly defective T cells, B cells, and natural killer cells. X-linked severe combined immunodeficiency is the most common form. In this report, we describe a 4-month-old male infant who was admitted to our hospital with progressive breathlessness and abdominal mass. He was diagnosed with hepatoblastoma and presented a pneumocystis jirovecii pneumonia at the beginning of chemotherapy. Definitive diagnosis of X-linked severe combined immunodeficiency was established by DNA analysis of the interleukin 2 receptor gamma chain gene. This case is the first report which describes an X-linked severe combined immunodeficiency patient with hepatoblastoma.


Subject(s)
Hepatoblastoma , Liver Neoplasms , Pneumocystis carinii , Pneumonia, Pneumocystis , X-Linked Combined Immunodeficiency Diseases , Hepatoblastoma/diagnosis , Hepatoblastoma/drug therapy , Humans , Infant , Liver Neoplasms/diagnosis , Liver Neoplasms/drug therapy , Male , Pneumonia, Pneumocystis/diagnosis , Pneumonia, Pneumocystis/drug therapy , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/drug therapy
8.
J Pediatr Hematol Oncol ; 39(8): e470-e472, 2017 11.
Article in English | MEDLINE | ID: mdl-28678090

ABSTRACT

Severe combined immunodeficiency (SCID) is a defect in the differentiation and function of T cells. An increased malignancy risk, mainly lymphatic malignancy, has been described in patients with SCID. We report a patient with X-linked SCID who developed acute myeloid leukemia, derived from the recipient with somatic NRAS mutation 4 months after cord blood transplantation (CBT). Loss of heterozygosity phenomenon of the recipient at 6q14 locus was observed at 2 months post-CBT and progressed to 6q deletion (6q-) chromosome abnormality. Somatic NRAS mutation was detected at 3 months post-CBT. Thus, 6q- and NRAS mutation were strongly associated with the leukemic transformation in our patient.


Subject(s)
Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/etiology , X-Linked Combined Immunodeficiency Diseases/complications , Chromosome Deletion , Chromosomes, Human, Pair 6 , Cord Blood Stem Cell Transplantation/adverse effects , Cord Blood Stem Cell Transplantation/methods , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Humans , Immunosuppressive Agents/therapeutic use , In Situ Hybridization, Fluorescence , Infant , Interleukin Receptor Common gamma Subunit/genetics , Loss of Heterozygosity , Male , Mutation , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/therapy
9.
Blood ; 123(14): 2148-52, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24550228

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic gammaherpesvirus that infects and persists in 95% of adults worldwide and has the potential to cause fatal disease, especially lymphoma, in immunocompromised hosts. Primary immunodeficiencies (PIDs) that predispose to EBV-associated malignancies have provided novel insights into the molecular mechanisms of immune defense against EBV. We have recently characterized a novel PID now named "X-linked immunodeficiency with magnesium defect, EBV infection, and neoplasia" (XMEN) disease characterized by loss-of-function mutations in the gene encoding magnesium transporter 1 (MAGT1), chronic high-level EBV with increased EBV-infected B cells, and heightened susceptibility to EBV-associated lymphomas. The genetic etiology of XMEN disease has revealed an unexpected quantitative role for intracellular free magnesium in immune functions and has led to novel diagnostic and therapeutic strategies. Here, we review the clinical presentation, genetic mutation spectrum, molecular mechanisms of pathogenesis, and diagnostic and therapeutic considerations for this previously unrecognized disease.


Subject(s)
Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/immunology , Immunity, Innate/drug effects , Magnesium Deficiency/complications , Magnesium/pharmacology , Neoplasms/complications , X-Linked Combined Immunodeficiency Diseases/complications , Adult , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/genetics , Humans , Magnesium Deficiency/diagnosis , Magnesium Deficiency/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Syndrome , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics
11.
J Clin Immunol ; 35(2): 112-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25504528

ABSTRACT

XMEN disease (X-linked immunodeficiency with Magnesium defect, Epstein-Barr virus infection and Neoplasia) is a novel primary immune deficiency caused by mutations in MAGT1 and characterised by chronic infection with Epstein-Barr virus (EBV), EBV-driven lymphoma, CD4 T-cell lymphopenia, and dysgammaglobulinemia [1]. Functional studies have demonstrated roles for magnesium as a second messenger in T-cell receptor signalling [1], and for NKG2D expression and consequently NK- and CD8 T-cell cytotoxicity [2]. 7 patients have been described in the literature; the oldest died at 45 years and was diagnosed posthumously [1-3]. We present the case of a 58-year-old Caucasian gentleman with a novel mutation in MAGT1 with the aim of adding to the phenotype of this newly described disease by detailing his clinical course over more than 20 years.


Subject(s)
Cation Transport Proteins/genetics , Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/etiology , Mutation , X-Linked Combined Immunodeficiency Diseases/complications , X-Linked Combined Immunodeficiency Diseases/genetics , Brain/pathology , DNA Mutational Analysis , Fluorodeoxyglucose F18 , Humans , Immunophenotyping , Lymph Nodes/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Phenotype , Positron-Emission Tomography , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tomography, X-Ray Computed , X-Linked Combined Immunodeficiency Diseases/diagnosis
12.
Curr Opin Pediatr ; 26(6): 713-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25313976

ABSTRACT

PURPOSE OF REVIEW: To describe the role of the magnesium transporter 1 (MAGT1) in the pathogenesis of 'X-linked immunodeficiency with magnesium defect, Epstein-Barr virus (EBV) infection, and neoplasia' (XMEN) disease and its clinical implications. RECENT FINDINGS: The magnesium transporter protein MAGT1 participates in the intracellular magnesium ion (Mg) homeostasis and facilitates a transient Mg influx induced by the activation of the T-cell receptor. Loss-of-function mutations in MAGT1 cause an immunodeficiency named 'XMEN syndrome', characterized by CD4 lymphopenia, chronic EBV infection, and EBV-related lymphoproliferative disorders. Patients with XMEN disease have impaired T-cell activation and decreased cytolytic function of natural killer (NK) and CD8 T cells because of decreased expression of the NK stimulatory receptor 'natural-killer group 2, member D' (NKG2D). Patients may have defective specific antibody responses secondary to T cell dysfunction, but B cells have not been shown to be directly affected by mutations in MAGT1. SUMMARY: XMEN disease has revealed a novel role for free intracellular magnesium in the immune system. Further understanding of the MAGT1 signaling pathway may lead to new diagnostic and therapeutic approaches.


Subject(s)
Epstein-Barr Virus Infections/complications , Magnesium Deficiency/complications , Neoplasms/complications , X-Linked Combined Immunodeficiency Diseases/complications , Cation Transport Proteins , Child , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/therapy , Humans , Magnesium Deficiency/diagnosis , Magnesium Deficiency/therapy , Male , Neoplasms/diagnosis , Neoplasms/therapy , Syndrome , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/therapy
13.
Front Cell Infect Microbiol ; 14: 1341236, 2024.
Article in English | MEDLINE | ID: mdl-38410723

ABSTRACT

Bacille Calmette-Guérin (BCG) is a live strain of Mycobacterium bovis (M.bovis) for use as an attenuated vaccine to prevent tuberculosis (TB) infection, while it could also lead to an infection in immunodeficient patients. M.bovis could infect patients with immunodeficiency via BCG vaccination. Disseminated BCG disease (BCGosis) is extremely rare and has a high mortality rate. This article presents a case of a 3-month-old patient with disseminated BCG infection who was initially diagnosed with hemophagocytic syndrome (HPS) and eventually found to have X-linked severe combined immunodeficiency (X-SCID). M.bovis and its drug resistance genes were identified by metagenomics next-generation sequencing (mNGS) combined with targeted next-generation sequencing (tNGS) in blood and cerebrospinal fluid. Whole exome sequencing (WES) revealed a pathogenic variant in the common γ-chain gene (IL2RG), confirming X-SCID. Finally, antituberculosis therapy and umbilical cord blood transplantation were given to the patient. He was successfully cured of BCGosis, and his immune function was restored. The mNGS combined with the tNGS provided effective methods for diagnosing rare BCG infections in children. Their combined application significantly improved the sensitivity and specificity of the detection of M.bovis.


Subject(s)
Immunologic Deficiency Syndromes , Latent Tuberculosis , Mycobacterium bovis , Tuberculosis , X-Linked Combined Immunodeficiency Diseases , Male , Infant , Child , Humans , Mycobacterium bovis/genetics , BCG Vaccine/adverse effects , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/complications , Tuberculosis/microbiology , Immunologic Deficiency Syndromes/complications , High-Throughput Nucleotide Sequencing
14.
Beijing Da Xue Xue Bao Yi Xue Ban ; 45(6): 852-8, 2013 Dec 18.
Article in Zh | MEDLINE | ID: mdl-24343061

ABSTRACT

OBJECTIVE: To explore the application of multiple displacement amplification (MDA) combined with short tandem repeats (STRs) in preimplantation genetic diagnosis (PGD). METHODS: MDA was applied to amplify the whole genome of a single cell and to retrieve and assemble the highly heterogeneous STR loci among human population. Haplotype analytic system was established with aiming at diagnosis of the single gene diseases by selecting the STR loci located within the pathogenic genes or on both bounding sides of the pathogenic genes. At the same time, allele specific amplification, PCR-reverse dot-blotting hybridization methods and gene sequencing methods were employed for direct detection of the pathogenic genes. The STR loci located at related chromosomes were selected to carry out allele number analysis on the basis of chromosome number and structural abnormality. RESULTS: In the study, 12 PGD systems were set up including 6 different monogenic diseases (spinal muscular atrophy, Duchenne muscular dystrophy, X-linked chronic granulomatous disease, osteopetrosis, achondroplasia, X-linked severe combined immunodeficiency), Robertsonian translocations, α-thalassemia combined with Robertsonian translocation, α- and ß-double thalassemia, ß-thalassemia with HLA typing and DMD with HLA typing. Then 44 PGD cycles were performed for 35 couples with different kinds of inherited diseases, which resulted in 20 healthy liveborns (12 singletons and 4 twins) and 5 ongoing pregnancies. The clinical pregnancy rate was 47.7% (21/44) per PGD cycle. The overall diagnostic rate was 94.6% (367/388). The MDA failed in 3.6% (14/388) single blastomeres. The amplification rate of the subsequent PCR was 97.1% and the average allele drop out (ADO) rate was 12.6% (range: 0-47.5%). CONCLUSION: The application of MDA combined with STRs provided a generic PGD approach for different genetic disorders, especially for simultaneous diagnosis of two or more hereditary statuses. The method could greatly shorten the time of developing PGD system of new diseases, which broadens the indications of PGD.


Subject(s)
Microsatellite Repeats , Nucleic Acid Amplification Techniques , Preimplantation Diagnosis/methods , Achondroplasia/diagnosis , Achondroplasia/genetics , Adult , Female , Granulomatous Disease, Chronic/diagnosis , Granulomatous Disease, Chronic/genetics , Humans , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/genetics , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/genetics , Osteopetrosis/diagnosis , Osteopetrosis/genetics , Pregnancy , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics , alpha-Thalassemia/diagnosis , alpha-Thalassemia/genetics , beta-Thalassemia/diagnosis , beta-Thalassemia/genetics
15.
Emerg Infect Dis ; 18(6): 992-4, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22607733

ABSTRACT

Patients with primary immunodeficiency are prone to persistently excrete Sabin-like virus after administration of live-attenuated oral polio vaccine and have an increased risk for vaccine-derived paralytic polio. We report a case of type 3 immunodeficiency-associated vaccine-derived poliovirus in a child in South Africa who was born with X-linked immunodeficiency syndrome.


Subject(s)
Poliomyelitis/diagnosis , Poliovirus Vaccine, Oral/adverse effects , Poliovirus/genetics , X-Linked Combined Immunodeficiency Diseases/diagnosis , Capsid Proteins/genetics , Humans , Infant , Male , Poliomyelitis/etiology , Poliomyelitis/virology , Sequence Analysis, DNA , South Africa , Vaccines, Attenuated/adverse effects , X-Linked Combined Immunodeficiency Diseases/complications
16.
J Clin Immunol ; 32(3): 449-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22350222

ABSTRACT

Adenosine deaminase deficiency is a disorder of purine metabolism manifesting severe combined immunodeficiency (ADA-SCID) and systemic abnormalities. Increased levels of the substrate deoxyadenosine triphosphate (dATP) lead to immunodeficiency and are associated in a murine model with pulmonary insufficiency. We compared a cohort of patients with ADA-SCID and X-linked SCID and found that despite similar radiological and respiratory findings, positive microbiology is significantly less frequent in ADA-SCID patients (p < 0.0005), suggesting a metabolic pathogenesis for the lung disease. Clinicians should be aware of this possibility and correct metabolic abnormalities either through enzyme replacement or haematopoietic stem cell transplant, in addition to treating infectious complications.


Subject(s)
Agammaglobulinemia/diagnosis , Lung Diseases/diagnosis , Severe Combined Immunodeficiency/diagnosis , X-Linked Combined Immunodeficiency Diseases/diagnosis , Adenosine Deaminase/deficiency , Adenosine Deaminase/therapeutic use , Agammaglobulinemia/microbiology , Agammaglobulinemia/therapy , Bronchoalveolar Lavage , Child, Preschool , Enzyme Replacement Therapy , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Infant, Newborn , Lung Diseases/microbiology , Lung Diseases/therapy , Male , Severe Combined Immunodeficiency/microbiology , Severe Combined Immunodeficiency/therapy , Tomography, X-Ray Computed , X-Linked Combined Immunodeficiency Diseases/microbiology , X-Linked Combined Immunodeficiency Diseases/therapy
17.
J Clin Immunol ; 31(5): 778-83, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21732012

ABSTRACT

Severe combined immunodeficiencies (SCID) are a heterogeneous group of genetic disorders characterized by a blockade or impairment of both cellular and humoral immunity. Several epidemiological studies in different geographic areas have shown that the most common type of SCID affecting almost half of these patients is the X-linked common γ-chain (γ(c)) deficiency. The objective of the study was to document the incidence and types of SCID in our area. We conducted a retrospective analysis of patients who were diagnosed with SCID in the major immunology center in Greece for a 20-year period. During the study period, 30 children from 27 unrelated families with final diagnosis of SCID were identified. The incidence of SCID in Greece is estimated at 1.7 cases per 100,000 live births. Out of 30 children, 19 were boys (63.3%) and 26 (86.7%) had Greek maternal origin. Lymphocyte immunophenotypes that were identified were T(-)B(-)NK(+) in 12 (40%) children, T(-)B(+)NK(-) in six (20%), T(-)B(+)NK(+) in three (10%), T(-)B(-)NK(-) in two (6.7%) and T(+)B(+/-)NK(+) in seven (23.4%) (among them, four [13.4%] females with Omenn's syndrome). Molecular diagnosis was available for 12 children: γ(c) (2) with non Greek maternal origin, Jak3 (2), Rag1 (2), Artemis (3), ADA deficiency (2), PNP deficiency (1). Out of the 26 children of Greek maternal origin diagnosed with SCID representing 23 distinct families, only two (8.7%) had lymphocyte immunophenotype compatible with γ(c)-chain gene mutation (no molecular testing or enough DNA was available for them at the time of diagnosis). Findings of the present study suggest that, for unknown reasons, mutations of the γ(c) chain of several cytokine receptors have a rare occurrence in our area.


Subject(s)
Interleukin Receptor Common gamma Subunit/metabolism , X-Linked Combined Immunodeficiency Diseases/epidemiology , X-Linked Combined Immunodeficiency Diseases/genetics , Child , Child, Preschool , DNA Mutational Analysis , Demography , Family , Female , Genetics, Population , Greece , Humans , Immunophenotyping , Incidence , Infant , Infant, Newborn , Interleukin Receptor Common gamma Subunit/genetics , Male , Mutation/genetics , Retrospective Studies , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/immunology
18.
J Clin Immunol ; 31(5): 773-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21710275

ABSTRACT

Despite our increasing characterization of the molecular basis for many primary immunodeficiency states, significant heterogeneity in clinical and immunological phenotype exists. Epigenetic alterations have been implicated in the pathogenesis of immune dysregulation and may provide a unique paradigm to help us understand the phenotypic heterogeneity in primary immunodeficiency. The occurrence of X-linked thrombocytopenia (XLT) and Wiskott-Aldrich syndrome (WAS) in monozygotic twins is a rare occurrence which allows for the exploration of epigenetic alterations and associated phenotypic heterogeneity. We describe a pair of monozygotic twin brothers with a missense mutation in the WAS gene consistent with reduced expression of the WAS protein, a XLT phenotype, and a good prognosis. Despite this genotype and anticipated mild phenotype in both twins, a discordant phenotype has evolved in which one twin demonstrates asymptomatic thrombocytopenia and the other symptomatic thrombocytopenia, infectious complications, and autoimmunity. Characterization of the potential epigenetic contribution to the spectrum of XLT and WAS is described and the implications of these findings are discussed.


Subject(s)
DNA Methylation , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/physiopathology , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/physiopathology , Asymptomatic Diseases , Autoantibodies/blood , Autoimmunity , Cells, Cultured , Child , CpG Islands/genetics , DNA Mutational Analysis , Down-Regulation , Epigenesis, Genetic/immunology , Genetic Association Studies , Genotype , Humans , Infant, Newborn , Infections , Male , Mutation, Missense/genetics , Promoter Regions, Genetic/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thrombocytopenia , Twins, Monozygotic/genetics , Wiskott-Aldrich Syndrome/diagnosis , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/immunology , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/immunology
19.
J Clin Immunol ; 31(5): 802-10, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21755389

ABSTRACT

BACKGROUND: Nuclear factor-κB essential modulator (NEMO) deficiency is a developmental and immunological disorder. The genetic and phenotypic correlation has been described. METHODS: We report a unique clinical presentation and the identification of a novel missense mutation in the NEMO gene in a 3-year-old boy with bacillus Calmette-Guerin (BCG) infection. RESULTS: The patient presented with fever, cervical lymphadenopathy, and abnormal anti-Epstein-Barr virus (EBV) antibody titers, suggestive of EBV-related diseases including chronic active EBV infection, X-linked lymphoproliferative syndrome, or nasopharyngeal carcinoma. Although the biopsy specimen from a nasopharyngeal lesion was initially diagnosed as squamous cell carcinoma, this was changed to disseminated BCG infection involving the nasopharynx, multiple systemic lymph nodes, and brain. A novel mutation (designated D311E) in the NEMO gene, located in the NEMO ubiquitin-binding (NUB) domain, was identified as the underlying cause of the immunodeficiency. Impaired immune responses which are characteristic of patients with NEMO deficiency were demonstrated. The patient underwent successful unrelated bone marrow transplantation at 4.9 years of age. CONCLUSION: This study suggests the importance of the NUB domain in host defense against mycobacteria. The unique presenting features in our patient indicate that a hypomorphic NEMO mutation can be associated with atypical pathological findings of the epithelial tissues in patients with BCG infection.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/immunology , I-kappa B Kinase/metabolism , Mycobacterium bovis/immunology , Nasopharyngeal Neoplasms/diagnosis , Nasopharyngeal Neoplasms/genetics , Tuberculosis/diagnosis , Tuberculosis/genetics , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/physiopathology , Child, Preschool , DNA Mutational Analysis , Diagnosis, Differential , Ectodermal Dysplasia , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/physiopathology , Herpesvirus 4, Human/pathogenicity , Humans , I-kappa B Kinase/genetics , Immunity, Innate , Lymphatic Diseases , Male , Mutation/genetics , Mycobacterium bovis/pathogenicity , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/physiopathology , Tuberculosis/immunology , Tuberculosis/pathology , Tuberculosis/physiopathology , Ubiquitination/genetics , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/pathology , X-Linked Combined Immunodeficiency Diseases/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL