Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 22(7): 893-903, 2021 07.
Article in English | MEDLINE | ID: mdl-34155405

ABSTRACT

In the present study, we report a human-inherited, impaired, adaptive immunity disorder, which predominantly manifested as a B cell differentiation defect, caused by a heterozygous IKZF3 missense variant, resulting in a glycine-to-arginine replacement within the DNA-binding domain of the encoded AIOLOS protein. Using mice that bear the corresponding variant and recapitulate the B and T cell phenotypes, we show that the mutant AIOLOS homodimers and AIOLOS-IKAROS heterodimers did not bind the canonical AIOLOS-IKAROS DNA sequence. In addition, homodimers and heterodimers containing one mutant AIOLOS bound to genomic regions lacking both canonical motifs. However, the removal of the dimerization capacity from mutant AIOLOS restored B cell development. Hence, the adaptive immunity defect is caused by the AIOLOS variant hijacking IKAROS function. Heterodimeric interference is a new mechanism of autosomal dominance that causes inborn errors of immunity by impairing protein function via the mutation of its heterodimeric partner.


Subject(s)
Adaptive Immunity , B-Lymphocytes/metabolism , Cell Differentiation , Ikaros Transcription Factor/metabolism , Primary Immunodeficiency Diseases/metabolism , T-Lymphocytes/metabolism , Animals , B-Lymphocytes/immunology , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Female , HEK293 Cells , Humans , Ikaros Transcription Factor/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense , NIH 3T3 Cells , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Signal Transduction , T-Lymphocytes/immunology
3.
J Clin Immunol ; 44(6): 128, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38773004

ABSTRACT

AIOLOS, encoded by the IKZF3 gene, belongs to the Ikaros zinc finger transcription factor family and plays a pivotal role in regulating lymphocyte development. Recently, heterozygous missense loss-of-function variants within the DNA-binding domain of the IKZF3 gene (G159R, N160S, and G191R) have been identified in patients with inborn errors of immunity (IEI). Additionally, a missense and a truncating variant (E82K and Q402X) leading to the AIOLOS haploinsufficiency have been documented. The majority of individuals with AIOLOS-associated IEI manifest recurrent sinopulmonary infections, as well as various bacterial and viral infections. The patients carrying the AIOLOSN160S variant exhibit severe immunodeficient phenotypes. In contrast, patients harboring AIOLOS haploinsufficient variants predominantly present with clinical phenotypes associated with immune dysregulation. A varying degree of B-lymphopenia and hypoimmunoglobulinemia was noted in approximately half of the patients. Mouse models of AIOLOSG159R and AIOLOSN160S variants (AiolosG158R and AiolosN159S in mice, respectively) recapitulated most of the immune abnormalities observed in the patients. Among these models, AiolosG158R mice prominently exhibited defects in early B cell differentiation resulting from mutant Aiolos interfering with Ikaros function through heterodimer formation. In contrast, AiolosN159S mice did not manifest early B cell differentiation defects. However, they displayed a distinct immune abnormality characterized by impaired induction of CD62L expression in lymphocytes, which is likely attributable to dysfunction of Ikaros, leading to defective lymphocyte homing to lymph nodes. Considering the diverse clinical phenotypes observed in the reported cases and the distinct molecular pathogenesis associated with each variant, further studies with more patients with AIOLOS-associated IEI would contribute to a better understanding of the clinical spectrum and underlying molecular mechanisms associated with this disorder.


Subject(s)
Ikaros Transcription Factor , Ikaros Transcription Factor/genetics , Humans , Animals , Haploinsufficiency , Phenotype , Mice , Mutation/genetics , Disease Models, Animal , Immunity/genetics , Genetic Predisposition to Disease
4.
Pediatr Blood Cancer ; 71(3): e30834, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38149846

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital anemia with erythroid cell aplasia. Most of the causative genes are ribosomal proteins. GATA1, a hematopoietic master transcription factor required for erythropoiesis, also causes DBA. GATA1 is located on Xp11.23; therefore, DBA develops only in males in an X-linked inheritance pattern. Here, we report a case of transient erythroblastopenia and moderate anemia in a female newborn infant with a de novo GATA1 variant. In this patient, increased methylation of the GATA1 wild-type allele was observed in erythroid cells. Skewed lyonization of GATA1 may cause mild transient erythroblastopenia in a female patient.


Subject(s)
Anemia, Aplastic , Anemia, Diamond-Blackfan , Anemia, Hemolytic, Congenital , Male , Infant , Infant, Newborn , Humans , Female , Ribosomal Proteins/genetics , Anemia, Diamond-Blackfan/genetics , Erythropoiesis , GATA1 Transcription Factor/genetics
5.
BMC Pediatr ; 24(1): 304, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704558

ABSTRACT

BACKGROUND: T-cell acute lymphoblastic leukemia (T-ALL) tends to involve central nervous system (CNS) infiltration at diagnosis. However, cases of residual CNS lesions detected at the end of induction and post early intensification have not been recorded in patients with T-ALL. Also, the ratio and prognosis of patients with residual intracranial lesions have not been defined. CASE PRESENTATION: A 9-year-old boy with T-ALL had multiple intracranial tumors, which were still detected post early intensification. To investigate residual CNS lesions, we used 11C-methionine (MET)-positron emission tomography. Negative MET uptake in CNS lesions and excellent MRD status in bone marrow allowed continuing therapies without hematopoietic cell transplantation. CONCLUSIONS: In cases with residual lesions on imaging studies, treatment strategies should be considered by the systemic response, direct assessment of spinal fluid, along with further development of noninvasive imaging methods in CNS. Further retrospective or prospective studies are required to determine the prognosis and frequency of cases with residual intracranial lesions after induction therapy.


Subject(s)
Neoplasm, Residual , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Male , Child , Brain Neoplasms/diagnostic imaging , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Positron-Emission Tomography , Methionine
6.
J Pediatr Hematol Oncol ; 45(2): e285-e289, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36757045

ABSTRACT

Artemis deficiency is characterized by DNA double-strand breaks repairing dysfunction and increased sensitivity to ionizing radiation and alkylating reagents. We describe the first successful case of T-cell receptor [TCR]αß/CD19-depleted hematopoietic cell transplantation [HCT] for Artemis deficiency in Japan. A 6-month-old Korean boy was diagnosed with Artemis-deficient severe combined immunodeficiency. He had no human leukocyte antigen (HLA)-matched sibling or unrelated donor. Therefore, TCRαß/CD19-depleted HCT from his haploidentical mother was performed. Despite mixed chimerism in whole blood, T cells achieved complete donor chimerism 6 months after HCT. TCRαß/CD19-depleted HCT could be an effective treatment for patients with radiation-sensitive severe combined immunodeficiency.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Humans , Infant , Male , Antigens, CD19 , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Receptors, Antigen, T-Cell, alpha-beta , Severe Combined Immunodeficiency/genetics , T-Lymphocytes , Transplantation Conditioning , Unrelated Donors
7.
J Infect Chemother ; 28(6): 814-818, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35125343

ABSTRACT

Patients with multisystem inflammatory syndrome in children (MIS-C) can develop clinical features resembling Kawasaki disease (KD). A full picture of MIS-C in East Asia which has higher incidence of KD than other regions remains unclear. We report on a 15-year-old Japanese boy with refractory MIS-C who was successfully treated with infliximab. A Japanese boy who was diagnosed with coronavirus disease 2019 (COVID-19) before a month developed MIS-C with fulfilling six principal symptoms of KD. Laboratory data showed extreme hyperferritinemia (11,404 ng/mL), besides lymphopenia and thrombocytopenia. The patient was refractory to initial therapy with intravenous immunoglobulin (IVIG; 2 g/kg), aspirin, and prednisolone. He was therefore administered a second IVIG (2 g/kg) and infliximab (5 mg/kg) on days 7 and 8 from the onset of fever, respectively, which resulted in an improvement of clinical symptoms. Only four Japanese cases with MIS-C were reported and all of them were responsive to IVIG. The hyperferritinemia in this case was distinctive from previously reported MIS-C cases in Japan and other cohorts and may be associated with refractoriness to IVIG therapy. Marked elevation of circulating ferritin levels is known to be induced by tumor necrosis factor-α, which plays a key role in the pathogenesis of both KD and MIS-C. Thus, for MIS-C patients with hyperferritinemia, early intervention with adjunctive infliximab may induce a more rapid resolution of inflammation and improve outcome. Because MIS-C may be heterogeneous with respect to immunopathology, genetic background, clinical phenotypes and response to therapies, optimized treatment strategies according to immunopathogenesis are required.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Connective Tissue Diseases , Hyperferritinemia , Mucocutaneous Lymph Node Syndrome , COVID-19/complications , Humans , Immunoglobulins, Intravenous/therapeutic use , Infliximab/therapeutic use , Japan , Male , Mucocutaneous Lymph Node Syndrome/complications , Mucocutaneous Lymph Node Syndrome/diagnosis , Mucocutaneous Lymph Node Syndrome/drug therapy , SARS-CoV-2 , Systemic Inflammatory Response Syndrome/drug therapy
8.
J Clin Immunol ; 41(5): 944-957, 2021 07.
Article in English | MEDLINE | ID: mdl-33527309

ABSTRACT

PURPOSE: The purpose of our study was to compare the safety and efficacy of hematopoietic cell transplantation (HCT) using fludarabine (Flu)-based reduced intensity conditioning (RIC) with busulfan (BU) or melphalan (Mel) for primary immunodeficiency diseases (PID). METHODS: We retrospectively analyzed transplant outcome, including engraftment, chimerism, immune reconstitution, and complications in 15 patients with severe combined immunodeficiency (SCID) and 27 patients with non-SCID PID. The patients underwent Flu-based RIC-HCT with BU (FluBU: 7 SCID, 16 non-SCID) or Mel (FluMel: 8 SCID, 11 non-SCID). The targeted low-dose BU with therapeutic drug monitoring was set to 30 mg hour/L for SCID. RESULTS: The 2-year overall survival of all patients was 79.6% and that of patients with SCID in the FluBU and FluMel groups was 100% and 62.5%, respectively. In the FluBU group, all seven patients achieved engraftment, good immune reconstitution, and long-term survival. All five patients receiving umbilical cord blood transplantation achieved complete or high-level mixed chimerism and sufficient specific IgG production. In the FluMel group, six of eight patients achieved complete or high-level mixed chimerism. Viral reactivation or new viral infection occurred in one FluBU group patient and four FluMel group patients. In the non-SCID group, 10 of 11 patients (91%) who received FluMel achieved complete or high-level mixed chimerism but had variable outcomes. Patients with WAS (2/2 patients), NEMO deficiency (2/2 patients), and X-linked hyper IgM syndrome (2/3 patients) who received FluBU achieved complete or high-level mixed chimerism and long-term survival. CONCLUSIONS: RIC-HCT with FluBU is a safe and effective strategy for obtaining high-level donor chimerism, immune reconstitution including B cell function, and long-term survival in patients with SCID. In patients with non-SCID PID, the results varied according to the subtype of the disease. Further prospective studies are required to optimize the conditioning regimen for non-SCID PID.


Subject(s)
Busulfan/therapeutic use , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/therapeutic use , Melphalan/therapeutic use , Primary Immunodeficiency Diseases/therapy , Transplantation Conditioning , Vidarabine/analogs & derivatives , Busulfan/pharmacokinetics , Child, Preschool , Drug Combinations , Female , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Leukocyte Count , Male , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/mortality , Retrospective Studies , Treatment Outcome , Vidarabine/therapeutic use
9.
J Allergy Clin Immunol ; 146(5): 1109-1120.e4, 2020 11.
Article in English | MEDLINE | ID: mdl-32298700

ABSTRACT

BACKGROUND: Interactions between the tumor necrosis factor (TNF) ligand superfamily and TNF receptor superfamily play critical roles in B-cell development and maturation. A proliferation-inducing ligand (APRIL), a member of the TNF ligand superfamily, is secreted from myeloid cells and known to induce the differentiation of memory B cells to plasmacytes. OBJECTIVE: We sought to elucidate the role of APRIL in B-cell differentiation and immunoglobulin production through the analysis of complete APRIL deficiency in human. METHODS: We performed whole exome sequencing in a patient with adult common variable immunodeficiency. His parents were in a consanguineous marriage. TNFSF13 mRNA and protein expression were analyzed in the primary cells and plasma from the patient and in cDNA-transfected cells and supernatants of the cultures in vitro. Immunologic analysis was performed by using flow cytometry and next-generation sequencing. Monocyte-derived dendritic cells differentiated from induced pluripotent stem cells (iPSC-moDCs) were cocultured with memory B cells from healthy controls to examine in vitro plasmacyte differentiation. RESULTS: We identified a homozygous frameshift mutation in TNFSF13, the gene encoding APRIL, in the patient. APRIL mRNA and protein were completely absent in the monocytes and iPSC-moDCs of the patient. In contrast to the results of previous animal model studies, the patient showed hypogammaglobulinemia with a markedly reduced level of plasmacytes in peripheral blood and a clearly increased level of circulating marginal zone B cells. Although iPSC-moDC-induced in vitro plasmacyte differentiation was reduced in the patient, recombinant APRIL supplementation corrected this abnormality. CONCLUSION: The first APRIL deficiency in an adult patient with common variable immunodeficiency revealed the role of APRIL in lifelong maintenance of plasmacytes and immunoglobulin production in humans.


Subject(s)
Agammaglobulinemia/genetics , Antibody Formation/genetics , B-Lymphocytes/immunology , Plasma Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Adult , Cell Differentiation , Cells, Cultured , Consanguinity , Humans , Immunologic Memory , Lymphocyte Activation , Male , Middle Aged , Mutation/genetics , Pedigree , Exome Sequencing
10.
J Clin Immunol ; 40(5): 729-740, 2020 07.
Article in English | MEDLINE | ID: mdl-32506361

ABSTRACT

PURPOSE: Owing to recent technological advancements, using next-generation sequencing (NGS) and the accumulation of clinical experiences worldwide, more than 420 genes associated with inborn errors of immunity (IEI) have been identified, which exhibit large genotypic and phenotypic variations. Consequently, NGS-based comprehensive genetic analysis, including whole-exome sequencing (WES), have become more valuable in the clinical setting and have contributed to earlier diagnosis, improved treatment, and prognosis. However, these approaches have the following disadvantages that need to be considered: a relatively low diagnostic rate, high cost, difficulties in the interpretation of each variant, and the risk of incidental findings. Thus, the objective of this study is to review our WES results of a large number of patients with IEI and to elucidate patient characteristics, which are related to the positive WES result. METHODS: We performed WES for 136 IEI patients with negative conventional screening results for candidate genes and classified these variants depending on validity of their pathogenicity. RESULTS: We identified disease-causing pathogenic mutations in 36 (26.5%) of the patients which were found in known IEI-causing genes. Although the overall diagnostic rate was not high and was not apparently correlated with the clinical subcategories and severity, we revealed that earlier onset with longer duration of diseases were associated with positive WES results, especially in pediatric cases. CONCLUSIONS: Most of the disease-causing germline mutations were located in the known IEI genes which could be predicted using patients' clinical characteristics. These results may be useful when considering appropriate genetic approaches in the clinical setting.


Subject(s)
Genotype , Germ-Line Mutation/genetics , Immunity/genetics , Mutation/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Genetic Association Studies , Genetic Diseases, Inborn , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Exome Sequencing , Young Adult
13.
J Infect Dis ; 218(5): 825-834, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29684201

ABSTRACT

Background: Some patients with genetic defects develop Epstein-Barr virus (EBV)-associated lymphoproliferative disorder (LPD)/lymphoma as the main feature. Hypomophic mutations can cause different clinical and laboratory manifestations from null mutations in the same genes. Methods: We sought to describe the clinical and immunologic phenotype of a 21-month-old boy with EBV-associated LPD who was in good health until then. A genetic and immunologic analysis was performed. Results: Whole-exome sequencing identified a novel compound heterozygous mutation of ZAP70 c.703-1G>A and c.1674G>A. A small amount of the normal transcript was observed. Unlike ZAP70 deficiency, which has been previously described as severe combined immunodeficiency with nonfunctional CD4+ T cells and absent CD8+ T cells, the patient had slightly low numbers of CD8+ T cells and a small amount of functional T cells. EBV-specific CD8+ T cells and invariant natural killer T (iNKT) cells were absent. The T-cell receptor repertoire, determined using next generation sequencing, was significantly restricted. Conclusions: Our patient showed that a hypomorphic mutation of ZAP70 can lead to EBV-associated LPD and that EBV-specific CD8+ T cells and iNKT cells are critically involved in immune response against EBV infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epstein-Barr Virus Infections/complications , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Mutant Proteins/genetics , Natural Killer T-Cells/immunology , ZAP-70 Protein-Tyrosine Kinase/genetics , Exome , Herpesvirus 4, Human/immunology , Heterozygote , Humans , Infant , Lymphoproliferative Disorders/genetics , Male , Mutant Proteins/metabolism , Mutation , Whole Genome Sequencing , ZAP-70 Protein-Tyrosine Kinase/metabolism
15.
J Clin Immunol ; 38(3): 300-306, 2018 04.
Article in English | MEDLINE | ID: mdl-29671114

ABSTRACT

OBJECTIVE: In the current study, we aimed to accurately evaluate donor/recipient or male/female chimerism in samples from patients who underwent hematopoietic stem cell transplantation (HSCT). METHODS: We designed the droplet digital polymerase chain reaction (ddPCR) for SRY and RPP30 to detect the male/female chimerism. We also developed mutation-specific ddPCR for four primary immunodeficiency diseases. RESULTS: The accuracy of the male/female chimerism analysis using ddPCR was confirmed by comparing the results with those of conventional methods (fluorescence in situ hybridization and short tandem repeat-PCR) and evaluating dilution assays. In particular, we found that this method was useful for analyzing small samples. Thus, this method could be used with patient samples, especially to sorted leukocyte subpopulations, during the early post-transplant period. Four mutation-specific ddPCR accurately detected post-transplant chimerism. CONCLUSION: ddPCR-based male/female chimerism analysis and mutation-specific ddPCR were useful for all HSCT, and these simple methods contribute to following the post-transplant chimerism, especially in disease-specific small leukocyte fractions.


Subject(s)
Chimerism , Immunologic Deficiency Syndromes/diagnosis , Transplantation Chimera/genetics , Alleles , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Deficiency Syndromes/therapy , In Situ Hybridization, Fluorescence , Male , Minisatellite Repeats , Mutation , Real-Time Polymerase Chain Reaction/methods , Transplantation, Homologous
16.
Pediatr Blood Cancer ; 65(2)2018 Feb.
Article in English | MEDLINE | ID: mdl-28960754

ABSTRACT

Autoimmune diseases in children are rare and can be difficult to diagnose.  Single causative genes have been identified for some pediatric autoimmune diseases. Such orphan diseases may not be diagnosed properly due to the variability of patients' phenotypes. Guidelines for the diagnostic process need to be developed. Fifteen patients with uncharacterized childhood autoimmune diseases with lymphoproliferation that had negative testing for autoimmune lymphoproliferative syndrome were subjected to whole-exome sequencing to identify genes associated with these conditions. Five causative genes, CTLA4, STAT3, TNFAIP3, IKZF1, and PSTPIP1, were identified. These genes should be considered as candidates for uncharacterized childhood autoimmune diseases with lymphoproliferation.


Subject(s)
Autoimmune Diseases/genetics , Lymphoproliferative Disorders/genetics , Neoplasm Proteins/genetics , Adolescent , Autoimmune Diseases/complications , Child , Child, Preschool , Female , Genome-Wide Association Study , Humans , Lymphoproliferative Disorders/complications , Male
18.
J Allergy Clin Immunol ; 140(1): 223-231, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27939403

ABSTRACT

BACKGROUND: Ikaros, which is encoded by IKZF1, is a transcriptional factor that play a critical role in hematopoiesis. Somatic IKZF1 alterations are known to be involved in the pathogenesis of leukemia in human subjects. Recently, immunodeficiency caused by germline IKZF1 mutation has been described. OBJECTIVE: We sought to describe the clinical and immunologic phenotypes of Japanese patients with heterozygous IKZF1 mutations. METHODS: We performed whole-exome sequencing in patients from a dysgammaglobulinemia or autoimmune disease cohort and used a candidate gene approach in 4 patients. Functional and laboratory studies, including detailed lymphopoiesis/hematopoiesis analysis in the bone marrow, were performed. RESULTS: Nine patients from 6 unrelated families were identified to have heterozygous germline mutations in IKZF1. Age of onset was 0 to 20 years (mean, 7.4 years). Eight of 9 patients presented with dysgammaglobulinemia accompanied by B-cell deficiency. Four of 9 patients had autoimmune disease, including immune thrombocytopenic purpura, IgA vasculitis, and systemic lupus erythematosus. Nonautoimmune pancytopenia was observed in 1 patient. All of the mutant Ikaros protein demonstrated impaired DNA binding to the target sequence and abnormal diffuse nuclear localization. Flow cytometric analysis of bone marrow revealed reduced levels of common lymphoid progenitors and normal development of pro-B to pre-B cells. CONCLUSIONS: Germline heterozygous IKZF1 mutations cause dysgammaglobulinemia; hematologic abnormalities, including B-cell defect; and autoimmune diseases.


Subject(s)
Autoimmune Diseases/genetics , Hematologic Diseases/genetics , Ikaros Transcription Factor/genetics , Adolescent , Adult , Autoimmune Diseases/immunology , Autoimmunity , B-Lymphocytes/immunology , Child , Female , Germ-Line Mutation , Hematologic Diseases/immunology , Hematopoiesis/genetics , Humans , Ikaros Transcription Factor/immunology , Lymphocyte Count , Male , T-Lymphocytes/immunology , Young Adult
19.
Allergol Int ; 67(1): 43-54, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28684198

ABSTRACT

Primary immunodeficiencies (PIDs) are a heterogeneous group of inherited diseases of the immune system. The definite diagnosis of PID is ascertained by genetic analysis; however, this takes time and is costly. Flow cytometry provides a rapid and highly sensitive tool for diagnosis of PIDs. Flow cytometry can evaluate specific cell populations and subpopulations, cell surface, intracellular and intranuclear proteins, biologic effects associated with specific immune defects, and certain functional immune characteristics, each being useful for the diagnosis and evaluation of PIDs. Flow cytometry effectively identifies major forms of PIDs, including severe combined immunodeficiency, X-linked agammaglobulinemia, hyper IgM syndromes, Wiskott-Aldrich syndrome, X-linked lymphoproliferative syndrome, familial hemophagocytic lymphohistiocytosis, autoimmune lymphoproliferative syndrome, IPEX syndrome, CTLA 4 haploinsufficiency and LRBA deficiency, IRAK4 and MyD88 deficiencies, Mendelian susceptibility to mycobacterial disease, chronic mucocuneous candidiasis, and chronic granulomatous disease. While genetic analysis is the definitive approach to establish specific diagnoses of PIDs, flow cytometry provides a tool to effectively evaluate patients with PIDs at relatively low cost.


Subject(s)
Flow Cytometry/methods , Immunologic Deficiency Syndromes/diagnosis , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology
20.
Clin Immunol ; 183: 112-120, 2017 10.
Article in English | MEDLINE | ID: mdl-28780374

ABSTRACT

X-linked severe combined immunodeficiency (X-SCID), caused by defects in the common gamma chain, is typically characterized by T and NK cell defects with the presence of B cells. T cell dysfunction and impaired class-switch recombination of B cells mean that patients typically have defects in class-switched immunoglobulins (IgG, IgA, and IgE) with detectable IgM. Here, we describe two patients with X-SCID with IgG1 gammopathy, in whom we identified maternal T and B cell engraftment. Exclusively, maternal B cells were found among the IgD-CD27+ class-switched memory B cells, whereas the patients' B cells remained naïve. In vitro stimulation with CD40L+IL-21 revealed that peripheral blood cells from both patients produced only IgG1. Class-switched maternal B cells had restricted receptor repertoires with various constant regions and few somatic hypermutations. In conclusion, engrafted maternal B cells underwent class-switch recombination and produced immunoglobulin, causing hypergammaglobulinemia in patients with X-SCID.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin G/immunology , Paraproteinemias/immunology , T-Lymphocytes/immunology , X-Linked Combined Immunodeficiency Diseases/immunology , Carrier Proteins/genetics , Flow Cytometry , Humans , Immunoglobulin Class Switching , Immunophenotyping , In Vitro Techniques , Infant , Infant, Newborn , Interleukin Receptor Common gamma Subunit/genetics , Male , Paraproteinemias/genetics , Reverse Transcriptase Polymerase Chain Reaction , X-Linked Combined Immunodeficiency Diseases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL