Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Nat Immunol ; 13(6): 612-20, 2012 May 13.
Article in English | MEDLINE | ID: mdl-22581261

ABSTRACT

The adaptors DOCK8 and MyD88 have been linked to serological memory. Here we report that DOCK8-deficient patients had impaired antibody responses and considerably fewer CD27(+) memory B cells. B cell proliferation and immunoglobulin production driven by Toll-like receptor 9 (TLR9) were considerably lower in DOCK8-deficient B cells, but those driven by the costimulatory molecule CD40 were not. In contrast, TLR9-driven expression of AICDA (which encodes the cytidine deaminase AID), the immunoglobulin receptor CD23 and the costimulatory molecule CD86 and activation of the transcription factor NF-κB, the kinase p38 and the GTPase Rac1 were intact. DOCK8 associated constitutively with MyD88 and the tyrosine kinase Pyk2 in normal B cells. After ligation of TLR9, DOCK8 became tyrosine-phosphorylated by Pyk2, bound the Src-family kinase Lyn and linked TLR9 to a Src-kinase Syk-transcription factor STAT3 cascade essential for TLR9-driven B cell proliferation and differentiation. Thus, DOCK8 functions as an adaptor in a TLR9-MyD88 signaling pathway in B cells.


Subject(s)
B-Lymphocytes/immunology , Guanine Nucleotide Exchange Factors/immunology , Immunologic Memory/immunology , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptor 9/immunology , Adolescent , Animals , Cell Differentiation/immunology , Child , Child, Preschool , Flow Cytometry , Focal Adhesion Kinase 2/immunology , Humans , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Phosphorylation , STAT3 Transcription Factor/immunology , src-Family Kinases/immunology
3.
J Allergy Clin Immunol ; 152(6): 1597-1606, 2023 12.
Article in English | MEDLINE | ID: mdl-37595757

ABSTRACT

BACKGROUND: Inborn errors of immunity are mostly monogenic. However, disease phenotype and outcome may be modified by the coexistence of a second gene defect. OBJECTIVE: We sought to identify the genetic basis of the disease in a patient who experienced bleeding episodes, pancytopenia, hepatosplenomegaly, and recurrent pneumonia that resulted in death. METHODS: Genetic analysis was done using next-generation sequencing. Protein expression and phosphorylation were determined by immunoblotting. T-cell proliferation and F-actin levels were studied by flow cytometry. RESULTS: The patient harbored 2 homozygous deletions in STX11 (c.369_370del, c.374_376del; p.V124fs60∗) previously associated with familial hemophagocytic lymphohistiocytosis and a novel homozygous missense variant in SLP76 (c.767C>T; p.T256I) that resulted in an approximately 85% decrease in SLP76 levels and absent T-cell proliferation. The patient's heterozygous family members showed an approximately 50% decrease in SLP76 levels but normal immune function. SLP76-deficient J14 Jurkat cells did not express SLP76 and had decreased extracellular signal-regulated kinase signaling, basal F-actin levels, and polymerization following T-cell receptor stimulation. Reconstitution of J14 cells with T256I mutant SLP76 resulted in low protein expression and abnormal extracellular signal-regulated kinase phosphorylation and F-actin polymerization after T-cell receptor activation compared with normal expression and J14 function when wild-type SLP76 was introduced. CONCLUSIONS: The hypomorphic mutation in SLP76 tones down the hyperinflammation due to STX11 deletion, resulting in a combined immunodeficiency that overshadows the hemophagocytic lymphohistiocytosis phenotype. To our knowledge, this study represents the first report of the opposing effects of 2 gene defects on the disease in a patient with an inborn error of immunity.


Subject(s)
Actins , Lymphohistiocytosis, Hemophagocytic , Humans , Extracellular Signal-Regulated MAP Kinases , Lymphohistiocytosis, Hemophagocytic/genetics , Mutation , Qa-SNARE Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Signal Transduction
4.
J Allergy Clin Immunol ; 152(1): 182-194.e7, 2023 07.
Article in English | MEDLINE | ID: mdl-36758835

ABSTRACT

BACKGROUND: Inborn errors of immunity have been implicated in causing immune dysregulation, including allergic diseases. STAT6 is a key regulator of allergic responses. OBJECTIVES: This study sought to characterize a novel gain-of-function STAT6 mutation identified in a child with severe allergic manifestations. METHODS: Whole-exome and targeted gene sequencing, lymphocyte characterization, and molecular and functional analyses of mutated STAT6 were performed. RESULTS: This study reports a child with a missense mutation in the DNA binding domain of STAT6 (c.1114G>A, p.E372K) who presented with severe atopic dermatitis, eosinophilia, and elevated IgE. Naive lymphocytes from the affected patient displayed increased TH2- and suppressed TH1- and TH17-cell responses. The mutation augmented both basal and cytokine-induced STAT6 phosphorylation without affecting dephosphorylation kinetics. Treatment with the Janus kinase 1/2 inhibitor ruxolitinib reversed STAT6 hyperresponsiveness to IL-4, normalized TH1 and TH17 cells, suppressed the eosinophilia, and improved the patient's atopic dermatitis. CONCLUSIONS: This study identified a novel inborn error of immunity due to a STAT6 gain-of-function mutation that gave rise to severe allergic dysregulation. Janus kinase inhibitor therapy could represent an effective targeted treatment for this disorder.


Subject(s)
Dermatitis, Atopic , Eosinophilia , Hypersensitivity , Child , Humans , Transcription Factors/genetics , Gain of Function Mutation , Dermatitis, Atopic/genetics , Hypersensitivity/genetics , Eosinophilia/genetics , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Th2 Cells
5.
Clin Immunol ; 257: 109813, 2023 12.
Article in English | MEDLINE | ID: mdl-37898412

ABSTRACT

BACKGROUND: RASGRP1-deficiency results in an immune dysregulation and immunodeficiency that manifest as autoimmunity, lymphoproliferation, lymphopenia, defective T cell function, and increased incidence of Epstein-Bar Virus infections and lymphomas. OBJECTIVE: To investigate the mechanism of autoimmune hemolytic anemia and infections in a male patient of consanguineous parents from Lebanon. METHODS: Genetic diagnosis was obtained using next generation and Sanger sequencing. Protein expression and phosphorylation were determined by immunoblotting. T and B cell development and function were studied by flow cytometry. Cytokine and immunoglobulin secretions were quantified by enzyme-linked immunosorbent assay. RESULTS: The patient suffered from severe lymphopenia especially affecting the T cell compartment. Genetic analysis revealed a homozygous insertion of adenine at position 1396_1397 in RASGRP1 that abolished protein expression and downstream Ras signaling. T cells from the patient showed severe activation defects resulting in uncontrolled Epstein-Bar Virus-induced B cell proliferation. B cells from the patient were normal. CONCLUSION: This report expands the spectrum of mutations in patients with RasGRP1 deficiency, and provides evidence for the important role RasGRP1 plays in the ability of T cells to control Epstein-Bar Virus-induced B cell proliferation. CLINICAL IMPLICATIONS: Following diagnosis, the patient will be maintained on oral valganciclovir and monitored regularly for Epstein-Bar Virus infections to avoid the development of Epstein-Bar Virus- induced B cell lymphoma. He is also candidate for hematopoietic stem cell transplantation.


Subject(s)
Epstein-Barr Virus Infections , Immunologic Deficiency Syndromes , Lymphopenia , Humans , Male , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Herpesvirus 4, Human , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Lymphopenia/complications , Lymphopenia/genetics , Mutation
6.
Br J Cancer ; 127(6): 976-987, 2022 10.
Article in English | MEDLINE | ID: mdl-35662275

ABSTRACT

Glioblastoma is the most common and aggressive primary malignant brain tumour. The prognosis of patients with glioblastoma is poor, and their overall survival averages at 1 year, despite advances made in cancer therapy. The emergence of immunotherapy, a strategy that targets the natural mechanisms of immune evasion by cancerous cells, has revolutionised the treatment of melanoma, lung cancer and other solid tumours; however, immunotherapy failed to improve the prognosis of patients with glioblastoma. This is attributed to the fact that glioblastoma is endowed with numerous mechanisms of resistance that include the intrinsic resistance, which refers to the location of the tumour within the brain and the nature of the blood-brain barrier, as well as the adaptive and acquired resistance that result from the tumour heterogeneity and its immunosuppressive microenvironment. Glioblastoma is notorious for its inter and intratumoral heterogeneity, which, coupled with its spatial and temporal evolution, limits its immunogenicity. In addition, the tumour microenvironment is enriched with immunosuppressive cells and molecules that hinder the reactivity of cytotoxic immune cells and the success of immunotherapies. In this article, we review the mechanisms of resistance of glioblastoma to immunotherapy and discuss treatment strategies to overcome them worthy of further exploration.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/therapy , Glioblastoma/therapy , Humans , Immunologic Factors , Immunotherapy , Tumor Microenvironment
7.
J Allergy Clin Immunol ; 146(1): 192-202, 2020 07.
Article in English | MEDLINE | ID: mdl-31862378

ABSTRACT

BACKGROUND: The gene AK2 encodes the phosphotransferase adenylate kinase 2 (AK2). Human variants in AK2 cause reticular dysgenesis, a severe combined immunodeficiency with agranulocytosis, lymphopenia, and sensorineural deafness that requires hematopoietic stem cell transplantation for survival. OBJECTIVE: We investigated the mechanisms underlying recurrent sinopulmonary infections and hypogammaglobulinemia in 15 patients, ranging from 3 to 34 years of age, from 9 kindreds. Only 2 patients, both of whom had mildly impaired T-cell proliferation, each had a single clinically significant opportunistic infection. METHODS: Patient cells were studied with next-generation DNA sequencing, tandem mass spectrometry, and assays of lymphocyte and mitochondrial function. RESULTS: We identified 2 different homozygous variants in AK2. AK2G100S and AK2A182D permit residual protein expression, enzymatic activity, and normal numbers of neutrophils and lymphocytes. All but 1 patient had intact hearing. The patients' B cells had severely impaired proliferation and in vitro immunoglobulin secretion. With activation, the patients' B cells exhibited defective mitochondrial respiration and impaired regulation of mitochondrial membrane potential and quality. Although activated T cells from the patients with opportunistic infections demonstrated impaired mitochondrial function, the mitochondrial quality in T cells was preserved. Consistent with the capacity of activated T cells to utilize nonmitochondrial metabolism, these findings revealed a less strict cellular dependence of T-cell function on AK2 activity. Chemical inhibition of ATP synthesis in control T and B cells similarly demonstrated the greater dependency of B cells on mitochondrial function. CONCLUSIONS: Our patients demonstrate the in vivo sequelae of the cell-specific requirements for the functions of AK2 and mitochondria, particularly in B-cell activation and antibody production.


Subject(s)
Adenylate Kinase/genetics , B-Lymphocytes/immunology , Homozygote , Lymphocyte Activation/genetics , Mutation, Missense , Severe Combined Immunodeficiency/genetics , Adenylate Kinase/immunology , Adult , Amino Acid Substitution , Child , Child, Preschool , Female , Humans , Male , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology
8.
Clin Immunol ; 219: 108573, 2020 10.
Article in English | MEDLINE | ID: mdl-32814211

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency disorder (PID) characterized by microthrombocytopenia, bloody diarrhea, eczema, recurrent infections, and a high incidence of autoimmunity and malignancy. OBJECTIVE: To investigate the mechanism of thrombocytopenia and infections in four boys of consanguineous parents from Lebanon. METHODS: Patient gDNA was studied using Next Generation Sequencing and Sanger Sequencing. Protein expression was determined by immunoblotting, and mRNA expression by semi-quantitative RT-PCR. F-actin polymerization and cellular proliferation were assayed by flow cytometry. RESULTS: We identified a threonine to a methionine change at position 45 (T45M) of the WAS protein (WASp) that abolished protein expression and disturbed F-actin polymerization and T cell proliferation, but not B cell proliferation. In addition, the levels of the WAS-interacting protein (WIP) were significantly decreased in the patients. CONCLUSION: The mutation identified severely destabilizes WASp and affects the downstream signaling events important for T cell function, but not B cell function. It was previously known that the stability of WASp depends on WIP. In this manuscript, we report that the stability of WIP also depends on WASp. Finally, it is important to suspect X-linked PIDs even in consanguineous families. CLINICAL IMPLICATIONS: The patients are above the optimal age for transplant in WAS, and it is difficult to identify one or more donors for four patients, therefore, they represent ideal candidates for gene therapy or interleukin-2 therapy.


Subject(s)
Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , B-Lymphocytes/immunology , Child , Child, Preschool , Consanguinity , Humans , Lebanon , Male , Mutation , Siblings , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome/immunology , X-Linked Combined Immunodeficiency Diseases/immunology
9.
J Allergy Clin Immunol ; 141(3): 1060-1073.e3, 2018 03.
Article in English | MEDLINE | ID: mdl-28629746

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 ß 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.


Subject(s)
Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/immunology , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Genotype , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Signal Transduction , Carrier Proteins/genetics , Carrier Proteins/immunology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Ectodermal Dysplasia/pathology , Genetic Diseases, X-Linked/pathology , HEK293 Cells , Humans , Immunologic Deficiency Syndromes/pathology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Interleukin-6/genetics , Interleukin-6/immunology , NF-KappaB Inhibitor alpha/genetics , NF-KappaB Inhibitor alpha/immunology , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/immunology , Point Mutation , Primary Immunodeficiency Diseases , Proto-Oncogene Mas , Signal Transduction/genetics , Signal Transduction/immunology
10.
Clin Immunol ; 187: 68-75, 2018 02.
Article in English | MEDLINE | ID: mdl-29051008

ABSTRACT

Defects in DNA Recombination due to mutations in RAG1/2 or DCLRE1C result in combined immunodeficiency (CID) with a range of disease severity. We present the clinical, immunologic and molecular characteristics of 21 patients with defects in RAG1, RAG2 or DCLRE1C, who accounted for 24% of combined immune deficiency cases in the Kuwait National Primary Immunodeficiency Disorders Registry. The distribution of the patients was as follow: 8 with RAG1 deficiency, 6 with RAG2 deficiency and 7 with DCLRE1C deficiency. Nine patients presented with SCID, 6 with OS, 2 with leaky SCID and 4 with CID and granuloma and/or autoimmunity (CID-G/AI). Eight patients [(7 SCID and 1 OS) (38%)] received hematopoietic stem cell transplant (HSCT). The median age of HSCT was 11.5months and the median time from diagnosis to HSCT was 6months. Fifty percent of the transplanted patients are alive while only 23% of the untransplanted ones are alive.


Subject(s)
Autoimmune Diseases/immunology , Granuloma/immunology , Severe Combined Immunodeficiency/immunology , V(D)J Recombination/genetics , Adolescent , Anti-Bacterial Agents/therapeutic use , Autoimmune Diseases/genetics , Autoimmune Diseases/therapy , Child , Child, Preschool , DNA-Binding Proteins/genetics , Endonucleases/genetics , Female , Genetic Profile , Granuloma/genetics , Granuloma/therapy , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/genetics , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/therapy , Immunologic Factors/therapeutic use , Infant , Kuwait , Male , Nuclear Proteins/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy
11.
N Engl J Med ; 372(25): 2409-22, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26083206

ABSTRACT

Background Combined immunodeficiencies are marked by inborn errors of T-cell immunity in which the T cells that are present are quantitatively or functionally deficient. Impaired humoral immunity is also common. Patients have severe infections, autoimmunity, or both. The specific molecular, cellular, and clinical features of many types of combined immunodeficiencies remain unknown. Methods We performed genetic and cellular immunologic studies involving five unrelated children with early-onset invasive bacterial and viral infections, lymphopenia, and defective T-cell, B-cell, and natural killer (NK)-cell responses. Two patients died early in childhood; after allogeneic hematopoietic stem-cell transplantation, the other three had normalization of T-cell function and clinical improvement. Results We identified biallelic mutations in the dedicator of cytokinesis 2 gene (DOCK2) in these five patients. RAC1 activation was impaired in the T cells. Chemokine-induced migration and actin polymerization were defective in the T cells, B cells, and NK cells. NK-cell degranulation was also affected. Interferon-α and interferon-λ production by peripheral-blood mononuclear cells was diminished after viral infection. Moreover, in DOCK2-deficient fibroblasts, viral replication was increased and virus-induced cell death was enhanced; these conditions were normalized by treatment with interferon alfa-2b or after expression of wild-type DOCK2. Conclusions Autosomal recessive DOCK2 deficiency is a new mendelian disorder with pleiotropic defects of hematopoietic and nonhematopoietic immunity. Children with clinical features of combined immunodeficiencies, especially with early-onset, invasive infections, may have this condition. (Supported by the National Institutes of Health and others.).


Subject(s)
Genetic Diseases, Inborn/genetics , Guanine Nucleotide Exchange Factors/genetics , Immunologic Deficiency Syndromes/genetics , Mutation , T-Lymphocytes/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Child, Preschool , Fatal Outcome , Female , GTPase-Activating Proteins , Genes, Recessive , Genetic Diseases, Inborn/therapy , Guanine Nucleotide Exchange Factors/deficiency , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Deficiency Syndromes/therapy , Infant , Killer Cells, Natural/immunology , Male , Pedigree , T-Lymphocytes/metabolism , rac1 GTP-Binding Protein/metabolism
12.
J Clin Immunol ; 37(8): 811-819, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29058101

ABSTRACT

PURPOSE: The dedicator of cytokinesis 8 (DOCK8) deficiency is an autosomal recessive-combined immunodeficiency whose clinical spectra include recurrent infections, autoimmunity, malignancies, elevated serum IgE, eczema, and food allergies. Here, we report on patients with loss of function DOCK8 mutations with profound immune dysregulation suggestive of an immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX)-like disorder. METHODS: Immunophenotyping of lymphocyte subpopulations and analysis of DOCK8 protein expression were evaluated by flow cytometry. T regulatory (Treg) cells were isolated by cell sorting, and their suppressive activity was analyzed by flow cytometry. Gene mutational analysis was performed by whole-exome and Sanger sequencing. RESULTS: Patient 1 (P1) presented at 10 months of age with chronic severe diarrhea and active colitis in the absence of an infectious trigger, severe eczema with elevated serum IgE, and autoimmune hemolytic anemia, suggestive of an IPEX-related disorder. Whole-exome sequencing revealed a homozygous nonsense mutation in DOCK8 at the DOCK-homology region (DHR)-1 (c.1498C>T; p. R500X). Patient P2, a cousin of P1 who carries the same DOCK8 nonsense mutation, presented with eczema and recurrent ear infections in early infancy, and she developed persistent diarrhea by 3 years of age. Patient P3 presented with lymphoproliferation, severe eczema with allergic dysregulation, and chronic diarrhea with colitis. She harbored a homozygous loss of function DOCK8 mutation (c.2402 -1G→A). Treg cell function was severely compromised by both DOCK8 mutations. CONCLUSION: DOCK8 deficiency may present severe immune dysregulation with features that may overlap with those of IPEX and other IPEX-like disorders.


Subject(s)
Chromosome Disorders/diagnosis , Diabetes Mellitus, Type 1/congenital , Diarrhea/diagnosis , Genetic Diseases, X-Linked/diagnosis , Guanine Nucleotide Exchange Factors/genetics , Hypersensitivity/diagnosis , Immune System Diseases/congenital , Infections/diagnosis , Mutation/genetics , T-Lymphocytes, Regulatory/immunology , Anemia, Hemolytic , Child, Preschool , Diabetes Mellitus, Type 1/diagnosis , Female , Genes, Recessive/genetics , Humans , Immune System Diseases/diagnosis , Immunoglobulin E/metabolism , Immunophenotyping , Infant
13.
J Immunol ; 194(10): 4750-8, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25870239

ABSTRACT

The Rho GTPase Cdc42 coordinates regulation of the actin and the microtubule cytoskeleton by binding and activating the Wiskott-Aldrich syndrome protein. We sought to define the role of intrinsic expression of Cdc42 by mature B cells in their activation and function. Mice with inducible deletion of Cdc42 in mature B cells formed smaller germinal centers and had a reduced Ab response, mostly of low affinity to T cell-dependent Ag, compared with wild-type (WT) controls. Spreading formation of long protrusions that contain F-actin, microtubules, and Cdc42-interacting protein 4, and assumption of a dendritic cell morphology in response to anti-CD40 plus IL-4 were impaired in Cdc42-deficient B cells compared with WT B cells. Cdc42-deficient B cells had an intact migratory response to chemokine in vitro, but their homing to the B cell follicles in the spleen in vivo was significantly impaired. Cdc42-deficient B cells induced a skewed cytokine response in CD4(+) T cells, compared with WT B cells. Our results demonstrate a critical role for Cdc42 in the motility of mature B cells, their cognate interaction with T cells, and their differentiation into Ab-producing cells.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Activation/immunology , cdc42 GTP-Binding Protein/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Cell Movement/immunology , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
14.
J Allergy Clin Immunol ; 137(3): 879-88.e2, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26476480

ABSTRACT

BACKGROUND: Coronin-1A (CORO1A) is a regulator of actin dynamics important for T-cell homeostasis. CORO1A deficiency causes T(-)B(+) natural killer-positive severe combined immunodeficiency or T-cell lymphopenia with severe viral infections. However, because all known human mutations in CORO1A abrogate protein expression, the role of the protein's functional domains in host immunity is unknown. OBJECTIVE: We sought to identify the cause of the primary immunodeficiency in 2 young adult siblings with a history of disseminated varicella, cutaneous warts, and CD4(+) T-cell lymphopenia. METHODS: We performed immunologic, genetic, and biochemical studies in the patients, family members, and healthy control subjects. RESULTS: Both patients had CD4(+) T-cell lymphopenia and decreased lymphocyte proliferation to mitogens. IgG, IgM, IgA, and specific antibody responses were normal. Whole-genome sequencing identified a homozygous frameshift mutation in CORO1A disrupting the last 2 C-terminal domains by replacing 61 amino acids with a novel 91-amino-acid sequence. The CORO1A(S401fs) mutant was expressed in the patients' lymphocytes at a level comparable with that of wild-type CORO1A in normal lymphocytes but did not oligomerize and had impaired cytoskeletal association. CORO1A(S401fs) was associated with increased filamentous actin accumulation in T cells, severely defective thymic output, and impaired T-cell survival but normal calcium flux and cytotoxicity, demonstrating the importance of CORO1A oligomerization and subcellular localization in T-cell homeostasis. CONCLUSIONS: We describe a truncating mutation in CORO1A that permits protein expression and survival into young adulthood. Our studies demonstrate the importance of intact CORO1A C-terminal domains in thymic egress and T-cell survival, as well as in defense against viral pathogens.


Subject(s)
Cytoskeleton/metabolism , Homozygote , Microfilament Proteins/genetics , Mutation , Protein Multimerization , Virus Diseases/etiology , Virus Diseases/metabolism , Actins/chemistry , Actins/metabolism , Adolescent , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Degranulation/genetics , Cell Degranulation/immunology , Cell Survival/genetics , DNA Mutational Analysis , Female , Frameshift Mutation , Humans , Immunoglobulins/blood , Immunoglobulins/immunology , Lymphocyte Count , Lymphopenia , Male , Mice , Microfilament Proteins/chemistry , Microfilament Proteins/metabolism , Pedigree , Phenotype , Protein Multimerization/genetics , Protein Transport , Siblings , Signal Transduction , Skin Diseases/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Virus Diseases/diagnosis , Warts/pathology
15.
J Allergy Clin Immunol ; 138(3): 852-859.e3, 2016 09.
Article in English | MEDLINE | ID: mdl-27130861

ABSTRACT

BACKGROUND: Dedicator of cytokinesis 8 (DOCK8) deficiency can be cured by allogeneic hematopoietic stem cell transplantation (HSCT). Reports of outcomes are still limited. OBJECTIVE: We sought to analyze the results of HSCT in patients with DOCK8 deficiency and report whether approaches resulting in mixed chimerism result in clinically relevant immune reconstitution. METHODS: We performed a retrospective chart review of 11 patients with DOCK8 deficiency and measured DOCK8 expression and cytokine production. RESULTS: Of 11 patients, 7 received HSCT from related and 4 from unrelated donors; 9 patients received busulfan-based conditioning regimens. Survival was excellent (10 [91%] of 11 patients alive), including a patient who had undergone liver transplantation. Patients showed significant improvements in the frequency and severity of infections. Although eczema resolved in all, food allergies and high IgE levels persisted in some patients. Lymphopenia, eosinophilia, low numbers of naive CD8(+) T cells and switched memory B cells, and TH1/TH2 cytokine imbalance improved in most patients. Although the 8 matched related or unrelated donor recipients had full donor chimerism, all 3 recipients of mismatched unrelated donor HSCT had high levels of donor T-cell chimerism and low B-cell and myeloid cell chimerism (0% to 46%). Almost all switched memory B cells were of donor origin. All patients, including those with mixed chimerism, mounted robust antibody responses to vaccination. CONCLUSION: Allogeneic HSCT ameliorated the infectious and atopic symptoms of patients with DOCK8 deficiency. In patients with mixed chimerism, selective advantage for donor-derived T cells and switched memory B cells promoted restoration of cellular and humoral immunity and protection against opportunistic infection.


Subject(s)
Guanine Nucleotide Exchange Factors/deficiency , Hematopoietic Stem Cell Transplantation , Immunologic Deficiency Syndromes/immunology , B-Lymphocytes/immunology , Child , Child, Preschool , Cytokines/immunology , Female , Humans , Leukocytes, Mononuclear/immunology , Male , T-Lymphocytes/immunology , Treatment Outcome
16.
Clin Immunol ; 166-167: 100-2, 2016 05.
Article in English | MEDLINE | ID: mdl-27063589

ABSTRACT

ORAI1 is the pore-forming subunit of the calcium release-activated calcium channel responsible for calcium influx into cells triggered by endoplasmic reticulum store depletion. We report here a patient with severe combined immunodeficiency and absent store-operated calcium entry due to a novel mutation in ORAI1 that results in the expression of a C-terminally truncated protein that abolishes ORAI1 binding to STIM1.


Subject(s)
Mutation , Neoplasm Proteins/genetics , ORAI1 Protein/genetics , Severe Combined Immunodeficiency/genetics , Stromal Interaction Molecule 1/genetics , Base Sequence , Exome/genetics , Fatal Outcome , Female , HEK293 Cells , Homozygote , Humans , Infant , Neoplasm Proteins/metabolism , ORAI1 Protein/metabolism , Protein Binding/genetics , Sequence Analysis, DNA , Sequence Deletion , Severe Combined Immunodeficiency/metabolism , Stromal Interaction Molecule 1/metabolism
17.
Clin Immunol ; 171: 65-66, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27538774

ABSTRACT

Whole exome sequencing is increasingly used in the diagnosis of primary immunodeficiencies due to the overlapping and atypical presentations of these disorders. We report two patients who presented with recurrent infections and early onset colitis. They were investigated by whole exome sequencing due to suspicion of primary immunodeficiency and found to have mutations in pyrin known to cause familial Mediterranean fever.


Subject(s)
Colitis/genetics , Familial Mediterranean Fever/genetics , Immunologic Deficiency Syndromes/genetics , Pyrin/genetics , Child, Preschool , Colitis/diagnosis , Consanguinity , Diagnosis, Differential , Familial Mediterranean Fever/diagnosis , Female , Humans , Immunologic Deficiency Syndromes/diagnosis , Infant , Male , Mutation
18.
J Allergy Clin Immunol ; 135(1): 217-27, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25468195

ABSTRACT

BACKGROUND: A number of heritable immune dysregulatory diseases result from defects affecting regulatory T (Treg) cell development, function, or both. They include immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, which is caused by mutations in forkhead box P3 (FOXP3), and IPEX-like disorders caused by mutations in IL-2 receptor α (IL2RA), signal transducer and activator of transcription 5b (STAT5b), and signal transducer and activator of transcription 1 (STAT1). However, the genetic defects underlying many cases of IPEX-like disorders remain unknown. OBJECTIVE: We sought to identify the genetic abnormalities in patients with idiopathic IPEX-like disorders. METHODS: We performed whole-exome and targeted gene sequencing and phenotypic and functional analyses of Treg cells. RESULTS: A child who presented with an IPEX-like syndrome and severe Treg cell deficiency was found to harbor a nonsense mutation in the gene encoding LPS-responsive beige-like anchor (LRBA), which was previously implicated as a cause of common variable immunodeficiency with autoimmunity. Analysis of subjects with LRBA deficiency revealed marked Treg cell depletion; profoundly decreased expression of canonical Treg cell markers, including FOXP3, CD25, Helios, and cytotoxic T lymphocyte-associated antigen 4; and impaired Treg cell-mediated suppression. There was skewing in favor of memory T cells and intense autoantibody production, with marked expansion of T follicular helper and contraction of T follicular regulatory cells. Whereas the frequency of recent thymic emigrants and the differentiation of induced Treg cells were normal, LRBA-deficient T cells exhibited increased apoptosis and reduced activities of the metabolic sensors mammalian target of rapamycin complexes 1 and 2. CONCLUSION: LRBA deficiency is a novel cause of IPEX-like syndrome and Treg cell deficiency associated with metabolic dysfunction and increased apoptosis of Treg cells.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , T-Lymphocytes, Regulatory/immunology , Adolescent , Autoantibodies/immunology , Child, Preschool , Diabetes Mellitus, Type 1/congenital , Diarrhea , Female , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Humans , Immune System Diseases/congenital , Interleukin-10/immunology , Male , Mutation
19.
J Allergy Clin Immunol ; 136(2): 402-12, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25724123

ABSTRACT

BACKGROUND: Mutations in dedicator of cytokinesis 8 (DOCK8) cause a combined immunodeficiency (CID) also classified as autosomal recessive (AR) hyper-IgE syndrome (HIES). Recognizing patients with CID/HIES is of clinical importance because of the difference in prognosis and management. OBJECTIVES: We sought to define the clinical features that distinguish DOCK8 deficiency from other forms of HIES and CIDs, study the mutational spectrum of DOCK8 deficiency, and report on the frequency of specific clinical findings. METHODS: Eighty-two patients from 60 families with CID and the phenotype of AR-HIES with (64 patients) and without (18 patients) DOCK8 mutations were studied. Support vector machines were used to compare clinical data from 35 patients with DOCK8 deficiency with those from 10 patients with AR-HIES without a DOCK8 mutation and 64 patients with signal transducer and activator of transcription 3 (STAT3) mutations. RESULTS: DOCK8-deficient patients had median IgE levels of 5201 IU, high eosinophil levels of usually at least 800/µL (92% of patients), and low IgM levels (62%). About 20% of patients were lymphopenic, mainly because of low CD4(+) and CD8(+) T-cell counts. Fewer than half of the patients tested produced normal specific antibody responses to recall antigens. Bacterial (84%), viral (78%), and fungal (70%) infections were frequently observed. Skin abscesses (60%) and allergies (73%) were common clinical problems. In contrast to STAT3 deficiency, there were few pneumatoceles, bone fractures, and teething problems. Mortality was high (34%). A combination of 5 clinical features was helpful in distinguishing patients with DOCK8 mutations from those with STAT3 mutations. CONCLUSIONS: DOCK8 deficiency is likely in patients with severe viral infections, allergies, and/or low IgM levels who have a diagnosis of HIES plus hypereosinophilia and upper respiratory tract infections in the absence of parenchymal lung abnormalities, retained primary teeth, and minimal trauma fractures.


Subject(s)
Bacterial Infections/complications , Guanine Nucleotide Exchange Factors/deficiency , Job Syndrome/complications , Phenotype , Skin Diseases/complications , Virus Diseases/complications , Adolescent , Adult , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Antigens, Viral/blood , Antigens, Viral/immunology , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/mortality , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Child , Child, Preschool , Eosinophils/immunology , Eosinophils/pathology , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Humans , Immunoglobulin E/blood , Immunoglobulin E/genetics , Immunoglobulin M/blood , Immunoglobulin M/genetics , Infant , Job Syndrome/genetics , Job Syndrome/immunology , Job Syndrome/mortality , Lymphocyte Count , Male , Middle Aged , Mutation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Skin Diseases/genetics , Skin Diseases/immunology , Skin Diseases/mortality , Support Vector Machine , Survival Analysis , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/mortality
20.
Clin Immunol ; 158(2): 167-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25869295

ABSTRACT

The Recombination Activating Genes (RAG) 1/2 are important for the development and function of T and B cells. Loss of RAG1/2 function results in severe combined immunodeficiency (SCID), which could lead to early death. We studied the prevalence of RAG1/2 mutations in ten SCID patients in Egypt. We identified two novel homozygous nonsense mutations in RAG1, a novel homozygous deletion, and a previously reported homozygous missense mutation from four patients, as well as two homozygous mutations in RAG2 from the same patient. Prenatal diagnosis performed in the mother of a patient with RAG1 deficiency determined that the fetus was heterozygous for the same mutation. This represents the first report on RAG1/2 mutations in SCID patients in Egypt. The early diagnosis dramatically affects the outcome of the disease by allowing bone marrow transplantation at an early age, and providing prenatal diagnosis and genetic counseling for families with a history of SCID.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Severe Combined Immunodeficiency/epidemiology , Severe Combined Immunodeficiency/genetics , Child, Preschool , DNA-Binding Proteins/genetics , Egypt/epidemiology , Female , Homeodomain Proteins/genetics , Humans , Infant , Male , Mutation , Nuclear Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL