Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Nat Immunol ; 25(5): 764-777, 2024 May.
Article in English | MEDLINE | ID: mdl-38609546

ABSTRACT

The linear ubiquitin assembly complex (LUBAC) consists of HOIP, HOIL-1 and SHARPIN and is essential for proper immune responses. Individuals with HOIP and HOIL-1 deficiencies present with severe immunodeficiency, autoinflammation and glycogen storage disease. In mice, the loss of Sharpin leads to severe dermatitis due to excessive keratinocyte cell death. Here, we report two individuals with SHARPIN deficiency who manifest autoinflammatory symptoms but unexpectedly no dermatological problems. Fibroblasts and B cells from these individuals showed attenuated canonical NF-κB responses and a propensity for cell death mediated by TNF superfamily members. Both SHARPIN-deficient and HOIP-deficient individuals showed a substantial reduction of secondary lymphoid germinal center B cell development. Treatment of one SHARPIN-deficient individual with anti-TNF therapies led to complete clinical and transcriptomic resolution of autoinflammation. These findings underscore the critical function of the LUBAC as a gatekeeper for cell death-mediated immune dysregulation in humans.


Subject(s)
Immunologic Deficiency Syndromes , Nerve Tissue Proteins , Ubiquitins , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Female , Male , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/genetics , Inflammation/immunology , Inflammation/genetics , B-Lymphocytes/immunology , Loss of Function Mutation , Fibroblasts/metabolism , Fibroblasts/immunology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Mice , Alleles
2.
N Engl J Med ; 390(20): 1873-1884, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38810185

ABSTRACT

BACKGROUND: Autoimmune polyendocrine syndrome type 1 (APS-1) is a life-threatening, autosomal recessive syndrome caused by autoimmune regulator (AIRE) deficiency. In APS-1, self-reactive T cells escape thymic negative selection, infiltrate organs, and drive autoimmune injury. The effector mechanisms governing T-cell-mediated damage in APS-1 remain poorly understood. METHODS: We examined whether APS-1 could be classified as a disease mediated by interferon-γ. We first assessed patients with APS-1 who were participating in a prospective natural history study and evaluated mRNA and protein expression in blood and tissues. We then examined the pathogenic role of interferon-γ using Aire-/-Ifng-/- mice and Aire-/- mice treated with the Janus kinase (JAK) inhibitor ruxolitinib. On the basis of our findings, we used ruxolitinib to treat five patients with APS-1 and assessed clinical, immunologic, histologic, transcriptional, and autoantibody responses. RESULTS: Patients with APS-1 had enhanced interferon-γ responses in blood and in all examined autoimmunity-affected tissues. Aire-/- mice had selectively increased interferon-γ production by T cells and enhanced interferon-γ, phosphorylated signal transducer and activator of transcription 1 (pSTAT1), and CXCL9 signals in multiple organs. Ifng ablation or ruxolitinib-induced JAK-STAT blockade in Aire-/- mice normalized interferon-γ responses and averted T-cell infiltration and damage in organs. Ruxolitinib treatment of five patients with APS-1 led to decreased levels of T-cell-derived interferon-γ, normalized interferon-γ and CXCL9 levels, and remission of alopecia, oral candidiasis, nail dystrophy, gastritis, enteritis, arthritis, Sjögren's-like syndrome, urticaria, and thyroiditis. No serious adverse effects from ruxolitinib were identified in these patients. CONCLUSIONS: Our findings indicate that APS-1, which is caused by AIRE deficiency, is characterized by excessive, multiorgan interferon-γ-mediated responses. JAK inhibition with ruxolitinib in five patients showed promising results. (Funded by the National Institute of Allergy and Infectious Diseases and others.).


Subject(s)
AIRE Protein , Interferon-gamma , Janus Kinase Inhibitors , Polyendocrinopathies, Autoimmune , Adult , Animals , Female , Humans , Male , Mice , AIRE Protein/deficiency , AIRE Protein/genetics , AIRE Protein/immunology , Autoantibodies/blood , Autoantibodies/immunology , Chemokine CXCL9/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Janus Kinase Inhibitors/therapeutic use , Mice, Knockout , Nitriles/therapeutic use , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/drug therapy , Polyendocrinopathies, Autoimmune/immunology , Pyrazoles/therapeutic use , Pyrazoles/pharmacology , Pyrimidines/therapeutic use , T-Lymphocytes/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Pilot Projects , Disease Models, Animal , Child , Adolescent , Middle Aged
3.
Nat Immunol ; 15(1): 88-97, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24165795

ABSTRACT

The p110δ subunit of phosphatidylinositol-3-OH kinase (PI(3)K) is selectively expressed in leukocytes and is critical for lymphocyte biology. Here we report fourteen patients from seven families who were heterozygous for three different germline, gain-of-function mutations in PIK3CD (which encodes p110δ). These patients presented with sinopulmonary infections, lymphadenopathy, nodular lymphoid hyperplasia and viremia due to cytomegalovirus (CMV) and/or Epstein-Barr virus (EBV). Strikingly, they had a substantial deficiency in naive T cells but an over-representation of senescent effector T cells. In vitro, T cells from patients exhibited increased phosphorylation of the kinase Akt and hyperactivation of the metabolic checkpoint kinase mTOR, enhanced glucose uptake and terminal effector differentiation. Notably, treatment with rapamycin to inhibit mTOR activity in vivo partially restored the abundance of naive T cells, largely 'rescued' the in vitro T cell defects and improved the clinical course.


Subject(s)
Cellular Senescence/genetics , Germ-Line Mutation , Immunologic Deficiency Syndromes/genetics , Phosphatidylinositol 3-Kinases/genetics , T-Lymphocytes/metabolism , Antibiotics, Antineoplastic/therapeutic use , Cell Differentiation/genetics , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Cytomegalovirus Infections/blood , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Female , Genes, Dominant , Humans , Immunoblotting , Immunologic Deficiency Syndromes/drug therapy , Male , Pedigree , Phosphatidylinositol 3-Kinases/chemistry , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Viremia/drug therapy , Viremia/genetics , Viremia/virology
4.
Clin Immunol ; 260: 109922, 2024 03.
Article in English | MEDLINE | ID: mdl-38320737

ABSTRACT

IKAROS, encoded by IKZF1, is a tumor suppressor and a key hematopoietic transcription factor responsible for lymphoid and myeloid differentiation. IKZF1 mutations result in inborn errors of immunity presenting with increased susceptibility to infections, immune dysregulation, and malignancies. In particular, patients carrying IKZF1 gain-of-function (GOF) mutations mostly exhibit symptoms of immune dysregulation and polyclonal plasma cell proliferation. Herein, we describe seven new IKAROS GOF cases from two unrelated families, presenting with novel infectious, immune dysregulation and hematologic diseases. Two of the patients underwent allogeneic hematopoietic cell transplantation (HCT) due to poorly responsive complications. HCT was well-tolerated achieving full engraftment in both patients receiving reduced intensity, matched unrelated donor grafts, with no severe acute or chronic graft-vs-host-disease, and in remission from their diseases 2.5 and 4 years post-HCT, respectively. These results suggest that HCT is a valid and curative option in patients with IKAROS GOF disease and severe clinical manifestations.


Subject(s)
Hematologic Diseases , Hematopoietic Stem Cell Transplantation , Ikaros Transcription Factor , Humans , Gain of Function Mutation , Transplantation Conditioning/methods , Ikaros Transcription Factor/genetics
5.
J Allergy Clin Immunol ; 152(3): 736-747, 2023 09.
Article in English | MEDLINE | ID: mdl-37277074

ABSTRACT

BACKGROUND: TCF3 is a transcription factor contributing to early lymphocyte differentiation. Germline monoallelic dominant negative and biallelic loss-of-function (LOF) null TCF3 mutations cause a fully penetrant severe immunodeficiency. We identified 8 individuals from 7 unrelated families with monoallelic LOF TCF3 variants presenting with immunodeficiency with incomplete clinical penetrance. OBJECTIVE: We sought to define TCF3 haploinsufficiency (HI) biology and its association with immunodeficiency. METHODS: Patient clinical data and blood samples were analyzed. Flow cytometry, Western blot analysis, plasmablast differentiation, immunoglobulin secretion, and transcriptional activity studies were conducted on individuals carrying TCF3 variants. Mice with a heterozygous Tcf3 deletion were analyzed for lymphocyte development and phenotyping. RESULTS: Individuals carrying monoallelic LOF TCF3 variants showed B-cell defects (eg, reduced total, class-switched memory, and/or plasmablasts) and reduced serum immunoglobulin levels; most but not all presented with recurrent but nonsevere infections. These TCF3 LOF variants were either not transcribed or translated, resulting in reduced wild-type TCF3 protein expression, strongly suggesting HI pathophysiology for the disease. Targeted RNA sequencing analysis of T-cell blasts from TCF3-null, dominant negative, or HI individuals clustered away from healthy donors, implying that 2 WT copies of TCF3 are needed to sustain a tightly regulated TCF3 gene-dosage effect. Murine TCF3 HI resulted in a reduction of circulating B cells but overall normal humoral immune responses. CONCLUSION: Monoallelic LOF TCF3 mutations cause a gene-dosage-dependent reduction in wild-type protein expression, B-cell defects, and a dysregulated transcriptome, resulting in immunodeficiency. Tcf3+/- mice partially recapitulate the human phenotype, underscoring the differences between TCF3 in humans and mice.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Haploinsufficiency , Immunologic Deficiency Syndromes , Animals , Humans , Mice , B-Lymphocytes , Basic Helix-Loop-Helix Transcription Factors/genetics , Immunoglobulins/genetics , Immunologic Deficiency Syndromes/genetics , T-Lymphocytes
6.
Clin Immunol ; 255: 109732, 2023 10.
Article in English | MEDLINE | ID: mdl-37562721

ABSTRACT

Subcutaneous panniculitis-like T-cell lymphoma (SPTCL) is a rare primary cutaneous non-Hodgkin lymphoma involving CD8+ T cells, the genetic underpinnings of which remain incompletely understood. Here we report two unrelated patients with B cell Expansion with NF-κB and T cell Anergy (BENTA) disease and a novel presentation of SPTCL. Patient 1 presented early in life with recurrent infections and B cell lymphocytosis, linked to a novel gain-of-function (GOF) CARD11 mutation (p.Lys238del). He developed SPTCL-like lesions and membranoproliferative glomerulonephritis by age 2, treated successfully with cyclosporine. Patient 2 presented at 13 months with splenomegaly, lymphadenopathy, and SPTCL with evidence of hemophagocytic lymphohistiocytosis. Genetic analysis revealed two in cis germline GOF CARD11 variants (p.Glu121Asp/p.Gly126Ser). Autologous bone marrow transplant resulted in SPTCL remission despite persistent B cell lymphocytosis. These cases illuminate an unusual pathological manifestation for BENTA disease, suggesting that CARD11 GOF mutations can manifest in cutaneous CD4+and CD8+ T cell malignancies.


Subject(s)
Immunologic Deficiency Syndromes , Lymphocytosis , Lymphoma, T-Cell , Panniculitis , Male , Humans , Child, Preschool , CD8-Positive T-Lymphocytes/pathology , Panniculitis/genetics , Panniculitis/pathology , Panniculitis/therapy , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/therapy
7.
Blood ; 137(3): 349-363, 2021 01 21.
Article in English | MEDLINE | ID: mdl-32845957

ABSTRACT

IKAROS is a transcription factor forming homo- and heterodimers and regulating lymphocyte development and function. Germline mutations affecting the IKAROS N-terminal DNA binding domain, acting in a haploinsufficient or dominant-negative manner, cause immunodeficiency. Herein, we describe 4 germline heterozygous IKAROS variants affecting its C-terminal dimerization domain, via haploinsufficiency, in 4 unrelated families. Index patients presented with hematologic disease consisting of cytopenias (thrombocytopenia, anemia, neutropenia)/Evans syndrome and malignancies (T-cell acute lymphoblastic leukemia, Burkitt lymphoma). These dimerization defective mutants disrupt homo- and heterodimerization in a complete or partial manner, but they do not affect the wild-type allele function. Moreover, they alter key mechanisms of IKAROS gene regulation, including sumoylation, protein stability, and the recruitment of the nucleosome remodeling and deacetylase complex; none affected in N-terminal DNA binding defects. These C-terminal dimerization mutations are largely associated with hematologic disorders, display dimerization haploinsufficiency and incomplete clinical penetrance, and differ from previously reported allelic variants in their mechanism of action. Dimerization mutants contribute to the growing spectrum of IKAROS-associated diseases displaying a genotype-phenotype correlation.


Subject(s)
Germ Cells/metabolism , Haploinsufficiency/genetics , Hematologic Neoplasms/pathology , Ikaros Transcription Factor/metabolism , Protein Multimerization , Adolescent , Adult , Aged , Amino Acid Sequence , Base Sequence , Centromere/metabolism , Chromosome Segregation/genetics , DNA/metabolism , Female , Gene Expression Regulation , Heterochromatin/metabolism , Histone Deacetylase 1/metabolism , Humans , Ikaros Transcription Factor/chemistry , Ikaros Transcription Factor/genetics , Male , Middle Aged , Mutant Proteins/metabolism , Mutation/genetics , Pedigree , Protein Binding , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sumoylation , Transcription, Genetic
8.
Blood ; 138(12): 1019-1033, 2021 09 23.
Article in English | MEDLINE | ID: mdl-33876203

ABSTRACT

Sterile alpha motif (SAM) and Src homology-3 (SH3) domain-containing 3 (SASH3), also called SH3-containing lymphocyte protein (SLY1), is a putative adaptor protein that is postulated to play an important role in the organization of signaling complexes and propagation of signal transduction cascades in lymphocytes. The SASH3 gene is located on the X-chromosome. Here, we identified 3 novel SASH3 deleterious variants in 4 unrelated male patients with a history of combined immunodeficiency and immune dysregulation that manifested as recurrent sinopulmonary, cutaneous, and mucosal infections and refractory autoimmune cytopenias. Patients exhibited CD4+ T-cell lymphopenia, decreased T-cell proliferation, cell cycle progression, and increased T-cell apoptosis in response to mitogens. In vitro T-cell differentiation of CD34+ cells and molecular signatures of rearrangements at the T-cell receptor α (TRA) locus were indicative of impaired thymocyte survival. These patients also manifested neutropenia and B-cell and natural killer (NK)-cell lymphopenia. Lentivirus-mediated transfer of the SASH3 complementary DNA-corrected protein expression, in vitro proliferation, and signaling in SASH3-deficient Jurkat and patient-derived T cells. These findings define a new type of X-linked combined immunodeficiency in humans that recapitulates many of the abnormalities reported in mice with Sly1-/- and Sly1Δ/Δ mutations, highlighting an important role of SASH3 in human lymphocyte function and survival.


Subject(s)
Chromosomes, Human, X/genetics , Mutation , X-Linked Combined Immunodeficiency Diseases/genetics , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Child, Preschool , Chromosomes, Human, X/immunology , Genetic Loci , Humans , Jurkat Cells , Killer Cells, Natural/immunology , Lymphopenia/genetics , Lymphopenia/immunology , Male , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , X-Linked Combined Immunodeficiency Diseases/immunology
9.
J Allergy Clin Immunol ; 149(1): 302-314, 2022 01.
Article in English | MEDLINE | ID: mdl-34089750

ABSTRACT

BACKGROUND: Pediatric endogenous Cushing syndrome (eCs) is mainly caused by pituitary corticotropin-producing adenomas, and most glucocorticoid-dependent effects progressively regress upon tumor removal. eCs reproduces long-term, high-dose glucocorticoid therapy, representing a clean, natural, and unbiased model in which to study glucocorticoid bona fide effects on immunity. OBJECTIVE: We performed extensive immunologic studies in otherwise healthy pediatric patients with eCs before and 6 to 13 months after tumor resection, as well as in in vitro glucocorticoid-treated control cells. METHODS: Flow cytometry, immunoblotting, enzyme-linked immunosorbent assay, real-time quantitative PCR, and RNA-Seq techniques were used to characterize patients' and in vitro glucocorticoid treated cells. RESULTS: Reduced thymic output, decreased naive T cells, diminished proliferation, and increased T-cell apoptosis were detected before surgery; all these defects eventually normalized after tumor removal in patients. In vitro studies also showed increased T-cell apoptosis, with correspondingly diminished NF-κB signaling and IL-21 levels. In this setting, IL-21 addition upregulated antiapoptotic BCL2 expression and rescued T-cell apoptosis in a PI3K pathway-dependent manner. Similar and reproducible findings were confirmed in eCs patient cells as well. CONCLUSIONS: We identified decreased thymic output and lymphocyte proliferation, together with increased apoptosis, as the underlying causes to T-cell lymphopenia in eCs patients. IL-21 was decreased in both natural and in vitro long-term, high-dose glucocorticoid environments, and in vitro addition of IL-21 counteracted the proapoptotic effects of glucocorticoid therapy. Thus, our results suggest that administration of IL-21 in patients receiving long-term, high-dose glucocorticoid therapy may contribute to ameliorate lymphopenia and the complications associated to it.


Subject(s)
Cushing Syndrome/immunology , Cytokines/immunology , Glucocorticoids/pharmacology , Lymphopenia/immunology , T-Lymphocytes/drug effects , Adolescent , Apoptosis/drug effects , Child , Cushing Syndrome/blood , Cushing Syndrome/genetics , Cytokines/blood , Cytokines/genetics , Female , Humans , Leukocyte Count , Lymphopenia/blood , Lymphopenia/genetics , Male , T-Lymphocytes/immunology
10.
J Clin Immunol ; 42(2): 336-349, 2022 02.
Article in English | MEDLINE | ID: mdl-34791587

ABSTRACT

BACKGROUND: CARD9 deficiency is an autosomal recessive primary immunodeficiency underlying increased susceptibility to fungal infection primarily presenting as invasive CNS Candida and/or cutaneous/invasive dermatophyte infections. More recently, a rare heterozygous dominant negative CARD9 variant c.1434 + 1G > C was reported to be protective from inflammatory bowel disease. OBJECTIVE: We studied two siblings carrying homozygous CARD9 variants (c.1434 + 1G > C) and born to heterozygous asymptomatic parents. One sibling was asymptomatic and the other presented with candida esophagitis, upper respiratory infections, hypogammaglobulinemia, and low class-switched memory B cells. METHODS AND RESULTS: The CARD9 c.1434 + 1G > C variant generated two mutant transcripts confirmed by mRNA and protein expression: an out-of-frame c.1358-1434 deletion/ ~ 55 kDa protein (CARD9Δex.11) and an in-frame c.1417-1434 deletion/ ~ 61 kDa protein (CARD9Δ18 nt.). Neither transcript was able to form a complete/functional CBM complex, which includes TRIM62. Based on the index patient's CVID-like phenotype, CARD9 expression was tested and detected in lymphocytes and monocytes from humans and mice. The functional impact of different CARD9 mutations and gene dosage conditions was evaluated in heterozygous and homozygous c.1434 + 1 G > C members of the index family, and in WT (two WT alleles), haploinsufficiency (one WT, one null allele), and null (two null alleles) individuals. CARD9 gene dosage impacted lymphocyte and monocyte functions including cytokine generation, MAPK activation, T-helper commitment, transcription, plasmablast differentiation, and immunoglobulin production in a differential manner. CONCLUSIONS: CARD9 exon 11 integrity is critical to CBM complex function. CARD9 is expressed and affects particular T and B cell functions in a gene dosage-dependent manner, which in turn may contribute to the phenotype of CARD9 deficiency.


Subject(s)
Candidiasis, Chronic Mucocutaneous , Alleles , Animals , CARD Signaling Adaptor Proteins/genetics , Gene Dosage , Homozygote , Humans , Mice , Phenotype
11.
J Clin Immunol ; 42(1): 119-129, 2022 01.
Article in English | MEDLINE | ID: mdl-34657245

ABSTRACT

Rare, biallelic loss-of-function mutations in DOCK8 result in a combined immune deficiency characterized by severe and recurrent cutaneous infections, eczema, allergies, and susceptibility to malignancy, as well as impaired humoral and cellular immunity and hyper-IgE. The advent of next-generation sequencing technologies has enabled the rapid molecular diagnosis of rare monogenic diseases, including inborn errors of immunity. These advances have resulted in the implementation of gene-guided treatments, such as hematopoietic stem cell transplant for DOCK8 deficiency. However, putative disease-causing variants revealed by next-generation sequencing need rigorous validation to demonstrate pathogenicity. Here, we report the eventual diagnosis of DOCK8 deficiency in a consanguineous family due to a novel homozygous intronic deletion variant that caused aberrant exon splicing and subsequent loss of expression of DOCK8 protein. Remarkably, the causative variant was not initially detected by clinical whole-genome sequencing but was subsequently identified and validated by combining advanced genomic analysis, RNA-seq, and flow cytometry. This case highlights the need to adopt multipronged confirmatory approaches to definitively solve complex genetic cases that result from variants outside protein-coding exons and conventional splice sites.


Subject(s)
Job Syndrome , Consanguinity , Guanine Nucleotide Exchange Factors/genetics , Homozygote , Humans , Job Syndrome/diagnosis , Job Syndrome/genetics , Mutation/genetics , Exome Sequencing
12.
Clin Infect Dis ; 73(7): e1624-e1631, 2021 10 05.
Article in English | MEDLINE | ID: mdl-32910141

ABSTRACT

BACKGROUND: Diethylcarbamazine citrate (DEC) treatment of loiasis is complicated by adverse reactions that are correlated with the number of circulating microfilariae (mf). The cause of these reactions is unknown, but they are accompanied by a dramatic interleukin-5 (IL-5)-dependent increase in eosinophilia and evidence of eosinophil activation. METHODS: To explore the role of IL-5 driven eosinophilia in post-DEC reactions, 8 adults with confirmed loiasis and <5000 mf/mL blood were enrolled in a randomized, double-blind, placebo-controlled trial of the humanized anti-IL-5 antibody, reslizumab, (1.0 mg/kg IV) administered 3 to 7 days prior to initiation of DEC treatment (9 mg/kg/day for 21 days). The primary endpoint was the reduction in absolute eosinophil count (AEC) during the first week of DEC treatment. RESULTS: Baseline characteristics were comparable between the two groups. Single dose reslizumab lowered the AEC by 77% prior to initiation of DEC therapy (vs. 12% in the placebo group, P < .05). More importantly, AEC remained below baseline in the first week of DEC treatment in all subjects who received reslizumab and in none of the placebo subjects. Mf clearance occurred within 2 days of initiation of DEC in all 7 mf-positive subjects. Mild to moderate adverse events were seen in all 8 subjects and were not significantly different between the groups. CONCLUSIONS: In summary, although reslizumab was able to blunt peripheral eosinophilia post-DEC treatment in subjects with loiasis and had no effect on microfilarial clearance, the reduction in AEC appeared to have been insufficient to prevent post-treatment AEs.


Subject(s)
Eosinophilia , Loiasis , Adult , Animals , Antibodies, Monoclonal, Humanized , Diethylcarbamazine/adverse effects , Double-Blind Method , Eosinophilia/drug therapy , Humans , Interleukin-5 , Loa , Loiasis/drug therapy , Pilot Projects
13.
J Infect Dis ; 222(7): 1170-1179, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32386415

ABSTRACT

BACKGROUND: Chronic active Epstein-Barr virus (CAEBV) presents with high levels of viral genomes in blood and tissue infiltration with Epstein-Barr virus (EBV)-positive lymphocytes. The pathogenesis of CAEBV is poorly understood. METHODS: We evaluated 2 patients with natural killer (NK) cell CAEBV and studied their NK cell phenotype and signaling pathways in cells. RESULTS: Both patients had increased numbers of NK cells, EBV predominantly in NK cells, and immature NK cells in the blood. Both patients had increased phosphorylation of Akt, S6, and STAT1 in NK cells, and increased total STAT1. Treatment of 1 patient with sirolimus reduced phosphorylation of S6 in T and B cells, but not in NK cells and did not reduce levels of NK cells or EBV DNA in the blood. Treatment of both patients' cells with JAK inhibitors in vitro reduced phosphorylated STAT1 to normal. Patients with T- or B-cell CAEBV had increased phosphorylation of Akt and S6 in NK cells, but no increase in total STAT1. CONCLUSIONS: The increase in phosphorylated Akt, S6, and STAT1, as well as immature NK cells describe a new phenotype for NK cell CAEBV. The reduction of STAT1 phosphorylation in their NK cells with JAK inhibitors suggests a novel approach to therapy.


Subject(s)
Epstein-Barr Virus Infections/diagnosis , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphoproliferative Disorders/diagnosis , Signal Transduction , Adolescent , Adult , B-Lymphocytes/immunology , B-Lymphocytes/virology , Chronic Disease , Epstein-Barr Virus Infections/virology , Female , Humans , Killer Cells, Natural/virology , Lymphoproliferative Disorders/virology , Male , Phosphorylation , Prospective Studies , Proto-Oncogene Proteins c-akt/metabolism , STAT1 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/virology
14.
J Clin Immunol ; 40(8): 1093-1101, 2020 11.
Article in English | MEDLINE | ID: mdl-32813180

ABSTRACT

The noncanonical NF-κB pathway is implicated in diverse biological and immunological processes. Monoallelic C-terminus loss-of-function and gain-of-function mutations of NFKB2 have been recently identified as a cause of immunodeficiency manifesting with common variable immunodeficiency (CVID) or combined immunodeficiency (CID) phenotypes. Herein we report a family carrying a heterozygous nonsense mutation in NFKB2 (c.809G > A, p.W270*). This variant is associated with increased mRNA decay and no mutant NFKB2 protein expression, leading to NFKB2 haploinsufficiency. Our findings demonstrate that bona fide NFKB2 haploinsufficiency, likely caused by mutant mRNA decay and protein instability leading to the transcription and expression of only the wild-type allele, is associated with clinical immunodeficiency, although with incomplete clinical penetrance. Abnormal B cell development, hypogammaglobulinemia, poor antibody response, and abnormal noncanonical (but normal canonical) NF-κB pathway signaling are the immunologic hallmarks of this disease. This adds a third allelic variant to the pathophysiology of NFKB2-mediated immunodeficiency disorders.


Subject(s)
Agammaglobulinemia/diagnosis , Agammaglobulinemia/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Haploinsufficiency , Mutation , NF-kappa B p52 Subunit/genetics , Adolescent , Adult , Alleles , Female , Genetic Association Studies/methods , Genotype , Humans , Immunophenotyping , Lymphocytes/metabolism , Male , Middle Aged , Pedigree , Phenotype , Exome Sequencing , Young Adult
16.
Blood ; 130(13): 1553-1564, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28778864

ABSTRACT

NF-κB signaling through its NFKB1-dependent canonical and NFKB2-dependent noncanonical pathways plays distinctive roles in a diverse range of immune processes. Recently, mutations in these 2 genes have been associated with common variable immunodeficiency (CVID). While studying patients with genetically uncharacterized primary immunodeficiencies, we detected 2 novel nonsense gain-of-function (GOF) NFKB2 mutations (E418X and R635X) in 3 patients from 2 families, and a novel missense change (S866R) in another patient. Their immunophenotype was assessed by flow cytometry and protein expression; activation of canonical and noncanonical pathways was examined in peripheral blood mononuclear cells and transfected HEK293T cells through immunoblotting, immunohistochemistry, luciferase activity, real-time polymerase chain reaction, and multiplex assays. The S866R change disrupted a C-terminal NF-κΒ2 critical site affecting protein phosphorylation and nuclear translocation, resulting in CVID with adrenocorticotropic hormone deficiency, growth hormone deficiency, and mild ectodermal dysplasia as previously described. In contrast, the nonsense mutations E418X and R635X observed in 3 patients led to constitutive nuclear localization and activation of both canonical and noncanonical NF-κΒ pathways, resulting in a combined immunodeficiency (CID) without endocrine or ectodermal manifestations. These changes were also found in 2 asymptomatic relatives. Thus, these novel NFKB2 GOF mutations produce a nonfully penetrant CID phenotype through a different pathophysiologic mechanism than previously described for mutations in NFKB2.


Subject(s)
Codon, Nonsense , NF-kappa B p52 Subunit/genetics , Severe Combined Immunodeficiency/genetics , Adrenal Insufficiency/genetics , Common Variable Immunodeficiency/genetics , Ectodermal Dysplasia/genetics , Growth Hormone/deficiency , HEK293 Cells , Humans , Mutation, Missense , Phenotype
17.
N Engl J Med ; 370(17): 1615-1625, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24716661

ABSTRACT

Genetic defects in MOGS, the gene encoding mannosyl-oligosaccharide glucosidase (the first enzyme in the processing pathway of N-linked oligosaccharide), cause the rare congenital disorder of glycosylation type IIb (CDG-IIb), also known as MOGS-CDG. MOGS is expressed in the endoplasmic reticulum and is involved in the trimming of N-glycans. We evaluated two siblings with CDG-IIb who presented with multiple neurologic complications and a paradoxical immunologic phenotype characterized by severe hypogammaglobulinemia but limited clinical evidence of an infectious diathesis. A shortened immunoglobulin half-life was determined to be the mechanism underlying the hypogammaglobulinemia. Impaired viral replication and cellular entry may explain a decreased susceptibility to infections.


Subject(s)
Agammaglobulinemia/genetics , Congenital Disorders of Glycosylation/immunology , Disease Resistance/genetics , Virus Diseases/immunology , alpha-Glucosidases/genetics , Agammaglobulinemia/immunology , Antibodies, Viral/blood , Child , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/metabolism , Female , Glycosylation , Humans , Immunoglobulins/metabolism , Male
SELECTION OF CITATIONS
SEARCH DETAIL