Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
J Pediatr Hematol Oncol ; 43(3): e351-e357, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33122583

ABSTRACT

OBJECTIVE: Mutations in IKZF1, which encodes Ikaros family zinc finger 1 (IKAROS) transcription factor, are associated with recurrent infections, cytopenia, autoimmune diseases, and hematologic malignancies. Diverse clinical phenotypes resulting from IKZF1 mutations include pulmonary fungal infections, cytopenia, autoimmune hemolytic anemia (AIHA), and malignancies. In this study, we aimed to assess the DNA-binding ability and pericentromeric (PC) localization of a variant of IKZF discovered in a patient. MATERIALS AND METHODS: DNA-binding ability of a pathogenic IKZF variant was tested using electrophoretic mobility shift assay and PC localization of the variant was assessed by immunofluorescent microscopy in NIH3T3 cells. RESULTS: Clinical features of a 3-month-old male infant who underwent hematopoietic stem cell transplantation because of an IKZF1 mutation-associated common variable immunodeficiency, AIHA, and pancytopenia are described. DNA studies revealed a heterozygous missense variant (IKZF1 NM_006060 c.427C>T; p.R143W). Cotransfection studies revealed that mutant R143W has a partial dominant-negative effect over PC targeting and DNA binding. CONCLUSIONS: IKZF1 mutation must be kept in mind if neonatal AIHA, common variable immunodeficiency, and pancytopenia are observed.


Subject(s)
Anemia, Hemolytic, Autoimmune/genetics , Common Variable Immunodeficiency/genetics , Ikaros Transcription Factor/genetics , Pancytopenia/genetics , Anemia, Hemolytic, Autoimmune/complications , Anemia, Hemolytic, Autoimmune/therapy , Animals , Common Variable Immunodeficiency/complications , Common Variable Immunodeficiency/therapy , Hematopoietic Stem Cell Transplantation , Humans , Infant , Male , Mice , NIH 3T3 Cells , Pancytopenia/complications , Pancytopenia/therapy , Point Mutation
7.
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
9.
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
10.
J Immunol ; 200(1): 110-118, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29187589

ABSTRACT

Understanding the control of Ag restimulation-induced T cell death (RICD), especially in cancer immunotherapy, where highly proliferating T cells will encounter potentially large amounts of tumor Ags, is important now more than ever. It has been known that growth cytokines make T cells susceptible to RICD, but the precise molecular mediators that govern this in T cell subsets is unknown until now. STAT proteins are a family of transcription factors that regulate gene expression programs underlying key immunological processes. In particular, STAT5 is known to favor the generation and survival of memory T cells. In this study, we report an unexpected role for STAT5 signaling in the death of effector memory T (TEM) cells in mice and humans. TEM cell death was prevented with neutralizing anti-IL-2 Ab or STAT5/JAK3 inhibitors, indicating that STAT5 signaling drives RICD in TEM cells. Moreover, we identified a unique patient with a heterozygous missense mutation in the coiled-coil domain of STAT5B that presented with autoimmune lymphoproliferative syndrome-like features. Similar to Stat5b-/- mice, this patient exhibited increased CD4+ TEM cells in the peripheral blood. The mutant STAT5B protein dominantly interfered with STAT5-driven transcriptional activity, leading to global downregulation of STAT5-regulated genes in patient T cells upon IL-2 stimulation. Notably, CD4+ TEM cells from the patient were strikingly resistant to cell death by in vitro TCR restimulation, a finding that was recapitulated in Stat5b-/- mice. Hence, STAT5B is a crucial regulator of RICD in memory T cells in mice and humans.


Subject(s)
Apoptosis , Autoimmune Lymphoproliferative Syndrome/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Survival , STAT5 Transcription Factor/metabolism , Animals , Antibodies, Neutralizing/metabolism , Autoimmune Lymphoproliferative Syndrome/genetics , Cells, Cultured , Female , Humans , Immunologic Memory , Interleukin-2/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense/genetics , STAT5 Transcription Factor/genetics , Signal Transduction , Transcription, Genetic
11.
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
12.
Ann Rheum Dis ; 77(4): 612-619, 2018 04.
Article in English | MEDLINE | ID: mdl-29358286

ABSTRACT

OBJECTIVES: To characterise the clinical features, immune manifestations and molecular mechanisms in a recently described autoinflammatory disease caused by mutations in TRNT1, a tRNA processing enzyme, and to explore the use of cytokine inhibitors in suppressing the inflammatory phenotype. METHODS: We studied nine patients with biallelic mutations in TRNT1 and the syndrome of congenital sideroblastic anaemia with immunodeficiency, fevers and developmental delay (SIFD). Genetic studies included whole exome sequencing (WES) and candidate gene screening. Patients' primary cells were used for deep RNA and tRNA sequencing, cytokine profiling, immunophenotyping, immunoblotting and electron microscopy (EM). RESULTS: We identified eight mutations in these nine patients, three of which have not been previously associated with SIFD. Three patients died in early childhood. Inflammatory cytokines, mainly interleukin (IL)-6, interferon gamma (IFN-γ) and IFN-induced cytokines were elevated in the serum, whereas tumour necrosis factor (TNF) and IL-1ß were present in tissue biopsies of patients with active inflammatory disease. Deep tRNA sequencing of patients' fibroblasts showed significant deficiency of mature cytosolic tRNAs. EM of bone marrow and skin biopsy samples revealed striking abnormalities across all cell types and a mix of necrotic and normal-appearing cells. By immunoprecipitation, we found evidence for dysregulation in protein clearance pathways. In 4/4 patients, treatment with a TNF inhibitor suppressed inflammation, reduced the need for blood transfusions and improved growth. CONCLUSIONS: Mutations of TRNT1 lead to a severe and often fatal syndrome, linking protein homeostasis and autoinflammation. Molecular diagnosis in early life will be crucial for initiating anti-TNF therapy, which might prevent some of the severe disease consequences.


Subject(s)
Anemia, Sideroblastic/genetics , Anti-Inflammatory Agents/therapeutic use , Genetic Diseases, X-Linked/genetics , Immunologic Deficiency Syndromes/genetics , Mutation , Nucleotidyltransferases/genetics , RNA, Transfer/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Anemia, Sideroblastic/blood , Child , Child, Preschool , Cytokines/blood , Cytokines/genetics , Developmental Disabilities/genetics , Female , Genetic Diseases, X-Linked/blood , Humans , Immunophenotyping , Male , Pedigree , Phenotype , Tumor Necrosis Factor-alpha/analysis , Exome Sequencing
13.
Blood ; 125(4): 591-9, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25359994

ABSTRACT

Germline loss-of-function mutations in the transcription factor signal transducer and activator of transcription 3 (STAT3) cause immunodeficiency, whereas somatic gain-of-function mutations in STAT3 are associated with large granular lymphocytic leukemic, myelodysplastic syndrome, and aplastic anemia. Recently, germline mutations in STAT3 have also been associated with autoimmune disease. Here, we report on 13 individuals from 10 families with lymphoproliferation and early-onset solid-organ autoimmunity associated with 9 different germline heterozygous mutations in STAT3. Patients exhibited a variety of clinical features, with most having lymphadenopathy, autoimmune cytopenias, multiorgan autoimmunity (lung, gastrointestinal, hepatic, and/or endocrine dysfunction), infections, and short stature. Functional analyses demonstrate that these mutations confer a gain-of-function in STAT3 leading to secondary defects in STAT5 and STAT1 phosphorylation and the regulatory T-cell compartment. Treatment targeting a cytokine pathway that signals through STAT3 led to clinical improvement in 1 patient, suggesting a potential therapeutic option for such patients. These results suggest that there is a broad range of autoimmunity caused by germline STAT3 gain-of-function mutations, and that hematologic autoimmunity is a major component of this newly described disorder. Some patients for this study were enrolled in a trial registered at www.clinicaltrials.gov as #NCT00001350.


Subject(s)
Autoimmune Diseases/genetics , Genetic Diseases, Inborn/genetics , Lymphoproliferative Disorders/genetics , STAT3 Transcription Factor/genetics , Adolescent , Adult , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Child , Child, Preschool , Female , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/pathology , Humans , Infant , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Male , Mutation , Phosphorylation/genetics , Phosphorylation/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
16.
Blood ; 123(13): 1989-99, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24398331

ABSTRACT

Autoimmune lymphoproliferative syndrome (ALPS) presents in childhood with nonmalignant lymphadenopathy and splenomegaly associated with a characteristic expansion of mature CD4 and CD8 negative or double negative T-cell receptor αß(+) T lymphocytes. Patients often present with chronic multilineage cytopenias due to autoimmune peripheral destruction and/or splenic sequestration of blood cells and have an increased risk of B-cell lymphoma. Deleterious heterozygous mutations in the FAS gene are the most common cause of this condition, which is termed ALPS-FAS. We report the natural history and pathophysiology of 150 ALPS-FAS patients and 63 healthy mutation-positive relatives evaluated in our institution over the last 2 decades. Our principal findings are that FAS mutations have a clinical penetrance of <60%, elevated serum vitamin B12 is a reliable and accurate biomarker of ALPS-FAS, and the major causes of morbidity and mortality in these patients are the overwhelming postsplenectomy sepsis and development of lymphoma. With longer follow-up, we observed a significantly greater relative risk of lymphoma than previously reported. Avoiding splenectomy while controlling hypersplenism by using corticosteroid-sparing treatments improves the outcome in ALPS-FAS patients. This trial was registered at www.clinicaltrials.gov as #NCT00001350.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/genetics , Autoimmune Lymphoproliferative Syndrome/therapy , Mutation , fas Receptor/genetics , Adolescent , Adult , Autoimmune Lymphoproliferative Syndrome/pathology , Cell Proliferation , Child , Disease Progression , Female , Follow-Up Studies , Humans , Lymphocytes/pathology , Lymphocytes/physiology , Male , Middle Aged , Penetrance , Young Adult
17.
J Clin Immunol ; 35(5): 445-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25947952

ABSTRACT

Severe chronic active Epstein-Barr virus (CAEBV) disease is defined as a severe progressive illness lasting 6 months or longer with infiltration of tissues with EBV-positive lymphocytes, markedly elevated levels of EBV DNA in the blood, and no known immunodeficiency such as HIV. These patients usually have fever, splenomegaly, lymphadenopathy, and may have markedly elevated EBV antibody titers to viral capsid antigen. Although the cause of most cases of severe CAEBV is unknown, one well-documented case was associated with compound heterozygous mutations in PRF1 (perforin 1). Here we report a patient with prolonged severe CAEBV who underwent bone marrow transplant for his disease and subsequently was found to have compound heterozygous mutations in STXBP2 (MUNC18-2) as well as a heterozygous mutation in PRF1 (perforin 1).


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Transplantation , Epstein-Barr Virus Infections/diagnosis , Herpesvirus 4, Human/isolation & purification , Lysosomal-Associated Membrane Protein 1/metabolism , Munc18 Proteins/genetics , Perforin/genetics , Adult , Age of Onset , B-Lymphocytes/virology , Biomarkers/metabolism , Chronic Disease , Cytotoxicity, Immunologic/genetics , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/therapy , Humans , Male , Mutation/genetics , Virus Activation/genetics , White People , Young Adult
18.
Blood ; 121(16): 3117-25, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23430113

ABSTRACT

Defective lymphocyte apoptosis results in chronic lymphadenopathy and/or splenomegaly associated with autoimmune phenomena. The prototype for human apoptosis disorders is the autoimmune lymphoproliferative syndrome (ALPS), which is caused by mutations in the FAS apoptotic pathway. Recently, patients with an ALPS-like disease called RAS-associated autoimmune leukoproliferative disorder, in which somatic mutations in NRAS or KRAS are found, also were described. Despite this progress, many patients with ALPS-like disease remain undefined genetically. We identified a homozygous, loss-of-function mutation in PRKCD (PKCδ) in a patient who presented with chronic lymphadenopathy, splenomegaly, autoantibodies, elevated immunoglobulins and natural killer dysfunction associated with chronic, low-grade Epstein-Barr virus infection. This mutation markedly decreased protein expression and resulted in ex vivo B-cell hyperproliferation, a phenotype similar to that of the PKCδ knockout mouse. Lymph nodes showed intense follicular hyperplasia, also mirroring the mouse model. Immunophenotyping of circulating lymphocytes demonstrated expansion of CD5+CD20+ B cells. Knockdown of PKCδ in normal mononuclear cells recapitulated the B-cell hyperproliferative phenotype in vitro. Reconstitution of PKCδ in patient-derived EBV-transformed B-cell lines partially restored phorbol-12-myristate-13-acetate-induced cell death. In summary, homozygous PRKCD mutation results in B-cell hyperproliferation and defective apoptosis with consequent lymphocyte accumulation and autoantibody production in humans, and disrupts natural killer cell function.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/genetics , B-Lymphocytes/pathology , Mutation , Protein Kinase C-delta/genetics , Animals , Apoptosis , Autoimmune Lymphoproliferative Syndrome/complications , Autoimmune Lymphoproliferative Syndrome/immunology , Autoimmune Lymphoproliferative Syndrome/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Cell Proliferation , Child , Cytokines/immunology , Epstein-Barr Virus Infections/complications , Gene Expression , Gene Knockdown Techniques , Herpesvirus 4, Human/isolation & purification , Homozygote , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphatic Diseases/complications , Male , Mice , Protein Kinase C-delta/immunology , Splenomegaly/complications
20.
J Clin Invest ; 134(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38015619

ABSTRACT

AIOLOS, also known as IKZF3, is a transcription factor that is highly expressed in the lymphoid lineage and is critical for lymphocyte differentiation and development. Here, we report on 9 individuals from 3 unrelated families carrying AIOLOS variants Q402* or E82K, which led to AIOLOS haploinsufficiency through different mechanisms of action. Nonsense mutant Q402* displayed abnormal DNA binding, pericentromeric targeting, posttranscriptional modification, and transcriptome regulation. Structurally, the mutant lacked the AIOLOS zinc finger (ZF) 5-6 dimerization domain, but was still able to homodimerize with WT AIOLOS and negatively regulate DNA binding through ZF1, a previously unrecognized function for this domain. Missense mutant E82K showed overall normal AIOLOS functions; however, by affecting a redefined AIOLOS protein stability domain, it also led to haploinsufficiency. Patients with AIOLOS haploinsufficiency showed hypogammaglobulinemia, recurrent infections, autoimmunity, and allergy, but with incomplete clinical penetrance. Altogether, these data redefine the AIOLOS structure-function relationship and expand the spectrum of AIOLOS-associated diseases.


Subject(s)
Haploinsufficiency , Trans-Activators , Humans , DNA , Gene Expression Regulation , Trans-Activators/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL