Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 24(3): 414-422, 2023 03.
Article in English | MEDLINE | ID: mdl-36732425

ABSTRACT

Interferon-γ (IFNγ) is an important mediator of cellular immune responses, but high systemic levels of this cytokine are associated with immunopathology. IFNγ binds to its receptor (IFNγR) and to extracellular matrix (ECM) via four positively charged C-terminal amino acids (KRKR), the ECM-binding domain (EBD). Across evolution, IFNγ is not well conserved, but the EBD is highly conserved, suggesting a critical function. Here, we show that IFNγ lacking the EBD (IFNγΔKRKR) does not bind to ECM but still binds to the IFNγR and retains bioactivity. Overexpression of IFNγΔKRKR in tumors reduced local ECM binding, increased systemic levels and induced sickness behavior, weight loss and toxicity. To analyze the function of the EBD during infection, we generated IFNγΔKRKR mice lacking the EBD by using CRISPR-Cas9. Infection with lymphocytic choriomeningitis virus resulted in higher systemic IFNγΔKRKR levels, enhanced sickness behavior, weight loss and fatal toxicity. We conclude that local retention of IFNγ is a pivotal mechanism to protect the organism from systemic toxicity during prolonged immune stimulation.


Subject(s)
Cytokines , Neoplasms , Mice , Animals , Cytokines/metabolism , Interferon-gamma/metabolism , Signal Transduction , Extracellular Matrix/metabolism
4.
J Allergy Clin Immunol ; 153(1): 243-255.e14, 2024 01.
Article in English | MEDLINE | ID: mdl-37595758

ABSTRACT

BACKGROUND: Hemophagocytic lymphohistiocytosis (HLH) is a hyperinflammatory disorder characterized by a life-threatening cytokine storm and immunopathology. Familial HLH type 3 (FHL3) accounts for approximately 30% of all inborn HLH cases worldwide. It is caused by mutations in the UNC13D gene that result in impaired degranulation of cytotoxic vesicles and hence compromised T-cell- and natural killer-cell-mediated killing. Current treatment protocols, including allogeneic hematopoietic stem cell (HSC) transplantation, still show high mortality. OBJECTIVE: We sought to develop and evaluate a curative genome editing strategy in the preclinical FHL3 Jinx mouse model. Jinx mice harbor a cryptic splice donor site in Unc13d intron 26 and develop clinical symptoms of human FHL3 upon infection with lymphocytic choriomeningitis virus (LCMV). METHODS: We employed clustered regularly interspaced short palindromic repeats (CRISPR)-Cas technology to delete the disease-causing mutation in HSCs and transplanted Unc13d-edited stem cells into busulfan-conditioned Jinx recipient mice. Safety studies included extensive genotyping and chromosomal aberrations analysis by single targeted linker-mediated PCR sequencing (CAST-Seq)-based off-target analyses. Cure from HLH predisposition was assessed by LCMV infection. RESULTS: Hematopoietic cells isolated from transplanted mice revealed efficient gene editing (>95%), polyclonality of the T-cell receptor repertoire, and neither signs of off-target effects nor leukemogenesis. Unc13d transcription levels of edited and wild-type cells were comparable. While LCMV challenge resulted in acute HLH in Jinx mice transplanted with mock-edited HSCs, Jinx mice grafted with Unc13d-edited cells showed rapid virus clearance and protection from HLH. CONCLUSIONS: Our study demonstrates that transplantation of CRISPR-Cas edited HSCs supports the development of a functional polyclonal T-cell response in the absence of genotoxicity-associated clonal outgrowth.


Subject(s)
Lymphohistiocytosis, Hemophagocytic , Humans , Mice , Animals , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/therapy , Lymphohistiocytosis, Hemophagocytic/diagnosis , T-Lymphocytes , Gene Editing , Mutation , Lymphocytic choriomeningitis virus , Hematopoietic Stem Cells , Membrane Proteins/genetics
5.
Eur J Immunol ; 50(12): 1952-1958, 2020 12.
Article in English | MEDLINE | ID: mdl-32734619

ABSTRACT

NK1.1+ cells found in salivary glands (SG) represent a unique cell population of innate lymphoid cells (ILC) with characteristics of both conventional NK cells and ILC1. Here, we demonstrate that these NK1.1+  cells limit the accumulation and differentiation of virus-specific tissue-resident memory CD8+ T cells (TRM  cells) in SG of mice infected with lymphocytic choriomeningitis virus (LCMV). The negative regulation of LCMV-specific CD8+ TRM  cells by NK1.1+  cells in SG is independent of NKG2D, NKp46, TRAIL, and perforin. Moreover, analysis of NKp46iCre+ Eomesfl/fl mice revealed that Eomes-dependent conventional NK cells are dispensable for negative regulation. Since the SG are prone to autoimmune reactions, regulation of TRM  cells by tissue-resident ILC may be particularly important to prevent immunopathology in this organ.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Innate/immunology , Immunologic Memory/immunology , Killer Cells, Natural/immunology , Salivary Glands/immunology , Animals , Cell Differentiation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/immunology , Natural Cytotoxicity Triggering Receptor 1/immunology , Perforin/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology
6.
Blood ; 131(13): 1442-1455, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29326099

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS) are life-threatening hyperferritinemic systemic inflammatory disorders. Although profound cytotoxic impairment causes familial HLH (fHLH), the mechanisms driving non-fHLH and MAS are largely unknown. MAS occurs in patients with suspected rheumatic disease, but the mechanistic basis for its distinction is unclear. Recently, a syndrome of recurrent MAS with infantile enterocolitis caused by NLRC4 inflammasome hyperactivity highlighted the potential importance of interleukin-18 (IL-18). We tested this association in hyperferritinemic and autoinflammatory patients and found a dramatic correlation of MAS risk with chronic (sometimes lifelong) elevation of mature IL-18, particularly with IL-18 unbound by IL-18 binding protein, or free IL-18. In a mouse engineered to carry a disease-causing germ line NLRC4T337S mutation, we observed inflammasome-dependent, chronic IL-18 elevation. Surprisingly, this NLRC4T337S-induced systemic IL-18 elevation derived entirely from intestinal epithelia. NLRC4T337S intestines were histologically normal but showed increased epithelial turnover and upregulation of interferon-γ-induced genes. Assessing cellular and tissue expression, classical inflammasome components such as Il1b, Nlrp3, and Mefv predominated in neutrophils, whereas Nlrc4 and Il18 were distinctly epithelial. Demonstrating the importance of free IL-18, Il18 transgenic mice exhibited free IL-18 elevation and more severe experimental MAS. NLRC4T337S mice, whose free IL-18 levels were normal, did not. Thus, we describe a unique connection between MAS risk and chronic IL-18, identify epithelial inflammasome hyperactivity as a potential source, and demonstrate the pathogenicity of free IL-18. These data suggest an IL-18-driven pathway, complementary to the cytotoxic impairment of fHLH, with potential as a distinguishing biomarker and therapeutic target in MAS.


Subject(s)
Interleukin-18/immunology , Macrophage Activation Syndrome/immunology , Signal Transduction/immunology , Amino Acid Substitution , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-18/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/pathology , Macrophage Activation Syndrome/genetics , Macrophage Activation Syndrome/pathology , Mice , Mice, Knockout , Mutation, Missense , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pyrin/genetics , Pyrin/immunology , Signal Transduction/genetics
7.
Eur J Immunol ; 47(2): 364-373, 2017 02.
Article in English | MEDLINE | ID: mdl-27925643

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening inflammatory syndrome characterized by hyperactivation of lymphocytes and histiocytes. T cells play a key role in HLH pathogenesis, but their differentiation pattern is not well characterized in patients with active HLH. We compared T-cell activation patterns between patients with familial HLH (1°HLH), 2°HLH without apparent infectious trigger (2°HLH) and 2°HLH induced by a viral infection (2°V-HLH). Polyclonal CD8+ T cells are highly activated in 1°HLH and 2°V-HLH, but less in 2°HLH as assessed by HLA-DR expression and marker combination with CD45RA, CCR7, CD127, PD-1 and CD57. Absence of increased HLA-DR expression on T cells excluded active 1° HLH with high sensitivity and specificity. A high proportion of polyclonal CD127- CD4+ T cells expressing HLA-DR, CD57, and perforin is a signature of infants with 1°HLH, much less prominent in virus-associated 2°HLH. The similar pattern and extent of CD8+ T-cell activation compared to 2° V-HLH is compatible with a viral trigger of 1°HLH. However, in most 1°HLH patients no triggering infection was documented and the unique activation of cytotoxic CD4+ T cells indicates that the overall T-cell response in 1°HLH is different. This may reflect different pathways of pathogenesis of these two HLH variants.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , Virus Diseases/immunology , CD57 Antigens/metabolism , Cell Differentiation , Cells, Cultured , Female , HLA-DR Antigens/metabolism , Humans , Infant , Infant, Newborn , Lymphocyte Activation , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/etiology , Male , Perforin/metabolism , Receptors, Antigen, T-Cell/genetics , Virus Diseases/complications , Virus Diseases/diagnosis
8.
Immunol Cell Biol ; 96(10): 1060-1071, 2018 11.
Article in English | MEDLINE | ID: mdl-29790605

ABSTRACT

Premature T-cell immunosenescence with CD57+ CD8+ T-cell accumulation has been linked to immunodeficiency and autoimmunity in primary immunodeficiencies including activated PI3 kinase delta syndrome (APDS). To address whether CD57 marks the typical senescent T-cell population seen in adult individuals or identifies a distinct population in APDS, we compared CD57+ CD8+ T cells from mostly pediatric APDS patients to those of healthy adults with similarly prominent senescent T cells. CD57+ CD8+ T cells from APDS patients were less differentiated with more CD27+ CD28+ effector memory T cells showing increased PD1 and Eomesodermin expression. In addition, transition of naïve to CD57+ CD8+ T cells was not associated with the characteristic telomere shortening. Nevertheless, they showed the increased interferon-gamma secretion, enhanced degranulation and reduced in vitro proliferation typical of senescent CD57+ CD8+ T cells. Thus, hyperactive PI3 kinase signaling favors premature accumulation of a CD57+ CD8+ T-cell population, which shows most functional features of typical senescent T cells, but is different in terms of differentiation and relative telomere shortening. Initial observations indicate that this specific differentiation state may offer the opportunity to revert premature T-cell immunosenescence and its potential contribution to inflammation and immunodeficiency in APDS.


Subject(s)
CD57 Antigens/metabolism , Cell Differentiation , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Telomere Shortening , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Senescence/genetics , Cellular Senescence/immunology , Class I Phosphatidylinositol 3-Kinases/metabolism , Cytokines/metabolism , Humans , Immunophenotyping , Lymphocyte Count , Primary Immunodeficiency Diseases , Sirolimus/pharmacology , T-Lymphocyte Subsets/drug effects
9.
Blood ; 127(8): 997-1006, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26744459

ABSTRACT

Genetic disorders affecting biogenesis and transport of lysosome-related organelles are heterogeneous diseases frequently associated with albinism. We studied a patient with albinism, neutropenia, immunodeficiency, neurodevelopmental delay, generalized seizures, and impaired hearing but with no mutation in genes so far associated with albinism and immunodeficiency. Whole exome sequencing identified a homozygous mutation in AP3D1 that leads to destabilization of the adaptor protein 3 (AP3) complex. AP3 complex formation and the degranulation defect in patient T cells were restored by retroviral reconstitution. A previously described hypopigmented mouse mutant with an Ap3d1 null mutation (mocha strain) shares the neurologic phenotype with our patient and shows a platelet storage pool deficiency characteristic of Hermansky-Pudlak syndrome (HPS) that was not studied in our patient because of a lack of bleeding. HPS2 caused by mutations in AP3B1A leads to a highly overlapping phenotype without the neurologic symptoms. The AP3 complex exists in a ubiquitous and a neuronal form. AP3D1 codes for the AP3δ subunit of the complex, which is essential for both forms. In contrast, the AP3ß3A subunit, affected in HPS2 patients, is substituted by AP3ß3B in the neuron-specific heterotetramer. AP3δ deficiency thus causes a severe neurologic disorder with immunodeficiency and albinism that we propose to classify as HPS10.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex delta Subunits/genetics , Hermanski-Pudlak Syndrome/classification , Hermanski-Pudlak Syndrome/genetics , Immunologic Deficiency Syndromes/genetics , Seizures/genetics , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Mutation , Transfection
10.
Blood ; 127(25): 3281-90, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27099148

ABSTRACT

Reduced-intensity conditioning has improved survival after hematopoietic stem cell transplantation (HSCT) for hemophagocytic lymphohistiocytosis (HLH) at the cost of more frequent mixed chimerism. The minimum level of donor chimerism (DC) required to prevent HLH reactivation in humans remains to be determined. In a multicenter retrospective study, 103 patients transplanted for hereditary HLH (2000-2013) and DC permanently or transiently <75% (overall, CD3(+), CD56(+)) were analyzed regarding DC, specific immunologic function, occurrence of systemic reactivations (≥5/8 HLH criteria), partial systemic flares (<5 criteria and HLH-directed treatment), isolated central nervous system reactivations, and management. Recurrence was reported in 18 patients (systemic reactivation n = 11, partial flare n = 3, isolated central nervous system reactivation n = 4). Ten events occurred during profound immune suppression before day 180 (median DC, 10%; range, 1-100%; CD3(+) if available, otherwise overall DC), which renders a differentiation between secondary post-HSCT HLH and HLH related to the genetic defect difficult. Eight events occurred between 0.5 and 6.7 years post-HSCT (median DC, 13%; range, 0-30%). In 5 patients, overall and lineage-specific DC were ≤10% for >6 months (median, 5.1; range, 1.1-10 years) without reactivation. A second HSCT was performed in 18 patients (median, DC 4%; range, 0-19%). Death from reactivation occurred in 4 patients (22% of recurrences). Six patients died of transplant complications following a second HSCT (33% of second HSCT). We conclude that a DC >20%-30% is protective against late reactivation. Lower levels do not, however, inescapably result in recurrences. The decision for or against second HSCT must be based on a thorough risk assessment.


Subject(s)
Chimerism , Lymphohistiocytosis, Hemophagocytic/immunology , Lymphohistiocytosis, Hemophagocytic/therapy , Tissue Donors , Adolescent , Adult , Child , Child, Preschool , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Infant, Newborn , Male , Recurrence , Retrospective Studies , Transplantation Conditioning/methods , Transplantation Immunology , Transplantation, Homologous/adverse effects , Treatment Outcome , Young Adult
11.
Pediatr Blood Cancer ; 65(11): e27344, 2018 11.
Article in English | MEDLINE | ID: mdl-30070073

ABSTRACT

In this report, we evaluate the hypothesis that hemophagocytic lymphohistiocytosis in patients with defects of lymphocyte cytotoxicity is usually triggered by infections. We show that in the majority of patients, extensive virus PCR panels performed in addition to routine microbiological investigations remain negative and summarize 25 patients with onset of hemophagocytic lymphohistiocytosis in utero or within the first 10 days of life, in none of which an associated bacterial or viral infection was reported. These observations, even though preliminary, invite to consider a key role of lymphocyte cytotoxicity in controlling T-cell homeostasis also in the absence of apparent infectious stimuli.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/etiology , Bacterial Infections/epidemiology , Child , Female , Fetus , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases/etiology , Male , Pregnancy , Virus Diseases/epidemiology , Young Adult
12.
J Clin Immunol ; 37(8): 770-780, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28936583

ABSTRACT

We report our experience in using flow cytometry-based immunological screening prospectively as a decision tool for the use of genetic studies in the diagnostic approach to patients with hemophagocytic lymphohistiocytosis (HLH). We restricted genetic analysis largely to patients with abnormal immunological screening, but included whole exome sequencing (WES) for those with normal findings upon Sanger sequencing. Among 290 children with suspected HLH analyzed between 2010 and 2014 (including 17 affected, but asymptomatic siblings), 87/162 patients with "full" HLH and 79/111 patients with "incomplete/atypical" HLH had normal immunological screening results. In 10 patients, degranulation could not be tested. Among the 166 patients with normal screening, genetic analysis was not performed in 107 (all with uneventful follow-up), while 154 single gene tests by Sanger sequencing in the remaining 59 patients only identified a single atypical CHS patient. Flow cytometry correctly predicted all 29 patients with FHL-2, XLP1 or 2. Among 85 patients with defective NK degranulation (including 13 asymptomatic siblings), 70 were Sanger sequenced resulting in a genetic diagnosis in 55 (79%). Eight patients underwent WES, revealing mutations in two known and one unknown cytotoxicity genes and one metabolic disease. FHL3 was the most frequent genetic diagnosis. Immunological screening provided an excellent decision tool for the need and depth of genetic analysis of HLH patients and provided functionally relevant information for rapid patient classification, contributing to a significant reduction in the time from diagnosis to transplantation in recent years.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/genetics , Lymphohistiocytosis, Hemophagocytic/diagnosis , Muscle Proteins/genetics , Transcription Factors/genetics , Asymptomatic Diseases , Cell Degranulation , Child , Flow Cytometry , Genetic Testing , Humans , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , LIM-Homeodomain Proteins/metabolism , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/immunology , Muscle Proteins/metabolism , Mutation/genetics , Organ Transplantation , Practice Guidelines as Topic , Prognosis , Prospective Studies , Siblings , Transcription Factors/metabolism , Exome Sequencing
13.
Eur J Immunol ; 46(11): 2639-2649, 2016 11.
Article in English | MEDLINE | ID: mdl-27615517

ABSTRACT

Tyrosine kinase 2 (TYK2) associates with interferon (IFN) alpha receptor, IL-10 receptor (IL-10R) beta and other cytokine receptor subunits for signal transduction, in response to various cytokines, including type-I and type-III IFNs, IL-6, IL-10, IL-12 and IL-23. Data on TYK2 dependence on cytokine responses and in vivo consequences of TYK2 deficiency are inconsistent. We investigated a TYK2 deficient patient, presenting with eczema, skin abscesses, respiratory infections and IgE levels >1000 U/mL, without viral or mycobacterial infections and a corresponding cellular model to analyze the role of TYK2 in type-III IFN mediated responses and NK-cell function. We established a novel simple diagnostic monocyte assay to show that the mutation completely abolishes the IFN-α mediated antiviral response. It also partly reduces IL-10 but not IL-6 mediated signaling associated with reduced IL-10Rß expression. However, we found almost normal type-III IFN signaling associated with minimal impairment of virus control in a TYK2 deficient human cell line. Contrary to observations in TYK2 deficient mice, NK-cell phenotype and function, including IL-12/IL-18 mediated responses, were normal in the patient. Thus, preserved type-III IFN responses and normal NK-cell function may contribute to antiviral protection in TYK2 deficiency leading to a surprisingly mild human phenotype.


Subject(s)
Interferons/immunology , Job Syndrome/immunology , Killer Cells, Natural/immunology , TYK2 Kinase/deficiency , TYK2 Kinase/metabolism , Animals , Cell Line , Child , Disease Susceptibility/immunology , Disease Susceptibility/virology , Eczema/etiology , Eczema/immunology , Humans , Immunoglobulin E/blood , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mutation , Receptors, Cytokine/immunology , Receptors, Interleukin-10/genetics , Receptors, Interleukin-10/immunology , Signal Transduction/immunology , Skin/pathology , TYK2 Kinase/genetics , TYK2 Kinase/immunology
14.
J Clin Immunol ; 36(7): 733-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27492372

ABSTRACT

The X-linked inhibitor of apoptosis (XIAP) deficiency is a primary immunodeficiency characterized by Epstein-Barr virus (EBV)-driven hemophagocytic lymphohistiocytosis (HLH), splenomegaly, and colitis. Here, we present, for the first time, granulomatous hepatitis and granulomatous and lymphocytic interstitial lung disease (GLILD) as manifestations of XIAP deficiency. We report successful treatment of GLILD in XIAP deficiency with rituximab and azathioprine and discuss the role of XIAP deficiency in immune dysregulation.


Subject(s)
Genetic Diseases, X-Linked/complications , Hepatitis/complications , Lung Diseases, Interstitial/complications , Lymphohistiocytosis, Hemophagocytic/complications , Lymphoproliferative Disorders/complications , Azathioprine/therapeutic use , Biomarkers , Biopsy , Bone Marrow/pathology , Child, Preschool , Female , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/therapy , Hepatitis/diagnosis , Hepatitis/therapy , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Immunophenotyping , Liver/pathology , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/drug therapy , Lymph Nodes/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/therapy , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/therapy , Male , Mutation , Pedigree , Phenotype , Respiratory Function Tests , Rituximab/therapeutic use , Tomography, X-Ray Computed , X-Linked Inhibitor of Apoptosis Protein/genetics
15.
Blood ; 124(6): 851-60, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24894771

ABSTRACT

Accumulation of CD3(+) T-cell receptor (TCR)αß(+)CD4(-)CD8(-) double-negative T cells (DNT) is a hallmark of autoimmune lymphoproliferative syndrome (ALPS). DNT origin and differentiation pathways remain controversial. Here we show that human ALPS DNT have features of terminally differentiated effector memory T cells reexpressing CD45RA(+) (TEMRA), but are CD27(+)CD28(+)KLRG1(-) and do not express the transcription factor T-bet. This unique phenotype was also detected among CD4(+) or CD8(+) ALPS TEMRA cells. T-cell receptor ß deep sequencing revealed a significant fraction of shared CDR3 sequences between ALPS DNT and both CD4(+) and CD8(+)TEMRA cells. Moreover, in ALPS patients with a germ line FAS mutation and somatic loss of heterozygosity, in whom biallelic mutant cells can be tracked by absent Fas expression, Fas-negative T cells accumulated not only among DNT, but also among CD4(+) and CD8(+)TEMRA cells. These data indicate that in human Fas deficiency DNT cannot only derive from CD8(+), but also from CD4(+) T cells. Furthermore, defective Fas signaling leads to aberrant transcriptional programs and differentiation of subsets of CD4(+) and CD8(+) T cells. Accumulation of these cells before their double-negative state appears to be an important early event in the pathogenesis of lymphoproliferation in ALPS patients.


Subject(s)
Autoimmune Lymphoproliferative Syndrome/immunology , Autoimmune Lymphoproliferative Syndrome/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , fas Receptor/deficiency , fas Receptor/genetics , Adolescent , Adult , Autoimmune Lymphoproliferative Syndrome/genetics , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Child , Child, Preschool , Genetic Association Studies , Germ-Line Mutation , Humans , Immunologic Memory , Leukocyte Common Antigens/metabolism , Loss of Heterozygosity , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Young Adult
16.
J Allergy Clin Immunol ; 136(2): 392-401, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25702838

ABSTRACT

BACKGROUND: Most patients with MHC class I (MHC-I) deficiency carry genetic defects in transporter associated with antigen processing 1 (TAP1) or TAP2. The clinical presentation can vary, and about half of the patients have severe skin disease. Previously, one report described ß2-microglobulin (ß2m) deficiency as another monogenetic cause of MHC-I deficiency, but no further immunologic evaluation was performed. OBJECTIVE: We sought to describe the molecular and immunologic features of ß2m deficiency in 2 Turkish siblings with new diagnoses. METHODS: Based on clinical and serologic findings, the genetic defect was detected by means of candidate gene analysis. The immunologic characterization comprises flow cytometry, ELISA, functional assays, and immunohistochemistry. RESULTS: Here we provide the first extensive clinical and immunologic description of ß2m deficiency in 2 siblings. The sister had recurrent respiratory tract infections and severe skin disease, whereas the brother was fairly asymptomatic but had bronchiectasis. Not only polymorphic MHC-I but also the related CD1a, CD1b, CD1c, and neonatal Fc receptor molecules were absent from the surfaces of ß2m-deficient cells. Absent neonatal Fc receptor surface expression led to low serum IgG and albumin levels in both siblings, whereas the heterozygous parents had normal results for all tested parameters except ß2m mRNA (B2M) expression. Similar to TAP deficiency in the absence of a regular CD8 T-cell compartment, CD8(+) γδ T cells were strongly expanded. Natural killer cells were normal in number but not "licensed to kill." CONCLUSION: The clinical presentation of patients with ß2m deficiency resembles that of patients with other forms of MHC-I deficiency, but because of the missing stabilizing effect of ß2m on other members of the MHC-I family, the immunologic defect is more extensive than in patients with TAP deficiency.


Subject(s)
Bronchiectasis/immunology , Immunologic Deficiency Syndromes/immunology , Respiratory Tract Infections/immunology , Skin Ulcer/immunology , beta 2-Microglobulin/immunology , Adaptive Immunity , Adolescent , Adult , Antigens, CD1/genetics , Antigens, CD1/immunology , Bronchiectasis/complications , Bronchiectasis/genetics , Bronchiectasis/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Consanguinity , Female , Gene Deletion , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Innate , Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Male , Pedigree , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/immunology , Receptors, Fc/deficiency , Receptors, Fc/genetics , Receptors, Fc/immunology , Recurrence , Respiratory Tract Infections/complications , Respiratory Tract Infections/genetics , Respiratory Tract Infections/pathology , Siblings , Skin Ulcer/complications , Skin Ulcer/genetics , Skin Ulcer/pathology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
18.
J Clin Immunol ; 35(5): 439-44, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25943627

ABSTRACT

PURPOSE: X-linked inhibitor of apoptosis (XIAP) deficiency caused by mutations in BIRC4 was originally described in male patients with X-linked lymphoproliferative syndrome type 2 (XLP2). Recent observations have highlighted a critical role of XIAP for the regulation of NOD2 signaling and are probably the molecular basis for increasingly recognized further immune dysregulatory symptoms of XIAP deficient patients, such as inflammatory bowel disease (IBD). We describe a large Caucasian family in which IBD and erythema nodosum (EN) also manifested in female carriers of XIAP mutations. METHODS: Clinical data and laboratory findings including flow cytometric analysis of XIAP protein expression and sequencing of the BIRC4 gene. NOD2 signaling was investigated by determination of TNFα production in monocytes upon L18-MDP stimulation in vitro. RESULTS: The BIRC4 nonsense mutation p.P225SfsX226 was identified as the genetic cause of XIAP deficiency in our family. Surprisingly, clinical symptoms were not restricted to male patients, but also occurred in several female carriers. The most severely affected carrier demonstrated random X-inactivation, leading to a significant expression of mutated XIAP protein in monocytes, and consequently to impaired NOD2 responses in vitro. CONCLUSION: Our report provides further evidence that clinical symptoms of XIAP deficiency are not restricted to male patients. Random X-inactivation may be associated with EN and mild IBD also in female carriers of BIRC4 mutations. Analysis of the X-inactivation pattern reflected by XIAP protein expression can identify such carriers and the analysis of NOD2 signaling by flow cytometry can confirm the functional significance. XIAP expression patterns should be investigated in female patients with a family history of EN and/or IBD.


Subject(s)
Erythema Nodosum/diagnosis , Genetic Diseases, X-Linked/diagnosis , Inflammatory Bowel Diseases/diagnosis , Lymphoproliferative Disorders/diagnosis , Monocytes/metabolism , Virus Diseases/diagnosis , X-Linked Inhibitor of Apoptosis Protein/genetics , Acetylmuramyl-Alanyl-Isoglutamine/analogs & derivatives , Acetylmuramyl-Alanyl-Isoglutamine/immunology , Adult , Aged , Aged, 80 and over , Carrier State , Cells, Cultured , Child , Child, Preschool , Erythema Nodosum/etiology , Erythema Nodosum/genetics , Fatal Outcome , Female , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/genetics , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/genetics , Lymphoproliferative Disorders/complications , Lymphoproliferative Disorders/genetics , Male , Middle Aged , Monocytes/immunology , Mutation/genetics , Nod2 Signaling Adaptor Protein/metabolism , Pedigree , Sex Factors , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism , Virus Diseases/etiology , Virus Diseases/genetics , White People
19.
J Clin Immunol ; 35(1): 22-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25491289

ABSTRACT

Familial hemophagocytic lymphohistiocytosis (FHL) is a genetically heterogeneous hyperinflammatory syndrome, caused by an uncontrolled and ineffective proliferation and activation of T-lymphocytes, NK-cells, and macrophages that infiltrate multiple organs. Herein, a patient is presented who suffered from hepatitis and atypical brain lesions. Genetic studies revealed a homozygous mutation in the STXP2 gene; and thus, the diagnosis of FHL5 was confirmed.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/genetics , Munc18 Proteins/genetics , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Age of Onset , Brain/pathology , Child , Fatal Outcome , Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/immunology , Homozygote , Humans , Killer Cells, Natural/immunology , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/immunology , Magnetic Resonance Imaging , Male , Mutation , T-Lymphocytes/immunology
20.
Blood ; 121(15): 2943-51, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23403622

ABSTRACT

Genetic disorders of lymphocyte cytotoxicity predispose patients to hemophagocytic lymphohistiocytosis (HLH). Reduced lymphocyte cytotoxicity has been demonstrated in Hermansky-Pudlak syndrome type 2 (HPS2), but only a single patient was reported who developed HLH. Because that patient also carried a potentially contributing heterozygous RAB27A mutation, the risk for HLH in HPS2 remains unclear. We analyzed susceptibility to HLH in the pearl mouse model of HPS2. After infection with lymphocytic choriomeningitis virus, pearl mice developed all key features of HLH, linked to impaired virus control caused by a moderate defect in CTL cytotoxicity in vivo. However, in contrast to perforin-deficient mice, the disease was transient, and all mice fully recovered and controlled the infection. An additional heterozygous Rab27a mutation did not aggravate the cytotoxicity defect or disease parameters. In the largest survey of 22 HPS2 patients covering 234 patient years, we identified only 1 additional patient with HLH and 2 with incomplete transient HLH-like episodes, although cytotoxicity or degranulation was impaired in all 16 patients tested. HPS2 confers a risk for HLH that is lower than in Griscelli or Chediak-Higashi syndrome, probably because of a milder defect in cytotoxicity. Preemptive hematopoietic stem cell transplantation does not appear justified in HPS2.


Subject(s)
Cytotoxicity, Immunologic/immunology , Hermanski-Pudlak Syndrome/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , T-Lymphocytes, Cytotoxic/immunology , Adaptor Protein Complex 3/deficiency , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex 3/immunology , Adaptor Protein Complex beta Subunits/deficiency , Adaptor Protein Complex beta Subunits/genetics , Adaptor Protein Complex beta Subunits/immunology , Adolescent , Adult , Animals , Child , Child, Preschool , Cytotoxicity, Immunologic/genetics , Flow Cytometry , Hermanski-Pudlak Syndrome/complications , Hermanski-Pudlak Syndrome/genetics , Humans , Lymphohistiocytosis, Hemophagocytic/etiology , Lymphohistiocytosis, Hemophagocytic/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Risk Factors , T-Lymphocytes, Cytotoxic/metabolism , Young Adult , rab GTP-Binding Proteins/deficiency , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/immunology , rab27 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL