32 results on '"Ehl, S."'
Search Results
2. Efficacy of T-cell assays for the diagnosis of primary defects in cytotoxic lymphocyte exocytosis.
- Author
-
Chiang SCC, Covill LE, Tesi B, Campbell TM, Schlums H, Nejati-Zendegani J, Mördrup K, Wood S, Theorell J, Sekine T, Al-Herz W, Akar HH, Belen FB, Chan MY, Devecioglu O, Aksu T, Ifversen M, Malinowska I, Sabel M, Unal E, Unal S, Introne WJ, Krzewski K, Gilmour KC, Ehl S, Ljunggren HG, Nordenskjöld M, Horne A, Henter JI, Meeths M, and Bryceson YT
- Subjects
- Humans, Adolescent, Child, Adult, Female, K562 Cells, Male, Child, Preschool, Middle Aged, Infant, Young Adult, Aged, Sensitivity and Specificity, Prospective Studies, Receptors, Antigen, T-Cell metabolism, Receptors, Antigen, T-Cell genetics, Exocytosis, T-Lymphocytes, Cytotoxic immunology, Lymphohistiocytosis, Hemophagocytic diagnosis, Lymphohistiocytosis, Hemophagocytic immunology, Lymphohistiocytosis, Hemophagocytic genetics, Lymphohistiocytosis, Hemophagocytic pathology, Killer Cells, Natural immunology, Killer Cells, Natural metabolism
- Abstract
Abstract: Primary hemophagocytic lymphohistiocytosis (HLH) is a life-threatening disorder associated with autosomal recessive variants in genes required for perforin-mediated lymphocyte cytotoxicity. A rapid diagnosis is crucial for successful treatment. Although defective cytotoxic T lymphocyte (CTL) function causes pathogenesis, quantification of natural killer (NK)-cell exocytosis triggered by K562 target cells currently represents a standard diagnostic procedure for primary HLH. We have prospectively evaluated different lymphocyte exocytosis assays in 213 patients referred for evaluation for suspected HLH and related hyperinflammatory syndromes. A total of 138 patients received a molecular diagnosis consistent with primary HLH. Assessment of Fc receptor-triggered NK-cell and T-cell receptor (TCR)-triggered CTL exocytosis displayed higher sensitivity and improved specificity for the diagnosis of primary HLH than routine K562 cell-based assays, with these assays combined providing a sensitivity of 100% and specificity of 98.3%. By comparison, NK-cell exocytosis after K562 target cell stimulation displayed a higher interindividual variability, in part explained by differences in NK-cell differentiation or large functional reductions after shipment. We thus recommend combined analysis of TCR-triggered CTL and Fc receptor-triggered NK-cell exocytosis for the diagnosis of patients with suspected familial HLH or atypical manifestations of congenital defects in lymphocyte exocytosis., (Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution.)
- Published
- 2024
- Full Text
- View/download PDF
3. Survival in primary hemophagocytic lymphohistiocytosis, 2016 to 2021: etoposide is better than its reputation.
- Author
-
Böhm S, Wustrau K, Pachlopnik Schmid J, Prader S, Ahlmann M, Yacobovich J, Beier R, Speckmann C, Behnisch W, Ifversen M, Jordan M, Marsh R, Naumann-Bartsch N, Mauz-Körholz C, Hönig M, Schulz A, Malinowska I, Hines M, Nichols KE, Gil-Herrera J, Talano JA, Crooks B, Formankova R, Jorch N, Bakhtiar S, Kühnle I, Streiter M, Nathrath M, Russo A, Dürken M, Lang P, Lindemans C, Henter JI, Lehmberg K, and Ehl S
- Subjects
- Infant, Newborn, Humans, Etoposide therapeutic use, Treatment Outcome, Lymphohistiocytosis, Hemophagocytic drug therapy, Lymphohistiocytosis, Hemophagocytic diagnosis, Hematopoietic Stem Cell Transplantation methods, Lymphoproliferative Disorders etiology
- Abstract
Abstract: Primary hemophagocytic lymphohistiocytosis (pHLH) is a life-threatening hyperinflammatory syndrome that develops mainly in patients with genetic disorders of lymphocyte cytotoxicity and X-linked lymphoproliferative syndromes. Previous studies with etoposide-based treatment followed by hematopoetic stem cell transplantation (HSCT) resulted in 5-year survival of 50% to 59%. Contemporary data are lacking. We evaluated 88 patients with pHLH documented in the international HLH registry from 2016-2021. In 12 of 88 patients, diagnosis was made without HLH activity, based on siblings or albinism. Major HLH-directed drugs (etoposide, antithymocyte globulin, alemtuzumab, emapalumab, ruxolitinib) were administered to 66 of 76 patients who were symptomatic (86% first-line etoposide); 16 of 57 patients treated with etoposide and 3 of 9 with other first-line treatment received salvage therapy. HSCT was performed in 75 patients; 7 patients died before HSCT. Three-year probability of survival (pSU) was 82% (confidence interval [CI], 72%-88%) for the entire cohort and 77% (CI, 64%-86%) for patients receiving first-line etoposide. Compared with the HLH-2004 study, both pre-HSCT and post-HSCT survival of patients receiving first-line etoposide improved, 83% to 91% and 70% to 88%. Differences to HLH-2004 included preferential use of reduced-toxicity conditioning and reduced time from diagnosis to HSCT (from 148 to 88 days). Three-year pSU was lower with haploidentical (4 of 9 patients [44%]) than with other donors (62 of 66 [94%]; P < .001). Importantly, early HSCT for patients who were asymptomatic resulted in 100% survival, emphasizing the potential benefit of newborn screening. This contemporary standard-of-care study of patients with pHLH reveals that first-line etoposide-based therapy is better than previously reported, providing a benchmark for novel treatment regimes., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
4. Germline STAT3 gain-of-function mutations in primary immunodeficiency: Impact on the cellular and clinical phenotype.
- Author
-
Faletti L, Ehl S, and Heeg M
- Subjects
- Germ Cells metabolism, Humans, Mutation genetics, Phenotype, Gain of Function Mutation, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism
- Abstract
Signal transducer and activator of transcription 3 (STAT3) is a key transcription factor involved in regulation of immune cell activation and differentiation. Recent discoveries highlight the role of germline activating STAT3 mutations in inborn errors of immunity characterized by early-onset multi-organ autoimmunity and lymphoproliferation. Much progress has been made in defining the clinical spectrum of STAT3 GOF disease and unraveling the molecular and cellular mechanisms underlying this disease. In this review, we summarize our current understanding of the disease and discuss the clinical phenotype, diagnostic approach, cellular and molecular effects of STAT3 GOF mutations and therapeutic concepts for these patients., Competing Interests: Conflicts of interest The authors declare no conflicts of interest., (Copyright © 2021 Chang Gung University. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
5. Germline TET2 loss of function causes childhood immunodeficiency and lymphoma.
- Author
-
Stremenova Spegarova J, Lawless D, Mohamad SMB, Engelhardt KR, Doody G, Shrimpton J, Rensing-Ehl A, Ehl S, Rieux-Laucat F, Cargo C, Griffin H, Mikulasova A, Acres M, Morgan NV, Poulter JA, Sheridan EG, Chetcuti P, O'Riordan S, Anwar R, Carter CR, Przyborski S, Windebank K, Cant AJ, Lako M, Bacon CM, Savic S, and Hambleton S
- Subjects
- Allografts, Apoptosis, B-Lymphocyte Subsets pathology, Cellular Reprogramming Techniques, Codon, Nonsense, DNA Methylation, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Dioxygenases, Fatal Outcome, Female, Hematopoietic Stem Cell Transplantation, Humans, Induced Pluripotent Stem Cells pathology, Infant, Newborn, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Lymphoma, T-Cell, Peripheral genetics, Lymphoma, T-Cell, Peripheral pathology, Male, Mutation, Missense, Neoplasms, Multiple Primary genetics, Pedigree, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins physiology, Severe Combined Immunodeficiency pathology, T-Lymphocyte Subsets pathology, Exome Sequencing, DNA-Binding Proteins deficiency, Germ-Line Mutation, Loss of Function Mutation, Lymphoproliferative Disorders genetics, Proto-Oncogene Proteins deficiency, Severe Combined Immunodeficiency genetics
- Abstract
Molecular dissection of inborn errors of immunity can help to elucidate the nonredundant functions of individual genes. We studied 3 children with an immune dysregulation syndrome of susceptibility to infection, lymphadenopathy, hepatosplenomegaly, developmental delay, autoimmunity, and lymphoma of B-cell (n = 2) or T-cell (n = 1) origin. All 3 showed early autologous T-cell reconstitution following allogeneic hematopoietic stem cell transplantation. By whole-exome sequencing, we identified rare homozygous germline missense or nonsense variants in a known epigenetic regulator of gene expression: ten-eleven translocation methylcytosine dioxygenase 2 (TET2). Mutated TET2 protein was absent or enzymatically defective for 5-hydroxymethylating activity, resulting in whole-blood DNA hypermethylation. Circulating T cells showed an abnormal immunophenotype including expanded double-negative, but depleted follicular helper, T-cell compartments and impaired Fas-dependent apoptosis in 2 of 3 patients. Moreover, TET2-deficient B cells showed defective class-switch recombination. The hematopoietic potential of patient-derived induced pluripotent stem cells was skewed toward the myeloid lineage. These are the first reported cases of autosomal-recessive germline TET2 deficiency in humans, causing clinically significant immunodeficiency and an autoimmune lymphoproliferative syndrome with marked predisposition to lymphoma. This disease phenotype demonstrates the broad role of TET2 within the human immune system., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
6. Interleukin-18 diagnostically distinguishes and pathogenically promotes human and murine macrophage activation syndrome.
- Author
-
Weiss ES, Girard-Guyonvarc'h C, Holzinger D, de Jesus AA, Tariq Z, Picarsic J, Schiffrin EJ, Foell D, Grom AA, Ammann S, Ehl S, Hoshino T, Goldbach-Mansky R, Gabay C, and Canna SW
- Subjects
- 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, Interleukin-18 immunology, Macrophage Activation Syndrome immunology, Signal Transduction immunology
- 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 NLRC4
T337S 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.- Published
- 2018
- Full Text
- View/download PDF
7. Etoposide for HLH: the limits of efficacy.
- Author
-
Ehl S
- Subjects
- Epstein-Barr Virus Infections, Humans, Etoposide, Lymphohistiocytosis, Hemophagocytic
- Abstract
Competing Interests: Conflict-of-interest disclosure: The author declares no competing financials interests.
- Published
- 2017
- Full Text
- View/download PDF
8. Hematopoietic stem cell transplantation in 29 patients hemizygous for hypomorphic IKBKG /NEMO mutations.
- Author
-
Miot C, Imai K, Imai C, Mancini AJ, Kucuk ZY, Kawai T, Nishikomori R, Ito E, Pellier I, Dupuis Girod S, Rosain J, Sasaki S, Chandrakasan S, Pachlopnik Schmid J, Okano T, Colin E, Olaya-Vargas A, Yamazaki-Nakashimada M, Qasim W, Espinosa Padilla S, Jones A, Krol A, Cole N, Jolles S, Bleesing J, Vraetz T, Gennery AR, Abinun M, Güngör T, Costa-Carvalho B, Condino-Neto A, Veys P, Holland SM, Uzel G, Moshous D, Neven B, Blanche S, Ehl S, Döffinger R, Patel SY, Puel A, Bustamante J, Gelfand EW, Casanova JL, Orange JS, and Picard C
- Subjects
- Child, Preschool, Cohort Studies, Heterozygote, Humans, Infant, Infant, Newborn, Inflammation pathology, Inflammatory Bowel Diseases etiology, NF-kappa B metabolism, Phenotype, Signal Transduction genetics, Survival Analysis, Tissue Donors, Transplantation Conditioning, Treatment Outcome, Hematopoietic Stem Cell Transplantation adverse effects, I-kappa B Kinase genetics, Mutation genetics
- Abstract
X-linked recessive ectodermal dysplasia with immunodeficiency is a rare primary immunodeficiency caused by hypomorphic mutations of the IKBKG gene encoding the nuclear factor κB essential modulator (NEMO) protein. This condition displays enormous allelic, immunological, and clinical heterogeneity, and therapeutic decisions are difficult because NEMO operates in both hematopoietic and nonhematopoietic cells. Hematopoietic stem cell transplantation (HSCT) is potentially life-saving, but the small number of case reports available suggests it has been reserved for only the most severe cases. Here, we report the health status before HSCT, transplantation outcome, and clinical follow-up for a series of 29 patients from unrelated kindreds from 11 countries. Between them, these patients carry 23 different hypomorphic IKBKG mutations. HSCT was performed from HLA-identical related donors (n = 7), HLA-matched unrelated donors (n = 12), HLA-mismatched unrelated donors (n = 8), and HLA-haploidentical related donors (n = 2). Engraftment was documented in 24 patients, and graft-versus-host disease in 13 patients. Up to 7 patients died 0.2 to 12 months after HSCT. The global survival rate after HSCT among NEMO-deficient children was 74% at a median follow-up after HSCT of 57 months (range, 4-108 months). Preexisting mycobacterial infection and colitis were associated with poor HSCT outcome. The underlying mutation does not appear to have any influence, as patients with the same mutation had different outcomes. Transplantation did not appear to cure colitis, possibly as a result of cell-intrinsic disorders of the epithelial barrier. Overall, HSCT can cure most clinical features of patients with a variety of IKBKG mutations.
- Published
- 2017
- Full Text
- View/download PDF
9. Hyperactive mTOR pathway promotes lymphoproliferation and abnormal differentiation in autoimmune lymphoproliferative syndrome.
- Author
-
Völkl S, Rensing-Ehl A, Allgäuer A, Schreiner E, Lorenz MR, Rohr J, Klemann C, Fuchs I, Schuster V, von Bueren AO, Naumann-Bartsch N, Gambineri E, Siepermann K, Kobbe R, Nathrath M, Arkwright PD, Miano M, Stachel KD, Metzler M, Schwarz K, Kremer AN, Speckmann C, Ehl S, and Mackensen A
- Subjects
- Adolescent, Adult, Autoimmune Lymphoproliferative Syndrome pathology, CD4-Positive T-Lymphocytes pathology, CD8-Positive T-Lymphocytes pathology, Child, Child, Preschool, Female, Humans, Lectins, C-Type immunology, Leukocyte Common Antigens immunology, Male, Proto-Oncogene Proteins c-akt immunology, Receptors, Antigen, T-Cell immunology, Receptors, Immunologic, Trans-Activators immunology, Autoimmune Lymphoproliferative Syndrome immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cell Differentiation immunology, Signal Transduction immunology, TOR Serine-Threonine Kinases immunology
- Abstract
Autoimmune lymphoproliferative syndrome (ALPS) is a human disorder characterized by defective Fas signaling, resulting in chronic benign lymphoproliferation and accumulation of TCRαβ(+) CD4(-) CD8(-) double-negative T (DNT) cells. Although their phenotype resembles that of terminally differentiated or exhausted T cells, lack of KLRG1, high eomesodermin, and marginal T-bet expression point instead to a long-lived memory state with potent proliferative capacity. Here we show that despite their terminally differentiated phenotype, human ALPS DNT cells exhibit substantial mitotic activity in vivo. Notably, hyperproliferation of ALPS DNT cells is associated with increased basal and activation-induced phosphorylation of serine-threonine kinases Akt and mechanistic target of rapamycin (mTOR). The mTOR inhibitor rapamycin abrogated survival and proliferation of ALPS DNT cells, but not of CD4(+) or CD8(+) T cells in vitro. In vivo, mTOR inhibition reduced proliferation and abnormal differentiation by DNT cells. Importantly, increased mitotic activity and hyperactive mTOR signaling was also observed in recently defined CD4(+) or CD8(+) precursor DNT cells, and mTOR inhibition specifically reduced these cells in vivo, indicating abnormal programming of Fas-deficient T cells before the DNT stage. Thus, our results identify the mTOR pathway as a major regulator of lymphoproliferation and aberrant differentiation in ALPS., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
10. The minimum required level of donor chimerism in hereditary hemophagocytic lymphohistiocytosis.
- Author
-
Hartz B, Marsh R, Rao K, Henter JI, Jordan M, Filipovich L, Bader P, Beier R, Burkhardt B, Meisel R, Schulz A, Winkler B, Albert MH, Greil J, Karasu G, Woessmann W, Corbacioglu S, Gruhn B, Holter W, Kühl JS, Lang P, Seidel MG, Veys P, Löfstedt A, Ammann S, Ehl S, Janka G, Müller I, and Lehmberg K
- Subjects
- 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, Chimerism, Lymphohistiocytosis, Hemophagocytic immunology, Lymphohistiocytosis, Hemophagocytic therapy, Tissue Donors
- 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., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
11. Disturbed B-lymphocyte selection in autoimmune lymphoproliferative syndrome.
- Author
-
Janda A, Schwarz K, van der Burg M, Vach W, Ijspeert H, Lorenz MR, Elgizouli M, Pieper K, Fisch P, Hagel J, Lorenzetti R, Seidl M, Roesler J, Hauck F, Traggiai E, Speckmann C, Rensing-Ehl A, Ehl S, Eibel H, and Rizzi M
- Subjects
- Apoptosis, Autoimmunity, Cell Line, Transformed, Cell Transformation, Neoplastic, Child, Codon, Nonsense, Female, Frameshift Mutation, Germ-Line Mutation, Heterozygote, Humans, Immunologic Memory, Loss of Heterozygosity, Male, Sequence Analysis, DNA, Somatic Hypermutation, Immunoglobulin, V(D)J Recombination, fas Receptor deficiency, fas Receptor genetics, Autoimmune Lymphoproliferative Syndrome immunology, B-Lymphocyte Subsets immunology, Clonal Selection, Antigen-Mediated, Mutation, fas Receptor physiology
- Abstract
Fas is a transmembrane receptor involved in the maintenance of tolerance and immune homeostasis. In murine models, it has been shown to be essential for deletion of autoreactive B cells in the germinal center. The role of Fas in human B-cell selection and in development of autoimmunity in patients carrying FAS mutations is unclear. We analyzed patients with either a somatic FAS mutation or a germline FAS mutation and somatic loss-of-heterozygosity, which allows comparing the fate of B cells with impaired vs normal Fas signaling within the same individual. Class-switched memory B cells showed: accumulation of FAS-mutated B cells; failure to enrich single V, D, J genes and single V-D, D-J gene combinations of the B-cell receptor variable region; increased frequency of variable regions with higher content of positively charged amino acids; and longer CDR3 and maintenance of polyreactive specificities. Importantly, Fas-deficient switched memory B cells showed increased rates of somatic hypermutation. Our data uncover a defect in B-cell selection in patients with FAS mutations, which has implications for the understanding of the pathogenesis of autoimmunity and lymphomagenesis of autoimmune lymphoproliferative syndrome., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
12. Mutations in AP3D1 associated with immunodeficiency and seizures define a new type of Hermansky-Pudlak syndrome.
- Author
-
Ammann S, Schulz A, Krägeloh-Mann I, Dieckmann NM, Niethammer K, Fuchs S, Eckl KM, Plank R, Werner R, Altmüller J, Thiele H, Nürnberg P, Bank J, Strauss A, von Bernuth H, Zur Stadt U, Grieve S, Griffiths GM, Lehmberg K, Hennies HC, and Ehl S
- Subjects
- Electrophoresis, Polyacrylamide Gel, Fluorescent Antibody Technique, Humans, Mutation, Transfection, 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
- 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 AP3β3A subunit, affected in HPS2 patients, is substituted by AP3β3B in the neuron-specific heterotetramer. AP3δ deficiency thus causes a severe neurologic disorder with immunodeficiency and albinism that we propose to classify as HPS10., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
13. Gray platelet syndrome can mimic autoimmune lymphoproliferative syndrome.
- Author
-
Rensing-Ehl A, Pannicke U, Zimmermann SY, Lorenz MR, Neven B, Fuchs I, Salzer U, Speckmann C, Strauss A, Maaβ E, Collet B, Enders A, Favier R, Alessi MC, Rieux-Laucat F, Zieger B, Schwarz K, and Ehl S
- Subjects
- Child, Preschool, Fas Ligand Protein genetics, Fas Ligand Protein metabolism, Female, Humans, Infant, Male, fas Receptor genetics, fas Receptor metabolism, Autoimmune Lymphoproliferative Syndrome diagnosis, Autoimmune Lymphoproliferative Syndrome genetics, Autoimmune Lymphoproliferative Syndrome metabolism, Autoimmune Lymphoproliferative Syndrome pathology, Blood Proteins genetics, Blood Proteins metabolism, Gray Platelet Syndrome diagnosis, Gray Platelet Syndrome genetics, Gray Platelet Syndrome metabolism, Gray Platelet Syndrome pathology, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism
- Published
- 2015
- Full Text
- View/download PDF
14. Omenn syndrome associated with a functional reversion due to a somatic second-site mutation in CARD11 deficiency.
- Author
-
Fuchs S, Rensing-Ehl A, Pannicke U, Lorenz MR, Fisch P, Jeelall Y, Rohr J, Speckmann C, Vraetz T, Farmand S, Schmitt-Graeff A, Krüger M, Strahm B, Henneke P, Enders A, Horikawa K, Goodnow C, Schwarz K, and Ehl S
- Subjects
- Animals, CARD Signaling Adaptor Proteins deficiency, CARD Signaling Adaptor Proteins immunology, Female, Flow Cytometry, Guanylate Cyclase deficiency, Guanylate Cyclase immunology, Humans, Immunoblotting, Immunohistochemistry, Immunophenotyping, Infant, Lymphocyte Activation immunology, Male, Mice, Real-Time Polymerase Chain Reaction, Severe Combined Immunodeficiency immunology, Siblings, CARD Signaling Adaptor Proteins genetics, Guanylate Cyclase genetics, Lymphocyte Activation genetics, Mutation, Severe Combined Immunodeficiency genetics, T-Lymphocytes, Regulatory immunology
- Abstract
Omenn syndrome (OS) is a severe immunodeficiency associated with erythroderma, lymphoproliferation, elevated IgE, and hyperactive oligoclonal T cells. A restricted T-cell repertoire caused by defective thymic T-cell development and selection, lymphopenia with homeostatic proliferation, and lack of regulatory T cells are considered key factors in OS pathogenesis. We report 2 siblings presenting with cytomegalovirus (CMV) and Pneumocystis jirovecii infections and recurrent sepsis; one developed all clinical features of OS. Both carried homozygous germline mutations in CARD11 (p.Cys150*), impairing NF-κB signaling and IL-2 production. A somatic second-site mutation reverting the stop codon to a missense mutation (p.Cys150Leu) was detected in tissue-infiltrating T cells of the OS patient. Expression of p.Cys150Leu in CARD11-deficient T cells largely reconstituted NF-κB signaling. The reversion likely occurred in a prethymic T-cell precursor, leading to a chimeric T-cell repertoire. We speculate that in our patient the functional advantage of the revertant T cells in the context of persistent CMV infection, combined with lack of regulatory T cells, may have been sufficient to favor OS. This first observation of OS in a patient with a T-cell activation defect suggests that severely defective T-cell development or homeostatic proliferation in a lymphopenic environment are not required for this severe immunopathology., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
15. Early-onset Evans syndrome, immunodeficiency, and premature immunosenescence associated with tripeptidyl-peptidase II deficiency.
- Author
-
Stepensky P, Rensing-Ehl A, Gather R, Revel-Vilk S, Fischer U, Nabhani S, Beier F, Brümmendorf TH, Fuchs S, Zenke S, Firat E, Pessach VM, Borkhardt A, Rakhmanov M, Keller B, Warnatz K, Eibel H, Niedermann G, Elpeleg O, and Ehl S
- Subjects
- Aminopeptidases deficiency, Aminopeptidases genetics, Anemia, Hemolytic, Autoimmune complications, Anemia, Hemolytic, Autoimmune immunology, Anemia, Hemolytic, Autoimmune pathology, Animals, Apoptosis, Base Sequence, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes pathology, Child, Child, Preschool, Consanguinity, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases deficiency, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases genetics, Female, Fibroblasts immunology, Fibroblasts metabolism, Fibroblasts pathology, Gene Expression, Humans, Immunologic Deficiency Syndromes complications, Immunologic Deficiency Syndromes immunology, Immunologic Deficiency Syndromes pathology, Male, Mice, Mice, Knockout, Molecular Sequence Data, Perforin genetics, Perforin immunology, Serine Endopeptidases deficiency, Serine Endopeptidases genetics, Siblings, T-Box Domain Proteins genetics, T-Box Domain Proteins immunology, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes pathology, Thrombocytopenia complications, Thrombocytopenia immunology, Thrombocytopenia pathology, Aging immunology, Aminopeptidases immunology, Anemia, Hemolytic, Autoimmune genetics, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases immunology, Frameshift Mutation, Immunologic Deficiency Syndromes genetics, Serine Endopeptidases immunology, Thrombocytopenia genetics
- Abstract
Autoimmune cytopenia is a frequent manifestation of primary immunodeficiencies. Two siblings presented with Evans syndrome, viral infections, and progressive leukopenia. DNA available from one patient showed a homozygous frameshift mutation in tripeptidyl peptidase II (TPP2) abolishing protein expression. TPP2 is a serine exopeptidase involved in extralysosomal peptide degradation. Its deficiency in mice activates cell death programs and premature senescence. Similar to cells from naïve, uninfected TPP2-deficient mice, patient cells showed increased major histocompatibility complex I expression and most CD8(+) T-cells had a senescent CCR7-CD127(-)CD28(-)CD57(+) phenotype with poor proliferative responses and enhanced staurosporine-induced apoptosis. T-cells showed increased expression of the effector molecules perforin and interferon-γ with high expression of the transcription factor T-bet. Age-associated B-cells with a CD21(-) CD11c(+) phenotype expressing T-bet were increased in humans and mice, combined with antinuclear antibodies. Moreover, markers of senescence were also present in human and murine TPP2-deficient fibroblasts. Telomere lengths were normal in patient fibroblasts and granulocytes, and low normal in lymphocytes, which were compatible with activation of stress-induced rather than replicative senescence programs. TPP2 deficiency is the first primary immunodeficiency linking premature immunosenescence to severe autoimmunity. Determination of senescent lymphocytes should be part of the diagnostic evaluation of children with refractory multilineage cytopenias., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
16. Abnormally differentiated CD4+ or CD8+ T cells with phenotypic and genetic features of double negative T cells in human Fas deficiency.
- Author
-
Rensing-Ehl A, Völkl S, Speckmann C, Lorenz MR, Ritter J, Janda A, Abinun M, Pircher H, Bengsch B, Thimme R, Fuchs I, Ammann S, Allgäuer A, Kentouche K, Cant A, Hambleton S, Bettoni da Cunha C, Huetker S, Kühnle I, Pekrun A, Seidel MG, Hummel M, Mackensen A, Schwarz K, and Ehl S
- Subjects
- 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, 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
- Abstract
Accumulation of CD3(+) T-cell receptor (TCR)αβ(+)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 β 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., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
17. Platelet secretion defect in a patient with stromal interaction molecule 1 deficiency.
- Author
-
Nakamura L, Sandrock-Lang K, Speckmann C, Vraetz T, Bührlen M, Ehl S, Heemskerk JW, and Zieger B
- Subjects
- Anemia, Hemolytic, Autoimmune blood, Anemia, Hemolytic, Autoimmune genetics, Calcium Signaling, Child, Preschool, Female, Homozygote, Humans, Point Mutation, Stromal Interaction Molecule 1, Syndrome, Thrombocytopenia blood, Thrombocytopenia genetics, Blood Platelets metabolism, Membrane Proteins deficiency, Membrane Proteins genetics, Neoplasm Proteins deficiency, Neoplasm Proteins genetics
- Published
- 2013
- Full Text
- View/download PDF
18. The risk of hemophagocytic lymphohistiocytosis in Hermansky-Pudlak syndrome type 2.
- Author
-
Jessen B, Bode SF, Ammann S, Chakravorty S, Davies G, Diestelhorst J, Frei-Jones M, Gahl WA, Gochuico BR, Griese M, Griffiths G, Janka G, Klein C, Kögl T, Kurnik K, Lehmberg K, Maul-Pavicic A, Mumford AD, Pace D, Parvaneh N, Rezaei N, de Saint Basile G, Schmitt-Graeff A, Schwarz K, Karasu GT, Zieger B, Zur Stadt U, Aichele P, and Ehl S
- Subjects
- 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, Cytotoxicity, Immunologic immunology, Hermanski-Pudlak Syndrome immunology, Lymphohistiocytosis, Hemophagocytic immunology, T-Lymphocytes, Cytotoxic immunology
- 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.
- Published
- 2013
- Full Text
- View/download PDF
19. Hemophagocytic lymphohistiocytosis in syntaxin-11-deficient mice: T-cell exhaustion limits fatal disease.
- Author
-
Kögl T, Müller J, Jessen B, Schmitt-Graeff A, Janka G, Ehl S, zur Stadt U, and Aichele P
- Subjects
- Animals, Antigens, CD metabolism, B7-H1 Antigen antagonists & inhibitors, Cell Degranulation genetics, Cell Degranulation immunology, Cytotoxicity, Immunologic genetics, Disease Models, Animal, Disease Progression, Interferon-gamma biosynthesis, Interferon-gamma immunology, Killer Cells, Natural immunology, Killer Cells, Natural metabolism, Lymphocyte Activation genetics, Lymphocyte Activation immunology, Lymphocytic choriomeningitis virus, Lymphohistiocytosis, Hemophagocytic immunology, Lymphohistiocytosis, Hemophagocytic virology, Mice, Mice, Knockout, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Cytotoxic metabolism, Lymphocyte Activation Gene 3 Protein, Lymphohistiocytosis, Hemophagocytic genetics, Qa-SNARE Proteins genetics
- Abstract
Syntaxin-11 (Stx11), an atypical member of the SNARE protein family, is part of the cytolytic machinery of T and NK cells and involved in the fusion of lytic granules with the plasmamembrane. Functional loss of syntaxin-11 in humans causes defective degranulation and impaired cytolytic activity of T and NK cells. Furthermore, patients with STX11 deficiency develop familial hemophagocytic lymphohistiocytosis type 4 (FHL4), a life-threatening disease of severe hyperinflammation. We established Stx11-deficient mice as an animal model for FHL4. Stx11-deficient mice exhibited severely reduced degranulation and cytolytic activity of CTL and NK cells and developed all clinical symptoms of hemophagocytic lymphohistiocytosis (HLH) after infection with lymphocytic choriomeningitis virus (LCMV). The HLH phenotype was further characterized by hyperactive CD8 T cells and continuous IFN-γ production. However, in contrast to perforin-deficient mice, which represent a model for FHL2, progression of HLH was not fatal. Survival of Stx11-deficient mice was determined by exhaustion of antigen-specific T cells, characterized by expression of inhibitory receptors, sequential loss of effector functions, and finally T-cell deletion. Blockade of inhibitory receptors on T cells in Stx11-deficient mice converted nonfatal disease course into fatal HLH, identifying T-cell exhaustion as an important factor for determination of disease severity in HLH.
- Published
- 2013
- Full Text
- View/download PDF
20. Distinct mutations in STXBP2 are associated with variable clinical presentations in patients with familial hemophagocytic lymphohistiocytosis type 5 (FHL5).
- Author
-
Pagel J, Beutel K, Lehmberg K, Koch F, Maul-Pavicic A, Rohlfs AK, Al-Jefri A, Beier R, Bomme Ousager L, Ehlert K, Gross-Wieltsch U, Jorch N, Kremens B, Pekrun A, Sparber-Sauer M, Mejstrikova E, Wawer A, Ehl S, zur Stadt U, and Janka G
- Subjects
- Adolescent, Adult, Basophil Degranulation Test, Child, Child, Preschool, Cohort Studies, DNA Mutational Analysis, Epistasis, Genetic, Female, Genetic Association Studies, Humans, Infant, Infant, Newborn, Lymphohistiocytosis, Hemophagocytic classification, Lymphohistiocytosis, Hemophagocytic ethnology, Male, Models, Biological, Munc18 Proteins physiology, Qa-SNARE Proteins genetics, Young Adult, Lymphohistiocytosis, Hemophagocytic genetics, Munc18 Proteins genetics, Mutation physiology
- Abstract
Familial hemophagocytic lymphohistiocytosis (FHL) is a genetically determined hyperinflammatory syndrome caused by uncontrolled immune response mediated by T-lymphocytes, natural killer (NK) cells, and macrophages. STXBP2 mutations have recently been associated with FHL5. To better characterize the genetic and clinical spectrum of FHL5, we analyzed a cohort of 185 patients with suspected FHL for mutations in STXBP2. We detected biallelic mutations in 37 patients from 28 families of various ethnic origins. Missense mutations and mutations affecting 1 of the exon 15 splice sites were the predominant changes detectable in this cohort. Patients with exon 15 splice-site mutations (n = 13) developed clinical manifestations significantly later than patients with other mutations (median age, 4.1 year vs 2 months) and showed less severe impairment of degranulation and cytotoxic function of NK cells and CTLs. Patients with FHL5 showed several atypical features, including sensorineural hearing deficit, abnormal bleeding, and, most frequently, severe diarrhea that was only present in early-onset disease. In conclusion, we report the largest cohort of patients with FHL5 so far, describe an extended disease spectrum, and demonstrate for the first time a clear genotype-phenotype correlation.
- Published
- 2012
- Full Text
- View/download PDF
21. A prospective evaluation of degranulation assays in the rapid diagnosis of familial hemophagocytic syndromes.
- Author
-
Bryceson YT, Pende D, Maul-Pavicic A, Gilmour KC, Ufheil H, Vraetz T, Chiang SC, Marcenaro S, Meazza R, Bondzio I, Walshe D, Janka G, Lehmberg K, Beutel K, zur Stadt U, Binder N, Arico M, Moretta L, Henter JI, and Ehl S
- Subjects
- Humans, Lymphohistiocytosis, Hemophagocytic immunology, Lymphohistiocytosis, Hemophagocytic metabolism, Lysosomal-Associated Membrane Protein 1, Prospective Studies, Sensitivity and Specificity, Time Factors, Cell Degranulation physiology, Immunologic Tests methods, Killer Cells, Natural physiology, Lymphohistiocytosis, Hemophagocytic diagnosis, T-Lymphocytes, Cytotoxic physiology
- Abstract
Familial hemophagocytic lymphohistiocytosis (FHL) is a life-threatening disorder of immune regulation caused by defects in lymphocyte cytotoxicity. Rapid differentiation of primary, genetic forms from secondary forms of hemophagocytic lymphohistiocytosis (HLH) is crucial for treatment decisions. We prospectively evaluated the performance of degranulation assays based on surface up-regulation of CD107a on natural killer (NK) cells and cytotoxic T lymphocytes in a cohort of 494 patients referred for evaluation for suspected HLH. Seventy-five of 77 patients (97%) with FHL3-5 and 11 of 13 patients (85%) with Griscelli syndrome type 2 or Chediak-Higashi syndrome had abnormal resting NK-cell degranulation. In contrast, NK-cell degranulation was normal in 14 of 16 patients (88%) with X-linked lymphoproliferative disease and in 8 of 14 patients (57%) with FHL2, who were identified by diminished intracellular SLAM-associated protein (SAP), X-linked inhibitor of apoptosis protein (XIAP), and perforin expression, respectively. Among 66 patients with a clinical diagnosis of secondary HLH, 13 of 59 (22%) had abnormal resting NK-cell degranulation, whereas 0 of 43 had abnormal degranulation using IL-2-activated NK cells. Active disease or immunosuppressive therapy did not impair the assay performance. Overall, resting NK-cell degranulation below 5% provided a 96% sensitivity for a genetic degranulation disorder and a specificity of 88%. Therefore, degranulation assays allow a rapid and reliable classification of patients, benefiting treatment decisions.
- Published
- 2012
- Full Text
- View/download PDF
22. Subtle differences in CTL cytotoxicity determine susceptibility to hemophagocytic lymphohistiocytosis in mice and humans with Chediak-Higashi syndrome.
- Author
-
Jessen B, Maul-Pavicic A, Ufheil H, Vraetz T, Enders A, Lehmberg K, Längler A, Gross-Wieltsch U, Bay A, Kaya Z, Bryceson YT, Koscielniak E, Badawy S, Davies G, Hufnagel M, Schmitt-Graeff A, Aichele P, Zur Stadt U, Schwarz K, and Ehl S
- Subjects
- Animals, Base Sequence, Cells, Cultured, Chediak-Higashi Syndrome genetics, Disease Models, Animal, Disease Susceptibility immunology, Genetic Predisposition to Disease, Humans, Individuality, Lymphocyte Activation genetics, Lymphocyte Activation immunology, Lymphohistiocytosis, Hemophagocytic genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Perforin genetics, T-Lymphocytes, Cytotoxic physiology, Vesicular Transport Proteins genetics, Chediak-Higashi Syndrome etiology, Chediak-Higashi Syndrome immunology, Lymphohistiocytosis, Hemophagocytic etiology, Lymphohistiocytosis, Hemophagocytic immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
Perforin-mediated cytotoxicity is important for controlling viral infections, but also for limiting immune reactions. Failure of this cytotoxic pathway leads to hemophagocytic lymphohistiocytosis (HLH), a life-threatening disorder of uncontrolled T-cell and macrophage activation. We studied susceptibility to HLH in 2 mouse strains (souris and beige(J)) and a cohort of patients with partial defects in perforin secretion resulting from different mutations in the LYST gene. Although both strains lacked NK-cell cytotoxicity, only souris mice developed all clinical and histopathologic signs of HLH after infection with lymphocytic choriomeningitis virus. The 2 strains showed subtle differences in CTL cytotoxicity in vitro that had a large impact on virus control in vivo. Whereas beige(J) CTLs eliminated lymphocytic choriomeningitis virus infection, souris CTLs failed to control the virus, which was associated with the development of HLH. In LYST-mutant patients with Chediak-Higashi syndrome, CTL cytotoxicity was reduced in patients with early-onset HLH, whereas it was retained in patients who later or never developed HLH. Thus, the risk of HLH development is set by a threshold that is determined by subtle differences in CTL cytotoxicity. Differences in the cytotoxic capacity of CTLs may be predictive for the risk of Chediak-Higashi syndrome patients to develop HLH.
- Published
- 2011
- Full Text
- View/download PDF
23. Platelet secretion defect in patients with familial hemophagocytic lymphohistiocytosis type 5 (FHL-5).
- Author
-
Sandrock K, Nakamura L, Vraetz T, Beutel K, Ehl S, and Zieger B
- Subjects
- Blood Platelets drug effects, Flow Cytometry, Hemostatics pharmacology, Humans, Lymphohistiocytosis, Hemophagocytic complications, Lymphohistiocytosis, Hemophagocytic genetics, Platelet Aggregation drug effects, Thrombin pharmacology, Blood Platelet Disorders etiology, Blood Platelets metabolism
- Published
- 2010
- Full Text
- View/download PDF
24. Clinical and immunologic consequences of a somatic reversion in a patient with X-linked severe combined immunodeficiency.
- Author
-
Speckmann C, Pannicke U, Wiech E, Schwarz K, Fisch P, Friedrich W, Niehues T, Gilmour K, Buiting K, Schlesier M, Eibel H, Rohr J, Superti-Furga A, Gross-Wieltsch U, and Ehl S
- Subjects
- B-Lymphocytes immunology, B-Lymphocytes pathology, Child, Preschool, Epithelial Cells immunology, Epithelial Cells pathology, Hematopoietic Stem Cell Transplantation, Humans, Interleukin Receptor Common gamma Subunit immunology, Killer Cells, Natural immunology, Killer Cells, Natural pathology, Lymphoid Progenitor Cells immunology, Lymphoid Progenitor Cells pathology, Lymphopenia immunology, Lymphopenia pathology, Lymphopenia therapy, Male, Receptors, Antigen, T-Cell, gamma-delta genetics, Receptors, Antigen, T-Cell, gamma-delta immunology, T-Lymphocytes immunology, T-Lymphocytes pathology, X-Linked Combined Immunodeficiency Diseases immunology, X-Linked Combined Immunodeficiency Diseases pathology, X-Linked Combined Immunodeficiency Diseases therapy, Antibody Formation genetics, Interleukin Receptor Common gamma Subunit genetics, Lymphopenia genetics, Point Mutation, X-Linked Combined Immunodeficiency Diseases genetics
- Abstract
X-linked severe combined immunodeficiency is a life-threatening disorder caused by mutations in the gene encoding the interleukin-2 receptor gamma chain (IL2RG). Hypomorphic mutations and reversion of mutations in subpopulations of cells can result in variant clinical phenotypes, making diagnosis and treatment difficult. We describe a 5-year-old boy with mild susceptibility to infection who was investigated for a mutation in IL2RG due to persistent natural killer (NK)- and T-cell lymphopenia. A functionally relevant novel T466C point mutation was found in B, NK, and epithelial cells, whereas alpha/beta and gamma/delta T cells showed the normal gene sequence, suggesting reversion of the mutation in a common T-cell precursor. This genetic correction in T cells resulted in a diverse T-cell repertoire and significant immunity despite failure to produce specific antibodies linked to an intrinsic defect of mutant B cells. These observations confirm the potential of revertant T-cell precursors to reconstitute immune function, but questions remain on the longevity of revertant cells implicating the need for careful follow up and early consideration of hematopoietic stem cell transplantation (HSCT).
- Published
- 2008
- Full Text
- View/download PDF
25. Recombinant Sendai virus induces T cell immunity against respiratory syncytial virus that is protective in the absence of antibodies.
- Author
-
Voges B, Vallbracht S, Zimmer G, Bossow S, Neubert WJ, Richter K, Hobeika E, Herrler G, and Ehl S
- Subjects
- Animals, Antibodies immunology, Mice, Mice, Inbred BALB C, Respiratory Syncytial Virus Infections pathology, Solubility, T-Lymphocytes drug effects, Viral Fusion Proteins genetics, Viral Fusion Proteins immunology, Genetic Engineering, Respiratory Syncytial Virus Infections immunology, Respiratory Syncytial Virus Infections prevention & control, Respiratory Syncytial Viruses immunology, Sendai virus genetics, Sendai virus immunology, T-Lymphocytes immunology
- Abstract
Respiratory syncytial virus (RSV) causes severe respiratory disease in infants and a vaccine is highly desirable. The fusion (F) protein of RSV is an important vaccine target, but the contribution of F-specific T cells to successful vaccination remains unclear. We studied the immune response to vaccination of mice with a recombinant Sendai virus expressing RSV F (rSeV F). rSeV F induced protective neutralizing antibody and RSV F-specific CTL responses. T cell immunity was stronger than that induced by recombinant vaccinia virus (rVV F), a well characterized reference vector. Vaccination of antibody-deficient mice showed that vaccine-induced RSV F-specific T cells were sufficient for protective immunity. rSeV F induced T cell immunity in the presence of neutralizing antibodies, which did not impair the vaccine response. Although the F protein only contains a subdominant CTL epitope, vaccination with rSeV F is sufficient to induce protective T cell immunity against RSV in mice.
- Published
- 2007
- Full Text
- View/download PDF
26. Lethal hemophagocytic lymphohistiocytosis in Hermansky-Pudlak syndrome type II.
- Author
-
Enders A, Zieger B, Schwarz K, Yoshimi A, Speckmann C, Knoepfle EM, Kontny U, Müller C, Nurden A, Rohr J, Henschen M, Pannicke U, Niemeyer C, Nurden P, and Ehl S
- Subjects
- Adaptor Protein Complex 3 genetics, Adaptor Protein Complex beta Subunits genetics, Bone Marrow pathology, Child, Preschool, Fatal Outcome, Flow Cytometry, Hermanski-Pudlak Syndrome genetics, Hermanski-Pudlak Syndrome therapy, Humans, Killer Cells, Natural immunology, Lymphohistiocytosis, Hemophagocytic genetics, Lymphohistiocytosis, Hemophagocytic pathology, Male, Mutation, Missense, Platelet Aggregation immunology, T-Lymphocytes, Cytotoxic immunology, Time Factors, Hermanski-Pudlak Syndrome complications, Hermanski-Pudlak Syndrome diagnosis, Lymphohistiocytosis, Hemophagocytic complications
- Abstract
Griscelli syndrome (GS) was diagnosed in a 2-year-old patient with oculocutaneous albinism and immunodeficiency, but sequencing of RAB27a revealed only a heterozygous mutation. Due to impaired natural killer (NK) and T-cell cytotoxicity implying a high risk of developing hemophagocytic lymphohistiocytosis (HLH), he was prepared for hematopoietic stem cell transplantation (HSCT). Unexpectedly, a severe bleeding episode occurred that led to the demonstration of disturbed platelet aggregation, reduced plateletdense granules, and impaired platelet degranulation. In combination with neutropenia, this suggested the diagnosis of Hermansky-Pudlak syndrome type II (HPSII) and a novel homozygous mutation in AP3B1 was detected. None of the 3 reported HPSII patients had developed HLH, and our patient seroconverted to Epstein-Barr virus (EBV) without clinical symptoms. HSCT was therefore withheld, and granulocyte-colony-stimulating factor (G-CSF) therapy was initiated and prevented further bacterial infections. At 3 years of age, however, the patient developed, without an obvious trigger, fulminant HLH that was resistant to therapy. This patient shows that careful clinical and molecular diagnosis is essential to differentiate the complex disorders of lysosomal trafficking. HPSII belongs to the group of familial hemophagocytic syndromes and may represent an indication for HSCT.
- Published
- 2006
- Full Text
- View/download PDF
27. Henoch-Schönlein purpura.
- Author
-
Mrusek S, Krüger M, Greiner P, Kleinschmidt M, Brandis M, and Ehl S
- Subjects
- Abdomen diagnostic imaging, Abdominal Pain diagnosis, Child, Diagnosis, Differential, Diarrhea diagnosis, Exanthema diagnosis, Humans, Inflammatory Bowel Diseases diagnosis, Male, Melena diagnosis, Tomography, X-Ray Computed, Ultrasonography, IgA Vasculitis diagnosis
- Published
- 2004
- Full Text
- View/download PDF
28. Modelling the dynamics of LCMV infection in mice: II. Compartmental structure and immunopathology.
- Author
-
Bocharov G, Klenerman P, and Ehl S
- Subjects
- Animals, Arenaviridae Infections prevention & control, Kidney immunology, Liver immunology, Mice, Mice, Inbred C57BL, Spleen immunology, Spleen virology, Thymus Gland immunology, Vaccination, Viremia, Virulence, Arenaviridae Infections immunology, Hepatitis, Viral, Animal immunology, Lymphocyte Activation, Lymphocytic choriomeningitis virus pathogenicity, Models, Immunological, T-Lymphocytes, Cytotoxic immunology
- Abstract
In this study, we develop a mathematical model for analysis of the compartmental aspects and immunopathology of lymphocytic choriomeningitis virus (LCMV) infection in mice. We used sets of original and published data on systemic (extrasplenic) virus distribution to estimate the parameters of virus growth and elimination for spleen and other anatomical compartments, such as the liver, kidney, thymus and lung as well as transfer rates between blood and the above organs. A mathematical model quantitatively integrating the virus distribution kinetics in the host, the specific cytotoxic T lymphocyte (CTL) response in spleen and the re-circulation of effector CTL between spleen, blood and liver is advanced to describe the CTL-mediated immunopathology (hepatitis) in mice infected with LCMV. For intravenous and "peripheral" routes of infection we examine the severity of the liver disease, as a function of the virus dose and the host's immune status characterized by the numbers of precursor and/or cytolytic effector CTL. The model is used to predict the efficacy of protection against virus persistence and disease in a localized viral infection as a function of the composition of CTL population. The modelling analysis suggests quantitative demands to CTL memory for maximal protection against a wide range of doses of infection with a primarily peripheral site of virus replication without the risk of favoring immunopathology. It specifies objectives for CTL vaccination to ensure virus elimination with minimal immunopathology vs. vaccination for disease., (Copyright 2003 Elsevier Science Ltd.)
- Published
- 2003
- Full Text
- View/download PDF
29. A cautionary note on experimental artefacts induced by fetal calf serum in a viral model of pulmonary eosinophilia.
- Author
-
Ostler T and Ehl S
- Subjects
- Animals, Artifacts, Cattle, Lung pathology, Mice, Mice, Inbred BALB C, T-Lymphocytes, Cytotoxic immunology, Th2 Cells immunology, Vaccination, Dendritic Cells immunology, Fetal Blood immunology, Pulmonary Eosinophilia immunology, Respiratory Syncytial Viruses immunology
- Abstract
In BALB/c mice, sensitization with the attachment protein (G) of respiratory syncytial virus (RSV) leads to CD4(+) T cell-mediated lung eosinophilia during subsequent challenge with RSV. In this study, we originally intended to test whether activation of RSV-specific cytotoxic T cells by peptide-pulsed dendritic cells (DC) after G protein sensitization could prevent this eosinophilic response. Peptide-pulsed dendritic cells activated CTL, which could mediate protective immunity to RSV. However, DC vaccination aggravated, rather than prevented, pulmonary eosinophilia in G-sensitized mice and also enhanced weight loss upon RSV infection. This was accompanied by preferential pulmonary recruitment of CD4(+) T cells secreting IL-5. The same enhanced Th2-mediated eosinophilic response could be observed in mice that received unloaded dendritic cells and this response occurred even in the absence of prior G sensitization. Since both dendritic cells and RSV were grown in fetal calf serum (FCS)-containing medium, we suspected that FCS had provoked this response. Indeed, neither eosinophilia nor enhanced pathology were observed in mice treated with DC raised in mouse serum. This observation calls for meticulous controls for artefacts induced by fetal calf serum particularly in mouse models of allergic responses of the respiratory tract.
- Published
- 2002
- Full Text
- View/download PDF
30. An improved protocol for measuring cytotoxic T cell activity in anatomic compartments with low cell numbers.
- Author
-
Ostler T, Schamel K, Hussell T, Openshaw P, Hausmann J, and Ehl S
- Subjects
- Animals, Borna Disease immunology, Borna Disease pathology, Brain cytology, Brain immunology, Bronchoalveolar Lavage Fluid cytology, Cell Line, Flow Cytometry, In Vitro Techniques, Killer Cells, Natural immunology, Lung cytology, Lung immunology, Lymphocyte Count, Lymphocytic Choriomeningitis immunology, Lymphocytic Choriomeningitis pathology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Organ Specificity, Respiratory Syncytial Virus Infections immunology, Respiratory Syncytial Virus Infections pathology, Cytotoxicity Tests, Immunologic methods, T-Lymphocytes, Cytotoxic immunology
- Abstract
The study of target cell lysis and cytokine production are valuable tools to characterize antigen-specific T and NK cell function during virus infections. After localized infections in compartments such as the lung or the brain, however, cell numbers isolated from these organs are too low to perform standard assays with individual mice. Here, we report a few simple modifications of the classical 51Cr release assay allowing reduction of the number of required effector cells by a factor of 10 without loosing sensitivity or specificity. Using not more than 4x10(5) effector cells, we were able to study ex vivo virus-specific CTL or NK activity from the lungs of individual mice after infection with respiratory syncytial virus (RSV) and from the brains of mice infected with Borna disease virus (BDV). Flow cytometric analysis of interferon-gamma production by virus-specific T cells including appropriate controls was achieved with as few as 10(5) effector cells.
- Published
- 2001
- Full Text
- View/download PDF
31. The impact of variation in the number of CD8(+) T-cell precursors on the outcome of virus infection.
- Author
-
Ehl S, Klenerman P, Zinkernagel RM, and Bocharov G
- Subjects
- Animals, Cell Count, Cell Line, Disease Models, Animal, Dogs, Lymphocytic Choriomeningitis prevention & control, Lymphocytic Choriomeningitis virology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Virus Latency, CD8-Positive T-Lymphocytes immunology, Hematopoietic Stem Cells immunology, Lymphocytic choriomeningitis virus immunology, T-Lymphocytes, Cytotoxic immunology
- Abstract
We investigated the role of varying the initial number of naive antiviral CTL precursors on the dynamics of LCMV-DOCILE infection. C57BL/6 mice, exhibiting LCMV-specific CTLp frequencies of about 50, are protected against virus persistence over a range of infectious doses up to 10(4) pfu. With 10-fold higher doses, a 100-fold increase in CTLp is required to restore virus control. With doses above 10(6) pfu, elevation of the initial CTLp number leads only to lethal immunopathology. Similarly, a 1000-fold increase in the number of initial naïve CTLp enhances the overall kinetics of virus elimination, but cannot limit early virus spread within the first 48 h after low-dose infection (500 pfu). Increases in initial naïve virus-specific CTLp numbers are of limited benefit in antiviral control. In addition to the number of virus-specific T cells, the time period needed to reach cytolytic effector function is a limiting parameter., (Copyright 1998 Academic Press.)
- Published
- 1998
- Full Text
- View/download PDF
32. A comparison of efficacy and specificity of three NK depleting antibodies.
- Author
-
Ehl S, Nuesch R, Tanaka T, Myasaka M, Hengartner H, and Zinkernagel R
- Subjects
- Animals, Antigens immunology, Antigens, Ly, Antigens, Surface, Cytotoxicity, Immunologic, G(M1) Ganglioside immunology, Immunity, Cellular, Lectins, C-Type, Lymphocytic choriomeningitis virus immunology, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, NK Cell Lectin-Like Receptor Subfamily B, Proteins immunology, Receptors, Interleukin-2 immunology, T-Lymphocytes, Cytotoxic immunology, Killer Cells, Natural immunology, Lymphocyte Depletion methods
- Abstract
This study compares in vivo efficacy and specificity of the three NK cell depleting antibodies anti-asialo GM1, anti-NK 1.1 and the recently described TM beta 1, which is directed against the interleukin-2 receptor beta chain. All three antibodies are equally efficacious as assessed by abolishing NK mediated cytolytic activity induced by a high dose virus infection or Poly IC against YAC-1 targets. Similarly, the generation of virus-specific cytotoxic T cells (CTL) was unimpaired after NK depletion in two different virus infections. However, if mice are treated with the antibodies several days after virus infection, when strong CTL responses have already been generated, anti-asialo GM1 and-to a lesser extent-also TM beta 1 have a significant effect on CTL activity. Only after treatment with anti-NK 1.1 antibody, CTL activity was not significantly impaired. We conclude, that of the NK depleting antibodies currently available, anti-NK 1.1 allows the best differentiation of activated CTL and NK cells in vivo.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.