Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ilka Fuchs is active.

Publication


Featured researches published by Ilka Fuchs.


The Journal of Allergy and Clinical Immunology | 2013

Deficiency of caspase recruitment domain family, member 11 (CARD11), causes profound combined immunodeficiency in human subjects

Polina Stepensky; Baerbel Keller; Mary Buchta; Anne-Kathrin Kienzler; Orly Elpeleg; Raz Somech; Sivan Cohen; Idit Shachar; Lisa A. Miosge; Michael Schlesier; Ilka Fuchs; Anselm Enders; Hermann Eibel; Bodo Grimbacher; Klaus Warnatz

BACKGROUND Profound combined immunodeficiency can present with normal numbers of T and B cells, and therefore the functional defect of the cellular and humoral immune response is often not recognized until the first severe clinical manifestation. Here we report a patient of consanguineous descent presenting at 13 months of age with hypogammaglobulinemia, Pneumocystis jirovecii pneumonia, and a suggestive family history. OBJECTIVE We sought to identify the genetic alteration in a patient with combined immunodeficiency and characterize human caspase recruitment domain family, member 11 (CARD11), deficiency. METHODS Molecular, immunologic, and functional assays were performed. RESULTS The immunologic characterization revealed only subtle changes in the T-cell and natural killer cell compartment, whereas B-cell differentiation, although normal in number, was distinctively blocked at the transitional stage. Genetic evaluation revealed a homozygous deletion of exon 21 in CARD11 as the underlying defect. This deletion abrogated protein expression and activation of the canonical nuclear factor κB (NF-κB) pathway in lymphocytes after antigen receptor or phorbol 12-myristate 13-acetate stimulation, whereas CD40 signaling in B cells was preserved. The abrogated activation of the canonical NF-κB pathway was associated with severely impaired upregulation of inducible T-cell costimulator, OX40, cytokine production, proliferation of T cells, and B cell-activating factor receptor expression on B cells. CONCLUSION Thus in patients with CARD11 deficiency, the combination of impaired activation and especially upregulation of inducible T-cell costimulator on T cells, together with severely disturbed peripheral B-cell differentiation, apparently leads to a defective T-cell/B-cell cooperation and probably germinal center formation and clinically results in severe immunodeficiency. This report discloses the crucial and nonredundant role of canonical NF-κB activation and specifically CARD11 in the antigen-specific immune response in human subjects.


Clinical Immunology | 2013

X-linked inhibitor of apoptosis (XIAP) deficiency: the spectrum of presenting manifestations beyond hemophagocytic lymphohistiocytosis.

Carsten Speckmann; Kai Lehmberg; Michael H. Albert; R.B. Damgaard; M. Fritsch; Mads Gyrd-Hansen; Anne Rensing-Ehl; Thomas Vraetz; Bodo Grimbacher; Ulrich Salzer; Ilka Fuchs; Heike Ufheil; Bernd H. Belohradsky; A. Hassan; C.M. Cale; M. Elawad; Brigitte Strahm; S. Schibli; M. Lauten; M. Kohl; J.J. Meerpohl; B. Rodeck; Reinhard Kolb; W. Eberl; J. Soerensen; H. von Bernuth; Myriam Ricarda Lorenz; Klaus Schwarz; U zur Stadt; Stephan Ehl

X-linked inhibitor of apoptosis (XIAP) deficiency caused by mutations in BIRC4 was initially described in patients with X-linked lymphoproliferative syndrome (XLP) who had no mutations in SH2D1A. In the initial reports, EBV-associated hemophagocytic lymphohistiocytosis (HLH) was the predominant clinical phenotype. Among 25 symptomatic patients diagnosed with XIAP deficiency, we identified 17 patients who initially presented with manifestations other than HLH. These included Crohn-like bowel disease (n=6), severe infectious mononucleosis (n=4), isolated splenomegaly (n=3), uveitis (n=1), periodic fever (n=1), fistulating skin abscesses (n=1) and severe Giardia enteritis (n=1). Subsequent manifestations included celiac-like disease, antibody deficiency, splenomegaly and partial HLH. Screening by flow cytometry identified 14 of 17 patients in our cohort. However, neither genotype nor protein expression nor results from cell death studies were clearly associated with the clinical phenotype. Only mutation analysis can reliably identify affected patients. XIAP deficiency must be considered in a wide range of clinical presentations.


Blood | 2016

Hyperactive mTOR pathway promotes lymphoproliferation and abnormal differentiation in autoimmune lymphoproliferative syndrome.

Simon Völkl; Anne Rensing-Ehl; Andrea Allgäuer; Elisabeth Schreiner; Myriam Ricarda Lorenz; Jan Rohr; Christian Klemann; Ilka Fuchs; Volker Schuster; André O. von Bueren; Nora Naumann-Bartsch; Eleonora Gambineri; Kathrin Siepermann; Robin Kobbe; Michaela Nathrath; Peter D. Arkwright; Maurizio Miano; Klaus-Daniel Stachel; Markus Metzler; Klaus Schwarz; Anita Kremer; Carsten Speckmann; Stephan Ehl; Andreas Mackensen

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.


Haematologica | 2013

Sequential decisions on FAS sequencing guided by biomarkers in patients with lymphoproliferation and autoimmune cytopenia

Anne Rensing-Ehl; Ales Janda; Myriam Ricarda Lorenz; Beryl Primrose Gladstone; Ilka Fuchs; Mario Abinun; Michael H. Albert; Karina Butler; Andrew J. Cant; Annamaria Cseh; Martin Ebinger; Sigune Goldacker; Sophie Hambleton; Holger Hebart; Leonora Houet; Karim Kentouche; Ingrid Kühnle; Kai Lehmberg; Ester Mejstrikova; Charlotte M. Niemeyer; Milen Minkov; Olaf Neth; Gregor Dückers; Stephen Owens; Joachim Rösler; Freimut H. Schilling; Volker Schuster; Markus G. Seidel; Petr Smisek; Martina Sukova

Clinical and genetic heterogeneity renders confirmation or exclusion of autoimmune lymphoproliferative syndrome difficult. To re-evaluate and improve the currently suggested diagnostic approach to patients with suspected FAS mutation, the most frequent cause of autoimmune lymphoproliferative syndrome, we prospectively determined 11 biomarkers in 163 patients with splenomegaly or lymphadenopathy and presumed or proven autoimmune cytopenia(s). Among 98 patients sequenced for FAS mutations in CD3+TCRα/β+CD4−CD8− “double negative” T cells, 32 had germline and six had somatic FAS mutations. The best a priori predictor of FAS mutations was the combination of vitamin B12 and soluble FAS ligand (cut-offs 1255 pg/mL and 559 pg/mL, respectively), which had a positive predictive value of 92% and a negative predictive value of 97%. We used these data to develop a web-based probability calculator for FAS mutations using the three most discriminatory biomarkers (vitamin B12, soluble FAS ligand, interleukin-10) of the 11 tested. Since more than 60% of patients with lymphoproliferation and autoimmune cytopenia(s) in our cohort did not harbor FAS mutations, 15% had somatic FAS mutations, and the predictive value of double-negative T-cell values was rather low (positive and negative predictive values of 61% and 77%, respectively), we argue that the previously suggested diagnostic algorithm based on determination of double-negative T cells and germline FAS sequencing, followed by biomarker analysis, is not efficient. We propose vitamin B12 and soluble FAS ligand assessment as the initial diagnostic step with subsequent decision on FAS sequencing supported by a probability-calculating tool.


European Journal of Immunology | 2017

Primary and secondary hemophagocytic lymphohistiocytosis have different patterns of T-cell activation, differentiation and repertoire

Sandra Ammann; Kai Lehmberg; Udo zur Stadt; Gritta Janka; Anne Rensing-Ehl; Christian Klemann; Maximilian Heeg; Sebastian F. N. Bode; Ilka Fuchs; Stephan Ehl

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.


Haematologica | 2017

Evolution of disease activity and biomarkers on and off rapamycin in 28 patients with autoimmune lymphoproliferative syndrome

Christian Klemann; Myrian Esquivel; Aude Magerus-Chatinet; Myriam Ricarda Lorenz; Ilka Fuchs; Nathalie Neveux; Martin Castelle; Jan Rohr; Claudia Bettoni da Cunha; Martin Ebinger; Robin Kobbe; Bernhard Kremens; Florian Kollert; Eleonora Gambineri; Kai Lehmberg; Markus G. Seidel; Kathrin Siepermann; Thomas Voelker; Volker Schuster; Sigune Goldacker; Klaus Schwarz; Carsten Speckmann; Capucine Picard; Alain Fischer; Frédéric Rieux-Laucat; Stephan Ehl; Anne Rensing-Ehl; Bénédicte Neven

Chronic benign lymphoproliferation and autoimmune cytopenias are the main features requiring treatment in FAS mutant patients with autoimmune lymphoproliferative syndrome (ALPS).[1][1],[2][2] Successful use of the mTOR inhibitor rapamycin was initially reported in the treatment of refractory


Journal of Clinical Immunology | 2017

Effective Immunological Guidance of Genetic Analyses Including Exome Sequencing in Patients Evaluated for Hemophagocytic Lymphohistiocytosis

Sandra Ammann; Kai Lehmberg; Udo zur Stadt; Christian Klemann; Sebastian F. N. Bode; Carsten Speckmann; Gritta Janka; Katharina Wustrau; Mirzokhid Rakhmanov; Ilka Fuchs; Hans Christian Hennies; Stephan Ehl

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.


Blood | 2015

Gray platelet syndrome can mimic autoimmune lymphoproliferative syndrome

Anne Rensing-Ehl; Ulrich Pannicke; Stefanie-Yvonne Zimmermann; Myriam Ricarda Lorenz; Bénédicte Neven; Ilka Fuchs; Ulrich Salzer; Carsten Speckmann; Anne Strauss; Eberhard Maaβ; Benedicte Collet; Anselm Enders; Rémi Favier; Marie Christine Alessi; Frédéric Rieux-Laucat; Barbara Zieger; Klaus Schwarz; Stephan Ehl

To the editor: The combination of splenomegaly and autoimmune cytopenia is frequently associated with primary immunodeficiencies. The most frequent disease is autoimmune lymphoproliferative syndrome (ALPS), caused by autosomal dominant germline or somatic mutations of the FAS gene.[1][1],[2][2] The


European Journal of Immunology | 2014

Patients with T+/low NK+ IL-2 receptor γ chain deficiency have differentially-impaired cytokine signaling resulting in severe combined immunodeficiency

Sebastian Fuchs; Anne Rensing-Ehl; Miriam Erlacher; Thomas Vraetz; Lara Hartjes; Ales Janda; Marta Rizzi; Myriam Ricarda Lorenz; Kimberly Gilmour; Geneviève de Saint-Basile; Chaim M. Roifman; Steven Cheuk; Andrew R. Gennery; Adrian J. Thrasher; Ilka Fuchs; Klaus Schwarz; Carsten Speckmann; Stephan Ehl

X‐linked severe combined immunodeficiency (X‐SCID) leads to a T−NK−B+ immunophenotype and is caused by mutations in the gene encoding the IL‐2 receptor γ‐chain (IL2RG). IL2RGR222C leads to atypical SCID with a severe early onset phenotype despite largely normal NK‐ and T‐cell numbers. To address this discrepancy, we performed a detailed analysis of T, B, and NK cells, including quantitative STAT phosphorylation and functional responses to the cytokines IL‐2, IL‐4, IL‐15, and IL‐21 in a patient with the IL2RGR222C mutation. Moreover, we identified nine additional unpublished patients with the same mutations, all with a full SCID phenotype, and confirmed selected immunological observations. T‐cell development was variably affected, but led to borderline T‐cell receptor excision circle (TREC) levels and a normal repertoire. T cells showed moderately reduced proliferation, failing enhancement by IL‐2. While NK‐cell development was normal, IL‐2 enhancement of NK‐cell degranulation and IL‐15‐induced cytokine production were absent. IL‐2 or IL‐21 failed to enhance B‐cell proliferation and plasmablast differentiation. These functional alterations were reflected by a differential impact of IL2RGR222C on cytokine signal transduction, with a gradient IL‐4


Journal of Clinical Immunology | 2015

Novel Patient with Late-Onset Familial Hemophagocytic Lymphohistiocytosis with STXBP2 Mutations Presenting with Autoimmune Hepatitis, Neurological Manifestations and Infections Associated with Hypogammaglobulinemia

Hossein Esmaeilzadeh; Mohammad Hasan Bemanian; Mohammad Nabavi; Saba Arshi; Morteza Fallahpour; Ilka Fuchs; Udo zur Stadt; Klaus Warnatz; Sandra Ammann; Stephan Ehl; Kai Lehmberg; Nima Rezaei

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.

Collaboration


Dive into the Ilka Fuchs's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge