Network


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

Hotspot


Dive into the research topics where Sebastian Fuchs is active.

Publication


Featured researches published by Sebastian Fuchs.


Proceedings of the National Academy of Sciences of the United States of America | 2011

ORAI1-mediated calcium influx is required for human cytotoxic lymphocyte degranulation and target cell lysis

Andrea Maul-Pavicic; Samuel C. C. Chiang; Anne Rensing-Ehl; Birthe Jessen; Cyril Fauriat; Stephanie M. Wood; Sebastian Sjöqvist; Markus Hufnagel; Ilka Schulze; Thilo Bass; Wolfgang W. A. Schamel; Sebastian Fuchs; Hanspeter Pircher; Christie-Ann McCarl; Katsuhiko Mikoshiba; Klaus Schwarz; Stefan Feske; Yenan T. Bryceson; Stephan Ehl

Lymphocytes mediate cytotoxicity by polarized release of the contents of cytotoxic granules toward their target cells. Here, we have studied the role of the calcium release-activated calcium channel ORAI1 in human lymphocyte cytotoxicity. Natural killer (NK) cells obtained from an ORAI1-deficient patient displayed defective store-operated Ca2+ entry (SOCE) and severely defective cytotoxic granule exocytosis leading to impaired target cell lysis. Similar findings were obtained using NK cells from a stromal interaction molecule 1-deficient patient. The defect occurred at a late stage of the signaling process, because activation of leukocyte functional antigen (LFA)-1 and cytotoxic granule polarization were not impaired. Moreover, pharmacological inhibition of SOCE interfered with degranulation and target cell lysis by freshly isolated NK cells and CD8+ effector T cells from healthy donors. In addition to effects on lymphocyte cytotoxicity, synthesis of the chemokine macrophage inflammatory protein-1β and the cytokines TNF-α and IFN-γ on target cell recognition was impaired in ORAI1-deficient NK cells, as previously described for T cells. By contrast, NK cell cytokine production induced by combinations of IL-12, IL-15, and IL-18 was not impaired by ORAI1 deficiency. Taken together, these results identify a critical role for ORAI1-mediated Ca2+ influx in granule exocytosis for lymphocyte cytotoxicity as well as for cytokine production induced by target cell recognition.


Journal of Immunology | 2012

Antiviral and Regulatory T Cell Immunity in a Patient with Stromal Interaction Molecule 1 Deficiency

Sebastian Fuchs; Anne Rensing-Ehl; Carsten Speckmann; Bertram Bengsch; Annette Schmitt-Graeff; Ilka Bondzio; Andrea Maul-Pavicic; Thilo Bass; Thomas Vraetz; Brigitte Strahm; Tobias Ankermann; Melina Benson; Almuth Caliebe; Regina Fölster-Holst; Petra Kaiser; Robert Thimme; Wolfgang W. A. Schamel; Klaus Schwarz; Stefan Feske; Stephan Ehl

Stromal interaction molecule 1 (STIM1) deficiency is a rare genetic disorder of store-operated calcium entry, associated with a complex syndrome including immunodeficiency and immune dysregulation. The link from the molecular defect to these clinical manifestations is incompletely understood. We report two patients with a homozygous R429C point mutation in STIM1 completely abolishing store-operated calcium entry in T cells. Immunological analysis of one patient revealed that despite the expected defect of T cell proliferation and cytokine production in vitro, significant antiviral T cell populations were generated in vivo. These T cells proliferated in response to viral Ags and showed normal antiviral cytotoxicity. However, antiviral immunity was insufficient to prevent chronic CMV and EBV infections with a possible contribution of impaired NK cell function and a lack of NKT cells. Furthermore, autoimmune cytopenia, eczema, and intermittent diarrhea suggested impaired immune regulation. FOXP3-positive regulatory T (Treg) cells were present but showed an abnormal phenotype. The suppressive function of STIM1-deficient Treg cells in vitro, however, was normal. Given these partial defects in cytotoxic and Treg cell function, impairment of other immune cell populations probably contributes more to the pathogenesis of immunodeficiency and autoimmunity in STIM1 deficiency than previously appreciated.


The New England Journal of Medicine | 2013

Deficiency of Innate and Acquired Immunity Caused by an IKBKB Mutation

Ulrich Pannicke; Bernd Baumann; Sebastian Fuchs; Philipp Henneke; Anne Rensing-Ehl; Marta Rizzi; Ales Janda; Katrin Hese; Michael Schlesier; Karlheinz Holzmann; Stephan Borte; Constanze Laux; Eva-Maria Rump; Alan Rosenberg; Teresa Zelinski; Hubert Schrezenmeier; Thomas Wirth; Stephan Ehl; Marlis L. Schroeder; Klaus Schwarz; Abstr Act

BACKGROUND Severe combined immunodeficiency (SCID) comprises a heterogeneous group of heritable deficiencies of humoral and cell-mediated immunity. Many patients with SCID have lymphocyte-activation defects that remain uncharacterized. METHODS We performed genetic studies in four patients, from four families of Northern Cree ancestry, who had clinical characteristics of SCID, including early onset of severe viral, bacterial, and fungal infections despite normal B-cell and T-cell counts. Genomewide homozygosity mapping was used to identify a candidate region, which was found on chromosome 8; all genes within this interval were sequenced. Immune-cell populations, signal transduction on activation, and effector functions were studied. RESULTS The patients had hypogammaglobulinemia or agammaglobulinemia, and their peripheral-blood B cells and T cells were almost exclusively of naive phenotype. Regulatory T cells and γδ T cells were absent. All patients carried a homozygous duplication--c.1292dupG in exon 13 of IKBKB, which encodes IκB kinase 2 (IKK2, also known as IKKβ)--leading to loss of expression of IKK2, a component of the IKK-nuclear factor κB (NF-κB) pathway. Immune cells from the patients had impaired responses to stimulation through T-cell receptors, B-cell receptors, toll-like receptors, inflammatory cytokine receptors, and mitogens. CONCLUSIONS A form of human SCID is characterized by normal lymphocyte development despite a loss of IKK2 function. IKK2 deficiency results in an impaired response to activation stimuli in a variety of immune cells, leading to clinically relevant impairment of adaptive and innate immunity. Although Ikk2 deficiency is lethal in mouse embryos, our observations suggest a more restricted, unique role of IKK2-NF-κB signaling in humans. (Funded by the German Federal Ministry of Education and Research and others.).


Clinical Immunology | 2010

Clinical and immunological overlap between autoimmune lymphoproliferative syndrome and common variable immunodeficiency

Anne Rensing-Ehl; Klaus Warnatz; Sebastian Fuchs; Michael Schlesier; Ulrich Salzer; R. Draeger; Ilka Bondzio; Y. Joos; Ales Janda; M. Gomes; Mario Abinun; Sophie Hambleton; Andrew J. Cant; F. Shackley; T Flood; Catherine Waruiru; K. Beutel; Kathrin Siepermann; Gregor Dueckers; Tim Niehues; Thomas Wiesel; Volker Schuster; Markus G. Seidel; Milen Minkov; K. Sirkiä; M.V. Kopp; M. Korhonen; Klaus Schwarz; Stephan Ehl; Carsten Speckmann

Autoimmune lymphoproliferative syndrome (ALPS) is mainly caused by defects in the CD95 pathway. Raised CD3+TCRαβ+CD4-CD8- double negative T cells and impaired T cell apoptosis are hallmarks of the disease. In contrast, the B cell compartment has been less well studied. We found an altered distribution of B cell subsets with raised transitional B cells and reduced marginal zone B cells, switched memory B cells and plasma blasts in most of 22 analyzed ALPS patients. Moreover, 5 out of 66 ALPS patients presented with low IgG and susceptibility to infection revealing a significant overlap between ALPS and common variable immunodeficiency (CVID). In patients presenting with lymphoproliferation, cytopenia, hypogammaglobulinemia and impaired B cell differentiation, serum biomarkers were helpful in addition to apoptosis tests for the identification of ALPS patients. Our observations may indicate a role for apoptosis defects in some diseases currently classified as CVID.


Blood | 2016

Mutations in AP3D1 associated with immunodeficiency and seizures define a new type of Hermansky-Pudlak syndrome

Sandra Ammann; Ansgar Schulz; I Krageloh-Mann; Nele M.G. Dieckmann; K Niethammer; Sebastian Fuchs; Katja-Martina Eckl; R Plank; R Werner; Janine Altmüller; Holger Thiele; Peter Nürnberg; J Bank; A Strauss; H. von Bernuth; U zur Stadt; Samantha Grieve; Gillian M. Griffiths; Kai Lehmberg; Hans Christian Hennies; Stephan Ehl

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.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Missense mutation in immunodeficient patients shows the multifunctional roles of coiled-coil domain 3 (CC3) in STIM1 activation

Mate Maus; Amit Jairaman; Peter B. Stathopulos; Martin Muik; Marc Fahrner; Carl Weidinger; Melina Benson; Sebastian Fuchs; Stephan Ehl; Christoph Romanin; Mitsuhiko Ikura; Murali Prakriya; Stefan Feske

Significance Stromal interaction molecule (STIM) 1 is an essential activator of the ORAI1 calcium (Ca2+) channel that mediates Ca2+ entry into many cell types. Patients with mutations in STIM1 or ORAI1 genes suffer from a severe immunodeficiency syndrome. Here, we studied a disease-causing mutant of STIM1 that is incapable of activating ORAI1 channels, thereby uncovering key molecular mechanisms of STIM1 function. The autosomal recessive R429C mutation interferes with the structural integrity of a protein–protein interaction domain in STIM1 and impairs the ability of STIM1 to multimerize and bind to ORAI1. Drugs targeting this domain may provide a means to selectively modulate STIM1–ORAI1 interaction and Ca2+ entry as a novel approach to treat autoimmune diseases and other disorders associated with abnormal ORAI1-mediated Ca2+ entry. Store-operated Ca2+ entry (SOCE) is a universal Ca2+ influx pathway that is important for the function of many cell types. SOCE occurs upon depletion of endoplasmic reticulum (ER) Ca2+ stores and relies on a complex molecular interplay between the plasma membrane (PM) Ca2+ channel ORAI1 and the ER Ca2+ sensor stromal interaction molecule (STIM) 1. Patients with null mutations in ORAI1 or STIM1 genes present with severe combined immunodeficiency (SCID)-like disease. Here, we describe the molecular mechanisms by which a loss-of-function STIM1 mutation (R429C) in human patients abolishes SOCE. R429 is located in the third coiled-coil (CC3) domain of the cytoplasmic C terminus of STIM1. Mutation of R429 destabilizes the CC3 structure and alters the conformation of the STIM1 C terminus, thereby releasing a polybasic domain that promotes STIM1 recruitment to ER–PM junctions. However, the mutation also impairs cytoplasmic STIM1 oligomerization and abolishes STIM1–ORAI1 interactions. Thus, despite its constitutive localization at ER–PM junctions, mutant STIM1 fails to activate SOCE. Our results demonstrate multifunctional roles of the CC3 domain in regulating intra- and intermolecular STIM1 interactions that control (i) transition of STIM1 from a quiescent to an active conformational state, (ii) cytoplasmic STIM1 oligomerization, and (iii) STIM1–ORAI1 binding required for ORAI1 activation.


Blood | 2015

Early-onset Evans syndrome, immunodeficiency and premature immunosenescence associated with tripeptidyl-peptidase II deficiency

Polina Stepensky; Anne Rensing-Ehl; Ruth Gather; Shoshana Revel-Vilk; Ute Fischer; Schafiq Nabhani; Fabian Beier; Tim H. Brümmendorf; Sebastian Fuchs; Simon Zenke; Elke Firat; Vered Molho Pessach; Arndt Borkhardt; Mirzokhid Rakhmanov; Bärbel Keller; Klaus Warnatz; Hermann Eibel; Gabriele Niedermann; Orly Elpeleg; Stephan Ehl

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.


Clinical Immunology | 2015

SYK expression endows human ZAP70-deficient CD8 T cells with residual TCR signaling.

Fabian Hauck; Britta Blumenthal; Sebastian Fuchs; Christelle Lenoir; Emmanuel Martin; Carsten Speckmann; Thomas Vraetz; Wilma Mannhardt-Laakmann; Nathalie Lambert; Marine Gil; Stephan Borte; Marie Audrain; Klaus Schwarz; Annick Lim; Wolfgang W. A. Schamel; Alain Fischer; Stephan Ehl; Anne Rensing-Ehl; Capucine Picard; Sylvain Latour

Autosomal recessive human ZAP70 deficiency is a rare cause of combined immunodeficiency (CID) characterized by defective CD4 T cells and profound CD8 T cell lymphopenia. Herein, we report two novel patients that extend the molecular genetics, the clinical and functional phenotypes associated with the ZAP70 deficiency. The patients presented as infant-onset CID with severe infections caused by varicella zoster virus and live vaccines. Retrospective TCR excision circle newborn screening was normal in both patients. One patient carried a novel non-sense mutation (p.A495fsX75); the other a previously described misense mutation (p.A507V). In contrast to CD4 T cells, the majority of the few CD8 T cells showed expression of the ZAP70-related tyrosine kinase SYK that correlated with residual TCR signaling including calcium flux and degranulation. Our findings highlight the differential requirements of ZAP70 and SYK during thymic development, peripheral homeostasis as well as effector functions of CD4 and CD8 T cells.


The Journal of Allergy and Clinical Immunology | 2017

Hematopoietic stem cell transplantation in patients with gain-of-function signal transducer and activator of transcription 1 mutations

Jennifer W. Leiding; Satoshi Okada; David Hagin; Mario Abinun; Anna Shcherbina; D.N. Balashov; Vy Hong-Diep Kim; Adi Ovadia; Stephen L. Guthery; Michael A. Pulsipher; Desa Lilic; Lisa Devlin; Sharon Christie; M Depner; Sebastian Fuchs; Annet van Royen-Kerkhof; Caroline A. Lindemans; Aleksandra Petrovic; Kathleen E. Sullivan; Nancy Bunin; Sara Sebnem Kilic; Fikret Arpaci; Oscar de la Calle-Martin; Laura Martinez-Martinez; Juan Carlos Aldave; Masao Kobayashi; Teppei Ohkawa; Kohsuke Imai; Akihiro Iguchi; Chaim M. Roifman

Background: Gain‐of‐function (GOF) mutations in signal transducer and activator of transcription 1 (STAT1) cause susceptibility to a range of infections, autoimmunity, immune dysregulation, and combined immunodeficiency. Disease manifestations can be mild or severe and life‐threatening. Hematopoietic stem cell transplantation (HSCT) has been used in some patients with more severe symptoms to treat and cure the disorder. However, the outcome of HSCT for this disorder is not well established. Objective: We sought to aggregate the worldwide experience of HSCT in patients with GOF‐STAT1 mutations and to assess outcomes, including donor engraftment, overall survival, graft‐versus‐host disease, and transplant‐related complications. Methods: Data were collected from an international cohort of 15 patients with GOF‐STAT1 mutations who had undergone HSCT using a variety of conditioning regimens and donor sources. Retrospective data collection allowed the outcome of transplantation to be assessed. In vitro functional testing was performed to confirm that each of the identified STAT1 variants was in fact a GOF mutation. Results: Primary donor engraftment in this cohort of 15 patients with GOF‐STAT1 mutations was 74%, and overall survival was only 40%. Secondary graft failure was common (50%), and posttransplantation event‐free survival was poor (10% by 100 days). A subset of patients had hemophagocytic lymphohistiocytosis before transplant, contributing to their poor outcomes. Conclusion: Our data indicate that HSCT for patients with GOF‐STAT1 mutations is curative but has significant risk of secondary graft failure and death.


Blood | 2015

Omenn syndrome associated with a functional reversion due to a somatic second-site mutation in CARD11 deficiency

Sebastian Fuchs; Anne Rensing-Ehl; Ulrich Pannicke; Myriam Ricarda Lorenz; Paul Fisch; Yogesh S. Jeelall; Jan Rohr; Carsten Speckmann; Thomas Vraetz; Susan Farmand; Annette Schmitt-Graeff; Marcus Krüger; Brigitte Strahm; Philipp Henneke; Anselm Enders; Keisuke Horikawa; Christopher C. Goodnow; Klaus Schwarz; Stephan Ehl

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.

Collaboration


Dive into the Sebastian Fuchs's collaboration.

Top Co-Authors

Avatar

Stephan Ehl

University of Freiburg

View shared research outputs
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

Tim Niehues

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge