Network


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

Hotspot


Dive into the research topics where Melanie Grusdat is active.

Publication


Featured researches published by Melanie Grusdat.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Natural killer cell activation enhances immune pathology and promotes chronic infection by limiting CD8+ T-cell immunity

Philipp A. Lang; Karl S. Lang; Haifeng C. Xu; Melanie Grusdat; Ian A. Parish; Mike Recher; Alisha R. Elford; Salim Dhanji; Namir Shaabani; Charles W. Tran; Dilan Dissanayake; Ramtin Rahbar; Magar Ghazarian; Anne Brüstle; Jason P. Fine; Peter W. Chen; Casey T. Weaver; Christoph S.N. Klose; Andreas Diefenbach; Dieter Häussinger; James R. Carlyle; Susan M. Kaech; Tak W. Mak; Pamela S. Ohashi

Infections with HIV, hepatitis B virus, and hepatitis C virus can turn into chronic infections, which currently affect more than 500 million patients worldwide. It is generally thought that virus-mediated T-cell exhaustion limits T-cell function, thus promoting chronic disease. Here we demonstrate that natural killer (NK) cells have a negative impact on the development of T-cell immunity by using the murine lymphocytic choriomeningitis virus. NK cell-deficient (Nfil3−/−, E4BP4−/−) mice exhibited a higher virus-specific T-cell response. In addition, NK cell depletion caused enhanced T-cell immunity in WT mice, which led to rapid virus control and prevented chronic infection in lymphocytic choriomeningitis virus clone 13- and reduced viral load in DOCILE-infected animals. Further experiments showed that NKG2D triggered regulatory NK cell functions, which were mediated by perforin, and limited T-cell responses. Therefore, we identified an important role of regulatory NK cells in limiting T-cell immunity during virus infection.


Nature Immunology | 2012

Enforced viral replication activates adaptive immunity and is essential for the control of a cytopathic virus.

Nadine Honke; Namir Shaabani; Giuseppe Cadeddu; Ursula R. Sorg; Dong-Er Zhang; Mirko Trilling; Karin Klingel; Martina Sauter; Reinhard Kandolf; Nicole Gailus; Nico van Rooijen; Christoph Burkart; Stephan Baldus; Melanie Grusdat; Max Löhning; Hartmut Hengel; Klaus Pfeffer; Masato Tanaka; Dieter Häussinger; Mike Recher; Philipp A. Lang; Karl S. Lang

The innate immune system limits viral replication via type I interferon and also induces the presentation of viral antigens to cells of the adaptive immune response. Using infection of mice with vesicular stomatitis virus, we analyzed how the innate immune system inhibits viral propagation but still allows the presentation of antigen to cells of the adaptive immune response. We found that expression of the gene encoding the inhibitory protein Usp18 in metallophilic macrophages led to lower type I interferon responsiveness, thereby allowing locally restricted replication of virus. This was essential for the induction of adaptive antiviral immune responses and, therefore, for preventing the fatal outcome of infection. In conclusion, we found that enforced viral replication in marginal zone macrophages was an immunological mechanism that ensured the production of sufficient antigen for effective activation of the adaptive immune response.


Proceedings of the National Academy of Sciences of the United States of America | 2013

The transcription factor Interferon Regulatory Factor 4 is required for the generation of protective effector CD8+ T cells

Friederike Raczkowski; Josephine Ritter; Kira Heesch; Valéa Schumacher; Anna Guralnik; Lena Höcker; Hartmann Raifer; Matthias Klein; Tobias Bopp; Hani Harb; Dörthe A. Kesper; Petra Ina Pfefferle; Melanie Grusdat; Philipp A. Lang; Hans-Willi Mittrücker; Magdalena Huber

Robust cytotoxic CD8+ T-cell response is important for immunity to intracellular pathogens. Here, we show that the transcription factor IFN Regulatory Factor 4 (IRF4) is crucial for the protective CD8+ T-cell response to the intracellular bacterium Listeria monocytogenes. IRF4-deficient (Irf4−/−) mice could not clear L. monocytogenes infection and generated decreased numbers of L. monocytogenes-specific CD8+ T cells with impaired effector phenotype and function. Transfer of wild-type CD8+ T cells into Irf4−/− mice improved bacterial clearance, suggesting an intrinsic defect of CD8+ T cells in Irf4−/− mice. Following transfer into wild-type recipients, Irf4−/− CD8+ T cells became activated and showed initial proliferation upon L. monocytogenes infection. However, these cells could not sustain proliferation, produced reduced amounts of IFN-γ and TNF-α, and failed to acquire cytotoxic function. Forced IRF4 expression in Irf4−/− CD8+ T cells rescued the defect. During acute infection, Irf4−/− CD8+ T cells demonstrated diminished expression of B lymphocyte-induced maturation protein-1 (Blimp-1), inhibitor of DNA binding (Id)2, and T-box expressed in T cells (T-bet), transcription factors programming effector-cell generation. IRF4 was essential for expression of Blimp-1, suggesting that altered regulation of Blimp-1 contributes to the defects of Irf4−/− CD8+ T cells. Despite increased levels of B-cell lymphoma 6 (BCL-6), Eomesodermin, and Id3, Irf4−/− CD8+ T cells showed impaired memory-cell formation, indicating additional functions for IRF4 in this process. As IRF4 governs B-cell and CD4+ T-cell differentiation, the identification of its decisive role in peripheral CD8+ T-cell differentiation, suggests a common regulatory function for IRF4 in adaptive lymphocytes fate decision.


Immunity | 2017

Glutathione Primes T Cell Metabolism for Inflammation

Tak W. Mak; Melanie Grusdat; Gordon S. Duncan; Catherine Dostert; Yannic Nonnenmacher; Maureen A. Cox; Carole Binsfeld; Zhenyue Hao; Anne Brüstle; Momoe Itsumi; Christian Jäger; Ying Chen; Olaf Pinkenburg; Bärbel Camara; Markus Ollert; Carsten Bindslev-Jensen; Vasilis Vasiliou; Chiara Gorrini; Philipp A. Lang; Michael Lohoff; Isaac S. Harris; Karsten Hiller; Dirk Brenner

&NA; Activated T cells produce reactive oxygen species (ROS), which trigger the antioxidative glutathione (GSH) response necessary to buffer rising ROS and prevent cellular damage. We report that GSH is essential for T cell effector functions through its regulation of metabolic activity. Conditional gene targeting of the catalytic subunit of glutamate cysteine ligase (Gclc) blocked GSH production specifically in murine T cells. Gclc‐deficient T cells initially underwent normal activation but could not meet their increased energy and biosynthetic requirements. GSH deficiency compromised the activation of mammalian target of rapamycin‐1 (mTOR) and expression of NFAT and Myc transcription factors, abrogating the energy utilization and Myc‐dependent metabolic reprogramming that allows activated T cells to switch to glycolysis and glutaminolysis. In vivo, T‐cell‐specific ablation of murine Gclc prevented autoimmune disease but blocked antiviral defense. The antioxidative GSH pathway thus plays an unexpected role in metabolic integration and reprogramming during inflammatory T cell responses. Graphical Abstract Figure. No caption available. HighlightsGlutathione (GSH) is not needed for early T cell activation but promotes T cell growthGSH supports mTOR and NFAT activity and drives glycolysis and glutaminolysisGclc‐derived GSH buffers ROS and regulates Myc‐dependent metabolic reprogrammingAblation of Gclc in T cells impairs inflammatory responses in vivo &NA; Upon activation, T cells adapt their metabolism to meet their increased bioenergetic and biosynthetic needs. Activated T cells produce ROS, which trigger the antioxidative GSH response to prevent cellular damage. Mak et al. report that the GSH pathway plays an unexpected role in metabolic integration during inflammatory T cell responses.


Cell Death & Differentiation | 2013

Reactive oxygen species delay control of lymphocytic choriomeningitis virus.

Philipp A. Lang; Haifeng C. Xu; Melanie Grusdat; David R. McIlwain; Aleksandra A. Pandyra; Isaac S. Harris; Namir Shaabani; Nadine Honke; S Kumar Maney; Elisabeth Lang; Vitaly I. Pozdeev; Mike Recher; B Odermatt; D Brenner; Dieter Häussinger; Pamela S. Ohashi; H Hengartner; R M Zinkernagel; Tak W. Mak; Karl S. Lang

Cluster of differentiation (CD)8+ T cells are like a double edged sword during chronic viral infections because they not only promote virus elimination but also induce virus-mediated immunopathology. Elevated levels of reactive oxygen species (ROS) have been reported during virus infections. However, the role of ROS in T-cell-mediated immunopathology remains unclear. Here we used the murine lymphocytic choriomeningitis virus to explore the role of ROS during the processes of virus elimination and induction of immunopathology. We found that virus infection led to elevated levels of ROS producing granulocytes and macrophages in virus-infected liver and spleen tissues that were triggered by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Lack of the regulatory subunit p47phox of the NADPH oxidase diminished ROS production in these cells. While CD8+ T cells exhibited ROS production that was independent of NADPH oxidase expression, survival and T-cell function was elevated in p47phox-deficient (Ncf1−/−) mice. In the absence of p47phox, enhanced T-cell immunity promoted virus elimination and blunted corresponding immunopathology. In conclusion, we find that NADPH-mediated production of ROS critically impairs the immune response, impacting elimination of virus and outcome of liver cell damage.


PLOS ONE | 2014

Recombinant p35 from Bacteria Can Form Interleukin (IL-)12, but Not IL-35

Samadhi Aparicio-Siegmund; Jens M. Moll; Juliane Lokau; Melanie Grusdat; Jutta Schröder; Svenja Plöhn; Stefan Rose-John; Joachim Grötzinger; Philipp A. Lang; Jürgen Scheller; Christoph Garbers

The Interleukin (IL)-12 family contains several heterodimeric composite cytokines which share subunits among each other. IL-12 consists of the subunits p40 (shared with IL-23) and p35. p35 is shared with the composite cytokine IL-35 which comprises of the p35/EBI3 heterodimer (EBI3 shared with IL-27). IL-35 signals via homo- or heterodimers of IL-12Rβ2, gp130 and WSX-1, which are shared with IL-12 and IL-27 receptor complexes, respectively. p35 was efficiently secreted in complex with p40 as IL-12 but not with EBI3 as IL-35 in several transfected cell lines tested which complicates the analysis of IL-35 signal transduction. p35 and p40 but not p35 and EBI3 form an inter-chain disulfide bridge. Mutation of the responsible cysteine residue (p40C197A) reduced IL-12 formation and activity only slightly. Importantly, the p40C197A mutation prevented the formation of antagonistic p40 homodimers which enabled the in vitro reconstitution of biologically active IL-12 with p35 produced in bacteria (p35bac). Reconstitution of IL-35 with p35bac and EBI3 did, however, fail to induce signal transduction in Ba/F3 cells expressing IL-12Rβ2 and gp130. In summary, we describe the in vitro reconstitution of IL-12, but fail to produce recombinant IL-35 by this novel approach.


Journal of Virology | 2015

Deficiency of the B Cell-Activating Factor Receptor Results in Limited CD169+ Macrophage Function during Viral Infection

Haifeng C. Xu; Jun Huang; Vishal Khairnar; Vikas Duhan; Aleksandra A. Pandyra; Melanie Grusdat; David R. McIlwain; Sathish Kumar Maney; Jennifer L. Gommerman; Max Löhning; Pamela S. Ohashi; Tak W. Mak; Kathrin Pieper; Heiko Sic; Matthaios Speletas; Hermann Eibel; Carl F. Ware; Alexei V. Tumanov; Andrey A. Kruglov; Sergei A. Nedospasov; Dieter Häussinger; Mike Recher; Karl S. Lang; Philipp A. Lang

ABSTRACT The B cell-activating factor (BAFF) is critical for B cell development and humoral immunity in mice and humans. While the role of BAFF in B cells has been widely described, its role in innate immunity remains unknown. Using BAFF receptor (BAFFR)-deficient mice, we characterized BAFFR-related innate and adaptive immune functions following infection with vesicular stomatitis virus (VSV) and lymphocytic choriomeningitis virus (LCMV). We identified a critical role for BAFFR signaling in the generation and maintenance of the CD169+ macrophage compartment. Consequently, Baffr − / − mice exhibited limited induction of innate type I interferon production after viral infection. Lack of BAFFR signaling reduced virus amplification and presentation following viral infection, resulting in highly reduced antiviral adaptive immune responses. As a consequence, BAFFR-deficient mice showed exacerbated and fatal disease after viral infection. Mechanistically, transient lack of B cells in Baffr − / − animals resulted in limited lymphotoxin expression, which is critical for maintenance of CD169+ cells. In conclusion, BAFFR signaling affects both innate and adaptive immune activation during viral infections. IMPORTANCE Viruses cause acute and chronic infections in humans resulting in millions of deaths every year. Innate immunity is critical for the outcome of a viral infection. Innate type I interferon production can limit viral replication, while adaptive immune priming by innate immune cells induces pathogen-specific immunity with long-term protection. Here, we show that BAFFR deficiency not only perturbed B cells, but also resulted in limited CD169+ macrophages. These macrophages are critical in amplifying viral particles to trigger type I interferon production and initiate adaptive immune priming. Consequently, BAFFR deficiency resulted in reduced enforced viral replication, limited type I interferon production, and reduced adaptive immunity compared to BAFFR-competent controls. As a result, BAFFR-deficient mice were predisposed to fatal viral infections. Thus, BAFFR expression is critical for innate immune activation and antiviral immunity.


European Journal of Immunology | 2015

T‐cell STAT3 is required for the maintenance of humoral immunity to LCMV

David R. McIlwain; Melanie Grusdat; Vitaly I. Pozdeev; Haifeng C. Xu; Colin Reardon; Zhenyue Hao; Marc Beyer; Andreas Bergthaler; Dieter Häussinger; Garry P. Nolan; Karl S. Lang; Philipp A. Lang

STAT3 is a critical transcription factor activated downstream of cytokine signaling and is integral for the function of multiple immune cell types. Human mutations in STAT3 cause primary immunodeficiency resulting in impaired control of a variety of infections, including reactivation of latent viruses. In this study, we investigate how T‐cell functions of STAT3 contribute to responses to viral infection by inducing chronic lymphocytic choriomeningitis virus (LCMV) infection in mice lacking STAT3 specifically in T cells. Although mice with conditional disruption of STAT3 in T cells were able to mount early responses to viral infection similar to control animals, including expansion of effector T cells, we found generation of T‐follicular helper (Tfh) cells to be impaired. As a result, STAT3 T cell deficient mice produced attenuated germinal center reactions, and did not accumulate bone marrow virus specific IgG‐secreting cells, resulting in failure to maintain levels of virus‐specific IgG or mount neutralizing responses to LCMV in the serum. These effects were associated with reduced control of viral replication and prolonged infection. Our results demonstrate the importance of STAT3 in T cells for the generation of functional long‐term humoral immunity to viral infections.


Trends in cancer | 2018

B-Cell Metabolic Remodeling and Cancer

Davide G. Franchina; Melanie Grusdat; Dirk Brenner

Cells of the immune system display varying metabolic profiles to fulfill their functions. B lymphocytes overcome fluctuating energy challenges as they transition from the resting state and recirculation to activation, rapid proliferation, and massive antibody production. Only through a controlled interplay between metabolism, extracellular stimuli, and intracellular signaling can successful humoral responses be mounted. Alterations to this balance can promote malignant transformation of B cells. The metabolic control of B-cell fate is only partially understood. Here, we provide a compelling overview of the current state of the art and describe the main metabolic features of B cells during normal development and oncogenesis, with emphasis on the major B-cell transcriptional and metabolic regulators, including myelocytomatosis virus oncogene cellular homolog (Myc) and hypoxia-inducible factor 1-α (HIF-1α).


Physiological Reviews | 2019

The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond

Catherine Dostert; Melanie Grusdat; Elisabeth Letellier; Dirk Brenner

The tumor necrosis factor (TNF) and TNF receptor (TNFR) superfamilies (TNFSF/TNFRSF) include 19 ligands and 29 receptors that play important roles in the modulation of cellular functions. The communication pathways mediated by TNFSF/TNFRSF are essential for numerous developmental, homeostatic, and stimulus-responsive processes in vivo. TNFSF/TNFRSF members regulate cellular differentiation, survival, and programmed death, but their most critical functions pertain to the immune system. Both innate and adaptive immune cells are controlled by TNFSF/TNFRSF members in a manner that is crucial for the coordination of various mechanisms driving either co-stimulation or co-inhibition of the immune response. Dysregulation of these same signaling pathways has been implicated in inflammatory and autoimmune diseases, highlighting the importance of their tight regulation. Investigation of the control of TNFSF/TNFRSF activities has led to the development of therapeutics with the potential to reduce chronic inflammation or promote anti-tumor immunity. The study of TNFSF/TNFRSF proteins has exploded over the last 30 yr, but there remains a need to better understand the fundamental mechanisms underlying the molecular pathways they mediate to design more effective anti-inflammatory and anti-cancer therapies.

Collaboration


Dive into the Melanie Grusdat's collaboration.

Top Co-Authors

Avatar

Philipp A. Lang

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karl S. Lang

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Haifeng C. Xu

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Tak W. Mak

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pamela S. Ohashi

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dirk Brenner

University of Southern Denmark

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge