Esther von Stebut
University of Mainz
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Esther von Stebut.
Journal of Experimental Medicine | 2006
Florian Woelbing; Susanna Lopez Kostka; Katharina Moelle; Yasmine Belkaid; Cord Sunderkoetter; Sjef Verbeek; Ari Waisman; Axel P. Nigg; Juergen Knop; Mark C. Udey; Esther von Stebut
Uptake of Leishmania major by dendritic cells (DCs) results in activation and interleukin (IL)-12 release. Infected DCs efficiently stimulate CD4− and CD8− T cells and vaccinate against leishmaniasis. In contrast, complement receptor 3–dependent phagocytosis of L. major by macrophages (MΦ) leads exclusively to MHC class II–restricted antigen presentation to primed, but not naive, T cells, and no IL-12 production. Herein, we demonstrate that uptake of L. major by DCs required parasite-reactive immunoglobulin (Ig)G and involved FcγRI and FcγRIII. In vivo, DC infiltration of L. major–infected skin lesions coincided with the appearance of antibodies in sera. Skin of infected B cell–deficient mice and Fcγ−/− mice contained fewer parasite-infected DCs in vivo. Infected B cell–deficient mice as well as Fcγ−/− mice (all on the C57BL/6 background) showed similarly increased disease susceptibility as assessed by lesion volumes and parasite burdens. The B cell–deficient mice displayed impaired T cell priming and dramatically reduced IFN-γ production, and these deficits were normalized by infection with IgG-opsonized parasites. These data demonstrate that DC and MΦ use different receptors to recognize and ingest L. major with different outcomes, and indicate that B cell–derived, parasite-reactive IgG and DC FcγRI and FcγRIII are essential for optimal development of protective immunity.
Journal of Immunology | 2009
Susanna Lopez Kostka; Stephanie Dinges; Klaus Griewank; Yoichiro Iwakura; Mark C. Udey; Esther von Stebut
Resistance to leishmaniasis in C57BL/6 mice depends on Th1/Tc1 cells. BALB/c mice preferentially develop Th2 immunity and succumb to infection. We now assessed the role of IL-17 in cutaneous leishmaniasis. During the course of Leishmania major infection, BALB/c CD4 cells and neutrophils produced increased amounts of IL-17 as compared with cells from C57BL/6 mice. This increase was associated with significantly increased IL-23 release from L. major-infected BALB/c dendritic cells (DC), whereas IL-6 and TGF-β1 production by BALB/c and C57BL/6 DC were comparable. Interestingly, lesion sizes in infected IL-17-deficient BALB/c mice were dramatically smaller and failed to progress as compared with those in control mice. Similar amounts of IL-4, IL-10, and IFN-γ were produced by T cells from IL-17-deficient mice and control mice consistent with development of Th2-predominant immunity in all animals. Improved disease outcome was associated with decreased CXCL2-accumulation in lesion sites and decreased neutrophil immigration into lesions of infected IL-17-deficient mice confirming prior observations that enhanced neutrophil recruitment contributes to disease susceptibility in BALB/c mice. This study excludes an important facilitating role for IL-17 in Th1/Th2 development in L. major-infected BALB/c mice, and suggests that IL-23 production by L. major-infected DC maintains IL-17+ cells that influence disease progression via regulation of neutrophil recruitment.
Journal of Experimental Medicine | 2011
Kordula Kautz-Neu; Madelon Noordegraaf; Stephanie Dinges; Clare L. Bennett; Dominik John; Bjoern E. Clausen; Esther von Stebut
Langerhans cells suppress the immune response to low-dose Leishmania major infection in part by inducing regulatory T cells.
Journal of Experimental Medicine | 2003
Esther von Stebut; Jan Ehrchen; Yasmine Belkaid; Susanna Lopez Kostka; Katharina Mölle; Jürgen Knop; Cord Sunderkötter; Mark C. Udey
Protective immunity against pathogens such as Leishmania major is mediated by interleukin (IL)-12–dependent Th1-immunity. We have shown previously that skin-dendritic cells (DCs) from both resistant C57BL/6 and susceptible BALB/c mice release IL-12 when infected with L. major, and infected BALB/c DCs effectively vaccinate against leishmaniasis. To determine if cytokines other than IL-12 might influence disease outcome, we surveyed DCs from both strains for production of a variety of cytokines. Skin-DCs produced significantly less IL-1α in response to lipopolysaccharide/interferon γ or L. major when expanded from BALB/c as compared with C57BL/6 mice. In addition, IL-1α mRNA accumulation in lymph nodes of L. major–infected BALB/c mice was ∼3-fold lower than that in C57BL/6 mice. Local injections of IL-1α during the first 3 d after infection led to dramatic, persistent reductions in lesion sizes. In L. major–infected BALB/c mice, IL-1α administration resulted in increased Th1- and strikingly decreased Th2-cytokine production. IL-1α and IL-12 treatments were similarly effective, and IL-1α efficacy was strictly IL-12 dependent. These data indicate that transient local administration of IL-1α acts in conjunction with IL-12 to influence Th-development in cutaneous leishmaniasis and prevents disease progression in susceptible BALB/c mice, perhaps by enhancing DC-induced Th1-education. Differential production of IL-1 by C57BL/6 and BALB/c mice may provide a partial explanation for the disparate outcomes of infection in these mouse strains.
The FASEB Journal | 2006
Marcus Maurer; Susanna Lopez Kostka; Frank Siebenhaar; Katharina Moelle; Martin Metz; Jürgen Knop; Esther von Stebut
Mast cells (MCs) initiate protective im‐ munity against bacteria. Here we demonstrate that MCs also contribute to the control of parasitic skin infections by Leishmania major. L. major‐infected MC‐defi‐cient KitW/KitW‐v mice developed markedly larger skin lesions than did normal Kit+/+ mice (>2‐fold), and cutaneous reconstitution with MCs resulted in normalization of lesion development. KitW/KitW‐v lesions contained significantly more parasites, and infections resulted in enhanced spreading of parasites to the spleens as compared to controls. In addition, recruitment of proinflammatory neutrophils, macrophages, and dendritic cells (DCs) in infected mice was MC dependent. In the absence of MCs, reduced numbers of lesional DCs were associated with decreased production of Th1‐promoting interleukin (IL)‐12. Antigen‐specific T cell priming was delayed in KitW/KitW‐v mice and cytokine responses were skewed towards Th2. Notably, local skin MC reconstitution at sites of infection was sufficient for the induction of systemic protection. Thus, MC‐mediated control of L. major infections is not limited to the induction of local inflammation. Instead, MCs contribute significantly to local DC recruitment, which mediates protective immunity. These findings extend the view of MCs as salient sentinels of innate immunity to complex host defense reactions against intracellular pathogens.—Maurer, M., Lopez Kostka, S., Siebenhaar, F., Moelle, K., Metz, M., Knop, J., von Stebut, E. Skin mast cells control T cell‐dependent host defense in Leishmania major infections. FASEB J. 20, 2460–2467 (2006)
European Journal of Immunology | 2011
Anne Dudeck; Cathleen A. Suender; Susanna Lopez Kostka; Esther von Stebut; Marcus Maurer
Mast cells (MCs) play an important role in the regulation of protective adaptive immune responses against pathogens. However, it is still unclear whether MCs promote such host defense responses via direct effects on T cells or rather by modifying the functions of antigen‐presenting cells. To identify the underlying mechanisms of the immunoregulatory capacity of MCs, we investigated the impact of MCs on dendritic cell (DC) maturation and function. We found that murine peritoneal MCs underwent direct crosstalk with immature DCs that induced DC maturation as evidenced by enhanced expression of costimulatory molecules. Furthermore, the MC/DC interaction resulted in the release of the T‐cell modulating cytokines IFN‐γ, IL‐2, IL‐6 and TGF‐β into coculture supernatants and increased the IL‐12p70, IFN‐γ, IL‐6 and TGF‐β secretion of LPS‐matured DCs. Such MC‐“primed” DCs subsequently induced efficient CD4+ T‐cell proliferation. Surprisingly, we observed that MC‐primed DCs stimulated CD4+ T cells to release high levels of IFN‐γ and IL‐17, demonstrating that MCs promote Th1 and Th17 responses. Confirming our in vitro findings, we found that the enhanced disease progression of MC‐deficient mice in Leishmania major infection is correlated with impaired induction of both Th1 and Th17 cells.
Journal of Investigative Dermatology | 2013
Khalifa El Malki; Susanne Karbach; Jula Huppert; Morad Zayoud; Sonja Reißig; Rebecca Schüler; Alexej Nikolaev; Khalad Karram; Thomas Münzel; Christoph R.W. Kuhlmann; Heiko J. Luhmann; Esther von Stebut; Simone Wörtge; Florian C. Kurschus; Ari Waisman
Topical application of imiquimod (IMQ) on the skin of mice induces inflammation with common features found in psoriatic skin. Recently, it was postulated that IL-17 has an important role both in psoriasis and in the IMQ model. To further investigate the impact of IL-17RA signaling in psoriasis, we generated IL-17 receptor A (IL-17RA)-deficient mice (IL-17RA(del)) and challenged these mice with IMQ. Interestingly, the disease was only partially reduced and delayed but not abolished when compared with controls. In the absence of IL-17RA, we found persisting signs of inflammation such as neutrophil and macrophage infiltration within the skin. Surprisingly, already in the naive state, the skin of IL-17RA(del) mice contained significantly elevated numbers of Th17- and IL-17-producing γδ T cells, assuming that IL-17RA signaling regulates the population size of Th17 and γδ T cells. Upon IMQ treatment of IL-17RA(del) mice, these cells secreted elevated amounts of tumor necrosis factor-α, IL-6, and IL-22, accompanied by increased levels of the chemokine CXCL2, suggesting an alternative pathway of neutrophil and macrophage skin infiltration. Hence, our findings have major implications in the potential long-term treatment of psoriasis by IL-17-targeting drugs.
Journal Der Deutschen Dermatologischen Gesellschaft | 2011
Gerhard Boecken; Cord Sunderkötter; Christian Bogdan; Thomas Weitzel; Marcellus Fischer; Andreas Müller; Micha Löbermann; Gerlind Anders; Esther von Stebut; Mirjam Schunk; Gerd D. Burchard; Martin P. Grobusch; Ralf Bialek; Gundel Harms-Zwingenberger; Bernhard Fleischer; Mathias Pietras; Michael Faulde; Kay Erkens
The incidence of cutaneous and mucocutaneous Leishmaniasis (CL/MCL) is increasing globally, also in Germany, although the cases are imported and still low in number. The current evidence for the different therapies has many limitations due to lack of sufficient studies on the different Leishmania species with differing virulence. So far there is no international gold standard for the optimal management. The aim of the German joint working group on Leishmaniasis, formed by the societies of Tropical Medicine (DTG), Chemotherapy (PEG) and Dermatology (DDG), was to establish a guideline for the diagnosis and treatment of CL and MCL in Germany, based on evidence (Medline search yielded 400 articles) and, where lacking, on consensus of the experts. As the clinical features do not necessarily reflect the involved Leishmania species and, as different parasite species and even geographically distinct strains of the same species may require different treatments or varying dosages or durations of therapy, the guidelines suggest for Germany to identify the underlying parasite prior to treatment. Because of relevant differences in prognosis and ensuing therapy species should be identified in i) New World CL/MCL (NWCL/ MCL) to distinguish between L. mexicana-complex and subgenus Viannia, ii) in suspected infections with L. mexicana-complex to distinguish from L. amazonensis, and iii) in Old World CL (OWCL) to distinguish between L. infantum and L. major, L. tropica, or L. aethiopica. A state-of-the-art diagnostic algorithm is presented. For recommendations on localized and systemic drug treatment and physical procedures, data from the accessible literature were adjusted according to the involved parasite species and a clinical differentiation into uncomplicated or complex lesions. Systemic therapy was strictly recommended for i) complex lesions (e. g. > 3 infected lesions, infections in functionally or cosmetically critical areas such as face or hands, presence of lymphangitis), ii) lesions refractory to therapy, iii) NWCL by the subgenus Viannia or by L. amazonensis, iv) in MCL and v) in recalcitrant, or disseminating or diffuse cutaneous courses. In e. g. infection with L. major it encompasses miltefosine, fluconazole and ketoconazole, while antimony or allopurinol were here considered second choice. Local therapy was considered appropriate for i) uncomplicated lesions of OWCL, ii) L. mexicana-complex and iii) pregnant women. In e. g. infection with L. major it encompasses perilesional antimony, combined with cryotherapy, paromomycin 15 %/in methylbenzethoniumchlorid 12 % and thermotherapy. The group also stated that there is an urgent need for improving the design and the way of publishing of clinical trials in leishmaniasis.
Arteriosclerosis, Thrombosis, and Vascular Biology | 2014
Susanne Karbach; Andrew L. Croxford; Matthias Oelze; Rebecca Schüler; Daniel Minwegen; Joanna Wegner; Lija Koukes; Nir Yogev; Alexei Nikolaev; Sonja Reißig; Alexander Ullmann; Maike Knorr; Maximilian J. Waldner; Markus F. Neurath; Huige Li; Zhixiong Wu; Christoph Brochhausen; Jürgen Scheller; Stefan Rose-John; Carolin Piotrowski; Ingo Bechmann; Markus P. Radsak; Philipp S. Wild; Andreas Daiber; Esther von Stebut; Philip Wenzel; Ari Waisman; Thomas Münzel
Objective— Interleukin (IL)-17A is regarded as an important cytokine to drive psoriasis, an inflammatory skin disease marked by increased cardiovascular mortality. We aimed to test the hypothesis that overproduction of IL-17A in the skin leading to dermal inflammation may systemically cause vascular dysfunction in psoriasis-like skin disease. Approach and Results— Conditional overexpression of IL-17A in keratinocytes caused severe psoriasis-like skin inflammation in mice (K14-IL-17Aind/+ mice), associated with increased reactive oxygen species formation and circulating CD11b+ inflammatory leukocytes in blood, with endothelial dysfunction, increased systolic blood pressure, left ventricular hypertrophy, and reduced survival compared with controls. In K14-IL-17Aind/+ mice, immunohistochemistry and flow cytometry revealed increased vascular production of the nitric oxide/superoxide reaction product peroxynitrite and infiltration of the vasculature with myeloperoxidase+CD11b+GR1+F4/80− cells accompanied by increased expression of the inducible nitric oxide synthase and the nicotinamide dinucleotide phosphate (NADPH) oxidase, nox2. Neutrophil depletion by anti-GR-1 antibody injections reduced oxidative stress in blood and vessels. Neutralization of tumor necrosis factor-&agr; and IL-6 (both downstream of IL-17A) reduced skin lesions, attenuated oxidative stress in heart and blood, and partially improved endothelial dysfunction in K14-IL-17Aind/+ mice. Conclusions— Dermal overexpression of IL-17A induces systemic endothelial dysfunction, vascular oxidative stress, arterial hypertension, and increases mortality mainly driven by myeloperoxidase+CD11b+GR1+F4/80− inflammatory cells. Depletion of the GR-1+ immune cells or neutralization of IL-17A downstream cytokines by biologicals attenuates the vascular phenotype in K14-IL-17Aind/+ mice.
European Journal of Immunology | 2005
Sabine Zahn; Stefan Wirtz; Mark Birkenbach; Richard S. Blumberg; Markus F. Neurath; Esther von Stebut
Protection against Leishmania major is dependent on IL‐12 release from L. major‐infected dendritic cells (DC) that induce IFN‐γ‐producing Th1/Tc1 cells. IL‐27, a novel member of the IL‐12 family, is a heterodimer composed of p28 and IL‐12p40‐related Epstein‐Barr virus‐induced gene 3 (EBI3), and was shown to be produced by DC. In this study, we utilized EBI3‐deficient mice to investigate the role of IL‐27 in leishmaniasis using physiological low‐dose infections that mimic natural transmissions. Lesions in EBI3–/– mice were significantly larger between weeks 3 and 10 post infection, reaching up to approximately threefold increased lesion volumes compared to wild types. In parallel, dermal lesions of EBI3–/– mice contained greater parasite numbers, reaching a peak load that was 2‐log higher than in C57BL/6 mice. However, lesions in EBI3–/– and wild‐type mice resolved after 12 weeks. At early time points, the antigen‐specific cytokine response in EBI3–/– lymph nodes showed increased levels of IL‐4, IL‐10 and IL‐13 and decreased IFN‐γ production. IL‐27 production was restricted to the DC population, since the majority of EBI3 expression in lymph nodes of infected mice was found in CD11c+ cells. In conclusion, our data show that DC‐derived IL‐27 is critical for the timely initiation of efficient anti‐parasite Th1 immunity early in infections.