Network


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

Hotspot


Dive into the research topics where Konrad Aden is active.

Publication


Featured researches published by Konrad Aden.


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

RNAi screening identifies mediators of NOD2 signaling: Implications for spatial specificity of MDP recognition

Simone Lipinski; Nils Grabe; Gunnar Jacobs; Susanne Billmann-Born; Andreas Till; Robert Häsler; Konrad Aden; Maren Paulsen; Alexander Arlt; Lars Kraemer; Nina Hagemann; Kai S. Erdmann; Stefan Schreiber; Philip Rosenstiel

The intracellular nucleotide-binding oligomerization domain-2 (NOD2) receptor detects bacteria-derived muramyl dipeptide (MDP) and activates the transcription factor NF-κB. Here we describe the regulatome of NOD2 signaling using a systematic RNAi screen. Using three consecutive screens, we identified a set of 20 positive NF-κB regulators including the known pathway members RIPK2, RELA, and BIRC4 (XIAP) as well as FRMPD2 (FERM and PDZ domain-containing 2). FRMPD2 interacts with NOD2 via leucine-rich repeats and forms a complex with the membrane-associated protein ERBB2IP. We demonstrate that FRMPD2 spatially assembles the NOD2-signaling complex, hereby restricting NOD2-mediated immune responses to the basolateral compartment of polarized intestinal epithelial cells. We show that genetic truncation of the NOD2 leucine-rich repeat domain, which is associated with Crohn disease, impairs the interaction with FRMPD2, and that intestinal inflammation leads to down-regulation of FRMPD2. These results suggest a structural mechanism for how polarity of epithelial cells acts on intestinal NOD-like receptor signaling to mediate spatial specificity of bacterial recognition and control of immune responses.


Gastroenterology | 2010

Reg IV Regulates Normal Intestinal and Colorectal Cancer Cell Susceptibility to Radiation-Induced Apoptosis

Kumar S. Bishnupuri; Qizhi Luo; Satheesh K. Sainathan; Kento Kikuchi; Sripathi M. Sureban; Mekala Sabarinathan; Jennifer Gross; Konrad Aden; Randal May; Courtney W. Houchen; Shrikant Anant; Brian K. Dieckgraefe

BACKGROUND & AIMS Regenerating (Reg) gene IV is predominantly expressed in gastrointestinal cells and highly up-regulated in many gastrointestinal malignancies, including colorectal cancer (CRC). Human CRC cells expressing higher levels of Reg IV gene and its protein product (Reg IV) are resistant to conventional therapies, including irradiation (IR). However, the underlying mechanism is not well defined. METHODS A murine model of IR-induced intestinal injury and in vitro and in vivo models of human CRC were used to determine the role of Reg IV in regulation of normal intestinal and colorectal cancer cell susceptibility to IR-induced apoptosis. RESULTS Treatments of recombinant human Reg IV (rhR4) protein protected normal intestinal crypt cells from IR-induced apoptosis by increasing the expression of antiapoptotic genes Bcl-2, Bcl-XL, and survivin. However, overexpression of Reg IV in human CRC cells was associated with increased resistance to IR-induced apoptosis. Therefore, we used antagonism of Reg IV as a tool to increase CRC cell susceptibility to IR-induced cell death. Two complementary approaches using specific monoclonal antibodies and small interfering RNAs were tested in both in vitro and in vivo models of human CRC. Both approaches resulted in increased apoptosis and decreased cell proliferation, leading to decreased tumor growth and increased animal survival. Furthermore, these approaches increased CRC cell susceptibility to IR-induced apoptosis. CONCLUSIONS These results implicate Reg IV as an important modulator of gastrointestinal cell susceptibility to IR; hence, it is a potential target for adjunctive treatments for human CRC and other gastrointestinal malignancies.


Journal of Biological Chemistry | 2014

Modulation of Nuclear Factor E2-related Factor-2 (Nrf2) Activation by the Stress Response Gene Immediate Early Response-3 (IER3) in Colonic Epithelial Cells A NOVEL MECHANISM OF CELLULAR ADAPTION TO INFLAMMATORY STRESS

Imke Stachel; Claudia Geismann; Konrad Aden; Florian Deisinger; Philip Rosenstiel; Stefan Schreiber; Susanne Sebens; Alexander Arlt

Background: Nrf2 has a dual role in tumorigenesis. Results: Nrf2 activation in colonic epithelial cells is controlled by the stress response gene IER3. Loss of IER3 expression causes enhanced Nrf2 activity, thereby conferring ROS protection and apoptosis resistance. Conclusion: By regulating Nrf2-dependent cytoprotection, IER3 exerts tumor suppressive activity. Significance: Loss of control by IER3 favors the protumorigenic action of Nrf2. Although nuclear factor E2-related factor-2 (Nrf2) protects from carcinogen-induced tumorigenesis, underlying the rationale for using Nrf2 inducers in chemoprevention, this antioxidative transcription factor may also act as a proto-oncogene. Thus, an enhanced Nrf2 activity promotes formation and chemoresistance of colon cancer. One mechanism causing persistent Nrf2 activation is the adaptation of epithelial cells to oxidative stress during chronic inflammation, e.g. colonocytes in inflammatory bowel diseases, and the multifunctional stress response gene immediate early response-3 (IER3) has a crucial role under these conditions. We now demonstrate that colonic tissue from Ier3−/− mice subject of dextran sodium sulfate colitis exhibit greater Nrf2 activity than Ier3+/+ mice, manifesting as increased nuclear Nrf2 protein level and Nrf2 target gene expression. Likewise, human NCM460 colonocytes subjected to shRNA-mediated IER3 knockdown exhibit greater Nrf2 activity compared with control cells, whereas IER3 overexpression attenuated Nrf2 activation. IER3-deficient NCM460 cells exhibited reduced reactive oxygen species levels, indicating increased antioxidative protection, as well as lower sensitivity to TRAIL or anticancer drug-induced apoptosis and greater clonogenicity. Knockdown of Nrf2 expression reversed these IER3-dependent effects. Further, the enhancing effect of IER3 deficiency on Nrf2 activity relates to the control of the inhibitory tyrosine kinase Fyn by the PI3K/Akt pathway. Thus, the PI3K inhibitor LY294002 or knockdown of Akt or Fyn expression abrogated the impact of IER3 deficiency on Nrf2 activity. In conclusion, the interference of IER3 with the PI3K/Akt-Fyn pathway represents a novel mechanism of Nrf2 regulation that may get lost in tumors and by which IER3 exerts its stress-adaptive and tumor-suppressive activity.


Human Molecular Genetics | 2017

Absence of RNase H2 triggers generation of immunogenic micronuclei removed by autophagy

Kareen Bartsch; Katharina Knittler; Christopher Borowski; Sönke Rudnik; Markus Damme; Konrad Aden; Martina E. Spehlmann; Norbert Frey; Paul Saftig; Athena Chalaris; Björn Rabe

&NA; Hypomorphic mutations in the DNA repair enzyme RNase H2 cause the neuroinflammatory autoimmune disorder Aicardi‐Goutières syndrome (AGS). Endogenous nucleic acids are believed to accumulate in patient cells and instigate pathogenic type I interferon expression. However, the underlying nucleic acid species amassing in the absence of RNase H2 has not been established yet. Here, we report that murine RNase H2 knockout cells accumulated cytosolic DNA aggregates virtually indistinguishable from micronuclei. RNase H2‐dependent micronuclei were surrounded by nuclear lamina and most of them contained damaged DNA. Importantly, they induced expression of interferon‐stimulated genes (ISGs) and co‐localized with the nucleic acid sensor cGAS. Moreover, micronuclei associated with RNase H2 deficiency were cleared by autophagy. Consequently, induction of autophagy by pharmacological mTOR inhibition resulted in a significant reduction of cytosolic DNA and the accompanied interferon signature. Autophagy induction might therefore represent a viable therapeutic option for RNase H2‐dependent disease. Endogenous retroelements have previously been proposed as a source of self‐nucleic acids triggering inappropriate activation of the immune system in AGS. We used human RNase H2‐knockout cells generated by CRISPR/Cas9 to investigate the impact of RNase H2 on retroelement propagation. Surprisingly, replication of LINE‐1 and Alu elements was blunted in cells lacking RNase H2, establishing RNase H2 as essential host factor for the mobilisation of endogenous retrotransposons.


Gut | 2013

Extracellular cathepsin K exerts antimicrobial activity and is protective against chronic intestinal inflammation in mice

Christian Sina; Simone Lipinski; Olga Gavrilova; Konrad Aden; Ateequr Rehman; Andreas Till; Andrea Rittger; Rainer Podschun; Ulf Meyer-Hoffert; Robert Haesler; Emilie Midtling; Katrin Pütsep; Michael A. McGuckin; Stefan Schreiber; Paul Saftig; Philip Rosenstiel

Objective Cathepsin K is a lysosomal cysteine protease that has pleiotropic roles in bone resorption, arthritis, atherosclerosis, blood pressure regulation, obesity and cancer. Recently, it was demonstrated that cathepsin K-deficient (Ctsk−/− ) mice are less susceptible to experimental autoimmune arthritis and encephalomyelitis, which implies a functional role for cathepsin K in chronic inflammatory responses. Here, the authors address the relevance of cathepsin K in the intestinal immune response during chronic intestinal inflammation. Design Chronic colitis was induced by administration of 2% dextran sodium sulphate (DSS) in distilled water. Mice were assessed for disease severity, histopathology and endoscopic appearance. Furthermore, DSS-exposed Ctsk−/− mice were treated by rectal administration of recombinant cathepsin K. Intestinal microflora was assessed by real-time PCR and 16srDNA molecular fingerprinting of ileal and colonic mucosal and faecal samples. Results Using Ctsk−/− mice, the authors demonstrate a protective role of cathepsin K against chronic DSS colitis. Dissecting the underlying mechanisms the authors found cathepsin K to be present in intestinal goblet cells and the mucin layer. Furthermore, a direct cathepsin K-mediated bactericidal activity against intestinal bacteria was demonstrated, which potentially explains the alteration of intestinal microbiota observed in Ctsk−/− mice. Rectal administration of recombinant cathepsin K in DSS-treated Ctsk−/− mice ameliorates the severity of intestinal inflammation. Conclusion These data identify extracellular cathepsin K as an intestinal antibacterial factor with anti-inflammatory potential and suggest that topical administration of cathepsin K might provide a therapeutic option for patients with inflammatory bowel disease.


Cell Reports | 2016

Epithelial IL-23R Signaling Licenses Protective IL-22 Responses in Intestinal Inflammation

Konrad Aden; Ateequr Rehman; Maren Falk-Paulsen; Thomas Secher; Jan W. P. Kuiper; Florian Tran; Steffen Pfeuffer; Raheleh Sheibani-Tezerji; Alexandra Breuer; Anne Luzius; Marlene Jentzsch; Robert Häsler; Susanne Billmann-Born; Olga Will; Simone Lipinski; Richa Bharti; Timon E. Adolph; Juan L. Iovanna; Richard S. Blumberg; Stefan Schreiber; Burkhard Becher; Mathias Chamaillard; Arthur Kaser; Philip Rosenstiel

A plethora of functional and genetic studies have suggested a key role for the IL-23 pathway in chronic intestinal inflammation. Currently, pathogenic actions of IL-23 have been ascribed to specific effects on immune cells. Herein, we unveil a protective role of IL-23R signaling. Mice deficient in IL-23R expression in intestinal epithelial cells (Il23R(ΔIEC)) have reduced Reg3b expression, show a disturbed colonic microflora with an expansion of flagellated bacteria, and succumb to DSS colitis. Surprisingly, Il23R(ΔIEC) mice show impaired mucosal IL-22 induction in response to IL-23. αThy-1 treatment significantly deteriorates colitis in Il23R(ΔIEC) animals, which can be rescued by IL-22 application. Importantly, exogenous Reg3b administration rescues DSS-treated Il23R(ΔIEC) mice by recruiting neutrophils as IL-22-producing cells, thereby restoring mucosal IL-22 levels. The study identifies a critical barrier-protective immune pathway that originates from, and is orchestrated by, IL-23R signaling in intestinal epithelial cells.


Gut | 2017

Uncoupling of mucosal gene regulation, mRNA splicing and adherent microbiota signatures in inflammatory bowel disease

Robert Häsler; Raheleh Sheibani-Tezerji; Anupam Sinha; Matthias Barann; Ateequr Rehman; Daniela Esser; Konrad Aden; Carolin Knecht; Berenice Brandt; Susanna Nikolaus; Sascha Schäuble; Christoph Kaleta; Andre Franke; Christoph Fretter; Werner Müller; Marc-Thorsten Hütt; Michael Krawczak; Stefan Schreiber; Philip Rosenstiel

Objective An inadequate host response to the intestinal microbiota likely contributes to the manifestation and progression of human inflammatory bowel disease (IBD). However, molecular approaches to unravelling the nature of the defective crosstalk and its consequences for intestinal metabolic and immunological networks are lacking. We assessed the mucosal transcript levels, splicing architecture and mucosa-attached microbial communities of patients with IBD to obtain a comprehensive view of the underlying, hitherto poorly characterised interactions, and how these are altered in IBD. Design Mucosal biopsies from Crohns disease and patients with UC, disease controls and healthy individuals (n=63) were subjected to microbiome, transcriptome and splicing analysis, employing next-generation sequencing. The three data levels were integrated by different bioinformatic approaches, including systems biology-inspired network and pathway analysis. Results Microbiota, host transcript levels and host splicing patterns were influenced most strongly by tissue differences, followed by the effect of inflammation. Both factors point towards a substantial disease-related alteration of metabolic processes. We also observed a strong enrichment of splicing events in inflamed tissues, accompanied by an alteration of the mucosa-attached bacterial taxa. Finally, we noted a striking uncoupling of the three molecular entities when moving from healthy individuals via disease controls to patients with IBD. Conclusions Our results provide strong evidence that the interplay between microbiome and host transcriptome, which normally characterises a state of intestinal homeostasis, is drastically perturbed in Crohns disease and UC. Consequently, integrating multiple OMICs levels appears to be a promising approach to further disentangle the complexity of IBD.


Oncogenesis | 2016

Classic IL-6R signalling is dispensable for intestinal epithelial proliferation and repair

Konrad Aden; A Breuer; Ateequr Rehman; H Geese; Florian Tran; Jan Sommer; Georg H. Waetzig; Torsten M. Reinheimer; Stefan Schreiber; Stefan Rose-John; Juergen Scheller; Philip Rosenstiel

Inflammatory bowel disease is characterized by disturbed cytokine signalling in the mucosa. Inhibition of the proinflammatory interleukin (IL)-6 pathway is a promising new therapeutic strategy, but safety concerns arise as IL-6 signalling also contributes to epithelial repair of the intestinal mucosa. To which extent IL-6 classic or trans-signalling contributes to intestinal repair remains elusive. We tested the influence of IL-6 classic signalling on intestinal repair and proliferation. Whereas IL-6 induced STAT3 phosphorylation in the colonic cancer cell lines, primary non-malignant intestinal organoids did not respond to IL-6 classic signalling. Mice deficient in intestinal IL-6R (IL-6RΔIEC mice) did not display increased susceptibility to acute dextran sulfate sodium (DSS)-induced colitis. In the azoxymethane DSS model IL-6RΔIEC mice were not protected from inflammation-induced carcinogenesis but showed comparable tumor load to wild-type mice. These data indicate that classic signalling is not the major pathway to transduce IL-6 stimuli into the intestinal epithelium.


Gut | 2018

Vedolizumab is associated with changes in innate rather than adaptive immunity in patients with inflammatory bowel disease

Sebastian Zeissig; Elisa Rosati; C Marie Dowds; Konrad Aden; Johannes Bethge; Berenice Schulte; Wei Hung Pan; Neha Mishra; Maaz Zuhayra; Marlies Marx; Maren Paulsen; A Strigli; Claudio Conrad; Dörthe Schuldt; Anupam Sinha; Henriette Ebsen; Sabin-Christin Kornell; Susanna Nikolaus; Alexander Arlt; Dieter Kabelitz; Mark Ellrichmann; Ulf Lützen; Philip Rosenstiel; Andre Franke; Stefan Schreiber

Objective Vedolizumab, a monoclonal antibody directed against the integrin heterodimer α4β7, is approved for the treatment of Crohn’s disease and ulcerative colitis. The efficacy of vedolizumab has been suggested to result from inhibition of intestinal T cell trafficking although human data to support this conclusion are scarce. We therefore performed a comprehensive analysis of vedolizumab-induced alterations in mucosal and systemic immunity in patients with inflammatory bowel disease (IBD), using anti-inflammatory therapy with the TNFα antibody infliximab as control. Design Immunophenotyping, immunohistochemistry, T cell receptor profiling and RNA sequencing were performed using blood and colonic biopsies from patients with IBD before and during treatment with vedolizumab (n=18) or, as control, the anti-TNFα antibody infliximab (n=20). Leucocyte trafficking in vivo was assessed using single photon emission computed tomography and endomicroscopy. Results Vedolizumab was not associated with alterations in the abundance or phenotype of lamina propria T cells and did not affect the mucosal T cell repertoire or leucocyte trafficking in vivo. Surprisingly, however, α4β7 antibody treatment was associated with substantial effects on innate immunity including changes in macrophage populations and pronounced alterations in the expression of molecules involved in microbial sensing, chemoattraction and regulation of the innate effector response. These effects were specific to vedolizumab, not observed in response to the TNFα antibody infliximab, and associated with inhibition of intestinal inflammation. Conclusion Our findings suggest that modulation of innate immunity contributes to the therapeutic efficacy of vedolizumab in IBD. Trial registration number NCT02694588


Journal of Experimental Medicine | 2018

ATG16L1 orchestrates interleukin-22-signaling in the intestinal epithelium via cGAS/STING

Konrad Aden; Florian Tran; Go Ito; Raheleh Sheibani-Tezerji; Simone Lipinski; Johannes Kuiper; Markus Tschurtschenthaler; Svetlana Saveljeva; Joya Bhattacharyya; Robert Häsler; Kareen Bartsch; Anne Luzius; Marlene Jentzsch; Maren Falk-Paulsen; Stephanie Stengel; Lina Welz; Robin Schwarzer; Björn Rabe; Winfried Barchet; Stefan Krautwald; Gunther Hartmann; Manolis Pasparakis; Richard S. Blumberg; Stefan Schreiber; Arthur Kaser; Philip Rosenstiel

A coding variant of the inflammatory bowel disease (IBD) risk gene ATG16L1 has been associated with defective autophagy and deregulation of endoplasmic reticulum (ER) function. IL-22 is a barrier protective cytokine by inducing regeneration and antimicrobial responses in the intestinal mucosa. We show that ATG16L1 critically orchestrates IL-22 signaling in the intestinal epithelium. IL-22 stimulation physiologically leads to transient ER stress and subsequent activation of STING-dependent type I interferon (IFN-I) signaling, which is augmented in Atg16l1&Dgr;IEC intestinal organoids. IFN-I signals amplify epithelial TNF production downstream of IL-22 and contribute to necroptotic cell death. In vivo, IL-22 treatment in Atg16l1&Dgr;IEC and Atg16l1&Dgr;IEC/Xbp1&Dgr;IEC mice potentiates endogenous ileal inflammation and causes widespread necroptotic epithelial cell death. Therapeutic blockade of IFN-I signaling ameliorates IL-22–induced ileal inflammation in Atg16l1&Dgr;IEC mice. Our data demonstrate an unexpected role of ATG16L1 in coordinating the outcome of IL-22 signaling in the intestinal epithelium.

Collaboration


Dive into the Konrad Aden'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