38 results on '"Simon Heidegger"'
Search Results
2. Experimental investigation of skin toxicity after immune checkpoint inhibition in combination with radiation therapy
- Author
-
Laura Lansink Rotgerink, Hannah Felchle, Annette Feuchtinger, Sophie M Nefzger, Caroline N Walther, Julia Gissibl, Katja Steiger, Thomas E Schmid, Simon Heidegger, Stephanie E Combs, and Julius C Fischer
- Subjects
Mice ,Original Article ,Original Articles ,immune checkpoint inhibition ,radiation therapy ,immune-related adverse events ,skin toxicity ,radiodermatitis ,epidermal hyperplasia ,dermal fibrosis ,adnexal atrophy ,Animals ,Immune Checkpoint Inhibitors ,Skin Diseases ,United Kingdom ,Skin ,ddc ,Pathology and Forensic Medicine - Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy. However, structured knowledge to mitigate a patient's specific risk of developing adverse events are limited. Nevertheless, there is an exponential growth of clinical studies combining conventional therapies such as radiation therapy (RT) with ICIs. Cutaneous reactions are among the most common adverse events after monotherapy with either ICIs or RT. So far, little is known about interindividual differences for the risk of developing severe tissue toxicity after the combination of RT with ICIs, and the underlying biological mechanisms are ill defined. We used experimental models of RT-induced skin injury to analyze skin toxicity after simultaneous application of ICIs. We compared different RT regimens such as fractionated or stereotactic RT with varying dose intensity. Strikingly, we found that simultaneous application of RT and ICIs did not significantly aggravate acute skin injury in two different mouse strains. Detailed examination of long-term tissue damage of the skin revealed similar signs of epidermal hyperplasia, dermal fibrosis, and adnexal atrophy. In summary, we here present the first experimental study demonstrating the excellent safety profiles of concurrent treatment with RT and ICIs. These findings will help to interpret the development of adverse events of the skin after radioimmunotherapy and guide the design of new clinical trials and clinical decision-making in individual cases. © 2022 The Authors. The Journal of Pathology published by John Wileyamp; Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2022
3. Supplementary Figure 1 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Author
-
Stefan Endres, David Anz, Cornelia Wurzenberger, Nadja Sandholzer, Bettina Storch, Laurin C. Roetzer, Simon Heidegger, Andreas Voelkl, Daniel Noerenberg, Christian Hotz, and Carole Bourquin
- Abstract
Supplementary Figure 1 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Published
- 2023
4. Supplementary Figure 3 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Author
-
Stefan Endres, David Anz, Cornelia Wurzenberger, Nadja Sandholzer, Bettina Storch, Laurin C. Roetzer, Simon Heidegger, Andreas Voelkl, Daniel Noerenberg, Christian Hotz, and Carole Bourquin
- Abstract
Supplementary Figure 3 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Published
- 2023
5. Supplementary Figure 2 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Author
-
Stefan Endres, David Anz, Cornelia Wurzenberger, Nadja Sandholzer, Bettina Storch, Laurin C. Roetzer, Simon Heidegger, Andreas Voelkl, Daniel Noerenberg, Christian Hotz, and Carole Bourquin
- Abstract
Supplementary Figure 2 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Published
- 2023
6. Supplementary Figure 4 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Author
-
Stefan Endres, David Anz, Cornelia Wurzenberger, Nadja Sandholzer, Bettina Storch, Laurin C. Roetzer, Simon Heidegger, Andreas Voelkl, Daniel Noerenberg, Christian Hotz, and Carole Bourquin
- Abstract
Supplementary Figure 4 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Published
- 2023
7. Supplementary Figure Legends 1-4 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Author
-
Stefan Endres, David Anz, Cornelia Wurzenberger, Nadja Sandholzer, Bettina Storch, Laurin C. Roetzer, Simon Heidegger, Andreas Voelkl, Daniel Noerenberg, Christian Hotz, and Carole Bourquin
- Abstract
Supplementary Figure Legends 1-4 from Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Published
- 2023
8. Identification of Bacterial and Viral Consortia for the Production of Intestinal Microbiota-Derived Metabolites Associated with Protection in Allogeneic Stem Cell Transplantation Patients
- Author
-
Erik Thiele Orberg, Elisabeth Meedt, Andreas Hiergeist, Jinling Xue, Paul Heinrich, Sakhila Ghimire, Melanie Tiefgraber, Sophia Göldel, Tina Eismann, Alix Schwarz, Sebastian Jarosch, Katja Steiger, Karin Kleigrewe, Julius Clemens Fischer, Klaus-Peter Janssen, Michael Quante, Simon Heidegger, Peter Herhaus, Mareike Verbeek, Jürgen Ruland, Christian Schulz, Daniela Weber, Matthias Edinger, Daniel Wolff, Dirk Busch, Wolfgang Herr, Florian Bassermann, Li Deng, Andre Gessner, Ernst Holler, and Hendrik Poeck
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
9. Negative feedback regulation of MAPK signaling is an important driver of CLL progression
- Author
-
Veronika Ecker, Lisa Brandmeier, Martina Stumpf, Piero Giansanti, Aida Varela Moreira, Lisa Pfeuffer, Marcel Fens, Junyan Lu, Bernhard Küster, Thomas Engleitner, Simon Heidegger, Ingo Ringshausen, Thorsten Zenz, Clemens Wendtner, Markus Müschen, Jürgen Ruland, and Maike Buchner
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Despite several potent targeted treatments for chronic lymphocytic leukemia (CLL), the clinical challenge of treating drug-resistant disease is emerging. In this study, we discovered that the dual-specific phosphatases DUSP1 and DUSP6 are required to negatively regulate Mitogen-activated protein kinases (MAPK) and thus counterbalance excessive MAPK activity to prevent apoptosis in CLL. We show that DUSP1 and DUSP6 are widely expressed in CLL and high expression of DUSP6 in CLL correlates with a poor clinical prognosis, which may reflect high levels of MAPK activity. Importantly, genetic deletion of the inhibitory phosphatase DUSP1 or DUSP6 and blocking DUSP1/6 function using a small molecule are toxic for CLL cells in vitro and in vivo. Analyzing downstream effects using global phospho-proteome approaches, we observed that acute activation of MAPK signaling by DUSP1/6 inhibition induces DNA damage response and thereby apoptotic cell death in CLL cells. This cell death is mediated by CHK kinases and can function independent of p53 and ATM, both effectors of DNA damage response, which are frequently deleted in CLL. Finally, we observed that DUSP1/6 inhibition is particularly effective against treatment-resistant CLL and therefore suggest transient DUSP1/6 inhibition as a promising novel treatment concept to eliminate drug-resistant CLL cells.
- Published
- 2023
10. The Microbial Metabolite Desaminotyrosine Enhances T-Cell Priming and Cancer Immunotherapy with Immune Checkpoint Inhibitors
- Author
-
Laura Joachim, Sascha Göttert, Anna Sax, Katja Steiger, Klaus Neuhaus, Kaiji Fan, Erik Thiele-Orberg, Karin Kleigrewe, Jürgen Ruland, Florian Bassermann, Wolfgang Herr, Christian Posch, Simon Heidegger, and Hendrik Poeck
- Published
- 2023
11. Tumor cell‐intrinsic RIG‐I signaling governs synergistic effects of immunogenic cancer therapies and checkpoint inhibitors in mice
- Author
-
Simon Heidegger, Sarah Dahl, Tobias Haas, Florian Bassermann, Alexander Wintges, and Hendrik Poeck
- Subjects
0301 basic medicine ,Programmed cell death ,viruses ,medicine.medical_treatment ,Immunology ,Melanoma, Experimental ,Receptors, Cell Surface ,chemical and pharmacologic phenomena ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,medicine ,Animals ,Humans ,Immunology and Allergy ,Receptor ,Immune Checkpoint Inhibitors ,Melanoma ,RIG-I ,RNA ,Cancer ,Drug Synergism ,Chemoradiotherapy ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,ddc ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Treatment Outcome ,030104 developmental biology ,Azacitidine ,Cancer research ,Immunotherapy ,biological phenomena, cell phenomena, and immunity ,Small nuclear RNA ,Epigenetic therapy ,Signal Transduction ,030215 immunology - Abstract
Immunogenic cancer therapies, including radiation and hypomethylating agents, such as 5-azacytidine, rely on tumor cell-intrinsic activation of the RNA receptor RIG-I for their synergism with immune checkpoint inhibitors. Possible RIG-I ligands are small nuclear RNA (snRNA) and endogenous retroviral elements (ERV) leaking from the nucleus during programmed cell death.
- Published
- 2021
12. Immunomodulatory Microbial Metabolites Identified in Allo-SCT Patients Protect from GvHD By Sting Dependent Modulation of Intestinal Tissue Homeostasis
- Author
-
Sascha Göttert, Erik Thiele-Orberg, Kaiji Fan, Sakhila Ghimire, Paul Heinrich, Wolfgang Herr, Simon Heidegger, Ernst Holler, and Hendrik Poeck
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
13. IFN-Gamma Is Crucial for the Counterbalance of T Cell-Mediated Injury to the Intestinal Stem Cell Compartment By Regulatory T Cells in Mice and Humans
- Author
-
Sascha Göttert, Julius Clemens Fischer, Gabriel Eisenkolb, Erik Thiele Orberg, Sebastian Jarosch, Ernst Holler, Dirk Busch, Simon Heidegger, and Hendrik Poeck
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
14. Identification of Protective, Metabolite-producing Bacterial and Viral Consortia in Allogeneic Stem Cell Transplantation Patients
- Author
-
Erik Thiele Orberg, Elisabeth Meedt, Andreas Hiergeist, Jinling Xue, Sakhila Ghimire, Melanie Tiefgraber, Sophia Göldel, Tina Eismann, Alix Schwarz, Sebastian Jarosch, Katja Steiger, Michael Gigl, Karin Kleigrewe, Julius Fischer, Klaus-Peter Janssen, Michael Quante, Simon Heidegger, Peter Herhaus, Mareike Verbeek, Jürgen Ruland, Daniela Weber, Daniel Wolff, Matthias Edinger, Dirk Busch, Wolfgang Herr, Florian Bassermann, André Gessner, Li Deng, Ernst Holler, and Hendrik Poeck
- Abstract
The intestinal bacteriome directly affects outcome in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-SCT). Besides bacteria, fungal and viral communities as well as microbiota-derived metabolites play a role. Yet, it is still unclear how dynamic shifts in these three communities contribute to (1) clinical outcome of allo-SCT patients, (2) production of metabolites and (3) how they are affected by microbiome modulation via antibiotics or fecal microbiota transplantation (FMT). Here, we performed a prospective, longitudinal study that combined transkingdom (bacteria, fungi, viruses) analysis of intestinal microbial communities with targeted metabolomics in allo-SCT patients (n=78) at two different transplantation centers. We uncovered a microbiome signature of metabolite-producing bacteria from the Lachnospiraceae and Oscillospiraceae families and their corresponding bacteriophages, which correlated with the production of immunomodulatory metabolites including short-chain fatty acids (SCFAs), metabolites associated with induction of type-I IFN signaling (IIMs) and bile acids. Sustained production of these “protective” metabolites after allo-SCT was associated with improved survival and reduced transplantation-related mortality, whereas antibiotic exposure significantly impaired metabolite expression. We demonstrate that single taxa domination and metabolite depletion in a patient suffering from graft versus host disease (GvHD) could be rescued by transfer of metabolite-producing bacterial consortia via fecal microbiota transplantation (FMT). FMT led to resolution of steroid refractory GvHD and was accompanied by an increase of bacterial and viral alpha diversity, restoration of SCFAs and IIMs and accumulation of regulatory T cells to the intestine. Our study demonstrates that microbiome modulation (via antibiotics or FMT) can affect the identified bacterial/bacteriophage network and their associated protective metabolites thereby determining clinical outcomes and provides a rationale for the development of engineered metabolite-producing consortia and defined metabolite combination drugs as novel microbiome-based therapies.
- Published
- 2022
15. In Vivo Immunogenicity Screening of Tumor-Derived Extracellular Vesicles by Flow Cytometry of Splenic T Cells
- Author
-
Florian, Stritzke, Hendrik, Poeck, and Simon, Heidegger
- Subjects
Extracellular Vesicles ,Mice ,Neoplasms ,Animals ,Flow Cytometry ,Spleen - Abstract
Immunogenic cell death of tumors, caused by chemotherapy or irradiation, can trigger tumor-specific T cell responses by releasing danger-associated molecular patterns and inducing the production of type I interferon. Immunotherapies, including checkpoint inhibition, primarily rely on preexisting tumor-specific T cells to unfold a therapeutic effect. Thus, synergistic therapeutic approaches that exploit immunogenic cell death as an intrinsic anti-cancer vaccine may improve their responsiveness. However, the spectrum of immunogenic factors released by cells under therapy-induced stress remains incompletely characterized, especially regarding extracellular vesicles (EVs). EVs, nano-scale membranous particles emitted from virtually all cells, are considered to facilitate intercellular communication and, in cancer, have been shown to mediate cross-priming against tumor antigens. To assess the immunogenic effect of EVs derived from tumors under various conditions, adaptable, scalable, and valid methods are sought-for. Therefore, herein a relatively easy and robust approach is presented to assess EVs' in vivo immunogenicity. The protocol is based on flow cytometry analysis of splenic T cells after in vivo immunization of mice with EVs, isolated by precipitation-based assays from tumor cell cultures under therapy or steady-state conditions. For example, this work shows that oxaliplatin exposure of B16-OVA murine melanoma cells resulted in the release of immunogenic EVs that can mediate the activation of tumor-reactive cytotoxic T cells. Hence, screening of EVs via in vivo immunization and flow cytometry identifies conditions under which immunogenic EVs can emerge. Identifying conditions of immunogenic EV release provides an essential prerequisite to testing EVs' therapeutic efficacy against cancer and exploring the underlying molecular mechanisms to ultimately unveil new insights into EVs' role in cancer immunology.
- Published
- 2021
16. In Vivo Immunogenicity Screening of Tumor-Derived Extracellular Vesicles by Flow Cytometry of Splenic T Cells
- Author
-
Florian Stritzke, Hendrik Poeck, and Simon Heidegger
- Subjects
medicine.diagnostic_test ,General Immunology and Microbiology ,Chemistry ,T cell ,General Chemical Engineering ,General Neuroscience ,General Biochemistry, Genetics and Molecular Biology ,Flow cytometry ,medicine.anatomical_structure ,Antigen ,In vivo ,Interferon ,medicine ,Cancer research ,Cytotoxic T cell ,Immunogenic cell death ,medicine.drug ,Cancer immunology - Abstract
Immunogenic cell death of tumors, caused by chemotherapy or irradiation, can trigger tumor-specific T cell responses by releasing danger-associated molecular patterns and inducing the production of type I interferon. Immunotherapies, including checkpoint inhibition, primarily rely on preexisting tumor-specific T cells to unfold a therapeutic effect. Thus, synergistic therapeutic approaches that exploit immunogenic cell death as an intrinsic anti-cancer vaccine may improve their responsiveness. However, the spectrum of immunogenic factors released by cells under therapy-induced stress remains incompletely characterized, especially regarding extracellular vesicles (EVs). EVs, nano-scale membranous particles emitted from virtually all cells, are considered to facilitate intercellular communication and, in cancer, have been shown to mediate cross-priming against tumor antigens. To assess the immunogenic effect of EVs derived from tumors under various conditions, adaptable, scalable, and valid methods are sought-for. Therefore, herein a relatively easy and robust approach is presented to assess EVs' in vivo immunogenicity. The protocol is based on flow cytometry analysis of splenic T cells after in vivo immunization of mice with EVs, isolated by precipitation-based assays from tumor cell cultures under therapy or steady-state conditions. For example, this work shows that oxaliplatin exposure of B16-OVA murine melanoma cells resulted in the release of immunogenic EVs that can mediate the activation of tumor-reactive cytotoxic T cells. Hence, screening of EVs via in vivo immunization and flow cytometry identifies conditions under which immunogenic EVs can emerge. Identifying conditions of immunogenic EV release provides an essential prerequisite to testing EVs' therapeutic efficacy against cancer and exploring the underlying molecular mechanisms to ultimately unveil new insights into EVs' role in cancer immunology.
- Published
- 2021
17. Innate Immune Stimulation in Cancer Therapy
- Author
-
Garcia-Foncillas J, Peter Duewell, Simon Heidegger, and Sebastian Kobold
- Subjects
Myeloid ,Adaptive Immunity ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Animals ,Humans ,Innate immune system ,Effector ,business.industry ,Pattern recognition receptor ,Hematology ,Acquired immune system ,Immunity, Innate ,3. Good health ,Immunosurveillance ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Immunotherapy ,business ,Neuroscience ,030215 immunology - Abstract
The innate immune system has evolved as a first line of defense against invading pathogens and acts via classes of germline-encoded receptor systems to respond with proinflammatory cytokines. Innate immune cells, predominantly cells of the myeloid compartment, are capable of providing a potent basis for boosting adaptive immunity in malignant diseases. The authors review their current understanding of the molecular mechanisms whereby innate pattern recognition receptors participate in immunosurveillance of cancer cells. They discuss how innate effector mechanisms are currently being targeted pharmacologically and how improved understanding of the biology of these pathways is leading to novel immunotherapies of cancer.
- Published
- 2019
18. IFN-Gamma Producing Regulatory T Cells Counterbalance T Cell-Mediated Injury to the Intestinal Stem Cell Compartment in Mice and Humans
- Author
-
Dirk Büsch, Caroline Walther, Julius C. Fischer, Thomas Engleitner, Stefanie E. Combs, Roland Rad, Simon Heidegger, Marianne Reiser, Laura Lansink Rotgerink, Hendrik Poeck, Gabriel Eisenkolb, Sascha Göttert, Sebastian Jarosch, Lena Klostermeier, Sophie Nefzger, Erik Thiele Orberg, Katja Steiger, Ernst Holler, and Florian Bassermann
- Subjects
medicine.anatomical_structure ,T cell ,Immunology ,medicine ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Stem cell compartment ,Ifn gamma ,Cell biology - Abstract
Background: Graft-versus-host disease (GVHD) is a dreaded complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Such inflammatory diseases are fostered by damage to the intestinal barrier after transplantation. Consequently, the integrity and regeneration of the intestinal barrier is a key factor in the prevention of GVHD. On one side the main driver for regeneration of damaged gut epithelium are intestinal stem cells (ISC), but on the other side these cells are themselves primary targets of donor-derived T cells. One known mechanism of T cell mediated damage to the stem cell compartment is through IFN-γ dependent ISC toxicity. Yet, little is known about how T cells are contributing to the regeneration of damaged tissue after allo-HSCT and GVHD. Methods: To address this, we used preclinical models for allo-HSCT and GVHD including transplantation of recipient mice with escalating doses of Wildtype or IFN-γ-deficient allogeneic T cells and in the presence or absence of the JAK-1/2 inhibitor ruxolitinib. Intestinal regeneration was assessed by RNA-seq, flow cytometry and a newly established ex vivo organoid recovery assay. GVHD outcome was assessed by clinical scoring, histology and survival. Additionally, we established an allogeneic co-culture system of murine or human intestinal organoids with CD4+ conventional T cells or T regs -/+ Ruxolitinib. Effects on organoid growth and cell death were assessed by size measurements and manual counting after passaging. Results: We here demonstrate that recipient mice with increasingly dense intestinal infiltration by allogeneic T cells not only developed more severe GVHD (Fig. 1A), but also showed augmented recovery potential early after allo-HSCT (Fig. 1B). This was associated with intestinal gene signatures related to epithelial regeneration and protection from GVHD. Utilizing ex vivo cultures of intestinal organoids generated from murine allo-HSCT recipients, we found that development of GVHD but also regenerative capacity of ISCs were dependent on interferon (IFN)-γ-producing T cells in the intestine (Fig. 2A-B). Mice with fulminant GVHD and enhanced organoid recovery showed accumulation of intestinal regulatory T cells (Tregs) (Fig. 2C). Ex vivo, T regs nurtured growth of intestinal organoids in an IFN-γ dependent manner (Fig. 2D-E). This effect was diminished in intestinal organoids lacking IFNγR signaling, but was independent of T reg intrinsic IFNγR signaling (Fig. 2E-F). Intriguingly, treatment of murine allo-HSCT recipients with the JAK-1/2 inhibitor ruxolitinib enhanced epithelial organoid regeneration and numbers of intestinal Tregs (Fig. 3A-B). Similarily, growth of human intestinal organoids co-cultured with allogeneic T cells could be augmented by ruxolitinib treatment (Fig. 3C). We thus propose that the level and differentiation of infiltrating intestinal T cells determines both ISC damage and epithelial regeneration during immune-mediated tissue injury, leading to a sensitive equilibrium that can be modulated by therapeutic intervention. We also provide evidence that ruxolitinib improves ISC regeneration via IFNγ-producing Treg cells. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
19. Microbial-Derived Metabolites Induce Epithelial Recovery Via the Sting Pathway in Mice and Men and Protect from Graft-Versus-Host Disease
- Author
-
Simon Heidegger, Wolfgang Herr, Florian Bassermann, Elisabeth Meedt, Sascha Göttert, Erik Thiele Orberg, Hendrik Poeck, Ernst Holler, Andreas Hiergeist, André Gessner, and Karin Kleigrewe
- Subjects
Sting ,Graft-versus-host disease ,business.industry ,Immunology ,medicine ,Cell Biology ,Hematology ,medicine.disease ,business ,Biochemistry - Abstract
Background: Graft-versus-host disease (GVHD) is a dreaded complication after allogeneic stem-cell transplantation (ASCT). Previously, we and others showed that activation of type I interferon (IFN-I) inducing pathways such as RIG-I/MAVS or cGAS-STING can promote the integrity of the intestinal barrier and limit GVHD (Fischer et al., Sci Transl Med, 2017). However, the signals that drive these protective IFN-I responses are poorly understood. Commensal microbiota can (i) have distant effects on immune responses through modulation of IFN-I signaling (Steed et al, Science, 2017; Swimm et al, Blood, 2018) and (ii) predict mortality in ASCT patients (Peled et al., N Engl J Med, 2020). We hypothesized that microbiota-derived products such as microbial metabolites engage IFN-I signaling in immune and non-immune cells poising them for induction of protective responses. We established a prospective, multi-centric clinical study in patients newly diagnosed with acute leukemia and performed longitudinal stool sampling to track changes in microbial community composition and metabolites expression levels. Submitted as a separate abstract to ASH 2021 (Orberg & Meedt et al.), we show that patients with high metabolite expression going into ASCT are less likely to develop GVHD. In this study, we translate our clinical observations to mouse models of acute GVHD and human and mouse intestinal organoids to uncover the molecular mechanisms via which metabolites protect the intestinal barrier during ASCT. Methods: Stool samples from ASCT patients were obtained in Munich and at Regensburg in accordance to IRB-approved study protocols. Patients were sampled at initial diagnosis (Dx), prior to conditioning and weekly after ASCT up to day 28. We analyzed samples by 16S rRNA sequencing and mass spectrometry to obtain a complete picture of microbiome composition and function. Next, we tested metabolites which we detected in patients (desaminotyrosine [DAT], indole-3-carboxaldehyde [ICA]) as treatment in preclinical models in ASCT and GVHD mouse models. Outcomes were assessed by a novel organoid recovery assay in addition to established read-outs. To obtain a mechanistic understanding of the signaling pathways involved, we stimulated WT or IFN-I-signaling-impaired mouse (incl. STING -/-, MAVS -/-, IFNαR -/-) as well as human intestinal crypt-derived organoids with metabolites. Results: Here, we present a 64-year-old female patient diagnosed with AML who received a 9/10 HLA-matched ASCT. At day 7, i.v. antibiotics were started due to fever and Enterococcus bacteraemia. At day 15, the patient developed skin and GI GVHD (Glucksberg III). At the timepoint initial diagnosis (Dx), i.e. the timepoint when we diagnosed AML but before therapy was initiated, we detected rich alpha diversity (Panel 1a) in the patient's stool. Flavonifractor plautii, a producer of the metabolite DAT, was detectable (Panel 1b). We observed high-level expression of metabolites including short-chain fatty acids, DAT, ICA and secondary bile acids (Panel 1c). Following ASCT, and especially at the early time-points day 0 and day 7, alpha diversity and metabolite expression declined drastically. We confirmed this trend in our multi-centric cohort of ASCT patients by comparing levels of DAT and ICA sampled at admission to the transplantation ward (Conditioning) versus at clinical diagnosis of GVHD: in patients with GVHD, metabolite levels were drastically reduced (Panel 2). Next, we prophylactically administered metabolites in a major mismatch mouse ASCT model. Metabolite-treated mice showed significantly showed better outcomes in our organoid recovery assay, which measures the ability of intestinal stem cells to recover after allogeneic injury (Panel 3). This effect that was abrogated in STING -/- recipients. Metabolite stimulation of mouse small intestinal organoids promoted organoid numbers and size, and required intact STING signaling (Panel 4a). Human colon organoids also responded to DAT and ICA, however the metabolite effect was lost when co-administered with the STING-inhibitor H151. Conclusions: We identify that microbial-derived metabolites detected in patients can engage the STING pathway in humans and mice to confer resistance from immune damage. Thus, prophylactic administration of metabolite cocktails or bacterial consortia that can produce these metabolites may reduce occurrence of GVHD in ASCT patients. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
20. A20 Restrains Thymic Regulatory T Cell Development
- Author
-
Simon Heidegger, Julius C. Fischer, Christian Peschel, Maike Kober, Xian Chang Li, Marc Schmidt-Supprian, Vera Otten, Hendrik Poeck, Marc Rosenbaum, Christoph Drees, Tobias Haas, Silvia Spoerl, Martina Schmickl, Geert van Loo, and Rudi Beyaert
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Regulatory T cell ,T cell ,Immunology ,Graft vs Host Disease ,Apoptosis ,chemical and pharmacologic phenomena ,Thymus Gland ,Biology ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Article ,Immune tolerance ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Glucocorticoid-Induced TNFR-Related Protein ,hemic and lymphatic diseases ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Tumor Necrosis Factor alpha-Induced Protein 3 ,NF-kappa B ,Transcription Factor RelA ,FOXP3 ,Cell Differentiation ,Forkhead Transcription Factors ,hemic and immune systems ,Flow Cytometry ,Natural killer T cell ,Proto-Oncogene Proteins c-rel ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Interleukin-2 ,Signal Transduction ,Stem Cell Transplantation ,030215 immunology - Abstract
Maintaining immune tolerance requires the production of Foxp3-expressing regulatory T (Treg) cells in the thymus. Activation of NF-κB transcription factors is critically required for Treg cell development, partly via initiating Foxp3 expression. NF-κB activation is controlled by a negative feedback regulation through the ubiquitin editing enzyme A20, which reduces proinflammatory signaling in myeloid cells and B cells. In naive CD4+ T cells, A20 prevents kinase RIPK3-dependent necroptosis. Using mice deficient for A20 in T lineage cells, we show that thymic and peripheral Treg cell compartments are quantitatively enlarged because of a cell-intrinsic developmental advantage of A20-deficient thymic Treg differentiation. A20-deficient thymic Treg cells exhibit reduced dependence on IL-2 but unchanged rates of proliferation and apoptosis. Activation of the NF-κB transcription factor RelA was enhanced, whereas nuclear translocation of c-Rel was decreased in A20-deficient thymic Treg cells. Furthermore, we found that the increase in Treg cells in T cell–specific A20-deficient mice was already observed in CD4+ single-positive CD25+ GITR+ Foxp3− thymic Treg cell progenitors. Treg cell precursors expressed high levels of the tumor necrosis factor receptor superfamily molecule GITR, whose stimulation is closely linked to thymic Treg cell development. A20-deficient Treg cells efficiently suppressed effector T cell–mediated graft-versus-host disease after allogeneic hematopoietic stem cell transplantation, suggesting normal suppressive function. Holding thymic production of natural Treg cells in check, A20 thus integrates Treg cell activity and increased effector T cell survival into an efficient CD4+ T cell response.
- Published
- 2017
21. Cutting Edge in IFN Regulation: Inflammatory Caspases Cleave cGAS
- Author
-
Tobias Haas, Simon Heidegger, and Hendrik Poeck
- Subjects
0301 basic medicine ,Inflammasomes ,Immunology ,Inflammation ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosol ,Interferon ,Cleave ,medicine ,Humans ,Immunology and Allergy ,Caspase ,biology ,biochemical phenomena, metabolism, and nutrition ,Nucleotidyltransferases ,Cell biology ,030104 developmental biology ,Infectious Diseases ,chemistry ,Apoptosis ,Caspases ,Interferon Type I ,biology.protein ,medicine.symptom ,DNA ,Interferon type I ,medicine.drug - Abstract
Caspases have important functions beyond their established role in driving inflammation and apoptosis. In this issue of Immunity, Wang et al. (2017) demonstrate that inflammasome-triggered caspases cleave and inactivate the DNA sensor cGAS, thus restricting the type I interferon response to cytosolic DNA.
- Published
- 2017
22. Type I interferon signaling before hematopoietic stem cell transplantation lowers donor T cell activation via reduced allogenicity of recipient cells
- Author
-
Simon Heidegger, Sascha Göttert, Michael Bscheider, Tobias Haas, Stephanie E. Combs, Erik Thiele Orberg, Julius C. Fischer, Hendrik Poeck, and Florian Bassermann
- Subjects
0301 basic medicine ,Transplantation Conditioning ,Bone marrow transplantation ,T cell ,medicine.medical_treatment ,T-Lymphocytes ,Graft vs Host Disease ,lcsh:Medicine ,chemical and pharmacologic phenomena ,Hematopoietic stem cell transplantation ,Lymphocyte Activation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Medicine ,Animals ,Transplantation, Homologous ,Interferon gamma ,lcsh:Science ,Cell Proliferation ,Immunosuppression Therapy ,Mice, Inbred BALB C ,Multidisciplinary ,Cell Death ,business.industry ,lcsh:R ,Hematopoietic Stem Cell Transplantation ,Total body irradiation ,Hematopoietic Stem Cells ,CD11c Antigen ,ddc ,Transplantation ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,surgical procedures, operative ,Preclinical research ,Interferon Type I ,Cancer research ,lcsh:Q ,business ,030215 immunology ,medicine.drug ,Signal Transduction - Abstract
Recent studies highlight immunoregulatory functions of type I interferons (IFN-I) during the pathogenesis of graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). We demonstrated that selective activation of IFN-I pathways including RIG-I/MAVS and cGAS/STING prior to allo-HSCT conditioning therapy can ameliorate the course of GVHD. However, direct effects of IFN-Is on immune cells remain ill characterized. We applied RIG-I agonists (3pRNA) to stimulate IFN-I production in murine models of conditioning therapy with total body irradiation (TBI) and GVHD. Using IFN-I receptor-deficient donor T cells and hematopoietic cells, we found that endogenous and RIG-I-induced IFN-Is do not reduce GVHD by acting on these cell types. However, 3pRNA applied before conditioning therapy reduced the ability of CD11c+ recipient cells to stimulate proliferation and interferon gamma expression of allogeneic T cells. Consistently, RIG-I activation before TBI reduced the proliferation of transplanted allogeneic T-cells. The reduced allogenicity of CD11c+ recipient cells was dependent on IFN-I signaling. Notably, this immunosuppressive function of DCs was restricted to a scenario where tissue damage occurs. Our findings uncover a context (damage by TBI) and IFN-I dependent modulation of T cells by DCs and extend the understanding about the cellular targets of IFN-I during allo-HSCT and GVHD.
- Published
- 2018
23. Regeneration After Radiation- and Immune-Mediated Tissue Injury Is Not Enhanced by Type III Interferon Signaling
- Author
-
Stephanie E. Combs, Matthias Beudert, Alexander Wintges, Martin Schlapschy, Simon Heidegger, Florian Bassermann, Julius C. Fischer, Arne Skerra, Hendrik Poeck, Tobias Haas, and Chia-Ching Lin
- Subjects
Cancer Research ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Context (language use) ,Hematopoietic stem cell transplantation ,Thymus Gland ,030218 nuclear medicine & medical imaging ,Interferon Lambda ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,immune system diseases ,Interferon ,medicine ,Cytotoxic T cell ,Animals ,Regeneration ,Radiology, Nuclear Medicine and imaging ,Intestinal Mucosa ,Radiation ,business.industry ,Regeneration (biology) ,Hematopoietic Stem Cell Transplantation ,Interleukin ,Total body irradiation ,Intestines ,surgical procedures, operative ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Interferons ,business ,Whole-Body Irradiation ,medicine.drug ,Signal Transduction - Abstract
Purpose Type I interferon (IFN-I) and interleukin (IL)-22 modulate regeneration of the thymus and intestinal epithelial cells (IECs) after cytotoxic stress such as irradiation. Radiation-induced damage to thymic tissues and IECs is a crucial aspect during the pathogenesis of inadequate immune reconstitution and acute graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT) with myeloablative total body irradiation (TBI), respectively. IL-22 and IFN-I reduce the severity of acute GVHD after allo-HSCT with myeloablative TBI. However, the role of biologically related type III interferon (IFN-III), also known as interferon lambda (IFN-λ) or IL-28, in this context is unclear. We therefore studied the role of the IFN-III pathway in thymic regeneration and GVHD after TBI and allo-HSCT. Methods and Materials Cohoused wild-type (WT) and IFN-III receptor–deficient (IL-28 receptor alpha subunit–deficient/IL-28Ra–/–) mice were analyzed in models of TBI-induced thymus damage and a model of GVHD after allo-HSCT with myeloablative TBI. PASylated IFN-III (PASylated IL-28A, XL-protein GmbH) was generated to prolong the plasma half-life of IFN-III. Pharmacologic activity and the effects of PASylated IL-28A on radiation-induced thymus damage and the course of GVHD after allo-HSCT with myeloablative TBI were tested. Results The course and severity of GVHD after myeloablative TBI and allo-HSCT in IL-28Ra–/– mice was comparable to those in WT mice. Activation of the IFN-III pathway by PASylated IL-28A did not significantly modulate GVHD after allo-HSCT with TBI. Furthermore, IL28Ra–/– mice and WT mice showed similar thymus regeneration after radiation, which could also not be significantly modulated by IFN-III receptor engagement using PASylated IL-28A. Conclusions We analyzed the role of IFN-III signaling during radiation-mediated acute tissue injury. Despite molecular and biologic homologies with IFN-I and IL-22, IFN-III signaling did not improve thymus regeneration after radiation or the course of GVHD after myeloablative TBI and allo-HSCT.
- Published
- 2018
24. TLR and RLR Signaling Are Reprogrammed in Opposite Directions after Detection of Viral Infection
- Author
-
Carole Bourquin, Thomas Huber, Simon Heidegger, Andreas Sailer, Marina Treinies, Anne Oberson, Christian Hotz, Laurin C.M. Roetzer, Stefan Endres, and Tina Herbst
- Subjects
Mice, 129 Strain ,Immunology ,Immunoblotting ,Gene Expression ,Inflammation ,Biology ,Sendai virus ,Cell Line ,DEAD-box RNA Helicases ,Immunity ,Cell Line, Tumor ,medicine ,Immunology and Allergy ,Animals ,Receptor ,DEAD Box Protein 58 ,Mice, Knockout ,Innate immune system ,Interleukin-6 ,Reverse Transcriptase Polymerase Chain Reaction ,Macrophages ,Toll-Like Receptors ,Interferon-alpha ,Interferon-beta ,Interleukin-10 ,Mice, Inbred C57BL ,Interleukin 10 ,RNA silencing ,Poly I-C ,Virus Diseases ,Receptors, Pattern Recognition ,Signal transduction ,medicine.symptom ,Signal Transduction - Abstract
Innate immune recognition of RNA is key for the initiation of immunity in response to viral infection. Although the factors controlling the detection of viral RNA by innate immune receptors in host cells are increasingly well understood, little is known about the dynamic changes in signaling after the initial triggering of these receptors. In this study, we report that preconditioning with the synthetic dsRNA polyinosinic-polycytidylic acid [poly(I:C)], a mimetic of viral RNA, rapidly reprograms murine APCs by simultaneously augmenting sensitivity of endosomal TLRs and inhibiting activation of RIG-I–like receptors (RLRs) in an IFN-β–dependent manner. These changes in receptor sensitivity were also seen in vivo after treatment of mice with poly(I:C). Mechanistically, the increased sensitivity of the TLR pathway was associated with elevated MAPK and NF-κB activity. The RLR response was inhibited downstream of TANK-binding kinase-1, resulting in decreased IFN regulatory factor 3 phosphorylation. Reprogramming of pattern-recognition receptor signaling also occurred after viral infection, because infection of host cells with Sendai virus or their exposure to supernatant from virus-infected cells induced the same changes in TLR and RLR sensitivity as poly(I:C). Thus, innate recognition of viral infection critically modifies responses to pattern-recognition receptor stimulation. These dynamic adaptations to infection may reinforce antiviral immunity and at the same time serve to limit pathological inflammation.
- Published
- 2015
- Full Text
- View/download PDF
25. Card9 controls Dectin-1-induced T-cell cytotoxicity and tumor growth in mice
- Author
-
Michael Bscheider, Julius C. Fischer, Tobias Haas, Alexander Wintges, Hendrik Poeck, Christian Peschel, Jürgen Ruland, Gabriel Eisenkolb, Sarah Bek, Martina Schmickl, Simon Heidegger, and Silvia Spoerl
- Subjects
0301 basic medicine ,Molecular immunology and signaling ,Inflammasomes ,medicine.medical_treatment ,Short Communication ,Immunology ,chemical and pharmacologic phenomena ,Biology ,03 medical and health sciences ,Mice ,Immune system ,Cross-Priming ,Neoplasms ,Cross‐priming ,Tumor immunity ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,CD8+ cytotoxic T cells ,Lectins, C-Type ,Basic ,Innate immune system ,Vaccination ,Inflammasome ,Immunotherapy ,Card9 ,Immunity, Innate ,3. Good health ,Cell biology ,ddc ,CARD Signaling Adaptor Proteins ,Mice, Inbred C57BL ,Short Communication|Basic ,CTL ,030104 developmental biology ,Signal transduction ,CD8 ,medicine.drug ,Signal Transduction ,T-Lymphocytes, Cytotoxic ,Dectin‐1 - Abstract
Activation of the C-type lectin receptor Dectin-1 by β-glucans triggers multiple signals within DCs that result in activation of innate immunity. While these mechanisms can potently prime CD8+ cytotoxic T-cell (CTL) responses without additional adjuvants, the Dectin-1 effector pathways that control CTL induction remain unclear. Here we demonstrate that Dectin-1-induced CTL cross-priming in mice does not require inflammasome activation but strictly depends on the adapter protein Card9 in vitro. In vivo, Dectin-1-mediated Card9 activation after vaccination drives both expansion and activation of Ag-specific CTLs, resulting in long-lasting CTL responses that are sufficient to protect mice from tumor challenge. This Dectin-1-induced antitumor immune response was independent of NK cell function and completely abrogated in Card9-deficient mice. Thus, our results demonstrate that Dectin-1-triggered Card9 signaling but not inflammasome activation can potently cross-prime Ag-specific CTLs, suggesting that this pathway would be a candidate for immunotherapy and vaccine development.
- Published
- 2017
26. Virus-associated activation of innate immunity induces rapid disruption of Peyer’s patches in mice
- Author
-
Susanne Radtke-Schuller, Christian Hotz, Carole Bourquin, Nicolas Stephan, Sebastian Kobold, Stefan Endres, Nadja Sandholzer, Bernadette Bohn, Wolfgang Peter Fendler, Simon Heidegger, Tina Herbst, David Anz, Katharina Eisenächer, and Nina Suhartha
- Subjects
Male ,Lymphocyte ,Immunology ,Biology ,Biochemistry ,Vesicular stomatitis Indiana virus ,Virus ,Mice ,Peyer's Patches ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Immunity ,Interferon ,medicine ,Animals ,Cells, Cultured ,030304 developmental biology ,Mice, Knockout ,B-Lymphocytes ,0303 health sciences ,Innate immune system ,Innate lymphoid cell ,Peyer's patch ,Cell Biology ,Hematology ,Acquired immune system ,Immunity, Innate ,3. Good health ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Interferon Type I ,RNA, Viral ,Female ,Vesicular Stomatitis ,030215 immunology ,medicine.drug - Abstract
Early in the course of infection, detection of pathogen-associated molecular patterns by innate immune receptors can shape the subsequent adaptive immune response. Here we investigate the influence of virus-associated innate immune activation on lymphocyte distribution in secondary lymphoid organs. We show for the first time that virus infection of mice induces rapid disruption of the Peyer's patches but not of other secondary lymphoid organs. The observed effect was not dependent on an active infectious process, but due to innate immune activation and could be mimicked by virus-associated molecular patterns such as the synthetic double-stranded RNA poly(I:C). Profound histomorphologic changes in Peyer's patches were associated with depletion of organ cellularity, most prominent among the B-cell subset. We demonstrate that the disruption is entirely dependent on type I interferon (IFN). At the cellular level, we show that virus-associated immune activation by IFN-α blocks B-cell trafficking to the Peyer's patches by downregulating expression of the homing molecule α4β7-integrin. In summary, our data identify a mechanism that results in type I IFN-dependent rapid but reversible disruption of intestinal lymphoid organs during systemic viral immune activation. We propose that such rerouted lymphocyte trafficking may impact the development of B-cell immunity to systemic viral pathogens.
- Published
- 2013
27. Interleukin-22 Is Frequently Expressed in Small- and Large-Cell Lung Cancer and Promotes Growth in Chemotherapy-Resistant Cancer Cells
- Author
-
Rudolf M. Huber, Stefanie Völk, Simon Rothenfußer, Natascha Jennifer Küpper, P Düwell, Sarah Minner, Simon Heidegger, Max Schnurr, Amanda Tufman, Waldemar Wilczak, Sebastian Kobold, Michael Lindner, Stefan Endres, Till S. Clauditz, and Ina Koch
- Subjects
Adult ,Male ,Large-cell lung cancer ,Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Adolescent ,Biology ,Interleukin-22 ,Small-cell lung cancer ,Flow cytometry ,Interleukin 22 ,Interleukin-22-receptor 1 ,Cell Line, Tumor ,medicine ,Carcinoma ,Humans ,Child ,Lung cancer ,Aged ,Aged, 80 and over ,medicine.diagnostic_test ,Cell growth ,Interleukins ,Receptors, Interleukin ,Middle Aged ,Prognosis ,medicine.disease ,Immunohistochemistry ,Small Cell Lung Carcinoma ,Oncology ,Drug Resistance, Neoplasm ,Apoptosis ,Cell culture ,Cancer cell ,Cancer research ,Carcinoma, Large Cell ,Female - Abstract
Introduction: In lung cancer, interleukin-22 (IL-22) expression within primary tissue has been demonstrated, but the frequency and the functional consequence of IL-22 signaling have not been addressed. This study aims at analyzing the cellular effects of IL-22 on lung carcinoma cell lines and the prognostic impact of IL-22 tis- sue expression in lung cancer patients. Methods: Biological effects of IL-22 signaling were investigated in seven lung cancer cell lines by Western blot, flow cytometry, real- time polymerase chain reaction, and proliferation assays. Tumor tissue specimens of two cohorts with a total of 2300 lung cancer patients were tested for IL-22 expression by immunohistochemistry. IL-22 serum concentrations were analyzed in 103 additional patients by enzyme-linked immunosorbent assay. Results: We found the IL-22 receptor 1 (IL-22-R1) to be expressed in six of seven lung cancer cell lines. However IL-22 signaling was functional in only four cell lines, where IL-22 induced signal trans- ducer activator of transcription 3 phosphorylation and increased cell proliferation. Furthermore, IL-22 induced the expression of anti- apoptotic B-cell lymphoma 2, but did not rescue tumor cells from carboplatin-induced apoptosis. Cisplatin-resistant cell lines showed a significant up-regulation of IL-22-R1 along with a stronger prolifera- tive response to IL-22 stimulation. IL-22 was preferentially expressed in small- and large-cell lung carcinoma (58% and 46% of cases, respectively). However, no correlation between IL-22 expression by immunohistochemistry and prognosis was observed. Conclusion: IL-22 is frequently expressed in lung cancer tissue. Enhanced IL-22-R1 expression and signaling in chemotherapy- refractory cell lines are indicative of a protumorigenic function of IL-22 and may contribute to a more aggressive phenotype.
- Published
- 2013
- Full Text
- View/download PDF
28. TLR Activation Excludes Circulating Naive CD8+ T Cells from Gut-Associated Lymphoid Organs in Mice
- Author
-
Carole Bourquin, Bärbel Stecher, Christian Hotz, David Anz, Bernadette Bohn, Holger Rüssmann, Simon Heidegger, Nicolas Stephan, Nina Suhartha, Nadja Sandholzer, Stefan Endres, and Sophie-Kathrin Kirchner
- Subjects
Salmonella typhimurium ,Integrins ,Ovalbumin ,Immunology ,Down-Regulation ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Systemic inflammation ,Mice ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,Immune system ,Cell Movement ,T-Lymphocyte Subsets ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Cell Proliferation ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Interleukin-12 Subunit p40 ,Interleukin-6 ,Effector ,Toll-Like Receptors ,Imidazoles ,CD28 ,Dendritic Cells ,Cell biology ,Mice, Inbred C57BL ,Salmonella Infections ,Female ,medicine.symptom ,CD8 ,Signal Transduction ,030215 immunology ,Homing (hematopoietic) - Abstract
The trafficking of effector T cells is tightly regulated by the expression of site-specific sets of homing molecules. In contrast, naive T cells are generally assumed to express a uniform pattern of homing molecules and to follow a random distribution within the blood and secondary lymphoid organs. In this study, we demonstrate that systemic infection fundamentally modifies the trafficking of circulating naive CD8+ T cells. We show that on naive CD8+ T cells, the constitutive expression of the integrin α4β7 that effects their entry into GALT is downregulated following infection of mice with Salmonella typhimurium. We further show that this downregulation is dependent on TLR signaling, and that the TLR-activated naive CD8+ T cells are blocked from entering GALT. This contrasts strongly with Ag-experienced effector T cells, for which TLR costimulation in the GALT potently upregulates α4β7 and enhances trafficking to intestinal tissues. Thus, TLR activation leads to opposite effects on migration of naive and effector CD8+ T cells. Our data identify a mechanism that excludes noncognate CD8+ T cells from selected immune compartments during TLR-induced systemic inflammation.
- Published
- 2013
29. Systemic Cancer Therapy with a Small Molecule Agonist of Toll-like Receptor 7 Can Be Improved by Circumventing TLR Tolerance
- Author
-
Christian Hotz, Cornelia Wurzenberger, Carole Bourquin, Bettina Storch, Nadja Sandholzer, Simon Heidegger, Andreas Voelkl, David Anz, Daniel Noerenberg, Stefan Endres, and Laurin C.M. Roetzer
- Subjects
Agonist ,Cancer Research ,medicine.drug_class ,medicine.medical_treatment ,Biology ,Mice ,Immune system ,Downregulation and upregulation ,Cancer immunotherapy ,Immune Tolerance ,medicine ,Animals ,Receptor ,Cells, Cultured ,Mice, Inbred BALB C ,Toll-like receptor ,Membrane Glycoproteins ,Interleukin-6 ,Carcinoma ,Imidazoles ,Interferon-alpha ,Interleukin ,Dendritic Cells ,TLR7 ,Interleukin-12 ,Mice, Inbred C57BL ,Interleukin-1 Receptor-Associated Kinases ,Toll-Like Receptor 7 ,Oncology ,Colonic Neoplasms ,Immunology ,Cytokines ,Female ,Tumor Escape ,Drug Screening Assays, Antitumor - Abstract
Topical application of small molecule Toll-like receptor 7 (TLR7) agonists is highly effective for the treatment of skin tumors, whereas their systemic application has been largely unsuccessful for cancer therapy. One reason may be that repeated systemic application of TLR ligands can induce a state of immune unresponsiveness, termed TLR tolerance. We show here that a single injection of the TLR7 agonist R848 in mice induces a short period of increased response to TLR stimulation followed by a state of hyporesponsiveness lasting several days. This state is characterized by inhibited secretion of the key cytokines interleukin (IL)-12p70 and IL-6 as well as by a block in IFN-α production. We show for the first time that at the cellular level, TLR7 tolerance occurs in both plasmacytoid and myeloid dendritic cells, two cell populations that play a critical role in the initiation and amplification of antitumor immune responses. We further show that TLR7 tolerance in plasmacytoid dendritic cells is accompanied by downregulation of the adaptor protein IL-1 receptor–associated kinase 1. On the basis of these findings, we have designed a novel strategy for the treatment of tumors by using cycles of repeated R848 injections separated by treatment-free intervals. We show in CT26 tumor-bearing mice that this protocol circumvents TLR7 tolerance and improves the efficacy of cancer immunotherapy. Cancer Res; 71(15); 5123–33. ©2011 AACR.
- Published
- 2011
30. Delivery of Immunostimulatory RNA Oligonucleotides by Gelatin Nanoparticles Triggers an Efficient Antitumoral Response
- Author
-
Nadja Sandholzer, Simon Heidegger, Sarah Weigel, Sebastian Fuchs, David Anz, Cornelia Wurzenberger, Gerhard Winter, Conrad Coester, Stefan Endres, and Carole Bourquin
- Subjects
Cancer Research ,Immunology ,Oligonucleotides ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Interferon-gamma ,Mice ,Drug Delivery Systems ,Immune system ,Gene expression ,Animals ,Immunology and Allergy ,Pharmacology ,Oligonucleotide ,RNA ,Dendritic Cells ,Neoplasms, Experimental ,Transfection ,TLR7 ,Interleukin-12 ,Molecular biology ,Immunity, Humoral ,Cell biology ,Mice, Inbred C57BL ,Toll-Like Receptor 7 ,Lipofectamine ,Nucleic acid ,Gelatin ,Nanoparticles ,Immunization - Abstract
RNA oligonucleotides have emerged as a new class of biologicals that can silence gene expression but also stimulate immune responses through specific pattern-recognition receptors. The development of effective delivery systems remains a major challenge for the therapeutic application of the RNA oligonucleotides. In this study, we have established a novel biodegradable carrier system that is highly effective for the delivery of immunostimulatory RNA oligonucleotides. Formulation of RNA oligonucleotides with cationized gelatin nanoparticles potentiates immune activation through the Toll-like receptor 7 (TLR7) in both myeloid and plasmacytoid dendritic cells. Further, nanoparticle-delivered RNA oligonucleotides trigger production of the antitumoral cytokines IL-12 and IFN-α. Binding to gelatin nanoparticles protects RNA oligonucleotides from degradation by nucleases, facilitates their uptake by dendritic cells, and targets these nucleic acids to the endosomal compartment in which they are recognized by TLR7. In these effects, the nanoparticles are superior to the conventional transfection reagents lipofectamine, polyethylenimine, and DOTAP. In vivo, the delivery of TLR7-activating RNA oligonucleotides by gelatin nanoparticles triggers antigen-specific CD8+ T-cell and antibody responses. Indeed, immunization with RNA-loaded nanoparticles leads to an efficient antitumoral immune response in two different mouse tumor models. Thus, gelatin-based nanoparticles represent a novel delivery system for immunostimulatory RNA oligonucleotides that is both effective and nontoxic.
- Published
- 2010
31. Role of melanoma cell-intrinsic RIG-I and STING signaling for checkpoint inhibitor-mediated anticancer immunity
- Author
-
Alexander Wintges, Simon Heidegger, Sarah Bek, Paul-Albert Koenig, Hendrik Poeck, Julius C. Fischer, Martina Schmickl, and Tobias Haas
- Subjects
Anticancer immunity ,Cancer Research ,RIG-I ,business.industry ,Immune checkpoint inhibitors ,Melanoma ,Cell ,chemical and pharmacologic phenomena ,medicine.disease ,Sting ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,business - Abstract
3081Background: Strong inter-individual variation in clinical response to immune checkpoint inhibitors (ICB) including anti-CTLA-4 remains a major challenge, but the molecular pathways that modulat...
- Published
- 2018
32. Tumor-Intrinsic RIG-I Signaling Promotes Anti-CTLA-4 Checkpoint Inhibitor-Mediated Anticancer Immunity
- Author
-
Martina Schmickl, Simon Heidegger, Tobias Haas, Christian Peschel, Alexander Wintges, Sarah Bek, Hendrik Poeck, Marcel R.M. van den Brink, and Jürgen Ruland
- Subjects
Cell cycle checkpoint ,RIG-I ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Cell Biology ,Hematology ,Immunotherapy ,Biology ,Biochemistry ,Immune system ,Antigen ,Interferon ,medicine ,Cancer research ,Signal transduction ,Interferon type I ,medicine.drug - Abstract
Strong inter-individual variation in clinical response to checkpoint inhibitors such as anti-CTLA-4 remains a major challenge. In a retrospective analysis, expression of viral defense genes in human melanomas including the cytosolic RNA receptor RIG-I (DDx58) has been associated with clinical benefit to CTLA-4 blockade. However, possible interconnections of these two distinct pathways remain unknown. Using CRISPR/Cas9 technology to generate B16 melanoma cells lines that lack nucleic acid receptors or downstream signaling molecules (RIG-I, STING, IRF3/7) together with available genetically deficient mouse models, we addressed the importance of nucleic acid receptor signaling in both tumor and host cells for the efficacy of anti-CTLA-4 immunotherapy. We here provide experimental proof that anti-CTLA-4 immunotherapy relies on tumor cell-intrinsic RIG-I but not STING signaling. Following anti-CTLA-4 treatment, tumor-intrinsic RIG-I signaling critically impacts on cross-presentation of tumor-associated antigen by CD103+ dendritic cells, the expansion of tumor antigen-specific CD8+ T cells and ultimately the accumulation of CD8+ T cells within tumor tissue. Consistently, therapeutic targeting of RIG-I with 5'-phosphorylated-RNA in both tumor and non-malignant cells potently augmented the efficacy of CTLA-4 checkpoint blockade. These processes were additionally dependent on host STING, MAVS and type I interferon signaling and were closely linked to RIG-I-mediated tumor cell death. In summary, our study identifies both tumor- and host-intrinsic RIG-I signaling as fundamental requirements for anti-CTLA-4-mediated antitumor immunity. Our data predict that targeting RIG-I may serve as a basis for the development of new combination strategies to increase the response rate of checkpoint inhibitor-based immunotherapy of malignancy including lymphoma, particularly in individuals that do not have a sufficient spontaneous antitumor T-cell immune response. Disclosures van den Brink: Jazz Pharmaceuticals: Consultancy; Seres: Research Funding; PureTech Health: Consultancy; Therakos Institute: Other: Speaking engagement.
- Published
- 2017
33. Mycoplasma hyorhinis-Contaminated Cell Lines Activate Primary Innate Immune Cells via a Protease-Sensitive Factor
- Author
-
Christian Hotz, Carole Bourquin, Stefan Endres, Simon Heidegger, Alexander Jarosch, and Martina Schmickl
- Subjects
Mycoplasma hyorhinis ,Cell ,lcsh:Medicine ,medicine.disease_cause ,Microbiology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Mycoplasma Infections ,lcsh:Science ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Innate immune system ,biology ,Interleukin-6 ,lcsh:R ,Interferon-alpha ,Mycoplasma ,Dendritic Cells ,biology.organism_classification ,Coculture Techniques ,Immunity, Innate ,Toll-Like Receptor 2 ,3. Good health ,ddc ,Mice, Inbred C57BL ,TLR2 ,medicine.anatomical_structure ,Cell culture ,Myeloid Differentiation Factor 88 ,Female ,lcsh:Q ,030215 immunology ,Research Article - Abstract
Mycoplasma are a frequent and occult contaminant of cell cultures, whereby these prokaryotic organisms can modify many aspects of cell physiology, rendering experiments that are conducted with such contaminated cells problematic. Chronic Mycoplasma contamination in human monocytic cells lines has been associated with suppressed Toll-like receptor (TLR) function. In contrast, we show here that components derived from a Mycoplasma hyorhinis-infected cell line can activate innate immunity in non-infected primary immune cells. Release of pro-inflammatory cytokines such as IL-6 by dendritic cells in response to Mycoplasma hyorhinis-infected cell components was critically dependent on the adapter protein MyD88 but only partially on TLR2. Unlike canonical TLR2 signaling that is triggered in response to the detection of Mycoplasma infection, innate immune activation by components of Mycoplasma-infected cells was inhibited by chloroquine treatment and sensitive to protease treatment. We further show that in plasmacytoid dendritic cells, soluble factors from Mycoplasma hyorhinis-infected cells induce the production of large amounts of IFN-α. We conclude that Mycoplasma hyorhinis-infected cell lines release protein factors that can potently activate co-cultured innate immune cells via a previously unrecognized mechanism, thus limiting the validity of such co-culture experiments.
- Published
- 2015
34. The Role of Pattern-Recognition Receptors in Graft-Versus-Host Disease and Graft-Versus-Leukemia after Allogeneic Stem Cell Transplantation
- Author
-
Hendrik Poeck, Marcel R.M. van den Brink, Simon Heidegger, and Tobias Haas
- Subjects
lcsh:Immunologic diseases. Allergy ,Mini Review ,T cell ,medicine.medical_treatment ,Immunology ,danger molecules ,inflammsome ,Hematopoietic stem cell transplantation ,pattern-recognition receptors ,immune system diseases ,allogenic hematopoietic stem cell transplantation ,graft-versus-host disease ,microbiota ,medicine ,Immunology and Allergy ,Innate immune system ,business.industry ,Pattern recognition receptor ,medicine.disease ,Transplantation ,surgical procedures, operative ,medicine.anatomical_structure ,Graft-versus-host disease ,Stem cell ,lcsh:RC581-607 ,business ,Cytokine storm - Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only treatment with curative potential for certain aggressive hematopoietic malignancies. Its success is limited by acute graft-versus-host disease (GVHD), a life-threatening complication that occurs when allo-reactive donor T cells attack recipient organs. There is growing evidence that microbes and innate pattern-recognition receptors (PRRs) such as toll-like receptors (TLR) and nod-like receptors (NLR) are critically involved in the pathogenesis of acute GVHD. Currently, a widely accepted model postulates that intensive chemotherapy and/or total-body irradiation during pre-transplant conditioning results in tissue damage and a loss of epithelial barrier function. Subsequent translocation of bacterial components as well as release of endogenous danger molecules stimulate PRRs of host antigen-presenting cells to trigger the production of pro-inflammatory cytokines (cytokine storm) that modulate T cell allo-reactivity against host tissues, but eventually also the beneficial graft-versus-leukemia (GVL) effect. Given the limitations of existing immunosuppressive therapies, a better understanding of the molecular mechanisms that govern GVHD versus GVL is urgently needed. This may ultimately allow to design modulators, which protect from GvHD but preserve donor T-cell attack on hematologic malignancies. Here, we will briefly summarize current knowledge about the role of innate immunity in the pathogenesis of GVHD and GVL following allo-HSCT.
- Published
- 2014
35. Abstract A005: Tumor- and host-intrinsic RIG-I signaling promote anticancer immunity by CTLA-4 blockade
- Author
-
Diana Kreppel, Martina Schmickl, Michael Bscheider, Alexander Wintges, Marcel R.M. van den Brink, Simon Heidegger, Hendrik Poeck, Sarah Bek, Tobias Haas, and Christian Peschel
- Subjects
Cancer Research ,RIG-I ,medicine.medical_treatment ,Immunology ,Cancer ,chemical and pharmacologic phenomena ,Immunotherapy ,Biology ,medicine.disease ,Blockade ,Cancer immunotherapy ,CTLA-4 ,medicine ,Immunogenic cell death ,CD8 - Abstract
Introduction: Targeting inhibitory T-cell receptors such as CTLA-4 has shown great promise in cancer therapy. However, strong inter-individual variation in clinical response to checkpoint inhibitors remains a major challenge. Expression of viral defense genes in human melanomas including the cytosolic RNA receptor RIG-I (DDx58) has recently been associated with clinical benefit to CTLA-4 blockade. Methods: Using CRISPR/Cas9 technology to generate B16 melanoma cells lines that lack nucleic acid receptors or downstream signaling molecules (RIG-I, STING, IRF3/7) together with available genetically deficient mouse models, we address the importance of nucleic acid receptor signaling for the efficacy of anti-CTLA-4 immunotherapy. Results: We provide experimental proof that anti-CTLA-4 immunotherapy indeed relies on tumor cell-intrinsic RIG-I/MAVS but not cGAS/STING signaling. Consistently, therapeutic targeting of RIG-I with the specific ligand 5'‑triphosphate-RNA (3pRNA) potently augments the efficacy of CTLA-4 checkpoint blockade. In situ vaccination by intratumoral 3pRNA injection enhanced cross-presentation of tumor-associated antigens by CD103+ migratory DCs in local draining lymph nodes. Subsequent expansion of tumor-specific CD8+ T cells resulted in control of local and distant tumors, otherwise resistant to anti-CTLA‑4 monotherapy. These processes were critically dependent on tumor cell-intrinsic RIG-I-mediated immunogenic cell death (ICD), but not tumor-derived type I IFN release. In such, the synergistic therapeutic effect of RIG-I activation and anti-CTLA-4 was restricted to tumor models that showed susceptibility to RIG‑I-mediated ICD. Furthermore, systemic antitumor immunity following anti-CTLA-4 +/- 3pRNA treatment required RIG-I signaling in both tumor and non-malignant host cells. Conclusion: Our study identifies a hitherto unrecognized role of RIG-I signaling in tumors and their microenvironment as a crucial component for checkpoint inhibitor-mediated immunotherapy of cancer. Therapeutic targeting of RIG-I using specific agonists strongly augments the efficacy of anti-CTLA-4 blockade and may thus serve as the basis for new combinatorial approaches. Citation Format: Simon Heidegger, Alexander Wintges, Diana Kreppel, Sarah Bek, Michael Bscheider, Martina Schmickl, Marcel R.M. van den Brink, Christian Peschel, Tobias Haas, Hendrik Poeck. Tumor- and host-intrinsic RIG-I signaling promote anticancer immunity by CTLA-4 blockade [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr A005.
- Published
- 2016
36. The RIG-I Agonist 3pRNA Synergizes with Checkpoint Blockade in Cancer Immunotherapy
- Author
-
Christian Peschel, Martina Schmickl, Diana Kreppel, Michael Bscheider, Simon Heidegger, Jürgen Ruland, Tobias Haas, Marcel R.M. van den Brink, Alexander Wintges, Sarah Bek, and Hendrik Poeck
- Subjects
Regulatory T cell ,medicine.medical_treatment ,Immunology ,Pattern recognition receptor ,chemical and pharmacologic phenomena ,Cell Biology ,Hematology ,Biology ,Biochemistry ,medicine.anatomical_structure ,Immune system ,Antigen ,Cancer immunotherapy ,Interferon ,medicine ,Cytotoxic T cell ,CD8 ,medicine.drug - Abstract
Antibody-mediated targeting of regulatory T cell receptors such as CTLA-4 has been shown to enhance anti-tumor immune responses against several cancer entities including malignant melanoma. Yet, therapeutic success in patients remains variable underscoring the need for novel combinatorial approaches. Here we established a vaccination protocol that combines selective engagement of the nucleic acid-sensing pattern recognition receptor RIG-I, antigen and CTLA-4-blockade. We found that vaccination together with RIG-I ligation strongly synergized with CTLA-4 blockade to induce expansion and activation of antigen-specific CD8+ T cells and potent anti-tumor immunity. Cross-priming of cytotoxic T cells as well as anti-tumor immunity required the adapter protein MAVS and type I interferon (IFN) signaling and were mediated by dendritic cells. In addition, the benefit of the combined immunization with anti-CTLA-4 was reduced by systemic antibiotics pointing to the requisite of an intact commensal microbiota in this context. Together, our findings describe a novel combinatorial strategy that may form the basis for the design of new type I IFN-based regimens that enhance antigen-specific T cell reactivity against cancer. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
37. RIG-I-Induced Type I IFNs Promote Regeneration of the Intestinal Stem Cell Compartment during Acute Tissue Damage
- Author
-
Caroline A. Lindemans, Michael Bscheider, Alan M. Hanash, Julius C. Fischer, Sebastien Monette, Robert R. Jenq, Enrico Velardi, Melissa D. Docampo, Gabriel Eisenkolb, Simon Heidegger, Alexander Wintges, Marco Calafiore, Jarrod A Dudakov, Hendrik Poeck, Kori A. Porosnicu Rodriguez, Tobias Haas, Sophie Lieberman, Chen Liu, Marcel R.M. van den Brink, and Christian Peschel
- Subjects
RIG-I ,Regeneration (biology) ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Total body irradiation ,Biology ,medicine.disease ,Biochemistry ,Epithelium ,medicine.anatomical_structure ,Graft-versus-host disease ,Cytokine ,Cancer research ,medicine ,Stem cell - Abstract
Introduction: Although the role of Type I IFNs in initiating host defense against pathogens is well established, recent work highlights the regenerative function of this cytokine family. Yet, despite its involvement in tissue repair, the cellular targets and mechanisms of action by which Type I IFNs act during tissue regeneration are poorly understood. Results: Here, we describe a hitherto unrecognized regenerative function for the RIG-I/MAVS/IFN-I pathway through direct effects on epithelial regeneration. Mice deficient in the RIG-I adaptor MAVS were more sensitive to intestinal barrier damage after total body irradiation (TBI) and, like RIG-I deficient mice, developed worse graft-versus-host disease (GVHD) in a preclinical model for allogeneic hematopoietic stem cell transplantation (allo-HSCT). This phenotype was not associated with changes in the intestinal microbiota, but with a defect in epithelial regeneration. Moreover, in contrast to previous reports in steady-state conditions and after viral challenge, we found that interferon-α/β receptor (IFNAR) signaling in non-hematopoietic epithelial cells was crucial for tissue regeneration after acute damage. Importantly, we could demonstrate that this pathway could be therapeutically targeted with RIG-I agonists, which promoted barrier integrity and prevented GVHD. Mechanistically, Type I IFNs (either RIG-I-induced or recombinant) could promote intestinal stem cell (ISC) growth in crypt organoid cultures, suggesting that stimulation of the ISC compartment led to epithelial regeneration. Conclusion: Our findings suggest that activation of RIG-I and IFN-I can promote regeneration of intestinal epithelial cells and thus offers an innovative therapeutic strategy for the management of acute intestinal injury. Disclosures van den Brink: Merck: Honoraria; Boehringer Ingelheim: Consultancy, Other: Advisory board attendee; Regeneron: Honoraria; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Tobira Therapeutics: Other: Advisory board attendee.
- Published
- 2015
38. PS2-023. Following TLR Activation Naive CD8 T Cells Are Excluded From Gut-Associated Lymphoid Tissue In An IL-6-Dependent Manner
- Author
-
Nina Suhartha, Stefan Endres, Simon Heidegger, Bernadette Bohn, Sophie-Kathrin Kirchner, Nicolas Stephan, and Carole Bourquin
- Subjects
Dependent manner ,Gut-associated lymphoid tissue ,Immunology ,Innate lymphoid cell ,Hematology ,Biology ,Biochemistry ,medicine.anatomical_structure ,biology.protein ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Interleukin 6 ,Molecular Biology - Published
- 2011
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.