Percy A. Knolle
Technische Universität München
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Percy A. Knolle.
Hepatology | 2011
Christoph Roderburg; Gerd‐Willem Urban; Kira Bettermann; Mihael Vucur; Henning W. Zimmermann; Sabine Schmidt; Jörn Janssen; Christiane Koppe; Percy A. Knolle; Mirco Castoldi; Frank Tacke; Christian Trautwein; Tom Luedde
Liver fibrosis is orchestrated by a complex network of signaling pathways regulating the deposition of extracellular matrix proteins during fibrogenesis. MicroRNAs (miRNAs) represent a family of small noncoding RNAs controlling translation and transcription of many genes. Recently, miRNAs have been suggested to crucially modulate cellular processes in the liver such as hepatocarcinogenesis. However, their role in liver fibrosis is not well understood. We systematically analyzed the regulation of miRNAs in a mouse model of carbon tetrachloride–induced hepatic fibrogenesis (CCl4) by gene array analysis, which revealed a panel of miRNA that were specifically regulated in livers of mice undergoing hepatic fibrosis. Within those, all three members of the miR‐29‐family were significantly down‐regulated in livers of CCl4‐treated mice as well as in mice that underwent bile duct ligation. Specific regulation of miR‐29 members in murine fibrosis models correlated with lower expression of miR‐29 in livers from patients with advanced liver fibrosis. Moreover, patients with advanced liver cirrhosis showed significantly lower levels of miR‐29a in their serum when compared with healthy controls or patients with early fibrosis. On a cellular level, down‐regulation of miR‐29 in murine hepatic stellate cells (HSCs) was mediated by transforming growth factor beta (TGF‐β) as well as inflammatory signals, namely, lipopolysaccharide (LPS) and nuclear factor kappa B (NF‐κB). Furthermore, overexpression of miR‐29b in murine HSC resulted in down‐regulation of collagen expression. Conclusion: Our data indicate that miR‐29 mediates the regulation of liver fibrosis and is part of a signaling nexus involving TGF‐β‐ and NF‐κB–dependent down‐regulation of miR‐29 family members in HSC with subsequent up‐regulation of extracellular matrix genes. Thus they may represent targets for novel therapeutic strategies against hepatic fibrogenesis and also might evolve as biomarkers in the diagnosis of liver fibrosis. (HEPATOLOGY 2011.)
Nature Reviews Immunology | 2010
Angus W. Thomson; Percy A. Knolle
The demands that are imposed on the liver as a result of its function as a metabolic organ that extracts nutrients and clears gut-derived microbial products from the blood are met by a unique microanatomical and immunological environment. The inherent tolerogenicity of the liver and its role in the regulation of innate and adaptive immunity are mediated by parenchymal and non-parenchymal antigen-presenting cells (APCs), cell-autonomous molecular pathways and locally produced factors. Here, we review the central role of liver APCs in the regulation of hepatic immune function and also consider how recent insights may be applied in strategies to target liver tolerance for disease therapy.
Nature Reviews Immunology | 2010
Christian Kurts; Bruce W. S. Robinson; Percy A. Knolle
Cross-priming is an important mechanism to activate cytotoxic T lymphocytes (CTLs) for immune defence against viruses and tumours. Although it was discovered more than 25 years ago, we have only recently gained insight into the underlying cellular and molecular mechanisms, and we are just beginning to understand its physiological importance in health and disease. Here we summarize current concepts on the cross-talk between the immune cells involved in CTL cross-priming and on its role in antimicrobial and antitumour defence, as well as in immune-mediated diseases.
Nature Reviews Immunology | 2012
Ulrike Protzer; Mala K. Maini; Percy A. Knolle
The liver has vital metabolic and clearance functions that involve the uptake of nutrients, waste products and pathogens from the blood. In addition, its unique immunoregulatory functions mediated by local expression of co-inhibitory receptors and immunosuppressive mediators help to prevent inadvertent organ damage. However, these tolerogenic properties render the liver an attractive target site for pathogens. Although most pathogens that reach the liver via the blood are eliminated or controlled by local innate and adaptive immune responses, some pathogens (such as hepatitis viruses) can escape immune control and persist in hepatocytes, causing substantial morbidity and mortality worldwide. Here, we review our current knowledge of the mechanisms of liver targeting by pathogens and describe the interplay between pathogens and host factors that promote pathogen elimination and maintain organ integrity or that allow pathogen persistence.
Journal of Experimental Medicine | 2009
Luisa Klotz; Sven Burgdorf; Indra Dani; Kaoru Saijo; Juliane Flossdorf; Stephanie Hucke; Judith Alferink; Natalija Novak; Marc Beyer; Günter Mayer; Birgit Langhans; Thomas Klockgether; Ari Waisman; Gérard Eberl; Joachim L. Schultze; Michael Famulok; Waldemar Kolanus; Christopher K. Glass; Christian Kurts; Percy A. Knolle
T helper cells secreting interleukin (IL)-17 (Th17 cells) play a crucial role in autoimmune diseases like multiple sclerosis (MS). Th17 differentiation, which is induced by a combination of transforming growth factor (TGF)-β/IL-6 or IL-21, requires expression of the transcription factor retinoic acid receptor–related orphan receptor γt (RORγt). We identify the nuclear receptor peroxisome proliferator–activated receptor γ (PPARγ) as a key negative regulator of human and mouse Th17 differentiation. PPARγ activation in CD4+ T cells selectively suppressed Th17 differentiation, but not differentiation into Th1, Th2, or regulatory T cells. Control of Th17 differentiation by PPARγ involved inhibition of TGF-β/IL-6–induced expression of RORγt in T cells. Pharmacologic activation of PPARγ prevented removal of the silencing mediator for retinoid and thyroid hormone receptors corepressor from the RORγt promoter in T cells, thus interfering with RORγt transcription. Both T cell–specific PPARγ knockout and endogenous ligand activation revealed the physiological role of PPARγ for continuous T cell–intrinsic control of Th17 differentiation and development of autoimmunity. Importantly, human CD4+ T cells from healthy controls and MS patients were strongly susceptible to PPARγ-mediated suppression of Th17 differentiation. In summary, we report a PPARγ-mediated T cell–intrinsic molecular mechanism that selectively controls Th17 differentiation in mice and in humans and that is amenable to pharmacologic modulation. We therefore propose that PPARγ represents a promising molecular target for specific immunointervention in Th17-mediated autoimmune diseases such as MS.
Journal of Immunology | 2012
Franz Bauernfeind; Anna Rieger; Frank A. Schildberg; Percy A. Knolle; Jonathan L. Schmid-Burgk; Veit Hornung
Inflammasomes are multiprotein signaling platforms that form upon sensing microbe- or damage-associated molecular patterns. Upon their formation, caspase-1 is activated, leading to the processing of certain proinflammatory cytokines and the initiation of a special type of cell death, known as pyroptosis. Among known inflammasomes, NLRP3 takes on special importance because it appears to be a general sensor of cell stress. Moreover, unlike other inflammasome sensors, NLRP3 inflammasome activity is under additional transcriptional regulation. In this study, we identify the myeloid-specific microRNA miR-223 as another critical regulator of NLRP3 inflammasome activity. miR-223 suppresses NLRP3 expression through a conserved binding site within the 3′ untranslated region of NLRP3, translating to reduced NLRP3 inflammasome activity. Although miR-223 itself is not regulated by proinflammatory signals, its expression varies among different myeloid cell types. Therefore, given the tight transcriptional control of NLRP3 message itself, miR-223 functions as an important rheostat controlling NLRP3 inflammasome activity.
European Journal of Immunology | 2006
Gerhard Wingender; Natalio Garbi; Beatrix Schumak; Frank Jüngerkes; Elmar Endl; Dagmar von Bubnoff; Julia Steitz; Jörg Striegler; Gerd Moldenhauer; Thomas Tüting; Antje Heit; Katharina M. Huster; Osamu Takikawa; Shizuo Akira; Dirk H. Busch; Hermann Wagner; Günter J. Hämmerling; Percy A. Knolle; Andreas Limmer
CpG‐rich oligonucleotides (CpG‐ODN) bind to Toll‐like receptor 9 (TLR9) and are used as powerful adjuvants for vaccination. Here we report that CpG‐ODN not only act as immune stimulatory agents but can also induce strong immune suppression depending on the anatomical location of application. In agreement with the adjuvant effect, subcutaneous application of antigen plus CpG‐ODN resulted in antigen‐specific T cell activation in local lymph nodes. In contrast, systemic application of CpG‐ODN resulted in suppression of T cell expansion and CTL activity in the spleen. The suppressive effect was mediated by indoleamine 2,3‐dioxygenase (IDO) as indicated by the observation that CpG‐ODN induced IDO in the spleen and that T cell suppression could be abrogated by 1‐methyl‐tryptophan (1‐MT), an inhibitor of IDO. No expression of IDO was observed in lymph nodes after injection of CpG‐ODN, explaining why suppression was restricted to the spleen. Studies with a set of knockout mice demonstrated that the CpG‐ODN‐induced immune suppression is dependent on TLR9 stimulation and independent of type I and type II interferons. The present study shows that for the use of CpG‐ODN as an adjuvant in vaccines, the route of application is crucial and needs to be considered. In addition, the results indicate that down‐modulation of immune responses by CpG‐ODN may be possible in certain pathological conditions.
Hepatology | 2007
Linda Diehl; Anna Schurich; Regina Grochtmann; Silke Hegenbarth; Lieping Chen; Percy A. Knolle
Liver sinusoidal endothelial cells (LSEC) are unique organ‐resident antigen‐presenting cells capable of cross‐presentation and subsequent tolerization of naïve CD8+ T cells. We investigated the molecular mechanisms underlying this tolerance induction in naive CD8+ T cells. MHC class I–restricted antigen presentation by LSEC led to initial stimulation of naïve CD8+ T cells, which up‐regulated CD69, CD25, CD44, and programmed death (PD)‐1 and proliferated similar to dendritic cell (DC)–activated CD8+ T cells. Importantly, cognate interaction with naïve CD8+ T cells triggered increased expression of co‐inhibitory B7‐H1 but not co‐stimulatory CD80/86 molecules exclusively on LSEC but not DC. This matured phenotype of B7‐H1high CD80/86low was critical for induction of CD8+ T cell tolerance by LSEC: B7‐H1–deficient LSEC, that failed to interact with PD‐1 on stimulated T cells, were incapable of inducing CD8+ T cell tolerance. Moreover, increased costimulation via CD28 interfered with tolerance induction, indicating that the noninducible low expression levels of CD80/86 on LSEC supported B7‐H1–dependent tolerance induction. LSEC‐tolerized CD8+ T cells had a distinctive phenotype from naïve and activated T cells with CD25low, CD44high, CD62Lhigh. They also expressed the homeostatic cytokine receptors CD127, CD122, and high levels of Bcl‐2, indicating survival rather than deletion of tolerant CD8+ T cells. On adoptive transfer into congenic animals, tolerized CD8+ T cells failed to show specific cytotoxicity in vivo. Conclusion: Cognate interaction of LSEC with naïve CD8+ T cells elicits a unique tolerogenic maturation of LSEC and permissiveness of T cells for tolerogenic signals, demonstrating that LSEC‐induced tolerance is an active and dynamic process. (HEPATOLOGY 2007.)
Nature Immunology | 2014
Dominic De Nardo; Larisa I. Labzin; Hajime Kono; Reiko Seki; Susanne Schmidt; Marc Beyer; Dakang Xu; Sebastian Zimmer; Catharina Lahrmann; Frank A. Schildberg; Johanna Vogelhuber; Michael Kraut; Thomas Ulas; Anja Kerksiek; Wolfgang Krebs; Niklas Bode; Alena Grebe; Michael L. Fitzgerald; Nicholas J. Hernandez; Bryan R. G. Williams; Percy A. Knolle; Manfred Kneilling; Martin Röcken; Dieter Lütjohann; Samuel D. Wright; Joachim L. Schultze; Eicke Latz
High-density lipoprotein (HDL) mediates reverse cholesterol transport and is known to be protective against atherosclerosis. In addition, HDL has potent anti-inflammatory properties that may be critical for protection against other inflammatory diseases. The molecular mechanisms of how HDL can modulate inflammation, particularly in immune cells such as macrophages, remain poorly understood. Here we identify the transcriptional regulator ATF3, as an HDL-inducible target gene in macrophages that downregulates the expression of Toll-like receptor (TLR)-induced proinflammatory cytokines. The protective effects of HDL against TLR-induced inflammation were fully dependent on ATF3 in vitro and in vivo. Our findings may explain the broad anti-inflammatory and metabolic actions of HDL and provide the basis for predicting the success of new HDL-based therapies.
Gut | 2015
Mirjam B. Zeisel; Julie Lucifora; William S. Mason; Camille Sureau; Jürgen Beck; Massimo Levrero; Michael Kann; Percy A. Knolle; Monsef Benkirane; David Durantel; Marie Michel; Brigitte Autran; François-Loïc Cosset; Helene Strick-Marchand; Christian Trepo; Jia-Horng Kao; Fabrice Carrat; Karine Lacombe; Raymond F. Schinazi; Françoise Barré-Sinoussi; Jean François Delfraissy; Fabien Zoulim
HBV infection is a major cause of liver cirrhosis and hepatocellular carcinoma. Although HBV infection can be efficiently prevented by vaccination, and treatments are available, to date there is no reliable cure for the >240 million individuals that are chronically infected worldwide. Current treatments can only achieve viral suppression, and lifelong therapy is needed in the majority of infected persons. In the framework of the French National Agency for Research on AIDS and Viral Hepatitis ‘HBV Cure’ programme, a scientific workshop was held in Paris in June 2014 to define the state-of-the-art and unanswered questions regarding HBV pathobiology, and to develop a concerted strategy towards an HBV cure. This review summarises our current understanding of HBV host-interactions leading to viral persistence, as well as the roadblocks to be overcome to ultimately address unmet medical needs in the treatment of chronic HBV infection.