Frank A. Schildberg
Harvard University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Frank A. Schildberg.
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.
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.
Nature Immunology | 2014
Viviana Cremasco; Matthew Woodruff; Lucas Onder; Jovana Cupovic; Janice M. Nieves-Bonilla; Frank A. Schildberg; Jonathan L. Chang; Floriana Cremasco; Christopher J. Harvey; Kai W. Wucherpfennig; Burkhard Ludewig; Michael C. Carroll; Shannon J. Turley
Fibroblastic reticular cells (FRCs) are known to inhabit T cell–rich areas of lymphoid organs, where they function to facilitate interactions between T cells and dendritic cells. However, in vivo manipulation of FRCs has been limited by a dearth of genetic tools that target this lineage. Here, using a mouse model to conditionally ablate FRCs, we demonstrated their indispensable role in antiviral T cell responses. Unexpectedly, loss of FRCs also attenuated humoral immunity due to impaired B cell viability and follicular organization. Follicle-resident FRCs established a favorable niche for B lymphocytes via production of the cytokine BAFF. Thus, our study indicates that adaptive immunity requires an intact FRC network and identifies a subset of FRCs that control B cell homeostasis and follicle identity.
European Journal of Immunology | 2008
Frank A. Schildberg; Silke Hegenbarth; Beatrix Schumak; Andreas Limmer; Percy A. Knolle
The liver is known to induce tolerance rather than immunity through tolerogenic antigen presentation or elimination of effector T cells. In particular, hepatic dendritic cells (DC) are known to be little immunogenic for CD8 T cells. Here, we investigated whether this peculiar phenotype resulted from interaction with resident hepatic cell populations. Contact of DC with liver sinusoidal endothelial cells (LSEC) but not hepatocytes or B cells vetoed antigen‐presenting DC to fully activate naive CD8 T cells. This MHC‐independent regulatory effect of LSEC on DC function was not connected to soluble mediators but required physical contact. Because interaction with third‐party LSEC still allowed antigen‐presenting DC to stimulate expression of initial activation markers on naive CD8 T cells and to stimulate activated CD8 T cells, we hypothesize that LSEC controlled the DC costimulatory function. Indeed, contact with LSEC led to reduced DC expression levels of CD80/86 or IL‐12, but supplementation of these signals failed to rescue the ability to prime naive CD8 T cells, indicating involvement of further molecules. Taken together, our results reveal a novel principle operative in hepatic tolerance induction, in which LSEC not only tolerize T cells themselves but also suppress neighboring APC normally capable of inducing T cell immunity.
Journal of Hepatology | 2013
Bastian Höchst; Frank A. Schildberg; Pia Sauerborn; Yvonne A. Gäbel; Heidrun Gevensleben; Diane Goltz; Lukas C. Heukamp; Andreas Türler; Matthias Ballmaier; Friederike Gieseke; Ingo Müller; Jörg C. Kalff; Christian Kurts; Percy A. Knolle; Linda Diehl
BACKGROUND & AIMS Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells associated with the suppression of immunity. However, little is known about how or where MDSCs are induced and from which cells they originate. The liver is known for its immune regulatory functions. Here, we investigated the capacity of human hepatic stellate cells (HSCs) to transform peripheral blood monocytes into MDSCs. METHODS We cultured freshly isolated human monocytes from healthy donors on primary human HSCs or an HSC cell-line and characterized the phenotype and function of resulting CD14(+)HLA-DR(-/low) monocytes by flow cytometry, quantitative PCR, and functional assays. We analyzed the molecular mechanisms underlying the induction and function of the CD14(+)HLA-DR(-/low) cells by using blocking antibodies or knock-down technology. RESULTS Mature peripheral blood monocytes co-cultured with HSCs downregulated HLA-DR and developed a phenotypic and functional profile similar to MDSCs. Only activated but not freshly isolated HSCs were capable of inducing CD14(+)HLA-DR(-/low) cells. Such CD14(+)HLA-DR(-/low) monocyte-derived MDSCs suppressed T-cell proliferation in an arginase-1 dependent fashion. HSC-induced development of CD14(+)HLA-DR(-/low) monocyte-derived MDSCs was not mediated by soluble factors, but required physical interaction and was abrogated by blocking CD44. CONCLUSIONS Our study shows that activated human HSCs convert mature peripheral blood monocytes into MDSCs. As HSCs are activated during chronic inflammation, the subsequent local induction of MDSCs may prevent ensuing excessive liver injury. HSC-induced MDSCs functionally and phenotypically resemble those isolated from liver cancer patients. Thus, our data suggest that local generation of MDSCs by liver-resident HSCs may contribute to immune suppression during inflammation and cancer in the liver.
Hepatology | 2011
Frank A. Schildberg; Alexandra Wojtalla; Sören V. Siegmund; Elmar Endl; Linda Diehl; Zeinab Abdullah; Christian Kurts; Percy A. Knolle
The liver has a role in T cell tolerance induction, which is mainly achieved through the functions of tolerogenic hepatic antigen‐presenting cells (APCs) and regulatory T cells. Hepatic stellate cells (HSCs) are known to have various immune functions, which range from immunogenic antigen presentation to the induction of T cell apoptosis. Here we report a novel role for stellate cells in vetoing the priming of naive CD8 T cells. Murine and human HSCs and stromal cells (but not hepatocytes) prevented the activation of naive T cells by dendritic cells, artificial APCs, and phorbol 12‐myristate 13‐acetate/ionomycin by a cell contact–dependent mechanism. The veto function for inhibiting T cell activation was directly correlated with the activation state of HSCs and was most pronounced in HSCs from fibrotic livers. Mechanistically, high expression levels of CD54 simultaneously restricted the expression of interleukin‐2 (IL‐2) receptor and IL‐2 in T cells, and this was responsible for the inhibitory effect because exogenous IL‐2 overcame the HSC veto function. Conclusion: Our results demonstrate a novel function of HSCs in the local skewing of immune responses in the liver through the prevention of local stimulation of naive T cells. These results not only indicate a beneficial role in hepatic fibrosis, for which increased CD54 expression on HSCs could attenuate further T cell activation, but also identify IL‐2 as a key cytokine in mediating local T cell immunity to overcome hepatic tolerance. (HEPATOLOGY 2011;)
Hepatology | 2014
M Granzow; Robert Schierwagen; Sabine Klein; Benita Kowallick; Sebastian Huss; Markus Linhart; Irela Gretchen Reza Mazar; Jan Görtzen; A. Vogt; Frank A. Schildberg; Maria A. Gonzalez-Carmona; Alexandra Wojtalla; B Krämer; Jacob Nattermann; Sören V. Siegmund; Nikos Werner; Dieter O. Fürst; Wim Laleman; Percy A. Knolle; Vijay H. Shah; Tilman Sauerbruch; Jonel Trebicka
Activation of the renin angiotensin system resulting in stimulation of angiotensin‐II (AngII) type I receptor (AT1R) is an important factor in the development of liver fibrosis. Here, we investigated the role of Janus kinase 2 (JAK2) as a newly described intracellular effector of AT1R in mediating liver fibrosis. Fibrotic liver samples from rodents and humans were compared to respective controls. Transcription, protein expression, activation, and localization of JAK2 and downstream effectors were analyzed by real‐time polymerase chain reaction, western blotting, immunohistochemistry, and confocal microscopy. Experimental fibrosis was induced by bile duct ligation (BDL), CCl4 intoxication, thioacetamide intoxication or continuous AngII infusion. JAK2 was inhibited by AG490. In vitro experiments were performed with primary rodent hepatic stellate cells (HSCs), Kupffer cells (KCs), and hepatocytes as well as primary human and human‐derived LX2 cells. JAK2 expression and activity were increased in experimental rodent and human liver fibrosis, specifically in myofibroblastic HSCs. AT1R stimulation in wild‐type animals led to activation of HSCs and fibrosis in vivo through phosphorylation of JAK2 and subsequent RhoA/Rho‐kinase activation. These effects were prevented in AT1R−/− mice. Pharmacological inhibition of JAK2 attenuated liver fibrosis in rodent fibrosis models. In vitro, JAK2 and downstream effectors showed increased expression and activation in activated HSCs, when compared to quiescent HSCs, KCs, and hepatocytes isolated from rodents. In primary human and LX2 cells, AG490 blocked AngII‐induced profibrotic gene expression. Overexpression of JAK2 led to increased profibrotic gene expression in LX2 cells, which was blocked by AG490. Conclusion: Our study substantiates the important cell‐intrinsic role of JAK2 in HSCs for development of liver fibrosis. Inhibition of JAK2 might therefore offer a promising therapy for liver fibrosis. (Hepatology 2014;60:334–348)
PLOS Pathogens | 2015
Balthasar A. Heesters; Madelene Lindqvist; Parsia A. Vagefi; Eileen Scully; Frank A. Schildberg; Marcus Altfeld; Bruce D. Walker; Daniel E. Kaufmann; Michael C. Carroll
Despite the success of antiretroviral therapy (ART), it does not cure Human Immunodeficiency Virus (HIV) and discontinuation results in viral rebound. Follicular dendritic cells (FDC) are in direct contact with CD4+ T cells and they retain intact antigen for prolonged periods. We found that human FDC isolated from patients on ART retain infectious HIV within a non-degradative cycling compartment and transmit infectious virus to uninfected CD4 T cells in vitro. Importantly, treatment of the HIV+ FDC with a soluble complement receptor 2 purges the FDC of HIV virions and prevents viral transmission in vitro. Our results provide an explanation for how FDC can retain infectious HIV for extended periods and suggest a therapeutic strategy to achieve cure in HIV-infected humans.
Science Translational Medicine | 2014
Anne L. Fletcher; Jessica S Elman; Jillian L. Astarita; Ryan Murray; Nima Saeidi; Joshua D'Rozario; Konstantin Knoblich; Flavian D. Brown; Frank A. Schildberg; Janice M Nieves; Tracy S P Heng; Richard L. Boyd; Shannon J. Turley; Biju Parekkadan
Infusion of lymph node–derived fibroblastic reticular cells into septic mice increases survival at late time points after disease onset. A Swiss Army Knife for Treating Sepsis Sepsis is a complication of infection that kills ~7 million people a year, with no successful molecular therapy thus far. But cells are more versatile than molecules: They make products and respond to their environments. Now, Fletcher et al. investigate whether these multifunctional tools are better equipped to battle this multifocal disease. They showed that one injection of anti-inflammatory cells derived from the lymph nodes dramatically increased survival in two mouse models of sepsis, even when delivered late in the disease course and under conditions that mimic those in the clinic. These beneficial cells reduced the deadly sepsis-associated “cytokine storm” by dampening this response in a manner that required the induction of nitric oxide synthase 2. Sepsis is an aggressive inflammatory syndrome and a global health burden estimated to kill 7.3 million people annually. Single-target molecular therapies have not addressed the multiple disease pathways triggered by septic injury. Cell therapies might offer a broader set of mechanisms of action that benefit complex, multifocal disease processes. We describe a population of immune-specialized myofibroblasts derived from lymph node tissue, termed fibroblastic reticular cells (FRCs). Because FRCs have an immunoregulatory function in lymph nodes, we hypothesized that ex vivo–expanded FRCs would control inflammation when administered therapeutically. Indeed, a single injection of ex vivo–expanded allogeneic FRCs reduced mortality in mouse models of sepsis when administered at early or late time points after septic onset. Mice treated with FRCs exhibited lower local and systemic concentrations of proinflammatory cytokines and reduced bacteremia. When administered 4 hours after induction of lipopolysaccharide endotoxemia, or cecal ligation and puncture (CLP) sepsis in mice, FRCs reduced deaths by at least 70%. When administered late in disease (16 hours after CLP), FRCs still conveyed a robust survival advantage (44% survival compared to 0% for controls). FRC therapy was dependent on the metabolic activity of nitric oxide synthase 2 (NOS2) as the primary molecular mechanism of drug action in the mice. Together, these data describe a new anti-inflammatory cell type and provide preclinical evidence for therapeutic efficacy in severe sepsis that warrants further translational study.
Hepatology | 2012
Bastian Höchst; Frank A. Schildberg; Jan P. Böttcher; Christina Metzger; Sebastian Huss; Andreas Türler; Markus Overhaus; Andreas Knoblich; Berthold Schneider; Dimitrios Pantelis; Christian Kurts; Jörg C. Kalff; Percy A. Knolle; Linda Diehl
Immunity against cancer is impeded by local mechanisms promoting development of tumor‐specific T cell tolerance, such as regulatory T cells, myeloid‐derived suppressor cells, or immunosuppressive factors in the tumor microenvironment. The release of soluble antigens, such as carcinoembryonic antigen (CEA) from colorectal carcinoma (CRC) cells, has been investigated for diagnostic purposes, but not for its immunological consequences. Here, we address the question of whether soluble CEA influences tumor‐specific immunity. Mice were injected with soluble CEA protein, and CEA‐specific CD8 T cells were analyzed for their phenotype and functionality by means of restimulation ex vivo or antitumor efficacy in vivo. We furthermore characterized the CD8 T cell population in peripheral blood mononuclear cell (PBMCs) from healthy donors and colorectal carcinoma patients. In mice, circulating CEA was preferentially taken up in a mannose receptor–dependent manner and cross‐presented by liver sinusoidal endothelial cells, but not dendritic cells, to CD8 T cells. Such systemically circulating CEA promoted tolerization of CEA‐specific CD8 T cells in the endogenous T cell repertoire through the coinhibitory molecule B7H1. These CD8 T cells were not deleted but were rendered nonresponsive to antigen‐specific stimulation and failed to control growth of CEA‐expressing tumor cells. These nonresponsive CD8 T cells were phenotypically similar to central memory T cells being CD44highCD62LhighCD25neg. We found T cells with a similar phenotype in PBMCs of healthy donors and at increased frequency also in patients with colorectal carcinoma. Conclusion: Our results provide evidence for the existence of an unrecognized tumor immune escape involving cross‐presentation of systemically circulating tumor antigens that may influence immunotherapy of cancer. (HEPATOLOGY 2012;56:1924–1933)