TY - JOUR A1 - Leonhardt, Ines A1 - Spielberg, Steffi A1 - Weber, Michael A1 - Albrecht-Eckardt, Daniela A1 - Bläss, Markus A1 - Claus, Ralf A1 - Barz, Dagmar A1 - Scherlach, Kirstin A1 - Hertweck, Christian A1 - Löffler, Jürgen A1 - Hünniger, Kerstin A1 - Kurzai, Oliver T1 - The fungal quorum-sensing molecule farnesol activates innate immune cells but suppresses cellular adaptive immunity JF - mBio N2 - Farnesol, produced by the polymorphic fungus Candida albicans, is the first quorum-sensing molecule discovered in eukaryotes. Its main function is control of C. albicans filamentation, a process closely linked to pathogenesis. In this study, we analyzed the effects of farnesol on innate immune cells known to be important for fungal clearance and protective immunity. Farnesol enhanced the expression of activation markers on monocytes (CD86 and HLA-DR) and neutrophils (CD66b and CD11b) and promoted oxidative burst and the release of proinflammatory cytokines (tumor necrosis factor alpha [TNF-\(\alpha\)] and macrophage inflammatory protein 1 alpha [MIP-1 \(\alpha\)]). However, this activation did not result in enhanced fungal uptake or killing. Furthermore, the differentiation of monocytes to immature dendritic cells (iDC) was significantly affected by farnesol. Several markers important for maturation and antigen presentation like CD1a, CD83, CD86, and CD80 were significantly reduced in the presence of farnesol. Furthermore, farnesol modulated migrational behavior and cytokine release and impaired the ability of DC to induce T cell proliferation. Of major importance was the absence of interleukin 12 (IL-12) induction in iDC generated in the presence of farnesol. Transcriptome analyses revealed a farnesol-induced shift in effector molecule expression and a down-regulation of the granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor during monocytes to iDC differentiation. Taken together, our data unveil the ability of farnesol to act as a virulence factor of C. albicans by influencing innate immune cells to promote inflammation and mitigating the Th1 response, which is essential for fungal clearance. KW - human dendritic cells KW - Pseudomonas aeruginosa KW - induced apoptosis KW - cytokine production KW - biofilm formation KW - Candida albicans KW - mouse model KW - systemic candidiasis KW - oxidative stress KW - carcinoma cells Y1 - 2015 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-143756 VL - 6 IS - 2 ER - TY - JOUR A1 - Machata, Silke A1 - Sreekantapuram, Sravya A1 - Hünniger, Kerstin A1 - Kurzai, Oliver A1 - Dunker, Christine A1 - Schubert, Katja A1 - Krüger, Wibke A1 - Schulze-Richter, Bianca A1 - Speth, Cornelia A1 - Rambach, Günter A1 - Jacobsen, Ilse D. T1 - Significant Differences in Host-Pathogen Interactions Between Murine and Human Whole Blood JF - Frontiers in Immunology N2 - Murine infection models are widely used to study systemic candidiasis caused by C. albicans. Whole-blood models can help to elucidate host-pathogens interactions and have been used for several Candida species in human blood. We adapted the human whole-blood model to murine blood. Unlike human blood, murine blood was unable to reduce fungal burden and more substantial filamentation of C. albicans was observed. This coincided with less fungal association with leukocytes, especially neutrophils. The lower neutrophil number in murine blood only partially explains insufficient infection and filamentation control, as spiking with murine neutrophils had only limited effects on fungal killing. Furthermore, increased fungal survival is not mediated by enhanced filamentation, as a filament-deficient mutant was likewise not eliminated. We also observed host-dependent differences for interaction of platelets with C. albicans, showing enhanced platelet aggregation, adhesion and activation in murine blood. For human blood, opsonization was shown to decrease platelet interaction suggesting that complement factors interfere with fungus-to-platelet binding. Our results reveal substantial differences between murine and human whole-blood models infected with C. albicans and thereby demonstrate limitations in the translatability of this ex vivo model between hosts. KW - whole blood ex vivo model KW - host-pathogen interaction KW - neutrophils KW - mice Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-222575 SN - 1664-3224 VL - 11 ER - TY - JOUR A1 - Weiss, Esther A1 - Schlegel, Jan A1 - Terpitz, Ulrich A1 - Weber, Michael A1 - Linde, Jörg A1 - Schmitt, Anna-Lena A1 - Hünniger, Kerstin A1 - Marischen, Lothar A1 - Gamon, Florian A1 - Bauer, Joachim A1 - Löffler, Claudia A1 - Kurzai, Oliver A1 - Morton, Charles Oliver A1 - Sauer, Markus A1 - Einsele, Hermann A1 - Loeffler, Juergen T1 - Reconstituting NK Cells After Allogeneic Stem Cell Transplantation Show Impaired Response to the Fungal Pathogen Aspergillus fumigatus JF - Frontiers in Immunology N2 - Delayed natural killer (NK) cell reconstitution after allogeneic stem cell transplantation (alloSCT) is associated with a higher risk of developing invasive aspergillosis. The interaction of NK cells with the human pathogen Aspergillus (A.) fumigatus is mediated by the fungal recognition receptor CD56, which is relocated to the fungal interface after contact. Blocking of CD56 signaling inhibits the fungal mediated chemokine secretion of MIP-1α, MIP-1β, and RANTES and reduces cell activation, indicating a functional role of CD56 in fungal recognition. We collected peripheral blood from recipients of an allograft at defined time points after alloSCT (day 60, 90, 120, 180). NK cells were isolated, directly challenged with live A. fumigatus germ tubes, and cell function was analyzed and compared to healthy age and gender-matched individuals. After alloSCT, NK cells displayed a higher percentage of CD56\(^{bright}\)CD16\(^{dim}\) cells throughout the time of blood collection. However, CD56 binding and relocalization to the fungal contact side were decreased. We were able to correlate this deficiency to the administration of corticosteroid therapy that further negatively influenced the secretion of MIP-1α, MIP-1β, and RANTES. As a consequence, the treatment of healthy NK cells ex vivo with corticosteroids abrogated chemokine secretion measured by multiplex immunoassay. Furthermore, we analyzed NK cells regarding their actin cytoskeleton by Structured Illumination Microscopy (SIM) and flow cytometry and demonstrate an actin dysfunction of NK cells shown by reduced F-actin content after fungal co-cultivation early after alloSCT. This dysfunction remains until 180 days post-alloSCT, concluding that further actin-dependent cellular processes may be negatively influenced after alloSCT. To investigate the molecular pathomechansism, we compared CD56 receptor mobility on the plasma membrane of healthy and alloSCT primary NK cells by single-molecule tracking. The results were very robust and reproducible between tested conditions which point to a different molecular mechanism and emphasize the importance of proper CD56 mobility. KW - natural killer cell KW - stem cell transplantation KW - corticosteroids KW - CCL3 KW - CCL4 KW - CCL5 Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-212581 SN - 1664-3224 VL - 11 ER - TY - JOUR A1 - Prauße, Maria T. E. A1 - Lehnert, Teresa A1 - Timme, Sandra A1 - Hünniger, Kerstin A1 - Leonhardt, Ines A1 - Kurzai, Oliver A1 - Figge, Marc Thilo T1 - Predictive Virtual Infection Modeling of Fungal Immune Evasion in Human Whole Blood JF - Frontiers in Immunology N2 - Bloodstream infections by the human-pathogenic fungi Candida albicans and Candida glabrata increasingly occur in hospitalized patients and are associated with high mortality rates. The early immune response against these fungi in human blood comprises a concerted action of humoral and cellular components of the innate immune system. Upon entering the blood, the majority of fungal cells will be eliminated by innate immune cells, i.e., neutrophils and monocytes. However, recent studies identified a population of fungal cells that can evade the immune response and thereby may disseminate and cause organ dissemination, which is frequently observed during candidemia. In this study, we investigate the so far unresolved mechanism of fungal immune evasion in human whole blood by testing hypotheses with the help of mathematical modeling. We use a previously established state-based virtual infection model for whole-blood infection with C. albicans to quantify the immune response and identified the fungal immune-evasion mechanism. While this process was assumed to be spontaneous in the previous model, we now hypothesize that the immune-evasion process is mediated by host factors and incorporate such a mechanism in the model. In particular, we propose, based on previous studies that the fungal immune-evasion mechanism could possibly arise through modification of the fungal surface by as of yet unknown proteins that are assumed to be secreted by activated neutrophils. To validate or reject any of the immune-evasion mechanisms, we compared the simulation of both immune-evasion models for different infection scenarios, i.e., infection of whole blood with either C. albicans or C. glabrata under non-neutropenic and neutropenic conditions. We found that under non-neutropenic conditions, both immune-evasion models fit the experimental data from whole-blood infection with C. albicans and C. glabrata. However, differences between the immune-evasion models could be observed for the infection outcome under neutropenic conditions with respect to the distribution of fungal cells across the immune cells. Based on these predictions, we suggested specific experimental studies that might allow for the validation or rejection of the proposed immune-evasion mechanism. KW - immune evasion KW - state-based model KW - innate immune response KW - polymorphonuclear neutrophils KW - whole-blood infection assay Y1 - 2018 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-197493 SN - 1664-3224 VL - 9 IS - 560 ER - TY - JOUR A1 - Tappe, Beeke A1 - Lauruschkat, Chris D. A1 - Strobel, Lea A1 - Pantaleón García, Jezreel A1 - Kurzai, Oliver A1 - Rebhan, Silke A1 - Kraus, Sabrina A1 - Pfeuffer-Jovic, Elena A1 - Bussemer, Lydia A1 - Possler, Lotte A1 - Held, Matthias A1 - Hünniger, Kerstin A1 - Kniemeyer, Olaf A1 - Schäuble, Sascha A1 - Brakhage, Axel A. A1 - Panagiotou, Gianni A1 - White, P. Lewis A1 - Einsele, Hermann A1 - Löffler, Jürgen A1 - Wurster, Sebastian T1 - COVID-19 patients share common, corticosteroid-independent features of impaired host immunity to pathogenic molds JF - Frontiers in Immunology N2 - Patients suffering from coronavirus disease-2019 (COVID-19) are susceptible to deadly secondary fungal infections such as COVID-19-associated pulmonary aspergillosis and COVID-19-associated mucormycosis. Despite this clinical observation, direct experimental evidence for severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2)-driven alterations of antifungal immunity is scarce. Using an ex-vivo whole blood stimulation assay, we challenged blood from twelve COVID-19 patients with Aspergillus fumigatus and Rhizopus arrhizus antigens and studied the expression of activation, maturation, and exhaustion markers, as well as cytokine secretion. Compared to healthy controls, T-helper cells from COVID-19 patients displayed increased expression levels of the exhaustion marker PD-1 and weakened A. fumigatus- and R. arrhizus-induced activation. While baseline secretion of proinflammatory cytokines was massively elevated, whole blood from COVID-19 patients elicited diminished release of T-cellular (e.g., IFN-γ, IL-2) and innate immune cell-derived (e.g., CXCL9, CXCL10) cytokines in response to A. fumigatus and R. arrhizus antigens. Additionally, samples from COVID-19 patients showed deficient granulocyte activation by mold antigens and reduced fungal killing capacity of neutrophils. These features of weakened anti-mold immune responses were largely decoupled from COVID-19 severity, the time elapsed since diagnosis of COVID-19, and recent corticosteroid uptake, suggesting that impaired anti-mold defense is a common denominator of the underlying SARS-CoV-2 infection. Taken together, these results expand our understanding of the immune predisposition to post-viral mold infections and could inform future studies of immunotherapeutic strategies to prevent and treat fungal superinfections in COVID-19 patients. KW - COVID-19 KW - immune impairment KW - T cells KW - granulocytes KW - Aspergillus KW - Rhizopus Y1 - 2022 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:bvb:20-opus-283558 SN - 1664-3224 VL - 13 ER -