44 results on '"Mark S Gresnigt"'
Search Results
2. Human albumin enhances the pathogenic potential of Candida glabrata on vaginal epithelial cells.
- Author
-
Marina Pekmezovic, Ann-Kristin Kaune, Sophie Austermeier, Sophia U J Hitzler, Selene Mogavero, Hrant Hovhannisyan, Toni Gabaldón, Mark S Gresnigt, and Bernhard Hube
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The opportunistic pathogen Candida glabrata is the second most frequent causative agent of vulvovaginal candidiasis (VVC), a disease that affects 70-75% of women at least once during their life. However, C. glabrata is almost avirulent in mice and normally incapable of inflicting damage to vaginal epithelial cells in vitro. We thus proposed that host factors present in vivo may influence C. glabrata pathogenicity. We, therefore, analyzed the impact of albumin, one of the most abundant proteins of the vaginal fluid. The presence of human, but not murine, albumin dramatically increased the potential of C. glabrata to damage vaginal epithelial cells. This effect depended on macropinocytosis-mediated epithelial uptake of albumin and subsequent proteolytic processing. The enhanced pathogenicity of C. glabrata can be explained by a combination of beneficial effects for the fungus, which includes an increased access to iron, accelerated growth, and increased adhesion. Screening of C. glabrata deletion mutants revealed that Hap5, a key regulator of iron homeostasis, is essential for the albumin-augmented damage potential. The albumin-augmented pathogenicity was reversed by the addition of iron chelators and a similar increase in pathogenicity was shown by increasing the iron availability, confirming a key role of iron. Accelerated growth not only led to higher cell numbers, but also to increased fungal metabolic activity and oxidative stress resistance. Finally, the albumin-driven enhanced damage potential was associated with the expression of distinct C. glabrata virulence genes. Transcriptional responses of the epithelial cells suggested an unfolded protein response (UPR) and ER-stress responses combined with glucose starvation induced by fast growing C. glabrata cells as potential mechanisms by which cytotoxicity is mediated.Collectively, we demonstrate that albumin augments the pathogenic potential of C. glabrata during interaction with vaginal epithelial cells. This suggests a role for albumin as a key player in the pathogenesis of VVC.
- Published
- 2021
- Full Text
- View/download PDF
3. Candida albicans colonization of the gastrointestinal tract: A double-edged sword.
- Author
-
Rebeca Alonso-Monge, Mark S Gresnigt, Elvira Román, Bernhard Hube, and Jesús Pla
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Published
- 2021
- Full Text
- View/download PDF
4. Rewiring monocyte glucose metabolism via C-type lectin signaling protects against disseminated candidiasis.
- Author
-
Jorge Domínguez-Andrés, Rob J W Arts, Rob Ter Horst, Mark S Gresnigt, Sanne P Smeekens, Jacqueline M Ratter, Ekta Lachmandas, Lily Boutens, Frank L van de Veerdonk, Leo A B Joosten, Richard A Notebaart, Carlos Ardavín, and Mihai G Netea
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Monocytes are innate immune cells that play a pivotal role in antifungal immunity, but little is known regarding the cellular metabolic events that regulate their function during infection. Using complementary transcriptomic and immunological studies in human primary monocytes, we show that activation of monocytes by Candida albicans yeast and hyphae was accompanied by metabolic rewiring induced through C-type lectin-signaling pathways. We describe that the innate immune responses against Candida yeast are energy-demanding processes that lead to the mobilization of intracellular metabolite pools and require induction of glucose metabolism, oxidative phosphorylation and glutaminolysis, while responses to hyphae primarily rely on glycolysis. Experimental models of systemic candidiasis models validated a central role for glucose metabolism in anti-Candida immunity, as the impairment of glycolysis led to increased susceptibility in mice. Collectively, these data highlight the importance of understanding the complex network of metabolic responses triggered during infections, and unveil new potential targets for therapeutic approaches against fungal diseases.
- Published
- 2017
- Full Text
- View/download PDF
5. An integrative genomics approach identifies novel pathways that influence candidaemia susceptibility.
- Author
-
Vasiliki Matzaraki, Mark S Gresnigt, Martin Jaeger, Isis Ricaño-Ponce, Melissa D Johnson, Marije Oosting, Lude Franke, Sebo Withoff, John R Perfect, Leo A B Joosten, Bart Jan Kullberg, Frank L van de Veerdonk, Iris Jonkers, Yang Li, Cisca Wijmenga, Mihai G Netea, and Vinod Kumar
- Subjects
Medicine ,Science - Abstract
Candidaemia is a bloodstream infection caused by Candida species that primarily affects specific groups of at-risk patients. Because only small candidaemia patient cohorts are available, classical genome wide association cannot be used to identify Candida susceptibility genes. Therefore, we have applied an integrative genomics approach to identify novel susceptibility genes and pathways for candidaemia. Candida-induced transcriptome changes in human primary leukocytes were assessed by RNA sequencing. Genetic susceptibility to candidaemia was assessed using the Illumina immunochip platform for genotyping of a cohort of 217 patients. We then integrated genetics data with gene-expression profiles, Candida-induced cytokine production capacity, and circulating concentrations of cytokines. Based on the intersection of transcriptome pathways and genomic data, we prioritized 31 candidate genes for candidaemia susceptibility. This group of genes was enriched with genes involved in inflammation, innate immunity, complement, and hemostasis. We then validated the role of MAP3K8 in cytokine regulation in response to Candida stimulation. Here, we present a new framework for the identification of susceptibility genes for infectious diseases that uses an unbiased, hypothesis-free, systems genetics approach. By applying this approach to candidaemia, we identified novel susceptibility genes and pathways for candidaemia, and future studies should assess their potential as therapeutic targets.
- Published
- 2017
- Full Text
- View/download PDF
6. A synthetic peptide mimic kills Candida albicans and synergistically prevents infection
- Author
-
Sebastian Schaefer, Raghav Vij, Jakob L. Sprague, Sophie Austermeier, Hue Dinh, Peter R. Judzewitsch, Sven Müller-Loennies, Taynara Lopes Silva, Eric Seemann, Britta Qualmann, Christian Hertweck, Kirstin Scherlach, Thomas Gutsmann, Amy K. Cain, Nathaniel Corrigan, Mark S. Gresnigt, Cyrille Boyer, Megan D. Lenardon, and Sascha Brunke
- Subjects
Science - Abstract
Abstract More than two million people worldwide are affected by life-threatening, invasive fungal infections annually. Candida species are the most common cause of nosocomial, invasive fungal infections and are associated with mortality rates above 40%. Despite the increasing incidence of drug-resistance, the development of novel antifungal formulations has been limited. Here we investigate the antifungal mode of action and therapeutic potential of positively charged, synthetic peptide mimics to combat Candida albicans infections. Our data indicates that these synthetic polymers cause endoplasmic reticulum stress and affect protein glycosylation, a mode of action distinct from currently approved antifungal drugs. The most promising polymer composition damaged the mannan layer of the cell wall, with additional membrane-disrupting activity. The synergistic combination of the polymer with caspofungin prevented infection of human epithelial cells in vitro, improved fungal clearance by human macrophages, and significantly increased host survival in a Galleria mellonella model of systemic candidiasis. Additionally, prolonged exposure of C. albicans to the synergistic combination of polymer and caspofungin did not lead to the evolution of tolerant strains in vitro. Together, this work highlights the enormous potential of these synthetic peptide mimics to be used as novel antifungal formulations as well as adjunctive antifungal therapy.
- Published
- 2024
- Full Text
- View/download PDF
7. Alpha1-antitrypsin impacts innate host–pathogen interactions with Candida albicans by stimulating fungal filamentation
- Author
-
Martin Jaeger, Axel Dietschmann, Sophie Austermeier, Sude Dinçer, Pauline Porschitz, Larsen Vornholz, Ralph J.A. Maas, Evelien G.G. Sprenkeler, Jürgen Ruland, Stefan Wirtz, Tania Azam, Leo A.B. Joosten, Bernhard Hube, Mihai G. Netea, Charles A. Dinarello, and Mark S. Gresnigt
- Subjects
Immune escape ,immune evasion ,host–pathogen interactions ,fungal adaptation ,filamentous growth ,cell wall remodelling ,Infectious and parasitic diseases ,RC109-216 - Abstract
ABSTRACTOur immune system possesses sophisticated mechanisms to cope with invading microorganisms, while pathogens evolve strategies to deal with threats imposed by host immunity. Human plasma protein α1-antitrypsin (AAT) exhibits pleiotropic immune-modulating properties by both preventing immunopathology and improving antimicrobial host defence. Genetic associations suggested a role for AAT in candidemia, the most frequent fungal blood stream infection in intensive care units, yet little is known about how AAT influences interactions between Candida albicans and the immune system. Here, we show that AAT differentially impacts fungal killing by innate phagocytes. We observed that AAT induces fungal transcriptional reprogramming, associated with cell wall remodelling and downregulation of filamentation repressors. At low concentrations, the cell-wall remodelling induced by AAT increased immunogenic β-glucan exposure and consequently improved fungal clearance by monocytes. Contrastingly, higher AAT concentrations led to excessive C. albicans filamentation and thus promoted fungal immune escape from monocytes and macrophages. This underscores that fungal adaptations to the host protein AAT can differentially define the outcome of encounters with innate immune cells, either contributing to improved immune recognition or fungal immune escape.
- Published
- 2024
- Full Text
- View/download PDF
8. Protective host defense against disseminated candidiasis is impaired in mice expressing human interleukin-37
- Author
-
Frank L. Van De Veerdonk, Mark S Gresnigt, Jos WM Van Der Meer, Leo eJoosten, Mihai eNetea, and Charles eDinarello
- Subjects
Candida ,Neutrophils ,TNFa ,IL-37 ,antifungal host defense ,IL-1F7 ,Microbiology ,QR1-502 - Abstract
The effect of the anti-inflammatory cytokine interleukin-37 (IL 37) on host defense against Candida infections remains unknown. We assessed the role of IL 37 in a murine model of disseminated candidiasis using mice transgenic for human IL 37 (hIL 37Tg). Upon exposure to C. albicans pseudohyphae, macrophages from hIL-37Tg mice release 39% less TNFα compared to cells from wild-type mice (P=0.01). In vivo, hIL 37Tg mice displayed a decreased capacity to recruit neutrophils to the site of infection. These defects were associated with increased mortality and organ fungal growth in hIL-37Tg compared to wild-type mice. We conclude that IL-37 interferes with the innate protective anti-Candida host response by reducing the production of proinflammatory cytokines and suppressing neutrophil recruitment in response to Candida, resulting in an increased susceptibility to disseminated candidiasis.
- Published
- 2015
- Full Text
- View/download PDF
9. A polysaccharide virulence factor from Aspergillus fumigatus elicits anti-inflammatory effects through induction of Interleukin-1 receptor antagonist.
- Author
-
Mark S Gresnigt, Silvia Bozza, Katharina L Becker, Leo A B Joosten, Shahla Abdollahi-Roodsaz, Wim B van der Berg, Charles A Dinarello, Mihai G Netea, Thierry Fontaine, Antonella De Luca, Silvia Moretti, Luigina Romani, Jean-Paul Latge, and Frank L van de Veerdonk
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The galactosaminogalactan (GAG) is a cell wall component of Aspergillus fumigatus that has potent anti-inflammatory effects in mice. However, the mechanisms responsible for the anti-inflammatory property of GAG remain to be elucidated. In the present study we used in vitro PBMC stimulation assays to demonstrate, that GAG inhibits proinflammatory T-helper (Th)1 and Th17 cytokine production in human PBMCs by inducing Interleukin-1 receptor antagonist (IL-1Ra), a potent anti-inflammatory cytokine that blocks IL-1 signalling. GAG cannot suppress human T-helper cytokine production in the presence of neutralizing antibodies against IL-1Ra. In a mouse model of invasive aspergillosis, GAG induces IL-1Ra in vivo, and the increased susceptibility to invasive aspergillosis in the presence of GAG in wild type mice is not observed in mice deficient for IL-1Ra. Additionally, we demonstrate that the capacity of GAG to induce IL-1Ra could also be used for treatment of inflammatory diseases, as GAG was able to reduce severity of an experimental model of allergic aspergillosis, and in a murine DSS-induced colitis model. In the setting of invasive aspergillosis, GAG has a significant immunomodulatory function by inducing IL-1Ra and notably IL-1Ra knockout mice are completely protected to invasive pulmonary aspergillosis. This opens new treatment strategies that target IL-1Ra in the setting of acute invasive fungal infection. However, the observation that GAG can also protect mice from allergy and colitis makes GAG or a derivative structure of GAG a potential treatment compound for IL-1 driven inflammatory diseases.
- Published
- 2014
- Full Text
- View/download PDF
10. Nanobody-mediated neutralization of candidalysin prevents epithelial damage and inflammatory responses that drive vulvovaginal candidiasis pathogenesis
- Author
-
Marisa Valentine, Paul Rudolph, Axel Dietschmann, Antzela Tsavou, Selene Mogavero, Sejeong Lee, Emily L. Priest, Gaukhar Zhurgenbayeva, Nadja Jablonowski, Sandra Timme, Christian Eggeling, Stefanie Allert, Edward Dolk, Julian R. Naglik, Marc T. Figge, Mark S. Gresnigt, and Bernhard Hube
- Subjects
candidalysin ,vulvovaginal candidiasis ,inflammation ,cytotoxicity ,therapeutic strategy ,Microbiology ,QR1-502 - Abstract
ABSTRACT Candida albicans can cause mucosal infections in humans. This includes oropharyngeal candidiasis, which is commonly observed in human immunodeficiency virus infected patients, and vulvovaginal candidiasis (VVC), which is the most frequent manifestation of candidiasis. Epithelial cell invasion by C. albicans hyphae is accompanied by the secretion of candidalysin, a peptide toxin that causes epithelial cell cytotoxicity. During vaginal infections, candidalysin-driven tissue damage triggers epithelial signaling pathways, leading to hyperinflammatory responses and immunopathology, a hallmark of VVC. Therefore, we proposed blocking candidalysin activity using nanobodies to reduce epithelial damage and inflammation as a therapeutic strategy for VVC. Anti-candidalysin nanobodies were confirmed to localize around epithelial-invading C. albicans hyphae, even within the invasion pocket where candidalysin is secreted. The nanobodies reduced candidalysin-induced damage to epithelial cells and downstream proinflammatory responses. Accordingly, the nanobodies also decreased neutrophil activation and recruitment. In silico mathematical modeling enabled the quantification of epithelial damage caused by candidalysin under various nanobody dosing strategies. Thus, nanobody-mediated neutralization of candidalysin offers a novel therapeutic approach to block immunopathogenic events during VVC and alleviate symptoms.IMPORTANCEWorldwide, vaginal infections caused by Candida albicans (VVC) annually affect millions of women, with symptoms significantly impacting quality of life. Current treatments are based on anti-fungals and probiotics that target the fungus. However, in some cases, infections are recurrent, called recurrent VVC, which often fails to respond to treatment. Vaginal mucosal tissue damage caused by the C. albicans peptide toxin candidalysin is a key driver in the induction of hyperinflammatory responses that fail to clear the infection and contribute to immunopathology and disease severity. In this pre-clinical evaluation, we show that nanobody-mediated candidalysin neutralization reduces tissue damage and thereby limits inflammation. Implementation of candidalysin-neutralizing nanobodies may prove an attractive strategy to alleviate symptoms in complicated VVC cases.
- Published
- 2024
- Full Text
- View/download PDF
11. Editorial: Exploring novel experimental systems to study the mechanistic basis of fungal infections
- Author
-
Sara Gago, Mark S. Gresnigt, and Teresa Zelante
- Subjects
animal models ,fungal disease ,in vitro infection model ,Aspergillus ,sporotrichosis ,coccidioidomycosis ,Plant culture ,SB1-1110 - Published
- 2023
- Full Text
- View/download PDF
12. Uncoupling of IL-6 signaling and LC3-associated phagocytosis drives immunoparalysis during sepsis
- Author
-
Frank L. van de Veerdonk, Maria Venichaki, Georgios Chamilos, Jean-Paul Latgé, Dimitrios Georgopoulos, Eleni Diamantaki, Mark S. Gresnigt, Tonia Akoumianaki, Mihai G. Netea, Jamel El-Benna, Katerina Vaporidi, Kieu T. T. Le, Rémi Beau, George Samonis, Elias Drakos, Frédéric Pène, Vinod Kumar, Marina Gkountzinopulou, Lab Excellence Inflamex (CRI INSERM U1149 - Bichat Medical Faculty), Université Paris Diderot - Paris 7 (UPD7), and Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI)
- Subjects
MAPK/ERK pathway ,JAK2 TYROSINE KINASE ,Phagocytosis ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Biology ,Microbiology ,Monocytes ,Sepsis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Virology ,medicine ,NADPH OXIDASE ,Humans ,Autocrine signalling ,PHOSPHORYLATION ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Phagosome ,0303 health sciences ,Phagocytes ,NADPH oxidase ,IFN-GAMMA ,INDUCED IMMUNOSUPPRESSION ,Interleukin-6 ,Aspergillus fumigatus ,Macrophages ,Nuclear Proteins ,INHIBITOR ,Janus Kinase 2 ,medicine.disease ,PHAGOSOMES ,Cell biology ,Cytoskeletal Proteins ,MICE ,INFECTIONS ,biology.protein ,Phosphorylation ,Cytokines ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Parasitology ,AUTOPHAGY ,Microtubule-Associated Proteins ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Contains fulltext : 238107.pdf (Publisher’s version ) (Closed access) Immune deactivation of phagocytes is a central event in the pathogenesis of sepsis. Herein, we identify a master regulatory role of IL-6 signaling on LC3-associated phagocytosis (LAP) and reveal that uncoupling of these two processes during sepsis induces immunoparalysis in monocytes/macrophages. In particular, we demonstrate that activation of LAP by the human fungal pathogen Aspergillus fumigatus depends on ERK1/2-mediated phosphorylation of p47phox subunit of NADPH oxidase. Physiologically, autocrine IL-6/JAK2/Ninein axis orchestrates microtubule organization and dynamics regulating ERK recruitment to the phagosome and LC3(+) phagosome (LAPosome) formation. In sepsis, loss of IL-6 signaling specifically abrogates microtubule-mediated trafficking of ERK, leading to defective activation of LAP and impaired killing of bacterial and fungal pathogens by monocytes/macrophages, which can be selectively restored by IL-6 supplementation. Our work uncovers a molecular pathway linking IL-6 signaling with LAP and provides insight into the mechanisms underlying immunoparalysis in sepsis.
- Published
- 2021
- Full Text
- View/download PDF
13. Albumin Neutralizes Hydrophobic Toxins and Modulates Candida albicans Pathogenicity
- Author
-
David L. Moyes, Julian R. Naglik, Mark S. Gresnigt, Sophie Austermeier, Marina Pekmezovic, Nessim Kichik, Sejeong Lee, Natalia K. Kotowicz, Pauline Porschitz, J. Ho, and Bernhard Hube
- Subjects
0303 health sciences ,biology ,030306 microbiology ,Toxin ,Chemistry ,Albumin ,Serum albumin ,Inflammation ,medicine.disease_cause ,biology.organism_classification ,Blood proteins ,Microbiology ,Corpus albicans ,QR1-502 ,3. Good health ,03 medical and health sciences ,Virology ,medicine ,biology.protein ,medicine.symptom ,Candida albicans ,Candidalysin ,030304 developmental biology - Abstract
Albumin is abundant in serum but is also excreted at mucosal surfaces and enters tissues when inflammation increases vascular permeability. Host-associated opportunistic pathogens encounter albumin during commensalism and when causing infections. Considering the ubiquitous presence of albumin, we investigated its role in the pathogenesis of infections with the model human fungal pathogen, Candida albicans. Albumin was introduced in various in vitro models that mimic different stages of systemic or mucosal candidiasis, where it reduced the ability of C. albicans to damage host cells. The amphipathic toxin candidalysin mediates necrotic host cell damage induced by C. albicans. Using cellular and biophysical assays, we determined that albumin functions by neutralizing candidalysin through hydrophobic interactions. We discovered that albumin, similarly, can neutralize a variety of fungal (α-amanitin), bacterial (streptolysin O and staurosporin), and insect (melittin) hydrophobic toxins. These data suggest albumin as a defense mechanism against toxins, which can play a role in the pathogenesis of microbial infections. IMPORTANCE Albumin is the most abundant serum protein in humans. During inflammation, serum albumin levels decrease drastically, and low albumin levels are associated with poor patient outcome. Thus, albumin may have specific functions during infection. Here, we describe the ability of albumin to neutralize hydrophobic microbial toxins. We show that albumin can protect against damage induced by the pathogenic yeast C. albicans by neutralizing its cytolytic toxin candidalysin. These findings suggest that albumin is a toxin-neutralizing protein that may play a role during infections with toxin-producing microorganisms.
- Published
- 2021
- Full Text
- View/download PDF
14. Neuraminidase and SIGLEC15 modulate the host defense against pulmonary aspergillosis
- Author
-
Agostinho Carvalho, Lore Vanderbeke, Frank L. van de Veerdonk, Cláudia F. Campos, Martin Jaeger, Agustin Resendiz Sharpe, Greetje Vande Velde, Mihai G. Netea, Intan M.W. Dewi, Paul E. Verweij, Cristina Cunha, Fadel M. Garishah, Katrien Lagrou, Leo A. B. Joosten, André J. A. M. van der Ven, Joost Wauters, Mark S. Gresnigt, Marina E. Gkountzinopoulou, Quirijn de Mast, Roger J. M. Brüggemann, and Cláudio Duarte-Oliveira
- Subjects
Medicine (General) ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,neuraminidase ,Research & Experimental Medicine ,Aspergillosis ,Aspergillus fumigatus ,chemistry.chemical_compound ,SIGLEC15 ,aspergillosis ,Lung ,0303 health sciences ,ROLES ,biology ,3. Good health ,Aspergillus ,Medicine, Research & Experimental ,Life Sciences & Biomedicine ,Oseltamivir ,oseltamivir ,Immunoglobulins ,Peripheral blood mononuclear cell ,Antiviral Agents ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Immune system ,R5-920 ,All institutes and research themes of the Radboud University Medical Center ,Phagocytosis ,medicine ,Splenocyte ,Animals ,Humans ,030304 developmental biology ,Science & Technology ,FUMIGATUS ,030306 microbiology ,business.industry ,SIALIC ACIDS ,Membrane Proteins ,Cell Biology ,biology.organism_classification ,medicine.disease ,Sialic acid ,Mice, Inbred C57BL ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,chemistry ,Immunology ,biology.protein ,Leukocytes, Mononuclear ,Pulmonary Aspergillosis ,business ,Neuraminidase ,TAMIFLU(R) - Abstract
Summary Influenza-associated pulmonary aspergillosis (IAPA) has been reported increasingly since the advent of use of neuraminidase (NA) inhibitors following the 2009 influenza pandemic. We hypothesize that blocking host NA modulates the immune response against Aspergillus fumigatus. We demonstrate that NA influences the host response against A. fumigatus in vitro and that oseltamivir increases the susceptibility of mice to pulmonary aspergillosis. Oseltamivir impairs the mouse splenocyte and human peripheral blood mononuclear cell (PBMC) killing capacity of A. fumigatus, and adding NA restores this defect in PBMCs. Furthermore, the sialic acid-binding receptor SIGLEC15 is upregulated in PBMCs stimulated with A. fumigatus. Silencing of SIGLEC15 decrease PBMC killing of A. fumigatus. We provide evidence that host NA activity and sialic acid recognition are important for anti-Aspergillus defense. NA inhibitors might predispose individuals with severe influenza to invasive aspergillosis. These data shed light on the pathogenesis of invasive fungal infections and may identify potential therapeutic targets., Graphical abstract, Highlights Neuraminidase modulates the host immune response against A. fumigatus Oseltamivir increases the susceptibility of animal model to pulmonary aspergillosis SIGLEC15 is important for host defense against A. fumigatus, Dewi et al. show that neuraminidase plays an important role in host defense against A. fumigatus and that this effect could be mediated by SIGLEC15. Neuraminidase inhibition by oseltamivir might impair antifungal responses. These findings are important for understanding the pathogenesis of influenza-associated pulmonary aspergillosis (IAPA).
- Published
- 2021
15. Candida pathogens induce protective mitochondria-associated type I interferon signalling and a damage-driven response in vaginal epithelial cells
- Author
-
Sofía Siscar-Lewin, Sylvia Müller, Bernhard Hube, Elise Iracane, Toni Gabaldón, Sascha Brunke, Till Kalkreuter, Marina Pekmezovic, Hrant Hovhannisyan, Eric Seemann, Geraldine Butler, Britta Qualmann, Selene Mogavero, Mark S. Gresnigt, João Oliveira-Pacheco, Thomas Kamradt, and Barcelona Supercomputing Center
- Subjects
Microbiology (medical) ,Informàtica::Aplicacions de la informàtica::Bioinformàtica [Àrees temàtiques de la UPC] ,Immunology ,Library science ,Epithelial cells ,Applied Microbiology and Biotechnology ,Microbiology ,Fungal Proteins ,03 medical and health sciences ,Species Specificity ,Political science ,Candida albicans ,Genetics ,Humans ,media_common.cataloged_instance ,European union ,Transcriptomics ,Candidiasis, Vulvovaginal ,Candida ,030304 developmental biology ,media_common ,Fungal pathogenesis ,Innate immunity ,0303 health sciences ,Cèl·lules -- Biologia ,Virulence ,030306 microbiology ,Epithelial Cells ,Cell Biology ,Fungal host response ,Mitochondria ,3. Good health ,European molecular biology laboratory ,Research centre ,Interferon Type I ,Vagina ,Female ,Christian ministry - Abstract
Vaginal candidiasis is an extremely common disease predominantly caused by four phylogenetically diverse species: Candida albicans; Candida glabrata; Candida parapsilosis; and Candida tropicalis. Using a time course infection model of vaginal epithelial cells and dual RNA sequencing, we show that these species exhibit distinct pathogenicity patterns, which are defined by highly species-specific transcriptional profiles during infection of vaginal epithelial cells. In contrast, host cells exhibit a homogeneous response to all species at the early stages of infection, which is characterized by sublethal mitochondrial signalling inducing a protective type I interferon response. At the later stages, the transcriptional response of the host diverges in a species-dependent manner. This divergence is primarily driven by the extent of epithelial damage elicited by species-specific mechanisms, such as secretion of the toxin candidalysin by C. albicans. Our results uncover a dynamic, biphasic response of vaginal epithelial cells to Candida species, which is characterized by protective mitochondria-associated type I interferon signalling and a species-specific damage-driven response. M.P., H.H., E.I., J.O.P., T.G., G.B. and B.H. received funding from the European Union Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant no. 642095 (OPATHY). B.H. also received support from the German Research Foundation within the Collaborative Research Centre/Transregio 124 FungiNet (project C1). M.S.G. was supported by the German Research Foundation Emmy Noether Programme (project no. 434385622/GR 5617/1-1). We acknowledge the support of the Spanish Ministry of Science, Innovation and Universities (grant no. PGC2018-099921-B-I00) to the European Molecular Biology Laboratory partnership, the Centro de Excelencia Severo Ochoa and the CERCA Programme/Generalitat de Catalunya. We thank C. Kämnitz from the Electron Microscopy Center in Jena for the sample preparation for TEM. The schematic models in Figs. 4–6 were created with images adapted from Servier Medical Art (Servier).
- Published
- 2021
16. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives
- Author
-
Marisa Valentine, Mark S. Gresnigt, Carol A. Munro, Karine Roget, Ricardo Fróis-Martins, Salomé LeibundGut-Landmann, Pieter Van den Abbeele, Yoan Emritloll, Frédéric Bequet, Moran Morelli, Leovigildo-Rey Alaban, Nathaniel Cole, Ilse D. Jacobsen, Mihai G. Netea, Stéphanie Bornes, Sayoni Chakraborty, Alistair J. P. Brown, Margot Delavy, Ann-Kristin Kaune, Alan W. Walker, Diletta Rosati, Peter A Warn, Daria Kosmala, Marie-Elisabeth Bougnoux, Claudia Thoral, Chaysavanh Manichanh, Benoît Marsaux, Vincent Thomas, Zixuan Xie, Christophe d'Enfert, Bernhard Hube, Mélanie Legrand, Karla Queiroz, Biologie et Pathogénicité fongiques - Fungal Biology and Pathogenicity (BPF), Institut Pasteur [Paris] (IP)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), University of Aberdeen, BIOASTER Microbiology Technology Institute [Lyon], École Doctorale Bio Sorbonne Paris Cité [Paris] (ED562 - BioSPC), Université Sorbonne Paris Cité (USPC)-Université Paris Cité (UPCité), Leibniz Institute for Natural Product Research and Infection Biology (Hans Knoell Institute), Friedrich-Schiller-Universität = Friedrich Schiller University Jena [Jena, Germany], ProDigest BVBA, Partenaires INRAE, Universiteit Gent = Ghent University (UGENT), Universität Zürich [Zürich] = University of Zurich (UZH), Mimetas, Radboud University Medical Center [Nijmegen], Vall d'Hebron University Hospital [Barcelona], Magic Bullet Consulting, Unité Mixte de Recherche sur le Fromage (UMRF), VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Université Clermont Auvergne [2017-2020] (UCA [2017-2020])-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), NEXBIOME Therapeutics, University of Exeter, We received funding from the European Union's Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie action, Innovative Training Network: FunHoMic, grant N° 812969. CdE received funding from the French Government ‘Investissement d'Avenir’ program (Laboratoire d'Excellence Integrative Biology of Emerging Infectious Diseases, ANR-10-LABX-62-IBEID), the Agence Nationale de la Recherche (ERA-Net Infect-ERA, FUNCOMPATH, ANR-14-IFEC-0004), the EU Horizon2020 consortium ‘Host-Directed Medicine in invasive FUNgal infections’—HDM-FUN (Grant Agreement 847507). SLL and CdE received funding from the Swiss National Science Foundation (Sinergia program, #CRSII5_173863). BIOASTER received funding from the French Government ‘Investissement d'Avenir’ program (Grant No. ANR-10-AIRT-03). MSG was supported by a Humboldt Research Fellowship for Postdoctoral Researchers by the Alexander von Humboldt-Foundation and the Deutsche Forschungsgemeinschaft (DFG) Emmy Noether Program (project no. 434385622/GR 5617/1-1). BH was supported by the Deutsche Forschungsgemeinschaft (DFG) project Hu 532/20-1, project C1 within the Collaborative Research Centre (CRC)/Transregio 124 FungiNet and the Balance of the Microverse Cluster under Germany´s Excellence Strategy—EXC 2051–Project-ID 390713860, the EU Horizon2020 consortium ‘Host-Directed Medicine in invasive FUNgal infections’—HDM-FUN (Grant Agreement 847507), the Leibniz Association Campus InfectoOptics SAS-2015-HKI-LWC and the Wellcome Trust (215599/Z/19/Z). IDJ was supported by the Deutsche Forschungsgemeinschaft (DFG) project C5 within the Collaborative Research Centre (CRC)/Transregio 124 FungiNet and the Balance of the Microverse Cluster under Germany´s Excellence Strategy—EXC 2051–Project-ID 390713860, the Leibniz Association Campus InfectoOptics SAS-2015-HKI-LWC and the Wellcome Trust (Grant 215599/Z/19/Z). CM received funding from the the Instituto de Salud Carlos III/FEDER. MGN was supported by an ERC Advanced Grant (#833247) and a Spinoza grant of the Netherlands Organization for Scientific Research. CAM was supported by EU Horizon2020 consortium ‘Host-Directed Medicine in invasive FUNgal infections’—HDM-FUN (Grant Agreement 847507) and the Wellcome Trust Strategic Award for Medical Mycology and Fungal Immunology (097377/Z/11/Z). AWW receives core funding support from the Scottish Government's Rural and Environment Science and Analytical Services (RESAS). AJPB was supported by a programme grant from the UK Medical Research Council (MR/M026663/1) and by the Medical Research Council Centre for Medical Mycology at the University of Exeter (MR/N006364/1)., ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), ANR-14-IFEC-0004,FunComPath,From fungal commensalism to pathogenicity:dissection of the colonization-to-infection shift of Candida albicans(2014), ANR-10-AIRT-0003,BIOASTER,BIOASTER(2010), European Project: 812969,H2020-MSCA-ITN-2018,FunHoMic(2019), European Project: 847507,H2020-SC1-2019-Two-Stage-RTD,HDM-FUN(2020), Institut Pasteur [Paris]-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Doctorale Bio Sorbonne Paris Cité [Paris] (ED BioSPC), Université Sorbonne Paris Cité (USPC)-Université de Paris (UP), Universiteit Gent = Ghent University [Belgium] (UGENT), VetAgro Sup - Institut national d'enseignement supérieur et de recherche en alimentation, santé animale, sciences agronomiques et de l'environnement (VAS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Université Clermont Auvergne (UCA), Institut Català de la Salut, [d'Enfert C] Unite Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 75015 Paris, France. [Kaune AK] Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK. [Alaban LR] BIOASTER Microbiology Technology Institute, 69007 Lyon, France. Université de Paris, Sorbonne Paris Cité, 75015 Paris, France. [Chakraborty S] Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, 07745 Jena, Germany. Institute of Microbiology, Friedrich Schiller University, 07743 Jena, German. [Cole N] Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK. [Delavy M] Unite Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 75015 Paris, France. Université de Paris, Sorbonne Paris Cité, 75015 Paris, France. [Manichanh C] Grup de Recerca en Microbioma Intestinal, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Vall d’Hebron Hospital Universitari, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Eukaryota::hongos::Ascomycota::Saccharomycetales::Candida::Candida albicans [ORGANISMOS] ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Relacions hoste-paràsit ,CHRONIC MUCOCUTANEOUS CANDIDIASIS ,Review Article ,DESORPTION IONIZATION-TIME ,Candida albicans ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Chronic mucocutaneous candidiasis ,Microbiological Phenomena::Host Microbial Interactions [PHENOMENA AND PROCESSES] ,CROSS-KINGDOM INTERACTIONS ,Candida ,AcademicSubjects/SCI01150 ,0303 health sciences ,Eukaryota::Fungi::Ascomycota::Saccharomycetales::Candida::Candida albicans [ORGANISMS] ,biology ,ACTIVATED PROTEIN-KINASE ,Candidiasis ,Pattern recognition receptor ,Corpus albicans ,3. Good health ,CHAIN FATTY-ACIDS ,Infectious Diseases ,Candidiasi ,patient variability ,VULVO-VAGINAL CANDIDIASIS ,ARYL-HYDROCARBON RECEPTOR ,fungus-host-microbiota interactions ,digestive system ,Microbiology ,C-TYPE LECTIN ,03 medical and health sciences ,Immune system ,Immunity ,Bacterial Infections and Mycoses::Mycoses::Candidiasis [DISEASES] ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,medicine ,microbiota ,Humans ,fungal variability ,Microbiome ,PATTERN-RECOGNITION RECEPTOR ,030304 developmental biology ,TOLL-LIKE RECEPTORS ,Host Microbial Interactions ,030306 microbiology ,Biology and Life Sciences ,infecciones bacterianas y micosis::micosis::candidiasis [ENFERMEDADES] ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,medicine.disease ,biology.organism_classification ,Commensalism ,[SDV.MP.BAC]Life Sciences [q-bio]/Microbiology and Parasitology/Bacteriology ,stomatognathic diseases ,Immunology ,microbiota variability ,Microbial Interactions ,Candida infections ,mycobiota ,fenómenos microbiológicos::interacciones huésped-microorganismo [FENÓMENOS Y PROCESOS] ,antifungal immunity - Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients., The complexity and variability of FunHoMic interactions between the fungal pathogen, its human host and the Microbiota strongly influence the development and outcomes of the superficial and systemic Candida albicans infections that plague human health worldwide.
- Published
- 2021
- Full Text
- View/download PDF
17. Human albumin enhances the pathogenic potential of Candida glabrata on vaginal epithelial cells
- Author
-
Selene Mogavero, Bernhard Hube, Toni Gabaldón, Mark S. Gresnigt, Sophia U. J. Hitzler, Marina Pekmezovic, Ann Kristin Kaune, Sophie Austermeier, Hrant Hovhannisyan, and Barcelona Supercomputing Center
- Subjects
Cell ,Yeast and Fungal Models ,Candida glabrata ,Pathogenesis ,Epithelial cells ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Biochemistry ,Epithelium ,Mice ,Vulvovaginal candidiasis (VVC) ,Animal Cells ,Microorganismes patògens ,Medicine and Health Sciences ,Biology (General) ,Cytotoxicity ,Candida ,Fungal Pathogens ,0303 health sciences ,Organic Compounds ,Vulvovaginal candidiasis ,Monosaccharides ,Eukaryota ,3. Good health ,Chemistry ,medicine.anatomical_structure ,Experimental Organism Systems ,Medical Microbiology ,Pathogenic microorganisms ,Physical Sciences ,Female ,Cellular Types ,Anatomy ,Pathogens ,Research Article ,Informàtica::Aplicacions de la informàtica::Bioinformàtica [Àrees temàtiques de la UPC] ,QH301-705.5 ,Immunology ,Carbohydrates ,Virulence ,Mycology ,Biology ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Albumins ,Virology ,Genetics ,medicine ,Animals ,Humans ,Candida Albicans ,Pathogenicity ,Microbial Pathogens ,Molecular Biology ,Candidiasis, Vulvovaginal ,030304 developmental biology ,030306 microbiology ,Albumin ,Organic Chemistry ,Host Cells ,Chemical Compounds ,Organisms ,Fungi ,Biology and Life Sciences ,Proteins ,Epithelial Cells ,Cell Biology ,RC581-607 ,biology.organism_classification ,Yeast ,In vitro ,Oxidative Stress ,Biological Tissue ,Glucose ,Animal Studies ,Unfolded protein response ,Parasitology ,Immunologic diseases. Allergy ,Viral Transmission and Infection ,Oxidative stress - Abstract
The opportunistic pathogen Candida glabrata is the second most frequent causative agent of vulvovaginal candidiasis (VVC), a disease that affects 70–75% of women at least once during their life. However, C. glabrata is almost avirulent in mice and normally incapable of inflicting damage to vaginal epithelial cells in vitro. We thus proposed that host factors present in vivo may influence C. glabrata pathogenicity. We, therefore, analyzed the impact of albumin, one of the most abundant proteins of the vaginal fluid. The presence of human, but not murine, albumin dramatically increased the potential of C. glabrata to damage vaginal epithelial cells. This effect depended on macropinocytosis-mediated epithelial uptake of albumin and subsequent proteolytic processing. The enhanced pathogenicity of C. glabrata can be explained by a combination of beneficial effects for the fungus, which includes an increased access to iron, accelerated growth, and increased adhesion. Screening of C. glabrata deletion mutants revealed that Hap5, a key regulator of iron homeostasis, is essential for the albumin-augmented damage potential. The albumin-augmented pathogenicity was reversed by the addition of iron chelators and a similar increase in pathogenicity was shown by increasing the iron availability, confirming a key role of iron. Accelerated growth not only led to higher cell numbers, but also to increased fungal metabolic activity and oxidative stress resistance. Finally, the albumin-driven enhanced damage potential was associated with the expression of distinct C. glabrata virulence genes. Transcriptional responses of the epithelial cells suggested an unfolded protein response (UPR) and ER-stress responses combined with glucose starvation induced by fast growing C. glabrata cells as potential mechanisms by which cytotoxicity is mediated.Collectively, we demonstrate that albumin augments the pathogenic potential of C. glabrata during interaction with vaginal epithelial cells. This suggests a role for albumin as a key player in the pathogenesis of VVC., Author summary Candida glabrata is the overall second causative species of candidiasis in humans, but little is known about the pathogenicity mechanisms of this yeast. C. glabrata is capable of causing lethal systemic candidiasis mostly in elderly immunocompromised patients, but is also a frequent cause of vulvovaginal candidiasis. These clinical insights suggest that C. glabrata has a high virulence potential, yet little pathogenicity is observed in both in vitro and in vivo infection models. The finding that human albumin, the most abundant protein in the human body, is boosting C. glabrata pathogenicity in vitro provides novel insights into C. glabrata pathogenicity mechanisms and shows that the presence of distinct human factors can have a significant influence on the virulence potential of a pathogenic microbe.
- Published
- 2021
- Full Text
- View/download PDF
18. Comparative host transcriptome in response to pathogenic fungi identifies common and species-specific transcriptional antifungal host response pathways
- Author
-
Mihai G. Netea, Frank L. van de Veerdonk, Rob ter Horst, Jean-Paul Latgé, Laszlo Groh, Intan M.W. Dewi, Yang Li, Rutger J. Röring, Vinod Kumar, Marina Pekmezovic, Mariolina Bruno, Agostinho Carvalho, Berenice Rösler, Vicky Matzaraki, Mark S. Gresnigt, CiiM, Zentrum für individualisierte Infektionsmedizin, Feodor-Lynen-Str.7, 30625 Hannover., and Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI)
- Subjects
Pattern recognition receptors ,R. oryzae ,Biophysics ,Rhizopus oryzae ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Host immune response ,Antifungal core host response ,Aspergillosis ,Biochemistry ,A. fumigatus ,Aspergillus fumigatus ,Microbiology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Transcriptomics of pathogenic fungi ,Structural Biology ,C. albicans ,Genetics ,medicine ,Opportunistic infections ,Candida albicans ,030304 developmental biology ,ComputingMethodologies_COMPUTERGRAPHICS ,0303 health sciences ,Immunometabolism ,biology ,Pattern recognition receptor ,Vascular damage Radboud Institute for Molecular Life Sciences [Radboudumc 16] ,biology.organism_classification ,medicine.disease ,RNAseq ,Corpus albicans ,3. Good health ,Computer Science Applications ,030220 oncology & carcinogenesis ,Cytokines ,TP248.13-248.65 ,Biotechnology ,Research Article - Abstract
Graphical abstract, Candidiasis, aspergillosis, and mucormycosis cause the majority of nosocomial fungal infections in immunocompromised patients. Using an unbiased transcriptional profiling in PBMCs exposed to the fungal species causing these infections, we found a core host response in healthy individuals that may govern effective fungal clearance: it consists of 156 transcripts, involving canonical and non-canonical immune pathways. Systematic investigation of key steps in antifungal host defense revealed fungal-specific signatures. As previously demonstrated, Candida albicans induced type I and Type II interferon-related pathways. In contrast, central pattern recognition receptor, reactive oxygen species production, and host glycolytic pathways were down-regulated in response to Rhizopus oryzae, which was associated with an ER-stress response. TLR5 was identified to be uniquely regulated by Aspergillus fumigatus and to control cytokine release in response to this fungus. In conclusion, our data reveals the transcriptional profiles induced by C. albicans, A. fumigatus, and R. oryzae, and describes both the common and specific antifungal host responses that could be exploited for novel therapeutic strategies.
- Published
- 2020
19. A systems genomics approach identifies SIGLEC15 as a susceptibility factor in recurrent vulvovaginal candidiasis
- Author
-
Mirco Dindo, Marije Oosting, Martin Jaeger, Christian Büll, Peer Arts, Leo A. B. Joosten, F.L. van de Veerdonk, Luigina Romani, M. Feng, Vinod Kumar, Monica Borghi, Mihai G. Netea, Joris A. Veltman, Michele Pinelli, L. van Emst, Matteo Puccetti, N. Xu, C. Constantini, Mark S. Gresnigt, Bart Jan Kullberg, Isis Ricaño-Ponce, X. Wang, J. ten Oever, Marilena Pariano, Cor Jacobs, Jack D. Sobel, J. Gutierrez Achury, Gosse J. Adema, Christian Gilissen, Cisca Wijmenga, RS: GROW - R4 - Reproductive and Perinatal Medicine, and Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI)
- Subjects
0301 basic medicine ,Candidate gene ,GENE POLYMORPHISM ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,PATHOGENESIS ,030106 microbiology ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Peripheral blood mononuclear cell ,Pathogenesis ,OSTEOCLAST DIFFERENTIATION ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Immune system ,MANNOSE-BINDING LECTIN ,EPIDEMIOLOGY ,Medicine ,Gene silencing ,Mannan-binding lectin ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,business.industry ,ALBICANS ,WOMEN ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,ASSOCIATION ,General Medicine ,PREVALENCE ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,030104 developmental biology ,CELLS ,Immunology ,Recurrent vulvovaginal candidiasis ,Gene polymorphism ,business - Abstract
Candida vaginitis is a frequent clinical diagnosis with up to 8% of women experiencing recurrent vulvovaginal candidiasis (RVVC) globally. RVVC is characterized by at least three episodes per year. Most patients with RVVC lack known risk factors, suggesting a role for genetic risk factors in this condition. Through integration of genomic approaches and immunological studies in two independent cohorts of patients with RVVC and healthy individuals, we identified genes and cellular processes that contribute to the pathogenesis of RVVC, including cellular morphogenesis and metabolism, and cellular adhesion. We further identified SIGLEC15, a lectin expressed by various immune cells that binds sialic acid-containing structures, as a candidate gene involved in RVVC susceptibility. Candida stimulation induced SIGLEC15 expression in human peripheral blood mononuclear cells (PBMCs) and a polymorphism in the SIGLEC15 gene that was associated with RVVC in the patient cohorts led to an altered cytokine profile after PBMC stimulation. The same polymorphism led to an increase in IL1B and NLRP3 expression after Candida stimulation in HeLa cells in vitro. Last, Siglec15 expression was induced by Candida at the vaginal surface of mice, where in vivo silencing of Siglec15 led to an increase in the fungal burden. Siglec15 silencing was additionally accompanied by an increase in polymorphonuclear leukocytes during the course of infection. Identification of these pathways and cellular processes contributes to a better understanding of RVVC and may open new therapeutic avenues.
- Published
- 2019
20. The fungal peptide toxin Candidalysin activates the NLRP3 inflammasome and causes cytolysis in mononuclear phagocytes
- Author
-
Bernhard Hube, Rebecca A. Drummond, Annika König, Lydia Kasper, Johannes Westman, Michail S. Lionakis, Mark S. Gresnigt, Paul-Albert Koenig, Julian R. Naglik, Jürgen Ruland, and Olaf Groß
- Subjects
0301 basic medicine ,Inflammasomes ,Interleukin-1beta ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,General Physics and Astronomy ,Phagosomes ,Macrophage ,lcsh:Science ,Candida albicans ,Phagocytes ,Multidisciplinary ,Cell Death ,biology ,Chemistry ,Caspase 1 ,Pyroptosis ,Inflammasome ,Mononuclear phagocyte system ,3. Good health ,Female ,Candidalysin ,medicine.drug ,Science ,030106 microbiology ,Bone Marrow Cells ,Article ,General Biochemistry, Genetics and Molecular Biology ,Microbiology ,Fungal Proteins ,Necrosis ,03 medical and health sciences ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Humans ,Inflammation ,Innate immune system ,Macrophages ,Dendritic Cells ,General Chemistry ,Mycotoxins ,biology.organism_classification ,Actins ,Mice, Inbred C57BL ,Cytolysis ,030104 developmental biology ,Leukocytes, Mononuclear ,Potassium ,lcsh:Q - Abstract
Clearance of invading microbes requires phagocytes of the innate immune system. However, successful pathogens have evolved sophisticated strategies to evade immune killing. The opportunistic human fungal pathogen Candida albicans is efficiently phagocytosed by macrophages, but causes inflammasome activation, host cytolysis, and escapes after hypha formation. Previous studies suggest that macrophage lysis by C. albicans results from early inflammasome-dependent cell death (pyroptosis), late damage due to glucose depletion and membrane piercing by growing hyphae. Here we show that Candidalysin, a cytolytic peptide toxin encoded by the hypha-associated gene ECE1, is both a central trigger for NLRP3 inflammasome-dependent caspase-1 activation via potassium efflux and a key driver of inflammasome-independent cytolysis of macrophages and dendritic cells upon infection with C. albicans. This suggests that Candidalysin-induced cell damage is a third mechanism of C. albicans-mediated mononuclear phagocyte cell death in addition to damage caused by pyroptosis and the growth of glucose-consuming hyphae., Phagocytic cells of the innate immune system play critical roles in defence against invading pathogens including the opportunistic pathogen Candida albicans. Here the authors show that C. albicans derived Candidalysin in addition to being a cell-damaging toxin to mononuclear phagocytes is a trigger of NLRP3 inflammasome activation in these cells.
- Published
- 2018
- Full Text
- View/download PDF
21. The Multifaceted Role of T-Helper Responses in Host Defense against Aspergillus fumigatus
- Author
-
Mark S. Gresnigt, Frank L. van de Veerdonk, and Intan M.W. Dewi
- Subjects
0301 basic medicine ,Microbiology (medical) ,Inflammation ,Plant Science ,Review ,Aspergillosis ,T-helper cells ,Aspergillus fumigatus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunopathology ,medicine ,immunopathology ,aspergillosis ,lcsh:QH301-705.5 ,Ecology, Evolution, Behavior and Systematics ,Aspergillus ,Innate immune system ,biology ,Acquired immune system ,biology.organism_classification ,medicine.disease ,adaptive immune response ,030104 developmental biology ,lcsh:Biology (General) ,Immunology ,medicine.symptom ,030215 immunology - Abstract
The ubiquitous opportunistic fungal pathogen Aspergillus fumigatus rarely causes infections in immunocompetent individuals. A healthy functional innate immune system plays a crucial role in preventing Aspergillus-infection. This pivotal role for the innate immune system makes it a main research focus in studying the pathogenesis of aspergillosis. Although sometimes overshadowed by the innate immune response, the adaptive immune response, and in particular T-helper responses, also represents a key player in host defense against Aspergillus. Virtually all T-helper subsets have been described to play a role during aspergillosis, with the Th1 response being crucial for fungal clearance. However; morbidity and mortality of aspergillosis can also be partly attributed to detrimental immune responses resulting from adaptive immune activation. Th2 responses benefit fungal persistence; and are the foundation of allergic forms of aspergillosis. The Th17 response has two sides; although crucial for granulocyte recruitment, it can be involved in detrimental immunopathology. Regulatory T-cells, the endogenous regulators of inflammatory responses, play a key role in controlling detrimental inflammatory responses during aspergillosis. The current knowledge of the adaptive immune response against A. fumigatus is summarized in this review. A better understanding on how T-helper responses facilitate clearance of Aspergillus-infection and control inflammation can be the fundamental basis for understanding the pathogenesis of aspergillosis and for the development of novel host-directed therapies.
- Published
- 2017
22. Rewiring monocyte glucose metabolism via C-type lectin signaling protects against disseminated candidiasis
- Author
-
Mark S. Gresnigt, Sanne P. Smeekens, Carlos Ardavín, Frank L. van de Veerdonk, Rob ter Horst, Richard A. Notebaart, Jorge Domínguez-Andrés, Jacqueline M. Ratter, Mihai G. Netea, Ekta Lachmandas, Lily Boutens, Rob J.W. Arts, and Leo A. B. Joosten
- Subjects
Metabolic Processes ,0301 basic medicine ,Physiology ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Yeast and Fungal Models ,Pathology and Laboratory Medicine ,Biochemistry ,Monocytes ,Levensmiddelenmicrobiologie ,White Blood Cells ,Mice ,0302 clinical medicine ,Glucose Metabolism ,Animal Cells ,C-type lectin ,Immune Physiology ,Medicine and Health Sciences ,Biology (General) ,Candida albicans ,Candida ,Fungal Pathogens ,Innate Immune System ,biology ,Fungal Diseases ,Candidiasis ,Eukaryota ,3. Good health ,Infectious Diseases ,medicine.anatomical_structure ,Experimental Organism Systems ,Medical Microbiology ,Carbohydrate Metabolism ,Cytokines ,Pathogens ,Cellular Types ,Signal transduction ,Glycolysis ,Research Article ,Signal Transduction ,Cell Physiology ,QH301-705.5 ,Immune Cells ,Immunology ,Mycology ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Immunity ,Virology ,Genetics ,medicine ,Candida Albicans ,Animals ,Humans ,Life Science ,Lectins, C-Type ,Microbial Pathogens ,Molecular Biology ,Blood Cells ,Glutaminolysis ,Innate immune system ,Monocyte ,Organisms ,Fungi ,Biology and Life Sciences ,Cell Biology ,Molecular Development ,RC581-607 ,medicine.disease ,biology.organism_classification ,Yeast ,Immunity, Innate ,Cell Metabolism ,Metabolism ,Yeast Infections ,Glucose ,030104 developmental biology ,Immune System ,Food Microbiology ,Parasitology ,Systemic candidiasis ,Immunologic diseases. Allergy ,Developmental Biology ,030215 immunology - Abstract
Monocytes are innate immune cells that play a pivotal role in antifungal immunity, but little is known regarding the cellular metabolic events that regulate their function during infection. Using complementary transcriptomic and immunological studies in human primary monocytes, we show that activation of monocytes by Candida albicans yeast and hyphae was accompanied by metabolic rewiring induced through C-type lectin-signaling pathways. We describe that the innate immune responses against Candida yeast are energy-demanding processes that lead to the mobilization of intracellular metabolite pools and require induction of glucose metabolism, oxidative phosphorylation and glutaminolysis, while responses to hyphae primarily rely on glycolysis. Experimental models of systemic candidiasis models validated a central role for glucose metabolism in anti-Candida immunity, as the impairment of glycolysis led to increased susceptibility in mice. Collectively, these data highlight the importance of understanding the complex network of metabolic responses triggered during infections, and unveil new potential targets for therapeutic approaches against fungal diseases., Author summary Fungal infections are a major health concern for immunocompromised individuals due to the lack of success of the currently available antifungal therapies. Unveiling the metabolic processes involved in the immune function offers a promising opportunity for the development of new therapeutic approaches against these infections. In this report, we describe how changes in monocyte glucose metabolism are crucial for host defense against infections caused by the opportunistic pathogenic yeast Candida albicans. We report how the participation of various metabolic routes, such as glycolysis, oxidative phosphorylation and the pentose phosphate pathway, were differentially required after yeast or hyphal exposure, depending on the cellular energy requirements for each response. The proper control of metabolic reprogramming of immune cells was crucial to afford protection against fungal infections in vivo.
- Published
- 2017
23. A guiding map for inflammation
- Author
-
Luke A. J. O'Neill, Peter Pickkers, Tom van der Poll, Paul M. Ridker, David A. Schwartz, Michel Chonchol, Mihai G. Netea, Frank L. van de Veerdonk, Charles A. Dinarello, Jos W. M. van der Meer, Frances R. Balkwill, Thomas Mandrup-Poulsen, Mark S. Gresnigt, Britta Siegmund, Leo A. B. Joosten, Joost Schalkwijk, Kristen L. Nowak, Clifford J. Steer, Fabio Cominelli, Kingston H. G. Mills, Daniel L. Kastner, Peter Libby, Marc Y. Donath, Evangelos J. Giamarellos-Bourboulis, Martin Korte, Douglas T. Golenbock, Michael T. Heneka, Eicke Latz, Hal M. Hoffman, Alberto Mantovani, Herbert Tilg, Richard S. Hotchkiss, AII - Inflammatory diseases, Infectious diseases, Center of Experimental and Molecular Medicine, and Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7, 38124 Braunschweig, Germany.
- Subjects
0301 basic medicine ,Immunology ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Inflammation ,Cellular Immunology ,Bioinformatics ,Communicable Diseases ,Article ,Biological pathway ,immunology [Immunity, Cellular] ,03 medical and health sciences ,immunology [Inflammation] ,Immunity ,medicine ,Immunology and Allergy ,Humans ,ddc:610 ,Immunity, Cellular ,business.industry ,immunology [Communicable Diseases] ,3. Good health ,Immunity, Humoral ,030104 developmental biology ,Chronic disease ,immunology [Immunity, Humoral] ,Acute Disease ,Chronic Disease ,medicine.symptom ,business ,Inflammatory diseases Radboud Institute for Molecular Life Sciences [Radboudumc 5] - Abstract
Contains fulltext : 177132.pdf (Publisher’s version ) (Closed access) Biologists, physicians and immunologists have contributed to the understanding of the cellular participants and biological pathways involved in inflammation. Here, we provide a general guide to the cellular and humoral contributors to inflammation as well as to the pathways that characterize inflammation in specific organs and tissues.
- Published
- 2017
- Full Text
- View/download PDF
24. The Itaconate Pathway Is a Central Regulatory Node Linking Innate Immune Tolerance and Trained Immunity
- Author
-
Mark S. Gresnigt, Cisca Wijmenga, Henk Stunnenberg, Alessandro Michelucci, Peter Pickkers, Simone J.C.F.M. Moorlag, Marije Oosting, Leo A. B. Joosten, Rob J.W. Arts, Laszlo Groh, Brendon P. Scicluna, Jorge Domínguez-Andrés, Jelle Zwaag, Mihai G. Netea, Rebecca M. Koch, Vinod Kumar, Boris Novakovic, Tom van der Poll, Matthijs Kox, Rob ter Horst, Yang Li, Center of Experimental and Molecular Medicine, AII - Inflammatory diseases, AII - Infectious diseases, Epidemiology and Data Science, Infectious diseases, and Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI)
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Male ,Physiology ,Carboxy-Lyases ,animal diseases ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Stimulation ,Immune tolerance ,sepsis ,Mice ,trained immunity ,0302 clinical medicine ,immunoparalysis ,Escherichia coli Infections ,tolerance ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Healthy Volunteers ,Succinate Dehydrogenase ,monocytes ,Adult ,LPS ,Adolescent ,Secondary infection ,chemical and pharmacologic phenomena ,macromolecular substances ,Biology ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Sepsis ,03 medical and health sciences ,Young Adult ,Immune system ,All institutes and research themes of the Radboud University Medical Center ,Immunity ,medicine ,Immune Tolerance ,Animals ,Humans ,Epigenetics ,Molecular Biology ,Innate immune system ,epigenetics ,Succinates ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,succinate ,Endotoxemia ,Immunity, Innate ,030104 developmental biology ,RAW 264.7 Cells ,itaconate ,Immunology ,bacteria ,metabolism ,030217 neurology & neurosurgery - Abstract
Sepsis involves simultaneous hyperactivation of the immune system and immune paralysis, leading to both organ dysfunction and increased susceptibility to secondary infections. Acute activation of myeloid cells induced itaconate synthesis, which subsequently mediated innate immune tolerance in human monocytes. In contrast, induction of trained immunity by β-glucan counteracted tolerance induced in a model of human endotoxemia by inhibiting the expression of immune-responsive gene 1 (IRG1), the enzyme that controls itaconate synthesis. β-Glucan also increased the expression of succinate dehydrogenase (SDH), contributing to the integrity of the TCA cycle and leading to an enhanced innate immune response after secondary stimulation. The role of itaconate was further validated by IRG1 and SDH polymorphisms that modulate induction of tolerance and trained immunity in human monocytes. These data demonstrate the importance of the IRG1-itaconate-SDH axis in the development of immune tolerance and training and highlight the potential of β-glucan-induced trained immunity to revert immunoparalysis.
- Published
- 2019
25. Rewiring cellular metabolism via the AKT/mTOR pathway contributes to host defence against Mycobacterium tuberculosis in human and murine cells
- Author
-
Mark S. Gresnigt, Vinod Kumar, Mihai G. Netea, Rinke Stienstra, Xinhui Wang, Shih-Chin Cheng, Reinout van Crevel, Tom H. M. Ottenhoff, Ekta Lachmandas, Lily Boutens, Arjan van Laarhoven, Macarena Beigier-Bompadre, Anne Ammerdorffer, Thirumala-Devi Kanneganti, Cisca Wijmenga, Dirk J. de Jong, Stefan H. E. Kaufmann, Leo A. B. Joosten, and Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI)
- Subjects
0301 basic medicine ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Oxidative Phosphorylation ,Mice ,0302 clinical medicine ,Leukocytes ,Immunology and Allergy ,TLR2 ,Glycolysis ,Research Articles ,biology ,TOR Serine-Threonine Kinases ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,3. Good health ,Cell biology ,Anti-Bacterial Agents ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,mTOR ,Research Article|Basic ,Signal transduction ,Signal Transduction ,Immunology ,Mononuclear ,Immunity to infection ,Other Research Radboud Institute for Molecular Life Sciences [Radboudumc 0] ,Oxidative phosphorylation ,Mycobacterium tuberculosis ,03 medical and health sciences ,Journal Article ,Animals ,Humans ,Tuberculosis ,Basic ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Sirolimus ,Immunometabolism ,Gene Expression Profiling ,biology.organism_classification ,Toll-Like Receptor 2 ,Citric acid cycle ,030104 developmental biology ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,Glucose ,Anaerobic glycolysis ,Leukocytes, Mononuclear ,Proto-Oncogene Proteins c-akt - Abstract
Contains fulltext : 171426.pdf (Publisher’s version ) (Open Access) Cells in homeostasis metabolize glucose mainly through the tricarboxylic acid cycle and oxidative phosphorylation, while activated cells switch their basal metabolism to aerobic glycolysis. In this study, we examined whether metabolic reprogramming toward aerobic glycolysis is important for the host response to Mycobacterium tuberculosis (Mtb). Through transcriptional and metabolite analysis we show that Mtb induces a switch in host cellular metabolism toward aerobic glycolysis in human peripheral blood mononuclear cells (PBMCs). The metabolic switch is TLR2 dependent but NOD2 independent, and is mediated in part through activation of the AKT-mTOR (mammalian target of rapamycin) pathway. We show that pharmacological inhibition of the AKT/mTOR pathway inhibits cellular responses to Mtb both in vitro in human PBMCs, and in vivo in a model of murine tuberculosis. Our findings reveal a novel regulatory layer of host responses to Mtb that will aid understanding of host susceptibility to Mtb, and which may be exploited for host-directed therapy.
- Published
- 2016
- Full Text
- View/download PDF
26. Integrated transcriptomic and proteomic analyses of P. falciparum gametocytes: molecular insight into sex-specific processes and translational repression
- Author
-
Teun Bousema, Annet Italiaander, Edwin Lasonder, Ben C. L. van Schaijk, Chris J. Janse, Shahid M. Khan, Rob Woestenenk, Philip R. Kensche, Sanna R. Rijpma, Wieteke A. M. Hoeijmakers, Gunnar R. Mair, Martijn W. Vos, Richárd Bártfai, Robert W. Sauerwein, and Mark S. Gresnigt
- Subjects
0301 basic medicine ,DNA Replication ,Male ,Proteome ,030106 microbiology ,Plasmodium falciparum ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Data Resources and Analyses ,Biology ,Gametogenesis ,Transcriptome ,03 medical and health sciences ,parasitic diseases ,Genetics ,Gametocyte ,medicine ,Humans ,Malaria, Falciparum ,Gene ,Molecular Biology ,Sex Characteristics ,Sexual differentiation ,C100 ,biology.organism_classification ,C900 ,Chromatin ,3. Good health ,medicine.anatomical_structure ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,Gene Expression Regulation ,Protein Biosynthesis ,Gamete ,Female ,Metabolic Networks and Pathways - Abstract
Contains fulltext : 167311.pdf (Publisher’s version ) (Open Access) Sexual differentiation of malaria parasites into gametocytes in the vertebrate host and subsequent gamete fertilization in mosquitoes is essential for the spreading of the disease. The molecular processes orchestrating these transitions are far from fully understood. Here, we report the first transcriptome analysis of male and female Plasmodium falciparum gametocytes coupled with a comprehensive proteome analysis. In male gametocytes there is an enrichment of proteins involved in the formation of flagellated gametes; proteins involved in DNA replication, chromatin organization and axoneme formation. On the other hand, female gametocytes are enriched in proteins required for zygote formation and functions after fertilization; protein-, lipid- and energy-metabolism. Integration of transcriptome and proteome data revealed 512 highly expressed maternal transcripts without corresponding protein expression indicating large scale translational repression in P. falciparum female gametocytes for the first time. Despite a high degree of conservation between Plasmodium species, 260 of these 'repressed transcripts' have not been previously described. Moreover, for some of these genes, protein expression is only reported in oocysts and sporozoites indicating that repressed transcripts can be partitioned into short- and long-term storage. Finally, these data sets provide an essential resource for identification of vaccine/drug targets and for further mechanistic studies.
- Published
- 2016
- Full Text
- View/download PDF
27. Broad defects in the energy metabolism of leukocytes underlie immunoparalysis in sepsis
- Author
-
Matthijs Kox, Ganesh R. Manjeri, Jenneke Leentjens, Ekta Lachmandas, Shih-Chin Cheng, Anne Jan van der Meer, Frank L. van de Veerdonk, Olaf L. Cremer, Jori A. L. Wagenaars, Brendon P. Scicluna, Rob J.W. Arts, Evangelos J. Giamarellos-Bourboulis, Mark S. Gresnigt, Marcus J. Schultz, Tom van der Poll, Peter Pickkers, Marc J. M. Bonten, Mihai G. Netea, Leo A. B. Joosten, Peter H.G.M. Willems, AII - Amsterdam institute for Infection and Immunity, APH - Amsterdam Public Health, Epidemiology and Data Science, Intensive Care Medicine, Infectious diseases, and Center of Experimental and Molecular Medicine
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Male ,Antifungal Agents ,Lipopolysaccharide ,medicine.medical_treatment ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Monocytes ,Oxidative Phosphorylation ,chemistry.chemical_compound ,Mice ,Adenosine Triphosphate ,Leukocytes ,Immunology and Allergy ,Glycolysis ,Prospective Studies ,Non-U.S. Gov't ,Escherichia coli Infections ,Research Support, Non-U.S. Gov't ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Middle Aged ,Cytokine ,Cytokines ,Female ,Adult ,Immunoblotting ,Immunology ,Observational Study ,Oxidative phosphorylation ,Biology ,Research Support ,Sepsis ,03 medical and health sciences ,Interferon-gamma ,Young Adult ,Oxygen Consumption ,medicine ,Immune Tolerance ,Journal Article ,Animals ,Aspergillosis ,Humans ,Candidiasis, Invasive ,Lactic Acid ,Innate immune system ,Macrophages ,medicine.disease ,NAD ,Endotoxemia ,Immunity, Innate ,Metabolic pathway ,030104 developmental biology ,chemistry ,Anaerobic glycolysis ,Energy Metabolism ,Transcriptome - Abstract
Contains fulltext : 172049.pdf (Publisher’s version ) (Closed access) The acute phase of sepsis is characterized by a strong inflammatory reaction. At later stages in some patients, immunoparalysis may be encountered, which is associated with a poor outcome. By transcriptional and metabolic profiling of human patients with sepsis, we found that a shift from oxidative phosphorylation to aerobic glycolysis was an important component of initial activation of host defense. Blocking metabolic pathways with metformin diminished cytokine production and increased mortality in systemic fungal infection in mice. In contrast, in leukocytes rendered tolerant by exposure to lipopolysaccharide or after isolation from patients with sepsis and immunoparalysis, a generalized metabolic defect at the level of both glycolysis and oxidative metabolism was apparent, which was restored after recovery of the patients. Finally, the immunometabolic defects in humans were partially restored by therapy with recombinant interferon-gamma, which suggested that metabolic processes might represent a therapeutic target in sepsis.
- Published
- 2016
28. 967. Inhibition of Host Neuraminidase Increases Susceptibility to Invasive Pulmonary Aspergillosis
- Author
-
Intan M.W. Dewi, Lore vanderBeeke, Mark S. Gresnigt, Agostinho Carvalho, Greetje Vande Velde, Joost Wauters, Katrien Lagrou, Christina Cunha, Frank L. van de Veerdonk, Agustin Resendiz, and Martin Jaeger
- Subjects
0301 basic medicine ,Oseltamivir ,medicine.medical_treatment ,030106 microbiology ,Aspergillosis ,Microbiology ,Pathogenesis ,03 medical and health sciences ,chemistry.chemical_compound ,Abstracts ,Immune system ,A. Oral Abstracts ,medicine ,Aspergillus ,biology ,business.industry ,medicine.disease ,biology.organism_classification ,Infectious Diseases ,Cytokine ,Oncology ,chemistry ,biology.protein ,Immunocompetence ,business ,Neuraminidase - Abstract
Background Influenza-associated aspergillosis (IAA) is an emerging fungal infection with high mortality and morbidity and the pathogenesis of this disease is not well understood. Interestingly, the number of IAA case reports has increased since the widespread use of neuraminidase inhibitors, such as oseltamivir in 2009. We set out to determine whether oseltamivir could contribute to the pathogenesis of IAA by modulating host responses. Methods First, peripheral blood mononuclear cells (PBMCs) and neutrophils from healthy donors were stimulated with neuraminidase (NA)-treated A. fumigatus or were pre-exposed to NA prior to stimulation with Aspergillus conidia. In addition, PBMCs and neutrophils were pretreated with oseltamivir carboxylate prior to stimulation. Cytokines were measured from supernatants after 24 hours of incubation at 37°C. C57BL/6 and BALB/c mice were treated with oseltamivir prior to intranasal challenge with A. fumigatus. Immunosuppression was induced by corticosteroid or cyclophosphamide. Results We demonstrate that Aspergillus treated with NA induced an enhanced immune response. Moreover, PBMCs and neutrophils treated with NA produced increased cytokine responses. Blocking NA in vitro with oseltamivir reduced Aspergillus-induced cytokine responses. Next we investigated the effects of blocking neuraminidase activity with oseltamivir in vivo. Immunocompetent mice and mice treated with corticosteroids showed increased mortality, lung fungal burden, and decreased cytokine production when treated with oseltamivir. These effects were not observed in cyclophosphamide-treated mice, suggesting that the effects of NA activity in anti-Aspergillus host defense acts mainly via myeloid cells. Conclusion Our results provide evidence that host neuraminidase activity is important for protective anti-Aspergillus immune responses. Treatment with oseltamivir, thus blocking host NA activity, in a setting of corticosteroid use might therefore increase susceptibility to Aspergillus infection. These results warrant further study on the role of neuraminidase and the effects of oseltamivir on susceptibility to invasive pulmonary aspergillosis during active influenza infection. Disclosures All authors: No reported disclosures.
- Published
- 2018
29. Recognition of coxiella burnetii by toll-like receptors and nucleotide-binding oligomerization domain-like receptors
- Author
-
Marije Oosting, Mihai G. Netea, Tom Sprong, Johanna M.J. Rebel, Teske Schoffelen, Mark S. Gresnigt, Shahla Abdollahi-Roodsaz, Marcel van Deuren, Anne Ammerdorffer, Martijn H. den Brok, Hendrik I.J. Roest, Thirumala-Devi Kanneganti, Dirk J. de Jong, and Leo A. B. Joosten
- Subjects
Male ,Epidemiology ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Nod2 Signaling Adaptor Protein ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Nod1 Signaling Adaptor Protein ,NOD2 ,NOD-like receptors ,NOD1 ,Immunology and Allergy ,Receptor ,innate immunity ,Cells, Cultured ,Mice, Knockout ,Immunity, Cellular ,Toll-like receptor ,biology ,Middle Aged ,singlenucleotide polymorphism ,Infectious Diseases ,Coxiella burnetii ,Cytokines ,Female ,Inflammatory diseases Radboud Institute for Molecular Life Sciences [Radboudumc 5] ,Adult ,MAP Kinase Signaling System ,Bioinformatica & Diermodellen ,Other Research Radboud Institute for Molecular Life Sciences [Radboudumc 0] ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Microbiology ,Young Adult ,Bio-informatics & Animal models ,Animals ,Humans ,Epidemiology, Bio-informatics & Animal models ,Q fever ,Aged ,Epidemiologie ,Innate immune system ,pattern recognition ,biology.organism_classification ,bacterial infections and mycoses ,Virology ,Toll-like receptors ,Mice, Inbred C57BL ,TLR2 ,TLR6 ,Epidemiologie, Bioinformatica & Diermodellen ,bacteria - Abstract
Contains fulltext : 153819.pdf (Publisher’s version ) (Closed access) BACKGROUND: Infection with Coxiella burnetii can lead to acute and chronic Q fever. Toll-like receptor 1 (TLR1), TLR2, TLR4, TLR6, nucleotide-binding oligomerization domain receptor 1 (NOD1), NOD2, and the mitogen-activated protein kinases are central in the innate immune response against microorganisms, but little is known about their role in the recognition of C. burnetii in humans. METHODS: Human peripheral blood mononuclear cells (PBMCs) were stimulated with C. burnetii Nine Mile and the Dutch outbreak isolate C. burnetii 3262. TLRs were inhibited using specific antibodies or antagonists. Additionally, the influence of human polymorphisms in TLRs and Nod-like receptors (NLRs) on C. burnetii-induced cytokine production was assessed. RESULTS: Inhibition of TLR2, p38, JNK, and ERK led to decreased cytokine responses in C. burnetii-stimulated human PBMCs. Humans with polymorphisms in TLR1 and NOD2 had reduced cytokine production, compared with humans with wild-type genotypes, after stimulation. Interestingly, polymorphisms in TLR6 led to decreased cytokine production after C. burnetii 3262 stimulation but not after C. burnetii Nine Mile stimulation. CONCLUSIONS: The TLR1/TLR2 heterodimer and NOD2 are important recognition receptors for the induction of cytokine responses against C. burnetii in humans. Furthermore, an interesting finding was the divergent recognition of C. burnetii Nine Mile and C. burnetii 3262.
- Published
- 2015
30. A Polysaccharide Virulence Factor from Aspergillus fumigatus Elicits Anti-inflammatory Effects through Induction of Interleukin-1 Receptor Antagonist
- Author
-
Silvia Bozza, Leo A. B. Joosten, Mihai G. Netea, Katharina L. Becker, Mark S. Gresnigt, Thierry Fontaine, Silvia Moretti, Frank L. van de Veerdonk, Shahla Abdollahi-Roodsaz, Charles A. Dinarello, Wim B. van der Berg, Antonella De Luca, Jean-Paul Latgé, Luigina Romani, Radboud University Medical Center [Nijmegen], Università degli Studi di Perugia (UNIPG), Department of rheumatology, Radboud university [Nijmegen], University of Colorado [Denver], Aspergillus, Institut Pasteur [Paris], FLvdV was supported by a Veni grant of the Netherlands Organization for Scientific Research, and a NCMLS grant from RUNMC. MGN was supported by a Vici grant of the Netherlands Organization for Scientific Research. LR was supported by the Specific Targeted Research Project 'ALLFUN' FP7-HEALTH-2010-260338. TF and JPL were supported by grants from ESF (Fuminomics 06-RNP-132), FP7 (ALLFUN), EraNet Pathogenomics (ANR-08-PATH-009-02) Agence Nationale de la Recherche (ANR-06-EMPB-011-01), ANR-08-PATH-0009,ANTIFUN(2008), ANR-06-EMPB-0011,GALNACGAL,Développement d'un nouveau test de diagnostic de l'aspergillose(2006), European Project: 260338,EC:FP7:HEALTH,FP7-HEALTH-2010-single-stage,ALLFUN(2010), Università degli Studi di Perugia = University of Perugia (UNIPG), Radboud University [Nijmegen], and Institut Pasteur [Paris] (IP)
- Subjects
medicine.medical_treatment ,viruses ,Galactosaminogalactan ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Pathogenesis ,Aspergillosis ,MESH: Mice, Knockout ,Biochemistry ,Aspergillus fumigatus ,MESH: Fungal Polysaccharides ,chemistry.chemical_compound ,Mice ,MESH: Animals ,lcsh:QH301-705.5 ,[SDV.MP.MYC]Life Sciences [q-bio]/Microbiology and Parasitology/Mycology ,Mice, Knockout ,MESH: Cytokines ,Mice, Inbred BALB C ,biology ,MESH: Enzyme-Linked Immunosorbent Assay ,Receptor antagonist ,3. Good health ,MESH: Interleukin 1 Receptor Antagonist Protein ,MESH: Leukocytes, Mononuclear ,Cytokine ,Infectious Diseases ,Medical Microbiology ,Medicine ,Cytokines ,Female ,MESH: Aspergillus fumigatus ,Inflammatory diseases Radboud Institute for Molecular Life Sciences [Radboudumc 5] ,Research Article ,lcsh:Immunologic diseases. Allergy ,medicine.drug_class ,Virulence Factors ,Immunology ,MESH: Mice, Inbred BALB C ,Enzyme-Linked Immunosorbent Assay ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Mycology ,Microbiology ,Proinflammatory cytokine ,Immune system ,Polysaccharides ,Virology ,Genetics ,medicine ,Animals ,Humans ,MESH: Aspergillosis ,Molecular Biology ,MESH: Mice ,Biology ,MESH: Virulence Factors ,MESH: Humans ,Fungal Polysaccharides ,biology.organism_classification ,medicine.disease ,Disease Models, Animal ,Interleukin 1 Receptor Antagonist Protein ,Interleukin 1 receptor antagonist ,MESH: Polysaccharides ,lcsh:Biology (General) ,chemistry ,Leukocytes, Mononuclear ,Parasitology ,MESH: Disease Models, Animal ,lcsh:RC581-607 ,MESH: Female - Abstract
The galactosaminogalactan (GAG) is a cell wall component of Aspergillus fumigatus that has potent anti-inflammatory effects in mice. However, the mechanisms responsible for the anti-inflammatory property of GAG remain to be elucidated. In the present study we used in vitro PBMC stimulation assays to demonstrate, that GAG inhibits proinflammatory T-helper (Th)1 and Th17 cytokine production in human PBMCs by inducing Interleukin-1 receptor antagonist (IL-1Ra), a potent anti-inflammatory cytokine that blocks IL-1 signalling. GAG cannot suppress human T-helper cytokine production in the presence of neutralizing antibodies against IL-1Ra. In a mouse model of invasive aspergillosis, GAG induces IL-1Ra in vivo, and the increased susceptibility to invasive aspergillosis in the presence of GAG in wild type mice is not observed in mice deficient for IL-1Ra. Additionally, we demonstrate that the capacity of GAG to induce IL-1Ra could also be used for treatment of inflammatory diseases, as GAG was able to reduce severity of an experimental model of allergic aspergillosis, and in a murine DSS-induced colitis model. In the setting of invasive aspergillosis, GAG has a significant immunomodulatory function by inducing IL-1Ra and notably IL-1Ra knockout mice are completely protected to invasive pulmonary aspergillosis. This opens new treatment strategies that target IL-1Ra in the setting of acute invasive fungal infection. However, the observation that GAG can also protect mice from allergy and colitis makes GAG or a derivative structure of GAG a potential treatment compound for IL-1 driven inflammatory diseases., Author Summary Aspergillus fumigatus is an opportunistic pathogenic fungus that primarily causes infections in the immunocompromised host. It is known that Aspergillus employs various strategies to evade immune recognition by the host's immune system. Recently, galactosaminogalactan (GAG), a new component of the Aspergillus cell wall, was discovered to have potent anti-inflammatory effects in mice making them more susceptible to Aspergillosis. In the current study we found that this anti-inflammatory property of GAG was due to its capacity to induce the potent anti-inflammatory cytokine interleukin-1 Receptor antagonist. This cytokine interferes with IL-1 signalling and thereby can reduce IL-1–induced immune responses such as T-cell responses. We also found that the induction of this anti-inflammatory cytokine by GAG correlates with increased fungal burden, and mice deficient for this cytokine were protected against aspergillosis. Additionally, we show that the capacity of GAG to induce the natural regulator of IL-1 signalling could be used in the treatment of IL-1–mediated disease such as allergy and colitis. Our study provides new insights on the immunoregulatory activity of GAG and opens up possibilities to exploit the anti-inflammatory potential of GAG as a therapy for inflammatory diseases.
- Published
- 2014
- Full Text
- View/download PDF
31. Functional genomics identifies type I interferon pathway as central for host defense against Candida albicans
- Author
-
John R. Perfect, Marije Oosting, Aylwin Ng, Bart Jan Kullberg, Cisca Wijmenga, Alexander Hoischen, Theo S. Plantinga, William K. Scott, Cleo C. van Diemen, Eugène T P Verwiel, Vinod Kumar, Sanne P. Smeekens, Melissa D. Johnson, Shih-Chin Cheng, Suzanne van Sommeren, Karin Fransen, Ramnik J. Xavier, Jos W. M. van der Meer, Peer Arts, Mark S. Gresnigt, Mihai G. Netea, Leo A. B. Joosten, Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), Smeekens, Sanne P, Ng, Aylwin, Kumar, Vinod, Johnson, Melissa D, Arts, Peer, and Xavier, Ramnik J
- Subjects
Transcription, Genetic ,medicine.medical_treatment ,General Physics and Astronomy ,0302 clinical medicine ,Interferon ,Candida albicans ,EPIDEMIOLOGY ,Cluster Analysis ,RNA-SEQ ,Chronic mucocutaneous candidiasis ,MACROPHAGES ,LISTERIA-MONOCYTOGENES ,GENE-EXPRESSION ,0303 health sciences ,Multidisciplinary ,biology ,Candidiasis, Chronic Mucocutaneous ,Interferon type I ,Genomics ,3. Good health ,Pathogenesis and modulation of inflammation [N4i 1] ,Cytokine ,STAT1 Transcription Factor ,Host-Pathogen Interactions ,Interferon Type I ,medicine.symptom ,BLOOD-STREAM INFECTIONS ,Functional genomics ,medicine.drug ,Signal Transduction ,UNITED-STATES ,Inflammation ,Invasive mycoses and compromised host Infection and autoimmunity [N4i 2] ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,CLASSIFICATION ,Article ,Invasive mycoses and compromised host [N4i 2] ,03 medical and health sciences ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetics and epigenetic pathways of disease Translational research [NCMLS 6] ,030304 developmental biology ,RECOGNITION ,Candidemia ,Pathogenesis and modulation of inflammation Infection and autoimmunity [N4i 1] ,General Chemistry ,Th1 Cells ,biology.organism_classification ,medicine.disease ,Gene Expression Regulation ,HOSPITALS ,Case-Control Studies ,Immunology ,Mutation ,Th17 Cells ,Systemic candidiasis ,Genetics and epigenetic pathways of disease Genomic disorders and inherited multi-system disorders [NCMLS 6] ,030215 immunology - Abstract
Contains fulltext : 118453.pdf (Publisher’s version ) (Open Access) Candida albicans is the most common human fungal pathogen causing mucosal and systemic infections. However, human antifungal immunity remains poorly defined. Here by integrating transcriptional analysis and functional genomics, we identified Candida-specific host defence mechanisms in humans. Candida induced significant expression of genes from the type I interferon pathway in human peripheral blood mononuclear cells. This unexpectedly prominent role of type I interferon pathway in anti-Candida host defence was supported by additional evidence. Polymorphisms in type I interferon genes modulated Candida-induced cytokine production and were correlated with susceptibility to systemic candidiasis. In in vitro experiments, type I interferons skewed Candida-induced inflammation from a Th17 response towards a Th1 response. Patients with chronic mucocutaneous candidiasis displayed defective expression of genes in the type I interferon pathway. These findings indicate that the type I interferon pathway is a main signature of Candida-induced inflammation and has a crucial role in anti-Candida host defence in humans.
- Published
- 2013
32. Lactobacillus rhamnosus colonisation antagonizes Candida albicans by forcing metabolic adaptations that compromise pathogenicity
- Author
-
Raquel Alonso-Roman, Antonia Last, Mohammad H. Mirhakkak, Jakob L. Sprague, Lars Möller, Peter Großmann, Katja Graf, Rena Gratz, Selene Mogavero, Slavena Vylkova, Gianni Panagiotou, Sascha Schäuble, Bernhard Hube, and Mark S. Gresnigt
- Subjects
Science - Abstract
Commensal bacteria such as Lactobacillus rhamnosus can inhibit the pathogenicity of the fungus Candida albicans. Here, Alonso-Roman et al. investigate the interplay between C. albicans, L. rhamnosus and intestinal epithelial cells, showing that changes in the metabolic environment, induced by the bacteria, trigger adaptations in C. albicans that reduce fungal pathogenicity.
- Published
- 2022
- Full Text
- View/download PDF
33. Genetic determinants of fungi-induced ROS production are associated with the risk of invasive pulmonary aspergillosis
- Author
-
Vasiliki Matzaraki, Alexandra Beno, Martin Jaeger, Mark S. Gresnigt, Nick Keur, Collins Boahen, Cristina Cunha, Samuel M. Gonçalves, Luis Leite, João F. Lacerda, António Campos, Jr., Frank L. van de Veerdonk, Leo Joosten, Mihai G. Netea, Agostinho Carvalho, and Vinod Kumar
- Subjects
C. albicans ,A. fumigatus ,Reactive oxygen species ,QTLs ,Stem-cell transplant recipients ,Invasive aspergillosis ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Reactive oxygen species (ROS) are an essential component of the host defense against fungal infections. However, little is known about how common genetic variation affects ROS-mediated antifungal host defense. In the present study, we investigated the genetic factors that regulate ROS production capacity in response to the two human fungal pathogens: Candida albicans and Aspergillus fumigatus. We investigated fungal-stimulated ROS production by immune cells isolated from a population-based cohort of approximately 200 healthy individuals (200FG cohort), and mapped ROS-quantitative trait loci (QTLs). We identified several genetic loci that regulate ROS levels (P
- Published
- 2022
- Full Text
- View/download PDF
34. Comparative host transcriptome in response to pathogenic fungi identifies common and species-specific transcriptional antifungal host response pathways
- Author
-
Mariolina Bruno, Intan M.W. Dewi, Vicky Matzaraki, Rob ter Horst, Marina Pekmezovic, Berenice Rösler, Laszlo Groh, Rutger J. Röring, Vinod Kumar, Yang Li, Agostinho Carvalho, Mihai G. Netea, Jean-Paul Latgé, Mark S. Gresnigt, and Frank L. van de Veerdonk
- Subjects
Host immune response ,RNAseq ,Transcriptomics of pathogenic fungi ,Opportunistic infections ,C. albicans ,A. fumigatus ,Biotechnology ,TP248.13-248.65 - Abstract
Candidiasis, aspergillosis, and mucormycosis cause the majority of nosocomial fungal infections in immunocompromised patients. Using an unbiased transcriptional profiling in PBMCs exposed to the fungal species causing these infections, we found a core host response in healthy individuals that may govern effective fungal clearance: it consists of 156 transcripts, involving canonical and non-canonical immune pathways.Systematic investigation of key steps in antifungal host defense revealed fungal-specific signatures. As previously demonstrated, Candida albicans induced type I and Type II interferon-related pathways. In contrast, central pattern recognition receptor, reactive oxygen species production, and host glycolytic pathways were down-regulated in response to Rhizopus oryzae, which was associated with an ER-stress response. TLR5 was identified to be uniquely regulated by Aspergillus fumigatus and to control cytokine release in response to this fungus.In conclusion, our data reveals the transcriptional profiles induced by C. albicans, A. fumigatus, and R. oryzae, and describes both the common and specific antifungal host responses that could be exploited for novel therapeutic strategies.
- Published
- 2021
- Full Text
- View/download PDF
35. Albumin Neutralizes Hydrophobic Toxins and Modulates Candida albicans Pathogenicity
- Author
-
Sophie Austermeier, Marina Pekmezović, Pauline Porschitz, Sejeong Lee, Nessim Kichik, David L. Moyes, Jemima Ho, Natalia K. Kotowicz, Julian R. Naglik, Bernhard Hube, and Mark S. Gresnigt
- Subjects
Microbiology ,QR1-502 - Abstract
Albumin is the most abundant serum protein in humans. During inflammation, serum albumin levels decrease drastically, and low albumin levels are associated with poor patient outcome.
- Published
- 2021
- Full Text
- View/download PDF
36. Neuraminidase and SIGLEC15 modulate the host defense against pulmonary aspergillosis
- Author
-
Intan M.W. Dewi, Cristina Cunha, Martin Jaeger, Mark S. Gresnigt, Marina E. Gkountzinopoulou, Fadel M. Garishah, Cláudio Duarte-Oliveira, Cláudia F. Campos, Lore Vanderbeke, Agustin Resendiz Sharpe, Roger J. Brüggemann, Paul E. Verweij, Katrien Lagrou, Greetje Vande Velde, Quirijn de Mast, Leo A.B. Joosten, Mihai G. Netea, Andre J.A.M. van der Ven, Joost Wauters, Agostinho Carvalho, and Frank L. van de Veerdonk
- Subjects
aspergillosis ,neuraminidase ,oseltamivir ,SIGLEC15 ,Medicine (General) ,R5-920 - Abstract
Summary: Influenza-associated pulmonary aspergillosis (IAPA) has been reported increasingly since the advent of use of neuraminidase (NA) inhibitors following the 2009 influenza pandemic. We hypothesize that blocking host NA modulates the immune response against Aspergillus fumigatus. We demonstrate that NA influences the host response against A. fumigatus in vitro and that oseltamivir increases the susceptibility of mice to pulmonary aspergillosis. Oseltamivir impairs the mouse splenocyte and human peripheral blood mononuclear cell (PBMC) killing capacity of A. fumigatus, and adding NA restores this defect in PBMCs. Furthermore, the sialic acid-binding receptor SIGLEC15 is upregulated in PBMCs stimulated with A. fumigatus. Silencing of SIGLEC15 decrease PBMC killing of A. fumigatus. We provide evidence that host NA activity and sialic acid recognition are important for anti-Aspergillus defense. NA inhibitors might predispose individuals with severe influenza to invasive aspergillosis. These data shed light on the pathogenesis of invasive fungal infections and may identify potential therapeutic targets.
- Published
- 2021
- Full Text
- View/download PDF
37. Data of common and species-specific transcriptional host responses to pathogenic fungi
- Author
-
Mariolina Bruno, Robter Horst, Marina Pekmezovic, Vinod Kumar, Yang Li, Mihai G. Netea, Jean-Paul Latgé, Mark S. Gresnigt, and Frank L. van de Veerdonk
- Subjects
Immunology of fungal infections ,Opportunistic pathogenic fungi ,Candida albicans ,Aspergillus fumigatus ,Rhizopus. oryzae ,Coagulation ,Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
Using a comparative RNA-Sequencing based transcriptional profiling approach, responses of primary human peripheral blood mononuclear cells (PBMCs) to common human pathogenic fungi have been characterized (Bruno et al. Computational and Structural Biology Journal). Primary human PBMCs were stimulated in vitro with the fungi A. fumigatus, C. albicans, and R. oryzae after which RNA was isolated and sequenced. From raw sequencing reads differential expressed genes in response to the different fungi where calculated by comparison with unstimulated cells. By overlapping differentially expressed genes in response to the pathogenic fungi A. fumigatus, C. albicans, and R. oryzae a dataset was generated that encompasses a common response to these three distinct fungi as well as species-specific responses. Here we present datasets on these common and species-specific responses that complement the original study (Bruno et al. Computational and Structural Biology Journal). These data serve to facilitate further fundamental research on the immune response to opportunistic pathogenic fungi such as A. fumigatus, C. albicans, and R. oryzae.
- Published
- 2021
- Full Text
- View/download PDF
38. The fungal peptide toxin Candidalysin activates the NLRP3 inflammasome and causes cytolysis in mononuclear phagocytes
- Author
-
Lydia Kasper, Annika König, Paul-Albert Koenig, Mark S. Gresnigt, Johannes Westman, Rebecca A. Drummond, Michail S. Lionakis, Olaf Groß, Jürgen Ruland, Julian R. Naglik, and Bernhard Hube
- Subjects
Science - Abstract
Phagocytic cells of the innate immune system play critical roles in defence against invading pathogens including the opportunistic pathogen Candida albicans. Here the authors show that C. albicans derived Candidalysin in addition to being a cell-damaging toxin to mononuclear phagocytes is a trigger of NLRP3 inflammasome activation in these cells.
- Published
- 2018
- Full Text
- View/download PDF
39. Microbiological and immunological characteristics of a lethal pulmonary Aspergillus niger infection in a non-neutropenic patient
- Author
-
Jessica D. Workum, Suzanne W. de Jong, Mark S. Gresnigt, Katharina L. Becker, Peter Pickkers, Frank L. van de Veerdonk, Yvonne F. Heijdra, and Eva Kolwijck
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Invasive pulmonary aspergillosis is increasingly described in non-neutropenic patients, such as patients with COPD receiving corticosteroids and the critically ill. Here, we present a case of a lethal pulmonary Aspergillus niger infection in a COPD patient. Immunological tests showed an impaired innate and adaptive immune response to Aspergillus. A history of COPD, unresponsiveness to antibiotics and especially a suggestive CT-scan should trigger the clinician to consider diseases caused by Aspergillus. Keywords: Invasive pulmonary aspergillosis, Aspergillus niger, Immune system, Intensive care unit, Oxalate crystal
- Published
- 2018
- Full Text
- View/download PDF
40. Genetic deficiency of NOD2 confers resistance to invasive aspergillosis
- Author
-
Mark S. Gresnigt, Cristina Cunha, Martin Jaeger, Samuel M. Gonçalves, R. K. Subbarao Malireddi, Anne Ammerdorffer, Rosalie Lubbers, Marije Oosting, Orhan Rasid, Grégory Jouvion, Catherine Fitting, Dirk J. de Jong, João F. Lacerda, António Campos, Willem J. G. Melchers, Katrien Lagrou, Johan Maertens, Thirumala-Devi Kanneganti, Agostinho Carvalho, Oumaima Ibrahim-Granet, and Frank L. van de Veerdonk
- Subjects
Science - Abstract
NOD2 has been shown to be crucial for immune recognition of Aspergillus infection. Here the authors show that a common NOD2 genetic variant associated with Crohn’s disease is associated with reduced risk of disease due to enhanced antifungal activates of monocytes and macrophages.
- Published
- 2018
- Full Text
- View/download PDF
41. Adjuvant interferon-gamma immunotherapy in a patient with progressive cerebral Nocardia abscesses
- Author
-
Jenneke Leentjens, Mark S. Gresnigt, Frank L. van de Veerdonk, Matthijs Kox, Bart Jan Kullberg, Peter Pickkers, Annemarie E. Brouwer, and Mihai G. Netea
- Subjects
Nocardiosis ,Nocardia ,Cerebral abscess ,IFN-gamma ,Immunotherapy ,Infectious and parasitic diseases ,RC109-216 - Abstract
Despite advances in medical care, mortality due to cerebral Nocardia abscesses remains unacceptably high. The case of a typical immunocompromised patient, who deteriorated clinically despite optimal antimicrobial treatment, is reported here. Adjuvant immunotherapy with interferon-gamma resulted in partial restoration of the immune response and a corresponding clinical and radiographic recovery.
- Published
- 2017
- Full Text
- View/download PDF
42. Keeping Candida commensal: how lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model
- Author
-
Katja Graf, Antonia Last, Rena Gratz, Stefanie Allert, Susanne Linde, Martin Westermann, Marko Gröger, Alexander S. Mosig, Mark S. Gresnigt, and Bernhard Hube
- Subjects
Candida albicans ,Microbiota ,Commensalism ,Lactobacilli ,Antagonism ,In vitro model ,Medicine ,Pathology ,RB1-214 - Abstract
The intestine is the primary reservoir of Candida albicans that can cause systemic infections in immunocompromised patients. In this reservoir, the fungus exists as a harmless commensal. However, antibiotic treatment can disturb the bacterial microbiota, facilitating fungal overgrowth and favoring pathogenicity. The current in vitro gut models that are used to study the pathogenesis of C. albicans investigate the state in which C. albicans behaves as a pathogen rather than as a commensal. We present a novel in vitro gut model in which the fungal pathogenicity is reduced to a minimum by increasing the biological complexity. In this model, enterocytes represent the epithelial barrier and goblet cells limit C. albicans adhesion and invasion. Significant protection against C. albicans-induced necrotic damage was achieved by the introduction of a microbiota of antagonistic lactobacilli. We demonstrated a time-, dose- and species-dependent protective effect against C. albicans-induced cytotoxicity. This required bacterial growth, which relied on the presence of host cells, but was not dependent on the competition for adhesion sites. Lactobacillus rhamnosus reduced hyphal elongation, a key virulence attribute. Furthermore, bacterial-driven shedding of hyphae from the epithelial surface, associated with apoptotic epithelial cells, was identified as a main and novel mechanism of damage protection. However, host cell apoptosis was not the driving mechanism behind shedding. Collectively, we established an in vitro gut model that can be used to experimentally dissect commensal-like interactions of C. albicans with a bacterial microbiota and the host epithelial barrier. We also discovered fungal shedding as a novel mechanism by which bacteria contribute to the protection of epithelial surfaces. This article has an associated First Person interview with the joint first authors of the paper.
- Published
- 2019
- Full Text
- View/download PDF
43. The Absence of NOD1 Enhances Killing of Aspergillus fumigatus Through Modulation of Dectin-1 Expression
- Author
-
Mark S. Gresnigt, Martin Jaeger, R. K. Subbarao Malireddi, Orhan Rasid, Grégory Jouvion, Catherine Fitting, Willem J. G. Melchers, Thirumala-Devi Kanneganti, Agostinho Carvalho, Oumaima Ibrahim-Granet, and Frank L. van de Veerdonk
- Subjects
NOD1 ,Aspergillus fumigatus ,nucleotide-binding oligomerization domain ,dectin-1 ,fungal killing ,Immunologic diseases. Allergy ,RC581-607 - Abstract
One of the major life-threatening infections for which severely immunocompromised patients are at risk is invasive aspergillosis (IA). Despite the current treatment options, the increasing antifungal resistance and poor outcome highlight the need for novel therapeutic strategies to improve outcome of patients with IA. In the current study, we investigated whether and how the intracellular pattern recognition receptor NOD1 is involved in host defense against Aspergillus fumigatus. When exploring the role of NOD1 in an experimental mouse model, we found that Nod1−/− mice were protected against IA and demonstrated reduced fungal outgrowth in the lungs. We found that macrophages derived from bone marrow of Nod1−/− mice were more efficiently inducing reactive oxygen species and cytokines in response to Aspergillus. Most strikingly, these cells were highly potent in killing A. fumigatus compared with wild-type cells. In line, human macrophages in which NOD1 was silenced demonstrated augmented Aspergillus killing and NOD1 stimulation decreased fungal killing. The differentially altered killing capacity of NOD1 silencing versus NOD1 activation was associated with alterations in dectin-1 expression, with activation of NOD1 reducing dectin-1 expression. Furthermore, we were able to demonstrate that Nod1−/− mice have elevated dectin-1 expression in the lung and bone marrow, and silencing of NOD1 gene expression in human macrophages increases dectin-1 expression. The enhanced dectin-1 expression may be the mechanism of enhanced fungal killing of Nod1−/− cells and human cells in which NOD1 was silenced, since blockade of dectin-1 reversed the augmented killing in these cells. Collectively, our data demonstrate that NOD1 receptor plays an inhibitory role in the host defense against Aspergillus. This provides a rationale to develop novel immunotherapeutic strategies for treatment of aspergillosis that target the NOD1 receptor, to enhance the efficiency of host immune cells to clear the infection by increasing fungal killing and cytokine responses.
- Published
- 2017
- Full Text
- View/download PDF
44. The Multifaceted Role of T-Helper Responses in Host Defense against Aspergillus fumigatus
- Author
-
Intan M. W. Dewi, Frank L. van de Veerdonk, and Mark S. Gresnigt
- Subjects
aspergillosis ,T-helper cells ,adaptive immune response ,immunopathology ,Biology (General) ,QH301-705.5 - Abstract
The ubiquitous opportunistic fungal pathogen Aspergillus fumigatus rarely causes infections in immunocompetent individuals. A healthy functional innate immune system plays a crucial role in preventing Aspergillus-infection. This pivotal role for the innate immune system makes it a main research focus in studying the pathogenesis of aspergillosis. Although sometimes overshadowed by the innate immune response, the adaptive immune response, and in particular T-helper responses, also represents a key player in host defense against Aspergillus. Virtually all T-helper subsets have been described to play a role during aspergillosis, with the Th1 response being crucial for fungal clearance. However; morbidity and mortality of aspergillosis can also be partly attributed to detrimental immune responses resulting from adaptive immune activation. Th2 responses benefit fungal persistence; and are the foundation of allergic forms of aspergillosis. The Th17 response has two sides; although crucial for granulocyte recruitment, it can be involved in detrimental immunopathology. Regulatory T-cells, the endogenous regulators of inflammatory responses, play a key role in controlling detrimental inflammatory responses during aspergillosis. The current knowledge of the adaptive immune response against A. fumigatus is summarized in this review. A better understanding on how T-helper responses facilitate clearance of Aspergillus-infection and control inflammation can be the fundamental basis for understanding the pathogenesis of aspergillosis and for the development of novel host-directed therapies.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.