105 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. Human TH17 cells engage gasdermin E pores to release IL-1α on NLRP3 inflammasome activation
- Author
-
Ying-Yin Chao, Alisa Puhach, David Frieser, Mahima Arunkumar, Laurens Lehner, Thomas Seeholzer, Albert Garcia-Lopez, Marlot van der Wal, Silvia Fibi-Smetana, Axel Dietschmann, Thomas Sommermann, Tamara Ćiković, Leila Taher, Mark S. Gresnigt, Sebastiaan J. Vastert, Femke van Wijk, Gianni Panagiotou, Daniel Krappmann, Olaf Groß, and Christina E. Zielinski
- Subjects
Immunology ,Immunology and Allergy - Abstract
It has been shown that innate immune responses can adopt adaptive properties such as memory. Whether T cells utilize innate immune signaling pathways to diversify their repertoire of effector functions is unknown. Gasdermin E (GSDME) is a membrane pore-forming molecule that has been shown to execute pyroptotic cell death and thus to serve as a potential cancer checkpoint. In the present study, we show that human T cells express GSDME and, surprisingly, that this expression is associated with durable viability and repurposed for the release of the alarmin interleukin (IL)-1α. This property was restricted to a subset of human helper type 17 T cells with specificity for Candida albicans and regulated by a T cell-intrinsic NLRP3 inflammasome, and its engagement of a proteolytic cascade of successive caspase-8, caspase-3 and GSDME cleavage after T cell receptor stimulation and calcium-licensed calpain maturation of the pro-IL-1α form. Our results indicate that GSDME pore formation in T cells is a mechanism of unconventional cytokine release. This finding diversifies our understanding of the functional repertoire and mechanistic equipment of T cells and has implications for antifungal immunity.
- Published
- 2023
- Full Text
- View/download PDF
13. P342 Lactobacillus rhamnosus protection against Candida-induced vaginal epithelial cell damage is Candida albicans strain-dependent
- Author
-
Marisa Valentine, Raquel Alonso-Román, Shameema Abdul-Rahman, Marie-Elisabeth Bougnoux, Christophe d'Enfert, Bernhard Hube, and Mark S Gresnigt
- Subjects
Infectious Diseases ,General Medicine - Abstract
Poster session 3, September 23, 2022, 12:30 PM - 1:30 PM Microbial dysbiosis can lead to vulvovaginal candidiasis (VVC) which is characterized by a pathogenicity-induced inflammatory response progressing to neutrophil-driven immunopathology. Probiotic treatment has varied success rates and some women still experience VVC despite being colonized by lactobacilli. It is, therefore, imperative to identify factors that influence the success of probiotic treatment for VVC. We aimed to evaluate how Candida albicans strain differences contribute to the varying degrees to which probiotics such as Lactobacillus rhamnosus protect against Candida-induced epithelial tissue damage. We screened the commonly used highly virulent C. albicans laboratory strain SC5314, 24 C. albicans strains from different clades, and several vaginal C. albicans isolates. L. rhamnosus was used to colonize vaginal epithelial cells prior to C. albicans infection and vaginal epithelial cell damage was measured. Compared to SC5314, most C. albicans strains induced relatively low or no epithelial cell damage. Even increased multiplicities of infection did not increase epithelial damage to the level of SC5314. Three groups were identified based on the effect of L. rhamnosus on Candida-induced epithelial cell damage. Bacterial colonization decreased, did not affect, or even increased tissue damage during infection. The different C. albicans clades showed no correlation with the protective phenotypes. However, increased epithelial tissue damage in the presence of lactobacilli was generally observed with strains that alone were unable to damage the epithelium. Some strains had an enhanced potential to grow at low pH, yet growth at low pH alone was not able to distinguish the three groups. The protective potential of L. rhamnosus is highly C. albicans strain-dependent. Our data hint toward a potential multifactorial effect involving stress-resistance and metabolic interplay. Elucidating the processes that lead to epithelial protection or enhanced damage will be crucial to predict whether probiotic lactobacilli may be beneficial or detrimental for a patient and may help to design generally protective probiotics.
- Published
- 2022
- Full Text
- View/download PDF
14. Human T
- Author
-
Ying-Yin, Chao, Alisa, Puhach, David, Frieser, Mahima, Arunkumar, Laurens, Lehner, Thomas, Seeholzer, Albert, Garcia-Lopez, Marlot, van der Wal, Silvia, Fibi-Smetana, Axel, Dietschmann, Thomas, Sommermann, Tamara, Ćiković, Leila, Taher, Mark S, Gresnigt, Sebastiaan J, Vastert, Femke, van Wijk, Gianni, Panagiotou, Daniel, Krappmann, Olaf, Groß, and Christina E, Zielinski
- Abstract
It has been shown that innate immune responses can adopt adaptive properties such as memory. Whether T cells utilize innate immune signaling pathways to diversify their repertoire of effector functions is unknown. Gasdermin E (GSDME) is a membrane pore-forming molecule that has been shown to execute pyroptotic cell death and thus to serve as a potential cancer checkpoint. In the present study, we show that human T cells express GSDME and, surprisingly, that this expression is associated with durable viability and repurposed for the release of the alarmin interleukin (IL)-1α. This property was restricted to a subset of human helper type 17 T cells with specificity for Candida albicans and regulated by a T cell-intrinsic NLRP3 inflammasome, and its engagement of a proteolytic cascade of successive caspase-8, caspase-3 and GSDME cleavage after T cell receptor stimulation and calcium-licensed calpain maturation of the pro-IL-1α form. Our results indicate that GSDME pore formation in T cells is a mechanism of unconventional cytokine release. This finding diversifies our understanding of the functional repertoire and mechanistic equipment of T cells and has implications for antifungal immunity.
- Published
- 2022
15. I want to break free – macrophage strategies to recognize and kill Candida albicans, and fungal counter-strategies to escape
- Author
-
Mark S. Gresnigt, Lydia Kasper, Johannes Westman, and Sophie Austermeier
- Subjects
Microbiology (medical) ,Antifungal ,medicine.drug_class ,Biology ,Microbiology ,03 medical and health sciences ,Immune system ,Candida albicans ,Free macrophage ,medicine ,Animals ,Humans ,Macrophage ,Immune Evasion ,030304 developmental biology ,0303 health sciences ,Innate immune system ,030306 microbiology ,Macrophages ,Candidiasis ,biology.organism_classification ,Yeast ,Corpus albicans ,Infectious Diseases - Abstract
Candida albicans is a major cause of fungal nosocomial infections. Host defense against disseminated infections caused by this yeast strongly relies on myeloid cells of the innate immune system. Recently, several breakthroughs have been made that significantly improved our understanding of the role of macrophages during candidiasis and how C. albicans and macrophages interact. Resident tissue macrophages and macrophages derived from monocytes that infiltrate infected tissues are essential for the initiation of the antifungal immune response, as well as elimination of C. albicans from the bloodstream and infected organs. These cells engulf and try to eliminate the invading fungi through specialized mechanisms. Concurrently, C. albicans tries to survive the stresses imposed by the macrophage, acquires nutrients, and can break free from their captive environment. This review focuses on the most recent insights into the strategies of macrophages to eliminate C. albicans and the fungal counterstrategies to overcome these threats.
- Published
- 2020
- Full Text
- View/download PDF
16. The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine
- Author
-
Carol A. Kumamoto, Mark S. Gresnigt, and Bernhard Hube
- Subjects
Microbiology (medical) ,Population ,Opportunistic Infections ,Biology ,Microbiology ,Article ,03 medical and health sciences ,Immune system ,Candida albicans ,medicine ,Animals ,Humans ,Microbiome ,Symbiosis ,education ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,030306 microbiology ,Gastrointestinal Microbiome ,Candidiasis ,Commensalism ,biology.organism_classification ,medicine.disease ,Corpus albicans ,Gastrointestinal Tract ,Infectious Diseases ,Systemic candidiasis - Abstract
Candida albicans is a regular member of the intestinal microbiota in the majority of the human population. This underscores C. albicans' adaptation to life in the intestine without inducing competitive interactions with other microbes, or immune responses detrimental to its survival. However, specific conditions such as a dysbalanced microbiome, a suppression of the immune system, and an impaired intestinal barrier can predispose for invasive, mostly nosocomial, C. albicans infections. Colonization of the intestine and translocation through the intestinal barrier are fundamental aspects of the processes preceding life-threatening systemic candidiasis. Insights into C. albicans' commensal lifestyle and translocation can thus help us to understand how patients develop candidiasis, and may provide leads for therapeutic strategies aimed at preventing infection. In this review, we discuss the commensal lifestyle of C. albicans in the intestine, the role of morphology for commensalism, the influence of diet, and the interactions with bacteria of the microbiota.
- Published
- 2020
- Full Text
- View/download PDF
17. 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
Multidisciplinary ,Virulence ,Lacticaseibacillus rhamnosus ,Candida albicans ,Candidiasis ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology ,Candida - Abstract
Intestinal microbiota dysbiosis can initiate overgrowth of commensal Candida species – a major predisposing factor for disseminated candidiasis. Commensal bacteria such as Lactobacillus rhamnosus can antagonize Candida albicans pathogenicity. Here, we investigate the interplay between C. albicans, L. rhamnosus, and intestinal epithelial cells by integrating transcriptional and metabolic profiling, and reverse genetics. Untargeted metabolomics and in silico modelling indicate that intestinal epithelial cells foster bacterial growth metabolically, leading to bacterial production of antivirulence compounds. In addition, bacterial growth modifies the metabolic environment, including removal of C. albicans’ favoured nutrient sources. This is accompanied by transcriptional and metabolic changes in C. albicans, including altered expression of virulence-related genes. Our results indicate that intestinal colonization with bacteria can antagonize C. albicans by reshaping the metabolic environment, forcing metabolic adaptations that reduce fungal pathogenicity.
- Published
- 2021
18. 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
19. Candida albicans-induced leukotriene biosynthesis in neutrophils is restricted to the hyphal morphology
- Author
-
Ulrike Garscha, Oliver Werz, Mark S. Gresnigt, Bernhard Hube, and Jana Fischer
- Subjects
Leukotrienes ,Neutrophils ,Phagocytosis ,Hyphae ,Syk ,Inflammation ,Biochemistry ,Candida albicans ,Genetics ,medicine ,Humans ,Molecular Biology ,Leukotriene ,biology ,Chemistry ,fungi ,Chemotaxis ,biology.organism_classification ,Corpus albicans ,Cell biology ,Arachidonate 5-lipoxygenase ,Host-Pathogen Interactions ,biology.protein ,lipids (amino acids, peptides, and proteins) ,medicine.symptom ,Biotechnology ,Signal Transduction - Abstract
Neutrophils are the most abundant leukocytes in circulation playing a key role in acute inflammation during microbial infections. Phagocytosis, one of the crucial defence mechanisms of neutrophils against pathogens, is amplified by chemotactic leukotriene (LT)B4 , which is biosynthesized via 5-lipoxygenase (5-LOX). However, extensive liberation of LTB4 can be destructive by over-intensifying the inflammatory process. While enzymatic biosynthesis of LTB4 is well characterized, less is known about molecular mechanisms that activate 5-LOX and lead to LTB4 formation during host-pathogen interactions. Here, we investigated the ability of the common opportunistic fungal pathogen Candida albicans to induce LTB4 formation in neutrophils, and elucidated pathogen-mediated drivers and cellular processes that activate this pathway. We revealed that C. albicans-induced LTB4 biosynthesis requires both the morphological transition from yeast cells to hyphae and the expression of hyphae-associated genes, as exclusively viable hyphae or yeast-locked mutant cells expressing hyphae-associated genes stimulated 5-LOX by [Ca2+ ]i mobilization and p38 MAPK activation. LTB4 biosynthesis was orchestrated by synergistic activation of dectin-1 and Toll-like receptor 2, and corresponding signaling via SYK and MYD88, respectively. Conclusively, we report hyphae-specific induction of LTB4 biosynthesis in human neutrophils. This highlights an expanding role of neutrophils during inflammatory processes in the response to C. albicans infections.
- Published
- 2021
20. 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, Frank L. van de Veerdonk, Leo Joosten, Mihai G. Netea, Agostinho Carvalho, Vinod Kumar, Repositório da Universidade de Lisboa, and Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI)
- Subjects
All institutes and research themes of the Radboud University Medical Center ,C. albicans ,Organic Chemistry ,Clinical Biochemistry ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Invasive aspergillosis ,QTLs ,Reactive oxygen species ,A. fumigatus ,Biochemistry ,Stem-cell transplant recipients - Abstract
© 2022 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/)., 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 < 9.99 × 10-6), with some of these loci being pathogen-specific, and others shared between the two fungi. These ROS-QTLs were investigated for their influence on the risk of invasive pulmonary aspergillosis (IPA) in a disease relevant context. We stratified hematopoietic stem-cell transplant (HSCT) recipients based on the donor's SNP genotype and tested their impact on the risk of IPA. We identified rs4685368 as a ROS-QTL locus that was significantly associated with an increased risk of IPA after controlling for patient age and sex, hematological malignancy, type of transplantation, conditioning regimen, acute graft-versus-host-disease grades III-IV, and antifungal prophylaxis. Collectively, this data provides evidence that common genetic variation can influence ROS production capacity, and, importantly, the risk of developing IPA among HSCT recipients. This evidence warrants further research for patient stratification based on the genetic profiling that would allow the identifications of patients at high-risk for an invasive fungal infection, and who would benefit the most from a preventive strategy., This study was supported by the European Union's Horizon 2020 research and innovation programme under grant agreement no. 847507 (HDM-FUN). MGN was supported by an ERC Advanced grant (833247) and a Spinoza grant of the Netherlands Association for Scientific Research. VK was supported by a Research Grant [2017] of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and Hypatia tenure track grant. AC was supported by the Fundação para a Ciência e a Tecnologia (FCT) (UIDB/50026/2020 and UIDP/50026/2020), the Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, through the European Regional Development Fund (ERDF) (NORTE-01-0145-FEDER-000039), and the “la Caixa” Foundation (ID 100010434) and FCT under the agreement LCF/PR/HR17/52190003. CC was supported by FCT (CEECIND/04058/2018 and PTDC/SAU-SER/29,635/2017) and the Gilead Research Scholars Program – Antifungals. SMG was the recipient of a PhD fellowship funded by FCT (SFRH/BD/136,814/2018). MSG was supported by the German Research Foundation (Deutsche Forschungsgemeinschaft - DFG) Emmy Noether Program (project no. 434385622/GR 5617/1-1).
- Published
- 2022
- Full Text
- View/download PDF
21. Albumin Neutralizes Hydrophobic Toxins and Modulates
- 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
Mucous Membrane ,Virulence Factors ,Candidiasis ,toxins ,Cell Line ,Fungal Proteins ,virulence ,Albumins ,Candida albicans ,Host-Pathogen Interactions ,Vagina ,cytolysis ,Humans ,Female ,microbial cytotoxicity ,HT29 Cells ,Hydrophobic and Hydrophilic Interactions ,Cells, Cultured ,serum proteins ,Research Article - 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.
- Published
- 2021
22. 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
23. 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
24. 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
25. The anti-inflammatory cytokine interleukin-37 is an inhibitor of trained immunity
- Author
-
Davide Stefanoni, Silvia Giugliano, Eleonora Cantoni, Giulio Cavalli, Elan Z. Eisenmensser, Isak W. Tengesdal, Kiki Schraa, Lorenzo Dagna, Mihai G. Netea, Laura Cassina, Angelo D'Alessandro, Travis Nemkov, Leo A. B. Joosten, Eric M. Pietras, Jorge Domínguez-Andrés, Mark S. Gresnigt, Rob J.W. Arts, Taylor S. Mills, Raffaella Molteni, Alessandra Boletta, Charles A. Dinarello, Cavalli, Giulio, Tengesdal, I W, Gresnigt, M, Nemkov, T, Arts, R J W, Dominguez-Andres, J, Molteni, R, Stefanoni, D, Cantoni, E, Cassina, L, Giugliano, S, Schraa, K, Mills, T, Pietras, E M, Eisenmensser, E Z, Dagna, L, Boletta, A, D'Alessandro, A, Joosten, L A B, Netea, M G, and Dinarello, C A
- Subjects
0301 basic medicine ,Male ,Neutrophils ,medicine.medical_treatment ,immunometabolism ,Anti-Inflammatory Agents ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Epigenesis, Genetic ,trained immunity ,0302 clinical medicine ,Medicine ,lcsh:QH301-705.5 ,innate immunity ,biology ,cell energy metabolism ,Candidiasis ,Interleukin ,3. Good health ,Histone ,Cytokine ,regulatory cytokine ,Host-Pathogen Interactions ,medicine.symptom ,Glycolysis ,medicine.drug_class ,Inflammation ,General Biochemistry, Genetics and Molecular Biology ,Anti-inflammatory ,IL-1 family ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,Immunity ,Animals ,Humans ,Epigenetics ,Innate immune system ,epigenetics ,business.industry ,cytokines ,infection ,Immunity, Innate ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,inflammation ,Immunology ,biology.protein ,business ,030217 neurology & neurosurgery ,Interleukin-1 - Abstract
Summary Trained immunity (TI) is a de facto innate immune memory program induced in monocytes/macrophages by exposure to pathogens or vaccines, which evolved as protection against infections. TI is characterized by immunometabolic changes and histone post-translational modifications, which enhance production of pro-inflammatory cytokines. As aberrant activation of TI is implicated in inflammatory diseases, tight regulation is critical; however, the mechanisms responsible for this modulation remain elusive. Interleukin-37 (IL-37) is an anti-inflammatory cytokine that curbs inflammation and modulates metabolic pathways. In this study, we show that administration of recombinant IL-37 abrogates the protective effects of TI in vivo, as revealed by reduced host pro-inflammatory responses and survival to disseminated candidiasis. Mechanistically, IL-37 reverses the immunometabolic changes and histone post-translational modifications characteristic of TI in monocytes, thus suppressing cytokine production in response to infection. IL-37 thereby emerges as an inhibitor of TI and as a potential therapeutic target in immune-mediated pathologies.
- Published
- 2021
26. 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
27. 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
28. Human recombinant interleukin-38 suppresses inflammation in mouse models of local and systemic disease
- Author
-
Frank L. van de Veerdonk, Nicholas E. Powers, Dennis M. de Graaf, Leo A. B. Joosten, Sanne P. Smeekens, Mark S. Gresnigt, Jasmina S. Redzic, Suzhao Li, Monique M. Helsen, Charles A. Dinarello, Ralph J.A. Maas, Vassili Kalabokis, and Elan Z. Eisenmesser
- Subjects
Lipopolysaccharides ,Male ,0301 basic medicine ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,Anti-Inflammatory Agents ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Arthritis ,Inflammation ,Peritonitis ,Pharmacology ,Recombinant Interleukin ,Systemic inflammation ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,All institutes and research themes of the Radboud University Medical Center ,medicine ,Animals ,Humans ,Immunology and Allergy ,Amino Acid Sequence ,Molecular Biology ,Cells, Cultured ,Sequence Homology, Amino Acid ,Arthritis, Gouty ,Chemistry ,Interleukins ,Interleukin ,Hematology ,medicine.disease ,Recombinant Proteins ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cytokines ,Tumor necrosis factor alpha ,Bone marrow ,medicine.symptom ,Ex vivo ,Inflammatory diseases Radboud Institute for Molecular Life Sciences [Radboudumc 5] - Abstract
Interleukin (IL)–38 belongs to the IL–1 family and is part of the IL–36 subfamily due to its binding to the IL–36 Receptor (IL–1R6). In the current study, we assessed the anti-inflammatory properties of IL–38 in murine models of arthritis and systemic inflammation. First, the anti-inflammatory properties of mouse and human IL–38 precursors were compared to forms with a truncated N–terminus. In mouse bone marrow derived dendritic cells (BMDC), human and mouse IL–38 precursors with a truncation of the two N-terminal amino acids (3–152) suppressed LPS-induced IL–6. Recombinant human IL–38 (3–152) was further investigated for its immunomodulatory potential using four murine models of inflammatory disease: streptococcal cell wall (SCW)-induced arthritis, monosodium urate (MSU) crystal-induced arthritis, MSU crystal-induced peritonitis, and systemic endotoxemia. In each of these models IL–38 significantly reduced inflammation. In SCW and MSU crystal-induced arthritis, joint swelling, inflammatory cell influx, and synovial levels of IL–1β, IL–6, and KC were reduced by 50% or greater. These suppressive properties of IL–38 in SCW-induced arthritis were independent of the anti-inflammatory co-receptor IL–1R8, as IL–38 reduced arthritis equally in IL–1R8 deficient and WT mice. In MSU crystal-induced peritonitis, IL–38 reduced hypothermia, while plasma IL–6 and KC and peritoneal KC levels were reduced by 65–70%. In the LPS endotoxemia model, IL–38 pretreatment reduced systemic IL–6, TNFα and KC. Furthermore, in ex vivo cultured bone marrow, LPS-induced IL–6, TNFα and KC were reduced by 75–90%. Overall, IL–38 exhibits broad anti-inflammatory properties in models of systemic and local inflammation and therefore may be an effective cytokine therapy.
- Published
- 2021
29. 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
30. In vitro infection models to study fungal-host interactions
- Author
-
Michelle Maurer, Alexander S. Mosig, Bernhard Hube, and Mark S. Gresnigt
- Subjects
Histoplasma ,Cryptococcus ,Virulence ,Fungus ,Review Article ,Microbiology ,03 medical and health sciences ,Immune system ,medicine ,Humans ,030304 developmental biology ,Candida ,0303 health sciences ,Aspergillus ,AcademicSubjects/SCI01150 ,biology ,030306 microbiology ,Host (biology) ,Microbiota ,Fungi ,Mucous membrane ,fungal–host interaction ,biology.organism_classification ,Infectious Diseases ,medicine.anatomical_structure ,Mycoses ,in vitro model - Abstract
Fungal infections (mycoses) affect over a billion people per year. Approximately, two million of these infections are life-threatening, especially for patients with a compromised immune system. Fungi of the genera Aspergillus, Candida, Histoplasma and Cryptococcus are opportunistic pathogens that contribute to a substantial number of mycoses. To optimize the diagnosis and treatment of mycoses, we need to understand the complex fungal–host interplay during pathogenesis, the fungal attributes causing virulence and how the host resists infection via immunological defenses. In vitro models can be used to mimic fungal infections of various tissues and organs and the corresponding immune responses at near-physiological conditions. Furthermore, models can include fungal interactions with the host–microbiota to mimic the in vivo situation on skin and mucosal surfaces. This article reviews currently used in vitro models of fungal infections ranging from cell monolayers to microfluidic 3D organ-on-chip (OOC) platforms. We also discuss how OOC models can expand the toolbox for investigating interactions of fungi and their human hosts in the future., From basic to complex: in vitro models to study interactions between human fungal pathogens and their host.
- Published
- 2020
31. Decision letter: Chronic ethanol consumption compromises neutrophil function in acute pulmonary Aspergillus fumigatus infection
- Author
-
Agostinho Carvalho, Mark S. Gresnigt, and Frank L. van de Veerdonk
- Subjects
Consumption (economics) ,chemistry.chemical_compound ,Ethanol ,biology ,chemistry ,business.industry ,Immunology ,Medicine ,biology.organism_classification ,business ,Function (biology) ,Aspergillus fumigatus - Published
- 2020
- Full Text
- View/download PDF
32. Candida albicans colonization of the gastrointestinal tract: A double-edged sword
- Author
-
Bernhard Hube, Mark S. Gresnigt, Elvira Román, Rebeca Alonso-Monge, and Jesús Pla
- Subjects
Bacterial Diseases ,Yeast and Fungal Models ,Pathology and Laboratory Medicine ,Inflammatory bowel disease ,Pearls ,Gastrointestinal infections ,Medical Conditions ,Candida albicans ,Medicine and Health Sciences ,Gastrointestinal Infections ,Colonization ,Biology (General) ,Immune Response ,Candida ,Fungal Pathogens ,Gastrointestinal tract ,biology ,Gastrointestinal Microbiome ,Candidiasis ,Eukaryota ,Infectious Diseases ,Experimental Organism Systems ,Medical Microbiology ,Pathogens ,Anatomy ,Acinetobacter Infections ,QH301-705.5 ,Immunology ,Mycology ,Gastroenterology and Hepatology ,Research and Analysis Methods ,Microbiology ,Immune system ,Virology ,Genetics ,medicine ,Humans ,Microbial Pathogens ,Molecular Biology ,business.industry ,Inflammatory Bowel Disease ,Organisms ,Fungi ,Biology and Life Sciences ,RC581-607 ,medicine.disease ,biology.organism_classification ,Yeast ,Gastrointestinal Tract ,Animal Studies ,Parasitology ,Immunologic diseases. Allergy ,business ,Digestive System - Published
- 2021
- Full Text
- View/download PDF
33. Adjuvant interferon-gamma immunotherapy in a patient with progressive cerebral Nocardia abscesses
- Author
-
Annemarie E. Brouwer, Peter Pickkers, Bart Jan Kullberg, Jenneke Leentjens, Frank L. van de Veerdonk, Mark S. Gresnigt, Mihai G. Netea, and Matthijs Kox
- Subjects
Male ,Microbiology (medical) ,medicine.medical_treatment ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Brain Abscess ,Nocardia Infections ,Medical care ,Antiviral Agents ,Nocardia ,lcsh:Infectious and parasitic diseases ,IFN-gamma ,03 medical and health sciences ,Nocardiosis ,Immunocompromised Host ,Interferon-gamma ,0302 clinical medicine ,Immune system ,Adjuvants, Immunologic ,medicine ,Humans ,Interferon gamma ,lcsh:RC109-216 ,030212 general & internal medicine ,biology ,business.industry ,General Medicine ,Immunotherapy ,Middle Aged ,medicine.disease ,Antimicrobial ,biology.organism_classification ,Magnetic Resonance Imaging ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,Infectious Diseases ,030228 respiratory system ,Immunology ,Cerebral abscess ,business ,Adjuvant ,medicine.drug - Abstract
Contains fulltext : 174790.pdf (Publisher’s version ) (Open Access) 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
34. 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
35. A three-dimensional immunocompetent intestine-on-chip model as in vitro platform for functional and microbial interaction studies
- Author
-
Michelle Maurer, Mark S. Gresnigt, Antonia Last, Tony Wollny, Florian Berlinghof, Rebecca Pospich, Zoltan Cseresnyes, Anna Medyukhina, Katja Graf, Marko Gröger, Martin Raasch, Fatina Siwczak, Sandor Nietzsche, Ilse D. Jacobsen, Marc Thilo Figge, Bernhard Hube, Otmar Huber, and Alexander S. Mosig
- Subjects
Lipopolysaccharides ,Cell Membrane Permeability ,Biophysics ,Colony Count, Microbial ,Bioengineering ,Inflammation ,02 engineering and technology ,Biology ,Models, Biological ,law.invention ,Microbiology ,Immune tolerance ,Biomaterials ,Pathogenesis ,03 medical and health sciences ,Probiotic ,Immune system ,Lactobacillus rhamnosus ,law ,Antigens, CD ,Cell Movement ,Lab-On-A-Chip Devices ,medicine ,Human Umbilical Vein Endothelial Cells ,Humans ,Candida albicans ,030304 developmental biology ,0303 health sciences ,Innate immune system ,Microvilli ,Lacticaseibacillus rhamnosus ,Epithelial Cells ,021001 nanoscience & nanotechnology ,biology.organism_classification ,Cadherins ,Intestines ,Perfusion ,Mechanics of Materials ,Ceramics and Composites ,Zonula Occludens-1 Protein ,Cytokines ,Microbial Interactions ,medicine.symptom ,Caco-2 Cells ,0210 nano-technology ,Immunocompetence ,Biomarkers - Abstract
Alterations of the microbial composition in the gut and the concomitant dysregulation of the mucosal immune response are associated with the pathogenesis of opportunistic infections, chronic inflammation, and inflammatory bowel disease. To create a platform for the investigation of the underlying mechanisms, we established a three-dimensional microphysiological model of the human intestine. This model resembles organotypic microanatomical structures and includes tissue resident innate immune cells exhibiting features of mucosal macrophages and dendritic cells. The model displays the physiological immune tolerance of the intestinal lumen to microbial-associated molecular patterns and can, therefore, be colonised with living microorganisms. Functional studies on microbial interaction between probiotic Lactobacillus rhamnosus and the opportunistic pathogen Candida albicans show that pre-colonization of the intestinal lumen of the model by L. rhamnosus reduces C. albicans-induced tissue damage, lowers its translocation, and limits fungal burden. We demonstrate that microbial interactions can be efficiently investigated using the in vitro model creating a more physiological and immunocompetent microenvironment. The intestinal model allows a detailed characterisation of the immune response, microbial pathogenicity mechanisms, and quantification of cellular dysfunction attributed to alterations in the microbial composition.
- Published
- 2019
36. Keeping
- 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
Time Factors ,Hyphae ,Apoptosis ,Models, Biological ,Epithelium ,Species Specificity ,In vitro model ,Stress, Physiological ,Candida albicans ,Humans ,Cell Aggregation ,Antagonism ,Microbial Viability ,Microbiota ,Polysaccharides, Bacterial ,Adhesiveness ,Commensalism ,Gastrointestinal Tract ,Oxygen ,Lactobacillus ,Enterocytes ,Glucose ,Gene Expression Regulation ,Lactobacilli ,Host-Pathogen Interactions ,Lactates ,Caco-2 Cells ,Research Article - 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., Editor's choice: Using antagonistic lactobacilli, C. albicans pathogenicity was reduced to a minimum in an in vitro gut model consisting of enterocytes and goblet cells.
- Published
- 2019
37. P087 The anti-inflammatory cytokine interleukin 37 is an endogenous inhibitor of trained immunity
- Author
-
Angelo D'Alessandro, Mark S. Gresnigt, Mihai G. Netea, L. A. B. Joosten, Travis Nemkov, Charles A. Dinarello, Lorenzo Dagna, S Giugliano, E Eisenmensser, Giulio Cavalli, and Rob J.W. Arts
- Subjects
Innate immune system ,business.industry ,medicine.medical_treatment ,Interleukin ,Inflammation ,Proinflammatory cytokine ,Cytokine ,Immunity ,In vivo ,Immunology ,medicine ,medicine.symptom ,business ,Ex vivo - Abstract
Career situation of first and presenting author Assistant. Introduction Trained immunity (TI) is a de-facto innate immune memory program induced in monocytes/macrophages by exposure to pathogens or vaccines, which evolved as a protective mechanism against infections. TI is characterized by rewiring of functional, epigenetic and metabolic programs of innate immune cells such as monocytes and macrophages, which sustain enhanced production of pro-inflammatory cytokines. Since aberrant activation of TI is implicated in inflammatory diseases, tight regulatory mechanisms are likely in place, but the mechanisms responsible for this modulation remain elusive. Objectives Scope of this study was to evaluated the role of IL-37, an anti-inflammatory cytokine that curbs inflammation as well as modulates metabolic pathways, as an endogenous regulator of trained immunity. Methods The effects of recombinant IL-37 were evaluated in a mouse model of TI induced by the administration of beta-glucan in vivo (survival to a lethal inoculum of infectious agents, production of inflammatory cytokines, recruitment of inflammatory cells at the sites of infection). Subsequently, the effects of IL-37 were evaluated ex vivo on splenic and bone marrow monocytes (production of inflammatory cytokines, metabolomic analysis of the activation status of the main pathways of cellular energy metabolism). Finally, we evaluated the association between IL-37 gene polymorphisms and the induction of TI in monocytes of healthy donors with in vitro functional studies. Results The exogenous administration of IL-37 abrogates the pro-inflammatory effects of TI, significantly reducing the production of pro-inflammatory cytokines and the survival of experimental animals subjected to a disseminated infection model. The inhibitory effects of IL-37 on TI are also associated with reduced recruitment of neutrophils at sites of inflammation. IL-37 and TI programs have differential and opposite effects on the modulation of cellular energy metabolism of monocytes. In humans, polymorphisms in the IL-37 gene are associated with reduced activation of TI programs and reduced production of inflammatory cytokines by healthy donor monocytes. Conclusions In conclusion, IL-37 emerges as an endogenous regulator of TI, which makes this cytokine a potential therapeutic target in immune-mediated pathologies. Disclosure of Interest None declared.
- Published
- 2019
- Full Text
- View/download PDF
38. Keeping Candida commensal – How lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model
- Author
-
Mark S. Gresnigt, Martin Westermann, Marko Gröger, Rena Gratz, Alexander S. Mosig, Bernhard Hube, Susanne Linde, Stefanie Allert, and Katja Graf
- Subjects
0301 basic medicine ,Hypha ,Neuroscience (miscellaneous) ,lcsh:Medicine ,Medicine (miscellaneous) ,Virulence ,General Biochemistry, Genetics and Molecular Biology ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,In vitro model ,Immunology and Microbiology (miscellaneous) ,Lactobacillus rhamnosus ,Candida albicans ,lcsh:Pathology ,Pathogen ,Antagonism ,biology ,Microbiota ,lcsh:R ,biology.organism_classification ,Commensalism ,In vitro ,Corpus albicans ,3. Good health ,030104 developmental biology ,Lactobacilli ,030217 neurology & neurosurgery ,Bacteria ,lcsh: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
39. A Genome-Wide Functional Genomics Approach Identifies Susceptibility Pathways to Fungal Bloodstream Infection in Humans
- Author
-
Frank L. van de Veerdonk, Sebo Withoff, Mihai G. Netea, Bart Jan Kullberg, Iris Jonkers, Vinod Kumar, Sanne P. Smeekens, Mark S. Gresnigt, Martin Jaeger, Yang Li, Vasiliki Matzaraki, Cisca Wijmenga, Marije Oosting, Melissa D. Johnson, Raul Aguirre-Gamboa, Leo A. B. Joosten, John R. Perfect, Xiaojing Chu, Groningen Institute for Gastro Intestinal Genetics and Immunology (3GI), and Molecular Neuroscience and Ageing Research (MOLAR)
- Subjects
0301 basic medicine ,medicine.medical_treatment ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Single-nucleotide polymorphism ,Genome-wide association study ,Biology ,Quantitative trait locus ,Cohort Studies ,Cancer development and immune defence Radboud Institute for Health Sciences [Radboudumc 2] ,03 medical and health sciences ,Major Articles and Brief Reports ,0302 clinical medicine ,Candida albicans ,medicine ,Immunology and Allergy ,Homeostasis ,Humans ,Allele ,Gene ,Alleles ,Chromosomes, Human, Pair 15 ,Interleukin-6 ,Group IV Phospholipases A2 ,Candidemia ,Genomics ,biology.organism_classification ,3. Good health ,Oxidative Stress ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,030104 developmental biology ,Infectious Diseases ,Cytokine ,Genetic Loci ,Immunology ,Host-Pathogen Interactions ,Cytokines ,Disease Susceptibility ,Reactive Oxygen Species ,Functional genomics ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
BackgroundCandidemia, one of the most common causes of fungal bloodstream infection, leads to mortality rates up to 40% in affected patients. Understanding genetic mechanisms for differential susceptibility to candidemia may aid in designing host-directed therapies.MethodsWe performed the first genome-wide association study on candidemia, and we integrated these data with variants that affect cytokines in different cellular systems stimulated with Candida albicans.ResultsWe observed strong association between candidemia and a variant, rs8028958, that significantly affects the expression levels of PLA2G4B in blood. We found that up to 35% of the susceptibility loci affect in vitro cytokine production in response to Candida. Furthermore, potential causal genes located within these loci are enriched for lipid and arachidonic acid metabolism. Using an independent cohort, we also showed that the numbers of risk alleles at these loci are negatively correlated with reactive oxygen species and interleukin-6 levels in response to Candida. Finally, there was a significant correlation between susceptibility and allelic scores based on 16 independent candidemia-associated single-nucleotide polymorphisms that affect monocyte-derived cytokines, but not with T cell-derived cytokines.ConclusionsOur results prioritize the disturbed lipid homeostasis and oxidative stress as potential mechanisms that affect monocyte-derived cytokines to influence susceptibility to candidemia.
- Published
- 2019
40. Frontline Science: Endotoxin-induced immunotolerance is associated with loss of monocyte metabolic plasticity and reduction of oxidative burst
- Author
-
Matthijs Kox, Mihai G. Netea, Niklas Bruse, Peter Pickkers, Mark S. Gresnigt, Rob J.W. Arts, Jelle Gerretsen, Rob ter Horst, Rebecca M. Koch, Inge Grondman, Guus P. Leijte, and Rosalie W. M. Kempkes
- Subjects
0301 basic medicine ,Lipopolysaccharide ,Secondary infection ,CD14 ,medicine.medical_treatment ,immunometabolism ,Immunology ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Biology ,Monocytes ,sepsis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,endotoxin tolerance ,immunoparalysis ,Immune Tolerance ,medicine ,Humans ,Immunology and Allergy ,Respiratory Burst ,endotoxemia ,Monocyte ,Cell Biology ,Respiratory burst ,Endotoxins ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Highlighted Article ,chemistry ,030220 oncology & carcinogenesis ,Spotlight on Leading Edge Research ,Ex vivo ,Inflammatory diseases Radboud Institute for Molecular Life Sciences [Radboudumc 5] - Abstract
Secondary infections are a major complication of sepsis and associated with a compromised immune state, called sepsis‐induced immunoparalysis. Molecular mechanisms causing immunoparalysis remain unclear; however, changes in cellular metabolism of leukocytes have been linked to immunoparalysis. We investigated the relation of metabolic changes to antimicrobial monocyte functions in endotoxin‐induced immunotolerance, as a model for sepsis‐induced immunoparalysis. In this study, immunotolerance was induced in healthy males by intravenous endotoxin (2 ng/kg, derived from Escherichia coli O:113) administration. Before and after induction of immunotolerance, circulating CD14+ monocytes were isolated and assessed for antimicrobial functions, including cytokine production, oxidative burst, and microbial (Candida albicans) killing capacity, as well metabolic responses to ex vivo stimulation. Next, the effects of altered cellular metabolism on monocyte functions were validated in vitro. Ex vivo lipopolysaccharide stimulation induced an extensive rewiring of metabolism in naive monocytes. In contrast, endotoxin‐induced immunotolerant monocytes showed no metabolic plasticity, as they were unable to adapt their metabolism or mount cytokine and oxidative responses. Validation experiments showed that modulation of metabolic pathways, affected by immunotolerance, influenced monocyte cytokine production, oxidative burst, and microbial (C. albicans) killing in naive monocytes. Collectively, these data demonstrate that immunotolerant monocytes are characterized by a loss of metabolic plasticity and these metabolic defects impact antimicrobial monocyte immune functions. Further, these findings support that the changed cellular metabolism of immunotolerant monocytes might reveal novel therapeutic targets to reverse sepsis‐induced immunoparalysis., Endotoxin‐induced immunotolerant monocytes are characterized by a loss of metabolic plasticity and these metabolic defects impact antimicrobial monocyte immune functions.
- Published
- 2019
41. LC3-associated phagocytosis: a crucial mechanism for antifungal host defence againstAspergillus fumigatus
- Author
-
Mark S. Gresnigt, Evelien G. G. Sprenkeler, and Frank L. van de Veerdonk
- Subjects
0301 basic medicine ,Aspergillus ,Phagocytosis ,digestive, oral, and skin physiology ,030106 microbiology ,Immunology ,Autophagy ,Pattern recognition receptor ,Biology ,equipment and supplies ,biology.organism_classification ,Microbiology ,Aspergillus fumigatus ,03 medical and health sciences ,030104 developmental biology ,Immunity ,Virology ,Signal transduction ,human activities ,Phagosome - Abstract
LC3-associated phagocytosis (LAP) is a non-canonical autophagy pathway involved in the maturation of single-membrane phagosomes and subsequent killing of ingested pathogens by phagocytes. This pathway is initiated following recognition of pathogens by pattern recognition receptors and leads to the recruitment of LC3 into the phagosomal membrane. This form of phagocytosis is utilized for the antifungal host defence and is required for an efficient fungal killing. Here, we provide an overview of the LAP pathway and review the role of LAP in anti-Aspergillus host defence, as well as mechanisms induced by Aspergillus that modulate LAP to promote its survival in the host.
- Published
- 2016
- Full Text
- View/download PDF
42. Author Correction: A guiding map for inflammation
- Author
-
Daniel L. Kastner, Evangelos J. Giamarellos-Bourboulis, Marc Y. Donath, Thomas Mandrup-Poulsen, Mihai G. Netea, Hal M. Hoffman, Peter Libby, Frank L. van de Veerdonk, Britta Siegmund, Paul M. Ridker, Frances R. Balkwill, Alberto Mantovani, Martin Korte, Leo A. B. Joosten, Joost Schalkwijk, Eicke Latz, Jos W. M. van der Meer, Kingston H. G. Mills, Herbert Tilg, David A. Schwartz, Mark S. Gresnigt, Tom van der Poll, Luke A. J. O'Neill, Clifford J. Steer, Michel Chonchol, Peter Pickkers, Michael T. Heneka, Douglas T. Golenbock, Charles A. Dinarello, Fabio Cominelli, Richard S. Hotchkiss, and Kristen L. Nowak
- Subjects
Twilight ,Political science ,Published Erratum ,European research ,Immunology ,MEDLINE ,Immunology and Allergy ,Library science ,ddc:610 ,ComputingMilieux_MISCELLANEOUS ,GeneralLiterature_MISCELLANEOUS - Abstract
In the version of this article initially published, in the Acknowledgements, the grant number for the Start Grant TWILIGHT awarded to M.A.L. by the European Research Council was omitted. The correct wording is “Supported by...the European Research Council (...Start Grant TWILIGHT (637801) to M.A.L.).” The error has not been corrected in the original article.
- Published
- 2020
- Full Text
- View/download PDF
43. 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
44. Adjunctive interferon-γ immunotherapy in a pediatric case of Aspergillus terreus infection
- Author
-
Natasja Dors, Mark S. Gresnigt, Eemke L. Assendorp, Jan W. M. van der Linden, Stefanie S. V. Henriet, Jacques F. Meis, and Evelien G. G. Sprenkeler
- Subjects
0301 basic medicine ,Microbiology (medical) ,Antifungal Agents ,medicine.medical_treatment ,030106 microbiology ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Other Research Radboud Institute for Molecular Life Sciences [Radboudumc 0] ,Opportunistic Infections ,Aspergillosis ,Monocytes ,Aspergillus fumigatus ,Mannans ,03 medical and health sciences ,Interferon-gamma ,Immune system ,Amphotericin B ,medicine ,Humans ,Aspergillus terreus ,skin and connective tissue diseases ,Child ,Cells, Cultured ,biology ,business.industry ,Galactose ,General Medicine ,Immunotherapy ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,biology.organism_classification ,bacterial infections and mycoses ,Recombinant Proteins ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,Infectious Diseases ,Cytokine ,Aspergillus ,Treatment Outcome ,Immunology ,Cytokines ,Female ,business ,Adjuvant ,medicine.drug - Abstract
Item does not contain fulltext Aspergillus terreus causes invasive aspergillosis (IA) in immunocompromised patients. Treatment is complicated by intrinsic resistance to amphotericin B and thereby contributing to a high mortality. Therefore, we conducted in vitro studies to investigate the effectivity of adjunctive recombinant interferon-gamma immunotherapy. We describe a pediatric patient with A. terreus IA who received adjunctive recombinant interferon-gamma (rIFNgamma) immunotherapy. In vitro studies were conducted to investigate the capacity of rIFNgamma to improve antifungal host defense in terms of fungal killing ability and the release of pro-inflammatory cytokines in cells of the patient as well as healthy controls. An 8-year-old female pediatric patient with leukemia developed A. terreus IA. She clinically deteriorated and had high serum galactomannan levels despite broad antifungal therapy. Therefore, adjunctive immune stimulatory therapy with rIFNgamma was initiated. After 3 weeks of treatment, galactomannan levels decreased and the patient clinically showed improvement. Addition of rIFNgamma boosted the capacity of monocytes of healthy volunteers to mount TNFalpha and IL-1beta cytokine responses to Escherichia coli LPS, and increased TNFalpha response to both A. terreus and Aspergillus fumigatus. Monocytes isolated from the patient's blood demonstrated a similar augmented cytokine induction in response to rIFNgamma. In addition, rIFNgamma increased the capacity of monocytes from healthy volunteers as well as monocytes from the patient to kill A. terreus spores. Adjuvant immunotherapy with rIFNgamma might be a promising additional treatment strategy that could be used to improve outcome in patients with refractory invasive A. terreus infections or other resistant invasive Aspergillus infections.
- Published
- 2018
45. Microbiological and immunological characteristics of a lethal pulmonary Aspergillus niger infection in a non-neutropenic patient
- Author
-
Peter Pickkers, Katharina L. Becker, Eva Kolwijck, Yvonne F. Heijdra, Jessica D. Workum, Mark S. Gresnigt, Frank L. van de Veerdonk, and Suzanne W. de Jong
- Subjects
0301 basic medicine ,medicine.drug_class ,030106 microbiology ,Antibiotics ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Case Report ,Invasive pulmonary aspergillosis ,Microbiology ,Aspergillus niger infection ,law.invention ,03 medical and health sciences ,Immune system ,All institutes and research themes of the Radboud University Medical Center ,law ,medicine ,Intensive care unit ,lcsh:QH301-705.5 ,Aspergillus ,COPD ,lcsh:R5-920 ,biology ,business.industry ,Aspergillus niger ,biology.organism_classification ,Acquired immune system ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,Infectious Diseases ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,lcsh:Biology (General) ,Oxalate crystal ,Immunology ,Inflammatory diseases Radboud Institute for Health Sciences [Radboudumc 5] ,business ,lcsh:Medicine (General) - 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
46. Pattern recognition pathways leading to a Th2 cytokine bias in allergic bronchopulmonary aspergillosis patients
- Author
-
Martin Jaeger, Sanne P. Smeekens, Cecile Magis-Escurra, M. H. Reijers, Katharina L. Becker, Mihai G. Netea, Marije Oosting, X. Wang, R. Lubbers, F.L. van de Veerdonk, Mark S. Gresnigt, Cor Jacobs, and Leo A. B. Joosten
- Subjects
Adult ,Male ,medicine.medical_treatment ,Immunology ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Macrophage-1 Antigen ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Ligands ,Microbiology ,Aspergillus fumigatus ,Young Adult ,Th2 Cells ,Phagocytosis ,T-Lymphocyte Subsets ,medicine ,Humans ,Immunology and Allergy ,Lectins, C-Type ,skin and connective tissue diseases ,Interleukin 5 ,Antibodies, Fungal ,Aged ,biology ,Aspergillosis, Allergic Bronchopulmonary ,Pattern recognition receptor ,Immunoglobulin E ,Middle Aged ,Th1 Cells ,medicine.disease ,biology.organism_classification ,TLR2 ,lnfectious Diseases and Global Health Radboud Institute for Health Sciences [Radboudumc 4] ,Aspergillus ,Cytokine ,Case-Control Studies ,Immunoglobulin G ,Receptors, Pattern Recognition ,Mutation ,Interleukin 13 ,Inflammatory diseases Radboud Institute for Health Sciences [Radboudumc 5] ,Leukocytes, Mononuclear ,TLR4 ,Cytokines ,Female ,Allergic bronchopulmonary aspergillosis ,Signal Transduction - Abstract
Contains fulltext : 153612.pdf (Publisher’s version ) (Closed access) BACKGROUND: Allergic bronchopulmonary aspergillosis (ABPA) is characterised by an exaggerated Th2 response to Aspergillus fumigatus, but the immunological pathways responsible for this effect are unknown. OBJECTIVE: The aim of this study was to decipher the pattern recognition receptors (PRRs) and cytokines involved in the Aspergillus-specific Th2 response and to study Aspergillus-induced responses in healthy controls and ABPA patients. METHODS: Peripheral blood mononuclear cells (PBMCs) were stimulated with heat-killed Aspergillus conidia, various other pathogens, or PRR ligands. PRRs and cytokine pathways were blocked with PRR-blocking reagents, anti-TNF (Etanercept or Adalimumab), IL-1Ra (Anakinra) or IFNgamma (IFN-gamma). ELISA and FACS were used to analyse cytokine responses. RESULTS: Aspergillus was the only pathogen that stimulated the Th2 cytokines IL-5 and IL-13, while Gram-negative bacteria, Gram-positive bacteria, Candida albicans, chitin, beta-glucan or Toll-like receptor (TLR) ligands did not. Depletion of CD4(+) cells abolished IL-13 production. Blocking complement receptor 3 (CR3) significantly reduced IL-5 and IL-13, while blocking TLR2, TLR4 or dectin-1 had no effect. ABPA patients displayed increased Aspergillus-induced IL-5 and IL-13 and decreased IFNgamma production compared with healthy controls. All biological agents tested showed the capability to inhibit Th2 responses, but also decreased Aspergillus-induced IFNgamma. CONCLUSIONS AND CLINICAL RELEVANCE: Aspergillus conidia are unique in triggering Th2 responses in human PBMCs, through a CR3-dependent pathway. ABPA patients display a significantly increased Aspergillus-induced Th2/Th1 ratio that can be modulated by biologicals. These data provide a rationale to explore IFNgamma therapy in ABPA as a corticosteroid-sparing treatment option, by dampening Th2 responses and supplementing the IFNgamma deficiency at the same time.
- Published
- 2015
- Full Text
- View/download PDF
47. 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
48. Genetic deficiency of NOD2 confers resistance to invasive aspergillosis
- Author
-
Marije Oosting, Grégory Jouvion, António Campos, Katrien Lagrou, Willem J. G. Melchers, R. Lubbers, Cristina Cunha, Thirumala-Devi Kanneganti, Dirk J. de Jong, Frank L. van de Veerdonk, Mark S. Gresnigt, Anne Ammerdorffer, Agostinho Carvalho, Martin Jaeger, Catherine Fitting, Johan Maertens, Samuel M. Gonçalves, João F. Lacerda, Orhan Rasid, Oumaïma Ibrahim-Granet, R. K. Subbarao Malireddi, Universidade do Minho, and Repositório da Universidade de Lisboa
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Nod2 Signaling Adaptor Protein ,General Physics and Astronomy ,Hematopoietic stem cell transplantation ,Aspergillosis ,Risk Factors ,NOD2 ,Paranasal Sinuses ,lcsh:Science ,Lung ,Disease Resistance ,Multidisciplinary ,Women's cancers Radboud Institute for Molecular Life Sciences [Radboudumc 17] ,biology ,Hematopoietic Stem Cell Transplantation ,3. Good health ,Aspergillus ,Cytokines ,Female ,Phagocytosis ,Science ,Other Research Radboud Institute for Molecular Life Sciences [Radboudumc 0] ,Rare cancers Radboud Institute for Molecular Life Sciences [Radboudumc 9] ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,In vivo ,Genetic variation ,medicine ,Animals ,Humans ,Lectins, C-Type ,Science & Technology ,Microbial Viability ,General Chemistry ,medicine.disease ,biology.organism_classification ,digestive system diseases ,Transplantation ,Mice, Inbred C57BL ,030104 developmental biology ,Immunology ,lcsh:Q - Abstract
© The Author(s) 2018. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/., Invasive aspergillosis (IA) is a severe infection that can occur in severely immunocompromised patients. Efficient immune recognition of Aspergillus is crucial to protect against infection, and previous studies suggested a role for NOD2 in this process. However, thorough investigation of the impact of NOD2 on susceptibility to aspergillosis is lacking. Common genetic variations in NOD2 has been associated with Crohn's disease and here we investigated the influence of these genetic variations on the anti-Aspergillus host response. A NOD2 polymorphism reduced the risk of IA after hematopoietic stem-cell transplantation. Mechanistically, absence of NOD2 in monocytes and macrophages increases phagocytosis leading to enhanced fungal killing, conversely, NOD2 activation reduces the antifungal potential of these cells. Crucially, Nod2 deficiency results in resistance to Aspergillus infection in an in vivo model of pulmonary aspergillosis. Collectively, our data demonstrate that genetic deficiency of NOD2 plays a protective role during Aspergillus infection., F.L.v.d.V. was supported by the E-rare project EURO-CMC. M.O. was supported by the NWO, 016.176.006). A.C. and C.C. were supported by the Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, through the European Regional Development Fund (FEDER) (NORTE-01-0145-FEDER-000013), and the Fundação para a Ciência e Tecnologia (FCT) (IF/00735/2014 to A.C. and SFRH/BPD/96176/2013 to C.C.).
- Published
- 2017
49. Personalized medicine in influenza: a bridge too far or the near future?
- Author
-
Paul E. Verweij, Mihai G. Netea, Frank L. van de Veerdonk, and Mark S. Gresnigt
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Pathology ,medicine.medical_treatment ,Alternative medicine ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Context (language use) ,Disease ,Bridge (nautical) ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Intervention (counseling) ,Influenza, Human ,medicine ,Humans ,Precision Medicine ,Intensive care medicine ,business.industry ,Regimen ,030104 developmental biology ,Drug Design ,Personalized medicine ,business ,030215 immunology - Abstract
Item does not contain fulltext PURPOSE OF REVIEW: Personalized medicine is based on understanding mechanisms of disease and putting this in the context of an individual patient, which eventually helps to guide tailored diagnostic and therapeutic intervention. In this review we focus on one pulmonary infection that has major impact on society, namely influenza, and highlight the way we think personalized medicine could have an impact on the outcome of this pulmonary infection. RECENT FINDINGS: When a patient's defect is known, one could envision that restoring this defect in addition to the standard treatment regimen might result in a better clinical outcome. By highlighting the way one can explore mechanisms of disease, the recent progress in understanding influenza and its complications, and clinical observations, we have written a hypothesis-driven review that underscores in our opinion the way we could think about personalized medicine and realize its translation to the clinics. SUMMARY: This strategy will identify essential mechanisms that cause disease, design simple functional tests that have the ability to identify defects in these relevant mechanisms in individual patients, and explore targeted therapy to restore these defects, ideally with existing drugs.
- Published
- 2017
50. Toll-like receptor 2 induced cytotoxic T-lymphocyte-associated protein 4 regulates Aspergillus-induced regulatory T-cells with pro-inflammatory characteristics
- Author
-
Frank L. van de Veerdonk, Floor Aleva, Cor Jacobs, Ruud P. H. Raijmakers, Evelien G. G. Sprenkeler, Leo A. B. Joosten, Thirumala-Devi Kanneganti, and Mark S. Gresnigt
- Subjects
0301 basic medicine ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,lcsh:Medicine ,chemical and pharmacologic phenomena ,Aspergillosis ,Ligands ,T-Lymphocytes, Regulatory ,Article ,Aspergillus fumigatus ,Immunophenotyping ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Immunopathology ,medicine ,Cytotoxic T cell ,Animals ,Humans ,CTLA-4 Antigen ,lcsh:Science ,Mice, Knockout ,Toll-like receptor ,Multidisciplinary ,biology ,lcsh:R ,biology.organism_classification ,medicine.disease ,Phenotype ,Toll-Like Receptor 2 ,3. Good health ,TLR2 ,030104 developmental biology ,Aspergillus ,Case-Control Studies ,Receptors, Pattern Recognition ,Immunology ,Inflammatory diseases Radboud Institute for Health Sciences [Radboudumc 5] ,Cytokines ,Th17 Cells ,lcsh:Q ,Inflammation Mediators ,030215 immunology - Abstract
Patients with cystic fibrosis, chronic obstructive pulmonary disease, severe asthma, pre-existing pulmonary lesions, and severely immunocompromised patients are susceptible to develop infections with the opportunistic pathogenic fungus Aspergillus fumigatus, called aspergillosis. Infections in these patients are associated with persistent pro-inflammatory T-helper (TH)2 and TH17 responses. Regulatory T-cells, natural suppressor cells of the immune system, control pro-inflammatory T-cell responses, but can also contribute to disease by shifting to a pro-inflammatory TH17-like phenotype. Such a shift could play an important role in the detrimental immunopathology that is seen in aspergillosis. Our study demonstrates that Aspergillus fumigatus induces regulatory T-cells with a TH17-like phenotype. We also demonstrate that these regulatory T-cells with a pro-inflammatory TH17-like phenotype can be reprogrammed to their “classical” anti-inflammatory phenotype by activating Toll-like receptor 2 (TLR2), which regulates the induction of cytotoxic T-lymphocyte-associated protein 4 (CTLA4). Similarly, soluble CTLA4 could reverse the pro-inflammatory phenotype of Aspergillus-induced regulatory T-cells. In conclusion, our results suggest a role for regulatory T-cells with a pro-inflammatory TH17-like phenotype in Aspergillus-associated immunopathology, and identifies key players, i.e. TLR2 and CTLA4, involved in this mechanism.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.