Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Microbiol Spectr ; 12(5): e0425522, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38587411

RESUMO

tRNA modifications play important roles in maintaining translation accuracy in all domains of life. Disruptions in the tRNA modification machinery, especially of the anticodon stem loop, can be lethal for many bacteria and lead to a broad range of phenotypes in baker's yeast. Very little is known about the function of tRNA modifications in host-pathogen interactions, where rapidly changing environments and stresses require fast adaptations. We found that two closely related fungal pathogens of humans, the highly pathogenic Candida albicans and its much less pathogenic sister species, Candida dubliniensis, differ in the function of a tRNA-modifying enzyme. This enzyme, Hma1, exhibits species-specific effects on the ability of the two fungi to grow in the hypha morphology, which is central to their virulence potential. We show that Hma1 has tRNA-threonylcarbamoyladenosine dehydratase activity, and its deletion alters ribosome occupancy, especially at 37°C-the body temperature of the human host. A C. albicans HMA1 deletion mutant also shows defects in adhesion to and invasion into human epithelial cells and shows reduced virulence in a fungal infection model. This links tRNA modifications to host-induced filamentation and virulence of one of the most important fungal pathogens of humans.IMPORTANCEFungal infections are on the rise worldwide, and their global burden on human life and health is frequently underestimated. Among them, the human commensal and opportunistic pathogen, Candida albicans, is one of the major causative agents of severe infections. Its virulence is closely linked to its ability to change morphologies from yeasts to hyphae. Here, this ability is linked-to our knowledge for the first time-to modifications of tRNA and translational efficiency. One tRNA-modifying enzyme, Hma1, plays a specific role in C. albicans and its ability to invade the host. This adds a so-far unknown layer of regulation to the fungal virulence program and offers new potential therapeutic targets to fight fungal infections.


Assuntos
Candida albicans , Candidíase , Proteínas Fúngicas , Hifas , RNA de Transferência , Candida albicans/genética , Candida albicans/patogenicidade , Candida albicans/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo , Virulência/genética , Humanos , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Candidíase/microbiologia , Hifas/crescimento & desenvolvimento , Hifas/genética , Hifas/metabolismo , Animais , Candida/patogenicidade , Candida/genética , Candida/metabolismo , Interações Hospedeiro-Patógeno , Camundongos , Células Epiteliais/microbiologia
2.
Microorganisms ; 11(8)2023 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-37630490

RESUMO

Candida albicans is a pathobiont of the gastrointestinal tract. It can contribute to the diversity of the gut microbiome without causing harmful effects. When the immune system is compromised, C. albicans can damage intestinal cells and cause invasive disease. We hypothesize that a therapeutic approach against C. albicans infections can rely on the antimicrobial properties of probiotic bacteria. We investigated the impact of the probiotic strain Escherichia coli Nissle 1917 (EcN) on C. albicans growth and its ability to cause damage to intestinal cells. In co-culture kinetic assays, C. albicans abundance gradually decreased over time compared with C. albicans abundance in the absence of EcN. Quantification of C. albicans survival suggests that EcN exerts a fungicidal activity. Cell-free supernatants (CFS) collected from C. albicans-EcN co-culture mildly altered C. albicans growth, suggesting the involvement of an EcN-released compound. Using a model of co-culture in the presence of human intestinal epithelial cells, we further show that EcN prevents C. albicans from damaging enterocytes both distantly and through direct contact. Consistently, both C. albicans's filamentous growth and microcolony formation were altered by EcN. Taken together, our study proposes that probiotic-strain EcN can be exploited for future therapeutic approaches against C. albicans infections.

3.
J Immunol ; 211(5): 804-815, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37436030

RESUMO

Because of the growing numbers of immunocompromised patients, the incidence of life-threatening fungal infections caused by Candida albicans and Aspergillus fumigatus is increasing. We have recently identified enolase 1 (Eno1) from A. fumigatus as an immune evasion protein. Eno1 is a fungal moonlighting protein that mediates adhesion and invasion of human cells and also immune evasion through complement inactivation. We now show that soluble Eno1 has immunostimulatory activity. We observed that Eno1 from both C. albicans and A. fumigatus directly binds to the surface of lymphocytes, preferentially human and mouse B cells. Functionally, Eno1 upregulated CD86 expression on B cells and induced proliferation. Although the receptor for fungal Eno1 on B lymphocytes is still unknown, the comparison of B cells from wild-type and MyD88-deficient mice showed that B cell activation by Eno1 required MyD88 signaling. With respect to infection biology, we noted that mouse B cells stimulated by Eno1 secreted IgM and IgG2b. These Igs bound C. albicans hyphae in vitro, suggesting that Eno1-induced Ab secretion might contribute to protection from invasive fungal disease in vivo. Eno1 also triggered the release of proinflammatory cytokines from monocytes, particularly IL-6, which is a potent activator of B cells. Together, our data shed new light on the role of secreted Eno1 in infections with C. albicans and A. fumigatus. Eno1 secretion by these pathogenic microbes appears to be a double-edged sword by supporting fungal pathogenicity while triggering (antifungal) immunity.


Assuntos
Aspergillus fumigatus , Candida albicans , Fosfopiruvato Hidratase , Animais , Humanos , Camundongos , Aspergillus fumigatus/enzimologia , Aspergillus fumigatus/metabolismo , Candida albicans/enzimologia , Candida albicans/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Monócitos/metabolismo , Monócitos/microbiologia , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fosfopiruvato Hidratase/metabolismo , Linfócitos B/metabolismo , Linfócitos B/microbiologia
4.
Curr Clin Microbiol Rep ; 10(2): 55-65, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37151578

RESUMO

Purpose of Review: The fungus Candida albicans has evolved to live in close association with warm-blooded hosts and is found frequently on mucosal surfaces of healthy humans. As an opportunistic pathogen, C. albicans can also cause mucosal and disseminated infections (candidiasis). This review describes the features that differentiate the fungus in the commensal versus pathogenic state and the main factors underlying C. albicans commensal-to-pathogen transition. Recent Findings: Adhesion, invasion, and tissue damage are critical steps in the infection process. Especially invasion and damage require transcriptional and morphological changes that differentiate C. albicans in the pathogenic from the commensal state. While the commensal-to-pathogen transition has some conserved causes and features in the oral cavity, the female urogenital tract, and the gut, site-specific differences have been identified in recent years. Summary: This review highlights how specific factors in the different mucosal niches affect development of candidiasis. Recent evidence suggests that colonization of the gut is not only a risk factor for systemic candidiasis but might also provide beneficial effects to the host.

5.
Neuropharmacology ; 235: 109568, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37182790

RESUMO

Depression is highly prevalent (6% 1-year prevalence) and is the second leading cause of disability worldwide. Available treatment options for depression are far from optimal, with response rates only around 50%. This is most likely related to a heterogeneous clinical presentation of major depression disorder (MDD), suggesting different manifestations of underlying pathophysiological mechanisms. Poorer treatment outcomes to first-line antidepressants were reported in MDD patients endorsing an "atypical" symptom profile that is characterized by preserved reactivity in mood, increased appetite, hypersomnia, a heavy sensation in the limbs, and interpersonal rejection sensitivity. In recent years, evidence has emerged that immunometabolic biological dysregulation is an important underlying pathophysiological mechanism in depression, which maps more consistently to atypical features. In the last few years human microbial residents have emerged as a key influencing variable associated with immunometabolic dysregulations in depression. The microbiome plays a critical role in the training and development of key components of the host's innate and adaptive immune systems, while the immune system orchestrates the maintenance of key features of the host-microbe symbiosis. Moreover, by being a metabolically active ecosystem commensal microbes may have a huge impact on signaling pathways, involved in underlying mechanisms leading to atypical depressive symptoms. In this review, we discuss the interplay between the microbiome and immunometabolic imbalance in the context of atypical depressive symptoms. Although research in this field is in its infancy, targeting biological determinants in more homogeneous clinical presentations of MDD may offer new avenues for the development of novel therapeutic strategies for treatment-resistant depression. This article is part of the Special Issue on "Microbiome & the Brain: Mechanisms & Maladies".


Assuntos
Transtorno Depressivo Maior , Microbiota , Humanos , Transtorno Depressivo Maior/metabolismo , Encéfalo/metabolismo
6.
Methods Mol Biol ; 2667: 181-196, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37145285

RESUMO

Animal models have been crucial in understanding the pathogenesis and developing novel therapeutic approaches for fungal infections in general. This is especially true for mucormycosis, which has a low incidence but is often fatal or debilitating. Mucormycoses are caused by different species, via different routes of infections, and in patients differing in their underlying diseases and risk factors. Consequently, clinically relevant animal models use different types of immunosuppression and infection routes.This chapter describes how to induce different types of immunosuppression (high dose corticosteroids and induction of leukopenia, respectively) or diabetic ketoacidosis as underlying risk factors for mucormycosis. Furthermore, it provides details on how to perform intranasal application to establish pulmonary infection. Finally, some clinical parameters that can be used for developing scoring systems and define humane endpoints in mice are discussed.


Assuntos
Mucormicose , Animais , Camundongos , Mucormicose/microbiologia , Terapia de Imunossupressão , Modelos Animais de Doenças
7.
Aging Cell ; 22(7): e13856, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37101405

RESUMO

Macrophages adapt distinct pro-inflammatory (M1-like) and pro-resolving (M2-like) phenotypes with specific tasks in the immune response and tissue homeostasis. Altered macrophage responses with age are causative for unresolved inflammation, so-called inflammaging, and lead to higher infection susceptibility with unfavorable progression. Here, we reveal molecular determinants of age-related changes in phenotypic functions of murine peritoneal macrophages (PM) by employing comprehensive mass spectrometry-based proteomics (4746 protein groups) and metabololipidomics (>40 lipid mediators). Divergent expression of various macrophage-specific marker proteins and signaling pathways indicates aberrant PM phenotypes in old mice which detrimentally impact their capabilities to release immunomodulatory chemokines and cytokines. We show that aging strikingly compromises the polarization process of macrophages to adapt either pro-inflammatory or pro-resolving phenotypes, thereby yielding aberrant and afunctional macrophage subtypes that cannot be readily assigned to either a typical M1 or M2 phenotype. In particular, the phenotypic adaptation of the bacteria-challenged metabololipidome in macrophages related to inflammation is severely limited by age, which persists across ex vivo polarization towards M1 and M2a macrophages. Our results establish distinct age-associated PM phenotypes outside of the simplified M1 and M2 dichotomy and challenge the dogma of increased pro-inflammatory macrophage pre-activation due to aging by revealing maladaptive functions throughout all phases of inflammation, including resolution.


Assuntos
Ativação de Macrófagos , Proteômica , Camundongos , Animais , Inflamação/metabolismo , Envelhecimento , Imunidade
8.
Front Cell Infect Microbiol ; 12: 866416, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35651758

RESUMO

The human gut acts as the main reservoir of microbes and a relevant source of life-threatening infections, especially in immunocompromised patients. There, the opportunistic fungal pathogen Candida albicans adapts to the host environment and additionally interacts with residing bacteria. We investigated fungal-bacterial interactions by coinfecting enterocytes with the yeast Candida albicans and the Gram-negative bacterium Proteus mirabilis resulting in enhanced host cell damage. This synergistic effect was conserved across different P. mirabilis isolates and occurred also with non-albicans Candida species and C. albicans mutants defective in filamentation or candidalysin production. Using bacterial deletion mutants, we identified the P. mirabilis hemolysin HpmA to be the key effector for host cell destruction. Spatially separated coinfections demonstrated that synergism between Candida and Proteus is induced by contact, but also by soluble factors. Specifically, we identified Candida-mediated glucose consumption and farnesol production as potential triggers for Proteus virulence. In summary, our study demonstrates that coinfection of enterocytes with C. albicans and P. mirabilis can result in increased host cell damage which is mediated by bacterial virulence factors as a result of fungal niche modification via nutrient consumption and production of soluble factors. This supports the notion that certain fungal-bacterial combinations have the potential to result in enhanced virulence in niches such as the gut and might therefore promote translocation and dissemination.


Assuntos
Candida albicans , Coinfecção , Candida , Enterócitos , Humanos , Proteus mirabilis/genética
9.
Nat Commun ; 13(1): 1880, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35388002

RESUMO

Sensing of pathogens by pattern recognition receptors (PRR) is critical to initiate protective host defence reactions. However, activation of the immune system has to be carefully titrated to avoid tissue damage necessitating mechanisms to control and terminate PRR signalling. Dectin-1 is a PRR for fungal ß-glucans on immune cells that is rapidly internalised after ligand-binding. Here, we demonstrate that pathogen recognition by the Dectin-1a isoform results in the formation of a stable receptor fragment devoid of the ligand binding domain. This fragment persists in phagosomal membranes and contributes to signal transduction which is terminated by the intramembrane proteases Signal Peptide Peptidase-like (SPPL) 2a and 2b. Consequently, immune cells lacking SPPL2b demonstrate increased anti-fungal ROS production, killing capacity and cytokine responses. The identified mechanism allows to uncouple the PRR signalling response from delivery of the pathogen to degradative compartments and identifies intramembrane proteases as part of a regulatory circuit to control anti-fungal immune responses.


Assuntos
Lectinas Tipo C , Transdução de Sinais , Lectinas Tipo C/metabolismo , Ligantes , Proteólise , Receptores de Reconhecimento de Padrão/metabolismo
10.
PLoS Pathog ; 18(2): e1010283, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35108336

RESUMO

Protein kinases play central roles in virtually all signaling pathways that enable organisms to adapt to their environment. Microbial pathogens must cope with severely restricted iron availability in mammalian hosts to invade and establish themselves within infected tissues. To uncover protein kinase signaling pathways that are involved in the adaptation of the pathogenic yeast Candida albicans to iron limitation, we generated a comprehensive protein kinase deletion mutant library of a wild-type strain. Screening of this library revealed that the protein kinase Ire1, which has a conserved role in the response of eukaryotic cells to endoplasmic reticulum stress, is essential for growth of C. albicans under iron-limiting conditions. Ire1 was not necessary for the activity of the transcription factor Sef1, which regulates the response of the fungus to iron limitation, and Sef1 target genes that are induced by iron depletion were normally upregulated in ire1Δ mutants. Instead, Ire1 was required for proper localization of the high-affinity iron permease Ftr1 to the cell membrane. Intriguingly, iron limitation did not cause increased endoplasmic reticulum stress, and the transcription factor Hac1, which is activated by Ire1-mediated removal of the non-canonical intron in the HAC1 mRNA, was dispensable for Ftr1 localization to the cell membrane and growth under iron-limiting conditions. Nevertheless, expression of a pre-spliced HAC1 copy in ire1Δ mutants restored Ftr1 localization and rescued the growth defects of the mutants. Both ire1Δ and hac1Δ mutants were avirulent in a mouse model of systemic candidiasis, indicating that an appropriate response to endoplasmic reticulum stress is important for the virulence of C. albicans. However, the specific requirement of Ire1 for the functionality of the high-affinity iron permease Ftr1, a well-established virulence factor, even in the absence of endoplasmic reticulum stress uncovers a novel Hac1-independent essential role of Ire1 in iron acquisition and virulence of C. albicans.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Candida albicans/metabolismo , Candidíase/microbiologia , Ferro/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Candida albicans/genética , Candida albicans/patogenicidade , DNA Fúngico , Estresse do Retículo Endoplasmático , Feminino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Proteínas de Membrana Transportadoras/genética , Camundongos Endogâmicos BALB C , Proteínas Serina-Treonina Quinases/genética , Deleção de Sequência , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Virulência
11.
Front Fungal Biol ; 3: 940884, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37746206

RESUMO

Animal models are essential to understand the pathophysiology of infections, to test novel antifungal compounds, and to determine the potential of adjunctive therapies, e.g. immune modulation. The murine model of systemic candidiasis induced by intravenous infection is technically straightforward, highly reproducible, and well-characterized. However, intravenous inoculation circumvents the necessity for the fungus to translocate across mucosal barriers, and the use of SPF mice that are immunologically naïve to Candida does not reflect the situation in human patients, in whom adaptive immune responses have been induced by mucosal colonization prior to infection. Therefore, mouse models that combine intestinal colonization and systemic infection have been developed, resulting in novel insights into host-fungal interactions and immunity. In this review, we summarize the main findings, current questions, and discuss how these might impact the translatability of results from mice to humans.

12.
Virulence ; 12(1): 3137-3148, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34923920

RESUMO

Candida albicans is a common commensal on human mucosal surfaces, but can become pathogenic, e.g. if the host is immunocompromised. While neutrophils, macrophages and T cells are regarded as major players in the defense against pathogenic C. albicans, the role of B cells and the protective function of their antibodies are less well characterized. In this study, we show that human serum antibodies are able to enhance the association of human THP-1 monocyte-like cells with C. albicans cells. Human serum antibodies are also capable of inhibiting the adherence and damage dealt to epithelial cells. Furthermore, human serum antibodies impair C. albicans invasion of human oral epithelial cells by blocking induced endocytosis and consequently host cell damage. While aspartic proteases secreted by C. albicans are able to cleave human IgG, this process does not appear to affect the protective function of human antibodies. Thus, humans are equipped with a robust antibody response to C. albicans, which can enhance antifungal activities and prevent fungal-mediated epithelial damage.


Assuntos
Ácido Aspártico Proteases , Candida albicans , Formação de Anticorpos , Antifúngicos/farmacologia , Ácido Aspártico Endopeptidases , Humanos
13.
Front Immunol ; 12: 698849, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34819929

RESUMO

Candida albicans is usually a benign member of the human gut microbiota, but can become pathogenic under certain circumstances, for example in an immunocompromised host. The innate immune system, in particular neutrophils and macrophages, constitutes a crucial first line of defense against fungal invasion, however adaptive immunity may provide long term protection and thus allow vaccination of at risk patients. While TH1 and TH17 cells are important for antifungal responses, the role of B cells and antibodies in protection from C. albicans infection is less well defined. In this study, we show that C. albicans hyphae but not yeast, as well as fungal cell wall components, directly activate B cells via MyD88 signaling triggered by Toll- like receptor 2, leading to increased IgG1 production. While Dectin-1 signals and specific recognition by the B cell receptor are dispensable for B cell activation in this system, TLR2/MyD88 signals cooperate with CD40 signals in promoting B cell activation. Importantly, recognition of C. albicans via MyD88 signaling is also essential for induction of IL-6 secretion by B cells, which promotes TH17 polarization in T-B cell coculture experiments. B cells may thus be activated directly by C. albicans in its invasive form, leading to production of antibodies and T cell help for fungal clearance.


Assuntos
Linfócitos B/imunologia , Candida albicans/imunologia , Candidíase/imunologia , Diferenciação Celular , Hifas/imunologia , Imunoglobulina G/metabolismo , Interleucina-6/metabolismo , Células Th17/imunologia , Receptor 2 Toll-Like/metabolismo , Animais , Linfócitos B/metabolismo , Linfócitos B/microbiologia , Candida albicans/patogenicidade , Candidíase/metabolismo , Candidíase/microbiologia , Células Cultivadas , Técnicas de Cocultura , Interações Hospedeiro-Patógeno , Humanos , Hifas/patogenicidade , Ativação Linfocitária , Camundongos Endogâmicos C57BL , Fenótipo , Via Secretória , Transdução de Sinais , Células Th17/metabolismo , Células Th17/microbiologia
14.
Nat Commun ; 12(1): 3899, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162849

RESUMO

The ability of the fungal pathogen Candida albicans to undergo a yeast-to-hypha transition is believed to be a key virulence factor, as filaments mediate tissue damage. Here, we show that virulence is not necessarily reduced in filament-deficient strains, and the results depend on the infection model used. We generate a filament-deficient strain by deletion or repression of EED1 (known to be required for maintenance of hyphal growth). Consistent with previous studies, the strain is attenuated in damaging epithelial cells and macrophages in vitro and in a mouse model of intraperitoneal infection. However, in a mouse model of systemic infection, the strain is as virulent as the wild type when mice are challenged with intermediate infectious doses, and even more virulent when using low infectious doses. Retained virulence is associated with rapid yeast proliferation, likely the result of metabolic adaptation and improved fitness, leading to high organ fungal loads. Analyses of cytokine responses in vitro and in vivo, as well as systemic infections in immunosuppressed mice, suggest that differences in immunopathology contribute to some extent to retained virulence of the filament-deficient mutant. Our findings challenge the long-standing hypothesis that hyphae are essential for pathogenesis of systemic candidiasis by C. albicans.


Assuntos
Candida albicans/metabolismo , Candidíase/metabolismo , Proteínas Fúngicas/metabolismo , Hifas/metabolismo , Animais , Candida albicans/genética , Candida albicans/patogenicidade , Candidíase/microbiologia , Divisão Celular/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Humanos , Hifas/genética , Hifas/crescimento & desenvolvimento , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Mutação , Neutrófilos/metabolismo , Virulência/genética
15.
FEMS Microbiol Rev ; 45(3)2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33232448

RESUMO

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.


Assuntos
Candidíase/imunologia , Candidíase/microbiologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Interações Microbianas/fisiologia , Candida albicans/imunologia , Candida albicans/patogenicidade , Humanos
16.
Sci Rep ; 10(1): 21818, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33311597

RESUMO

Burn wounds are highly susceptible sites for colonization and infection by bacteria and fungi. Large wound surface, impaired local immunity, and broad-spectrum antibiotic therapy support growth of opportunistic fungi such as Candida albicans, which may lead to invasive candidiasis. Currently, it remains unknown whether depressed host defenses or fungal virulence drive the progression of burn wound candidiasis. Here we established an ex vivo burn wound model, where wounds were inflicted by applying preheated soldering iron to human skin explants, resulting in highly reproducible deep second-degree burn wounds. Eschar removal by debridement allowed for deeper C. albicans penetration into the burned tissue associated with prominent filamentation. Active migration of resident tissue neutrophils towards the damaged tissue and release of pro-inflammatory cytokine IL-1ß accompanied the burn. The neutrophil recruitment was further increased upon supplementation of the model with fresh immune cells. Wound area and depth decreased over time, indicating healing of the damaged tissue. Importantly, prominent neutrophil presence at the infected site correlated to the limited penetration of C. albicans into the burned tissue. Altogether, we established a reproducible burn wound model of candidiasis using ex vivo human skin explants, where immune responses actively control the progression of infection and promote tissue healing.


Assuntos
Queimaduras/imunologia , Candida albicans/imunologia , Candidíase/imunologia , Neutrófilos/imunologia , Pele/imunologia , Infecção dos Ferimentos/imunologia , Adulto , Queimaduras/microbiologia , Queimaduras/patologia , Candidíase/patologia , Feminino , Humanos , Interleucina-1beta/imunologia , Pessoa de Meia-Idade , Neutrófilos/patologia , Pele/microbiologia , Pele/patologia , Infecção dos Ferimentos/microbiologia , Infecção dos Ferimentos/patologia
17.
Virulence ; 11(1): 1337-1351, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33043780

RESUMO

Invasive pulmonary aspergillosis (IPA) is a severe infection that is difficult to diagnose due to the ubiquitous presence of fungal spores, the underlying diseases of risk patients, and limitations of currently available markers. In this study, we performed a comprehensive liquid chromatography tandem mass spectrometry (LC-MS/MS)-based identification of host and fungal proteins expressed during IPA in mice and humans. The proteomic analysis of bronchoalveolar lavage samples of individual IPA and control cases allowed the description of common host factors that had significantly increased abundance in both infected animals and IPA patients compared to their controls. Although increased levels of these individual host proteins might not be sufficient to distinguish bacterial from fungal infection, a combination of these markers might be beneficial to improve diagnosis. We also identified 16 fungal proteins that were specifically detected during infection and may be valuable candidates for biomarker evaluation.


Assuntos
Líquido da Lavagem Broncoalveolar/química , Proteínas Fúngicas/análise , Interações Hospedeiro-Patógeno , Aspergilose Pulmonar Invasiva/microbiologia , Proteínas/análise , Proteoma , Adulto , Idoso , Animais , Biomarcadores/análise , Líquido da Lavagem Broncoalveolar/microbiologia , Estudos de Casos e Controles , Cromatografia Líquida , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Organismos Livres de Patógenos Específicos , Espectrometria de Massas em Tandem
18.
mSphere ; 5(4)2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32817377

RESUMO

Typically, established lab strains are widely used to study host-pathogen interactions. However, to better reflect the infection process, the experimental use of clinical isolates has come more into focus. Here, we analyzed the interaction of multiple vaginal isolates of the opportunistic fungal pathogen Candida albicans, the most common cause of vulvovaginal candidiasis in women, with key players of the host immune system: macrophages. We tested several strains isolated from asymptomatic or symptomatic women with acute and recurrent infections. While all clinical strains showed a response similar to the commonly used lab strain SC5314 in various in vitro assays, they displayed remarkable differences during interaction with macrophages. This coincided with significantly reduced ß-glucan exposure on the cell surface, which appeared to be a shared property among the tested vaginal strains for yeast extract/peptone/dextrose-grown cells, which is partly lost when the isolates faced vaginal niche-like nutrient conditions. However, macrophage damage, survival of phagocytosis, and filamentation capacities were highly strain-specific. These results highlight the high heterogeneity of C. albicans strains in host-pathogen interactions, which have to be taken into account to bridge the gap between laboratory-gained data and disease-related outcomes in an actual patient.IMPORTANCE Vulvovaginal candidiasis is one of the most common fungal infections in humans with Candida albicans as the major causative agent. This study is the first to compare clinical vaginal isolates of defined patient groups in their interaction with macrophages, highlighting the vastly different outcomes in comparison to a laboratory strain using commonly applied virulence-determining assays.


Assuntos
Candida albicans/imunologia , Candidíase Vulvovaginal/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/microbiologia , Vagina/microbiologia , Animais , Doenças Assintomáticas , Candida albicans/patogenicidade , Linhagem Celular , Feminino , Humanos , Hifas/crescimento & desenvolvimento , Laboratórios , Macrófagos/imunologia , Camundongos , Fagocitose
19.
J Clin Invest ; 130(9): 4587-4600, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32484796

RESUMO

Th cells integrate signals from their microenvironment to acquire distinct specialization programs for efficient clearance of diverse pathogens or for immunotolerance. Ionic signals have recently been demonstrated to affect T cell polarization and function. Sodium chloride (NaCl) was proposed to accumulate in peripheral tissues upon dietary intake and to promote autoimmunity via the Th17 cell axis. Here, we demonstrate that high-NaCl conditions induced a stable, pathogen-specific, antiinflammatory Th17 cell fate in human T cells in vitro. The p38/MAPK pathway, involving NFAT5 and SGK1, regulated FoxP3 and IL-17A expression in high-NaCl conditions. The NaCl-induced acquisition of an antiinflammatory Th17 cell fate was confirmed in vivo in an experimental autoimmune encephalomyelitis (EAE) mouse model, which demonstrated strongly reduced disease symptoms upon transfer of T cells polarized in high-NaCl conditions. However, NaCl was coopted to promote murine and human Th17 cell pathogenicity, if T cell stimulation occurred in a proinflammatory and TGF-ß-low cytokine microenvironment. Taken together, our findings reveal a context-dependent, dichotomous role for NaCl in shaping Th17 cell pathogenicity. NaCl might therefore prove beneficial for the treatment of chronic inflammatory diseases in combination with cytokine-blocking drugs.


Assuntos
Microambiente Celular/efeitos dos fármacos , Citocinas/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Cloreto de Sódio na Dieta/farmacologia , Células Th17/imunologia , Animais , Microambiente Celular/imunologia , Citocinas/genética , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos Transgênicos , Células Th17/patologia
20.
Nat Commun ; 11(1): 2331, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-32393780

RESUMO

Extracellular vesicles have an important function in cellular communication. Here, we show that human and mouse monocytes release TGF-ß1-transporting vesicles in response to the pathogenic fungus Candida albicans. Soluble ß-glucan from C. albicans binds to complement receptor 3 (CR3, also known as CD11b/CD18) on monocytes and induces the release of TGF-ß1-transporting vesicles. CR3-dependence is demonstrated using CR3-deficient (CD11b knockout) monocytes generated by CRISPR-CAS9 genome editing and isolated from CR3-deficient (CD11b knockout) mice. These vesicles reduce the pro-inflammatory response in human M1-macrophages as well as in whole blood. Binding of the vesicle-transported TGF-ß1 to the TGF-ß receptor inhibits IL1B transcription via the SMAD7 pathway in whole blood and induces TGFB1 transcription in endothelial cells, which is resolved upon TGF-ß1 inhibition. Notably, human complement-opsonized apoptotic bodies induce production of similar TGF-ß1-transporting vesicles in monocytes, suggesting that the early immune response might be suppressed through this CR3-dependent anti-inflammatory vesicle pathway.


Assuntos
Imunomodulação , Antígeno de Macrófago 1/metabolismo , Monócitos/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Vesículas Transportadoras/metabolismo , Animais , Apoptose , Candida albicans/metabolismo , Candida albicans/ultraestrutura , Regulação para Baixo , Difusão Dinâmica da Luz , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/patologia , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Modelos Biológicos , Monócitos/microbiologia , Monócitos/ultraestrutura , Transporte Proteico , Solubilidade , Transcrição Gênica , Regulação para Cima , beta-Glucanas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...