Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
PLoS Pathog ; 20(6): e1012315, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38889192

RESUMEN

Invasive aspergillosis causes significant morbidity and mortality in immunocompromised patients. Natural killer (NK) cells are pivotal for antifungal defense. Thus far, CD56 is the only known pathogen recognition receptor on NK cells triggering potent antifungal activity against Aspergillus fumigatus. However, the underlying cellular mechanisms and the fungal ligand of CD56 have remained unknown. Using purified cell wall components, biochemical treatments, and ger mutants with altered cell wall composition, we herein found that CD56 interacts with the A. fumigatus cell wall carbohydrate galactosaminogalactan (GAG). This interaction induced NK-cell activation, degranulation, and secretion of immune-enhancing chemokines and cytotoxic effectors. Supernatants from GAG-stimulated NK cells elicited antifungal activity and enhanced antifungal effector responses of polymorphonuclear cells. In conclusion, we identified A. fumigatus GAG as a ligand of CD56 on human primary NK cells, stimulating potent antifungal effector responses and activating other immune cells.


Asunto(s)
Aspergilosis , Aspergillus fumigatus , Antígeno CD56 , Células Asesinas Naturales , Humanos , Aspergillus fumigatus/inmunología , Células Asesinas Naturales/inmunología , Antígeno CD56/metabolismo , Antígeno CD56/inmunología , Aspergilosis/inmunología , Aspergilosis/microbiología , Activación de Linfocitos/inmunología , Polisacáridos/metabolismo , Polisacáridos/inmunología , Pared Celular/inmunología , Pared Celular/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(6): e2212003120, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36719915

RESUMEN

While establishing an invasive infection, the dormant conidia of Aspergillus fumigatus transit through swollen and germinating stages, to form hyphae. During this morphotype transition, the conidial cell wall undergoes dynamic remodeling, which poses challenges to the host immune system and antifungal drugs. However, such cell wall reorganization during conidial germination has not been studied so far. Here, we explored the molecular rearrangement of Aspergillus fumigatus cell wall polysaccharides during different stages of germination. We took advantage of magic-angle spinning NMR to investigate the cell wall polysaccharides, without employing any destructive method for sample preparation. The breaking of dormancy was associated with a significant change in the molar ratio between the major polysaccharides ß-1,3-glucan and α-1,3-glucan, while chitin remained equally abundant. The use of various polarization transfers allowed the detection of rigid and mobile polysaccharides; the appearance of mobile galactosaminogalactan was a molecular hallmark of germinating conidia. We also report for the first time highly abundant triglyceride lipids in the mobile matrix of conidial cell walls. Water to polysaccharides polarization transfers revealed an increased surface exposure of glucans during germination, while chitin remained embedded deeper in the cell wall, suggesting a molecular compensation mechanism to keep the cell wall rigidity. We complement the NMR analysis with confocal and atomic force microscopies to explore the role of melanin and RodA hydrophobin on the dormant conidial surface. Exemplified here using Aspergillus fumigatus as a model, our approach provides a powerful tool to decipher the molecular remodeling of fungal cell walls during their morphotype switching.


Asunto(s)
Aspergillus fumigatus , Proteínas Fúngicas , Aspergillus fumigatus/metabolismo , Esporas Fúngicas/metabolismo , Proteínas Fúngicas/metabolismo , Polisacáridos/metabolismo , Quitina/metabolismo , Glucanos/metabolismo , Pared Celular/metabolismo
3.
PLoS Pathog ; 19(11): e1011841, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38033163

RESUMEN

Macrophages play a key role in disseminated cryptococcosis, a deadly fungal disease caused by Cryptococcus neoformans. This opportunistic infection can arise following the reactivation of a poorly characterized latent infection attributed to dormant C. neoformans. Here, we investigated the mechanisms underlying reactivation of dormant C. neoformans using an in vitro co-culture model of viable but non-culturable (VBNC; equivalent of dormant) yeast cells with bone marrow-derived murine macrophages (BMDMs). Comparative transcriptome analysis of BMDMs incubated with log, stationary phase or VBNC cells of C. neoformans showed that VBNC cells elicited a reduced transcriptional modification of the macrophage but retaining the ability to regulate genes important for immune response, such as NLRP3 inflammasome-related genes. We further confirmed the maintenance of the low immunostimulatory capacity of VBNC cells using multiplex cytokine profiling, and analysis of cell wall composition and dectin-1 ligands exposure. In addition, we evaluated the effects of classic (M1) or alternative (M2) macrophage polarization on VBNC cells. We observed that intracellular residence sustained dormancy, regardless of the polarization state of macrophages and despite indirect detection of pantothenic acid (or its derivatives), a known reactivator for VBNC cells, in the C. neoformans-containing phagolysosome. Notably, M0 and M2, but not M1 macrophages, induced extracellular reactivation of VBNC cells by the secretion of extracellular vesicles and non-lytic exocytosis. Our results indicate that VBNC cells retain the low immunostimulatory profile required for persistence of C. neoformans in the host. We also describe a pro-pathogen role of macrophage-derived extracellular vesicles in C. neoformans infection and reinforce the impact of non-lytic exocytosis and the macrophage profile on the pathophysiology of cryptococcosis.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Vesículas Extracelulares , Animales , Ratones , Cryptococcus neoformans/genética , Criptococosis/microbiología , Macrófagos , Exocitosis
4.
Mycopathologia ; 189(5): 86, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39302505

RESUMEN

Caspofungin, a lipopeptide, is an antifungal drug that belong to the class of echinocandin. It inhibits fungal cell wall ß-(1,3)-glucan synthase activity and is the second-line of drug for invasive aspergillosis, a fatal infection caused mainly by Aspergillus fumigatus. On the other hand, Enfumafungin is a natural triterpene glycoside also with a ß-(1,3)-glucan synthase inhibitory activity and reported to have antifungal potential. In the present study, we compared the growth as well as modifications in the A. fumigatus cell wall upon treatment with Caspofungin or Enfumafungin, consequentially their immunomodulatory capacity on human dendritic cells. Caspofungin initially inhibited the growth of A. fumigatus, but the effect was lost over time. By contrast, Enfumafungin inhibited this fungal growth for the duration investigated. Both Caspofungin and Enfumafungin caused a decrease in the cell wall ß-(1,3)-glucan content with a compensatory increase in the chitin, and to a minor extent they also affected cell wall galactose content. Treatment with these two antifungals did not result in the exposure of ß-(1,3)-glucan on A. fumigatus mycelial surface. Enzymatic digestion suggested a modification of ß-(1,3)-glucan structure, specifically its branching, upon Enfumafungin treatment. While there was no difference in the immunostimulatory capacity of antifungal treated A. fumigatus conidia, alkali soluble-fractions from Caspofungin treated mycelia weakly stimulated the dendritic cells, possibly due to an increased content of immunosuppressive polysaccharide galactosaminogalactan. Overall, we demonstrate a novel mechanism that Enfumafungin not only inhibits ß-(1,3)-glucan synthase activity, but also causes modifications in the structure of ß-(1,3)-glucan in the A. fumigatus cell wall.


Asunto(s)
Antifúngicos , Aspergillus fumigatus , Caspofungina , Pared Celular , Células Dendríticas , Equinocandinas , Glucosiltransferasas , Aspergillus fumigatus/efectos de los fármacos , Aspergillus fumigatus/enzimología , Humanos , Pared Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Antifúngicos/farmacología , Equinocandinas/farmacología , Caspofungina/farmacología , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/metabolismo , beta-Glucanos/farmacología , Lipopéptidos/farmacología , Células Cultivadas , Quitina/farmacología , Glicósidos , Triterpenos
5.
BMC Genomics ; 24(1): 684, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37964194

RESUMEN

BACKGROUND: Aspergillus fumigatus is a major fungal pathogen that causes severe problems due to its increasing resistance to many therapeutic agents. Fludioxonil is a compound that triggers a lethal activation of the fungal-specific High Osmolarity Glycerol pathway. Its pronounced antifungal activity against A. fumigatus and other pathogenic molds renders this agent an attractive lead substance for the development of new therapeutics. The group III hydride histidine kinase TcsC and its downstream target Skn7 are key elements of the multistep phosphorelay that represents the initial section of the High Osmolarity Glycerol pathway. Loss of tcsC results in resistance to fludioxonil, whereas a Δskn7 mutant is partially, but not completely resistant. RESULTS: In this study, we compared the fludioxonil-induced transcriptional responses in the ΔtcsC and Δskn7 mutant and their parental A. fumigatus strain. The number of differentially expressed genes correlates well with the susceptibility level of the individual strains. The wild type and, to a lesser extend also the Δskn7 mutant, showed a multi-faceted stress response involving genes linked to ribosomal and peroxisomal function, iron homeostasis and oxidative stress. A marked difference between the sensitive wild type and the largely resistant Δskn7 mutant was evident for many cell wall-related genes and in particular those involved in the biosynthesis of chitin. Biochemical data corroborate this differential gene expression that does not occur in response to hyperosmotic stress. CONCLUSIONS: Our data reveal that fludioxonil induces a strong and TcsC-dependent stress that affects many aspects of the cellular machinery. The data also demonstrate a link between Skn7 and the cell wall reorganizations that foster the characteristic ballooning and the subsequent lysis of fludioxonil-treated cells.


Asunto(s)
Antifúngicos , Aspergillus fumigatus , Dioxoles , Pirroles , Aspergillus fumigatus/genética , Aspergillus fumigatus/metabolismo , Antifúngicos/farmacología , Antifúngicos/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Glicerol/metabolismo , Pared Celular/metabolismo
6.
Infect Immun ; 88(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32571987

RESUMEN

Even though both cellular and humoral immunities contribute to host defense, the role played by humoral immunity against the airborne opportunistic fungal pathogen Aspergillus fumigatus has been underexplored. In this study, we aimed at deciphering the role of the complement system, the major humoral immune component, against A. fumigatus Mass spectrometry analysis of the proteins extracted from A. fumigatus conidial (asexual spores and infective propagules) surfaces opsonized with human serum indicated that C3 is the major complement protein involved. Flow cytometry and immunolabeling assays further confirmed C3b (activated C3) deposition on the conidial surfaces. Assays using cell wall components of conidia indicated that the hydrophobin RodAp, ß-(1,3)-glucan (BG) and galactomannan (GM) could efficiently activate C3. Using complement component-depleted sera, we showed that while RodAp activates C3 by the alternative pathway, BG and GM partially follow the classical and lectin pathways, respectively. Opsonization facilitated conidial aggregation and phagocytosis, and complement receptor (CR3 and CR4) blockage on phagocytes significantly inhibited phagocytosis, indicating that the complement system exerts a protective role against conidia by opsonizing them and facilitating their phagocytosis mainly through complement receptors. Conidial opsonization with human bronchoalveolar lavage fluid (BALF) confirmed C3 to be the major complement protein interacting with conidia. Nevertheless, complement C2 and mannose-binding lectin (MBL), the classical and lectin pathway components, respectively, were not identified, indicating that BALF activates the alternative pathway on the conidial surface. Moreover, the cytokine profiles were different upon stimulation of phagocytes with serum- and BALF-opsonized conidia, highlighting the importance of studying interaction of conidia with complement proteins in their biological niche.


Asunto(s)
Aspergillus fumigatus/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Complemento C3/inmunología , Polisacáridos Fúngicos/farmacología , Macrófagos/efectos de los fármacos , Suero/inmunología , Esporas Fúngicas/inmunología , Aspergilosis/genética , Aspergilosis/inmunología , Aspergilosis/microbiología , Aspergillus fumigatus/química , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/microbiología , Pared Celular/química , Pared Celular/inmunología , Activación de Complemento/efectos de los fármacos , Complemento C3/genética , Citocinas/biosíntesis , Citocinas/inmunología , Polisacáridos Fúngicos/inmunología , Polisacáridos Fúngicos/aislamiento & purificación , Galactosa/análogos & derivados , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunidad Celular , Inmunidad Humoral , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/inmunología , Antígeno de Macrófago-1/genética , Antígeno de Macrófago-1/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Mananos/inmunología , Mananos/aislamiento & purificación , Mananos/farmacología , Proteínas Opsoninas/farmacología , Fagocitosis/efectos de los fármacos , Cultivo Primario de Células , Unión Proteica , Especies Reactivas de Oxígeno , Suero/química , Suero/microbiología , Esporas Fúngicas/química , beta-Glucanos/inmunología , beta-Glucanos/aislamiento & purificación , beta-Glucanos/farmacología
7.
J Biol Chem ; 293(40): 15538-15555, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30139746

RESUMEN

Innate immunity in animals including humans encompasses the complement system, which is considered an important host defense mechanism against Aspergillus fumigatus, one of the most ubiquitous opportunistic human fungal pathogens. Previously, it has been shown that the alkaline protease Alp1p secreted from A. fumigatus mycelia degrades the complement components C3, C4, and C5. However, it remains unclear how the fungal spores (i.e. conidia) defend themselves against the activities of the complement system immediately after inhalation into the lung. Here, we show that A. fumigatus conidia contain a metalloprotease Mep1p, which is released upon conidial contact with collagen and inactivates all three complement pathways. In particular, Mep1p efficiently inactivated the major complement components C3, C4, and C5 and their activation products (C3a, C4a, and C5a) as well as the pattern-recognition molecules MBL and ficolin-1, either by directly cleaving them or by cleaving them to a form that is further broken down by other proteases of the complement system. Moreover, incubation of Mep1p with human serum significantly inhibited the complement hemolytic activity and conidial opsonization by C3b and their subsequent phagocytosis by macrophages. Together, these results indicate that Mep1p associated with and released from A. fumigatus conidia likely facilitates early immune evasion by disarming the complement defense in the human host.


Asunto(s)
Aspergillus fumigatus/inmunología , Complemento C3/genética , Complemento C4/genética , Complemento C5/genética , Aspergilosis Pulmonar Invasiva/inmunología , Metaloendopeptidasas/inmunología , Animales , Aspergillus fumigatus/crecimiento & desarrollo , Aspergillus fumigatus/patogenicidad , Colágeno/genética , Colágeno/inmunología , Complemento C3/metabolismo , Complemento C4/metabolismo , Complemento C5/metabolismo , Modelos Animales de Enfermedad , Proteínas Fúngicas/genética , Proteínas Fúngicas/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune , Inmunidad Innata , Aspergilosis Pulmonar Invasiva/genética , Aspergilosis Pulmonar Invasiva/microbiología , Aspergilosis Pulmonar Invasiva/patología , Lectinas/genética , Lectinas/inmunología , Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/inmunología , Metaloendopeptidasas/deficiencia , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fagocitosis , Esporas Fúngicas/crecimiento & desarrollo , Esporas Fúngicas/inmunología , Esporas Fúngicas/patogenicidad , Ficolinas
8.
J Biol Chem ; 293(13): 4901-4912, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29414772

RESUMEN

Surfactant protein D (SP-D), a C-type lectin and pattern-recognition soluble factor, plays an important role in immune surveillance to detect and eliminate human pulmonary pathogens. SP-D has been shown to protect against infections with the most ubiquitous airborne fungal pathogen, Aspergillus fumigatus, but the fungal surface component(s) interacting with SP-D is unknown. Here, we show that SP-D binds to melanin pigment on the surface of A. fumigatus dormant spores (conidia). SP-D also exhibited an affinity to two cell-wall polysaccharides of A. fumigatus, galactomannan (GM) and galactosaminogalactan (GAG). The immunolabeling pattern of SP-D was punctate on the conidial surface and was uniform on germinating conidia, in accordance with the localization of melanin, GM, and GAG. We also found that the collagen-like domain of SP-D is involved in its interaction with melanin, whereas its carbohydrate-recognition domain recognized GM and GAG. Unlike un-opsonized conidia, SP-D-opsonized conidia were phagocytosed more efficiently and stimulated the secretion of proinflammatory cytokines by human monocyte-derived macrophages. Furthermore, SP-D-/- mice challenged intranasally with wildtype conidia or melanin ghosts (i.e. hollow melanin spheres) displayed significantly reduced proinflammatory cytokines in the lung compared with wildtype mice. In summary, SP-D binds to melanin present on the dormant A. fumigatus conidial surface, facilitates conidial phagocytosis, and stimulates the host immune response.


Asunto(s)
Aspergillus fumigatus/inmunología , Polisacáridos Fúngicos/inmunología , Melaninas/inmunología , Fagocitosis , Aspergilosis Pulmonar/inmunología , Proteína D Asociada a Surfactante Pulmonar/inmunología , Esporas Fúngicas/inmunología , Animales , Aspergillus fumigatus/genética , Polisacáridos Fúngicos/genética , Melaninas/genética , Ratones , Ratones Noqueados , Aspergilosis Pulmonar/genética , Aspergilosis Pulmonar/patología , Proteína D Asociada a Surfactante Pulmonar/genética , Esporas Fúngicas/genética
9.
Bioconjug Chem ; 30(6): 1788-1797, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-31125199

RESUMEN

ß-(1,3)-Glucan is one of the antigenic components of the bacterial as well as fungal cell wall. We designed microcapsules (MCs) ligated with ß-(1,3)-glucan, to study its immunomodulatory effect. The MCs were obtained by interfacial polycondensation between diacyl chloride (sebacoyl chloride and terephtaloyl chloride) and diethylenetriamine in organic and aqueous phases, respectively. Planar films were first designed to optimize monomer compositions and to examine the kinetics of film formation. MCs with aqueous fluorescent core were then obtained upon controlled emulsification-polycondensation reactions using optimized monomer compositions and adding fluorescein into the aqueous phase. The selected MC-formulation was grafted with Curdlan, a linear ß-(1,3)-glucan from  Agrobacterium species or branched ß-(1,3)-glucan isolated from the cell wall of Aspergillus fumigatus. These ß-(1,3)-glucan grafted MCs were phagocytosed by human monocyte-derived macrophages, and stimulated cytokine secretion. Moreover, the blocking of dectin-1, a ß-(1,3)-glucan recognizing receptor, did not completely inhibit the phagocytosis of these ß-(1,3)-glucan grafted MCs, suggesting the involvement of other receptors in the recognition and uptake of ß-(1,3)-glucan. Overall, grafted MCs are a useful tool for the study of the mechanism of phagocytosis and immunomodulatory effect of the microbial polysaccharides.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Agrobacterium/química , Aspergillus fumigatus/química , Cápsulas , Pared Celular/química , Polisacáridos/farmacología , beta-Glucanos/química , Microscopía Electrónica de Rastreo , Reología
10.
Nat Commun ; 15(1): 6966, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39138196

RESUMEN

Pentraxin 3 (PTX3), a long pentraxin and a humoral pattern recognition molecule (PRM), has been demonstrated to be protective against Aspergillus fumigatus, an airborne human fungal pathogen. We explored its mode of interaction with A. fumigatus, and the resulting implications in the host immune response. Here, we demonstrate that PTX3 interacts with A. fumigatus in a morphotype-dependent manner: (a) it recognizes germinating conidia through galactosaminogalactan, a surface exposed cell wall polysaccharide of A. fumigatus, (b) in dormant conidia, surface proteins serve as weak PTX3 ligands, and (c) surfactant protein D (SP-D) and the complement proteins C1q and C3b, the other humoral PRMs, enhance the interaction of PTX3 with dormant conidia. SP-D, C3b or C1q opsonized conidia stimulated human primary immune cells to release pro-inflammatory cytokines and chemokines. However, subsequent binding of PTX3 to SP-D, C1q or C3b opsonized conidia significantly decreased the production of pro-inflammatory cytokines/chemokines. PTX3 opsonized germinating conidia also significantly lowered the production of pro-inflammatory cytokines/chemokines while increasing IL-10 (an anti-inflammatory cytokine) released by immune cells when compared to the unopsonized counterpart. Overall, our study demonstrates that PTX3 recognizes A. fumigatus either directly or by interplaying with other humoral PRMs, thereby restraining detrimental inflammation. Moreover, PTX3 levels were significantly higher in the serum of patients with invasive pulmonary aspergillosis (IPA) and COVID-19-associated pulmonary aspergillosis (CAPA), supporting previous observations in IPA patients, and suggesting that it could be a potential panel-biomarker for these pathological conditions caused by A. fumigatus.


Asunto(s)
Aspergillus fumigatus , Proteína C-Reactiva , Complemento C1q , Componente Amiloide P Sérico , Esporas Fúngicas , Aspergillus fumigatus/inmunología , Componente Amiloide P Sérico/metabolismo , Componente Amiloide P Sérico/inmunología , Humanos , Esporas Fúngicas/inmunología , Proteína C-Reactiva/metabolismo , Proteína C-Reactiva/inmunología , Complemento C1q/metabolismo , Complemento C1q/inmunología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/inmunología , Complemento C3b/inmunología , Complemento C3b/metabolismo , Citocinas/metabolismo , Citocinas/inmunología , Interleucina-10/metabolismo , Interleucina-10/inmunología , Aspergilosis/inmunología , Aspergilosis/microbiología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Humoral , Femenino , Polisacáridos
11.
Front Immunol ; 13: 978152, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36211424

RESUMEN

Invasive aspergillosis (IA) is a life-threatening fungal infection for immunocompromised hosts. It is, therefore, necessary to understand the immune pathways that control this infection. Although the primary infection site is the lungs, aspergillosis can disseminate to other organs through unknown mechanisms. Herein we have examined the in vivo role of various complement pathways as well as the complement receptors C3aR and C5aR1 during experimental systemic infection by Aspergillus fumigatus, the main species responsible for IA. We show that C3 knockout (C3-/-) mice are highly susceptible to systemic infection of A. fumigatus. Intriguingly, C4-/- and factor B (FB)-/- mice showed susceptibility similar to the wild-type mice, suggesting that either the complement pathways display functional redundancy during infection (i.e., one pathway compensates for the loss of the other), or complement is activated non-canonically by A. fumigatus protease. Our in vitro study substantiates the presence of C3 and C5 cleaving proteases in A. fumigatus. Examination of the importance of the terminal complement pathway employing C5-/- and C5aR1-/- mice reveals that it plays a vital role in the conidial clearance. This, in part, is due to the increased conidial uptake by phagocytes. Together, our data suggest that the complement deficiency enhances the susceptibility to systemic infection by A. fumigatus.


Asunto(s)
Aspergilosis , Aspergillus fumigatus , Animales , Complemento C5/genética , Complemento C5/metabolismo , Factor B del Complemento/genética , Pulmón , Ratones , Esporas Fúngicas
12.
J Colloid Interface Sci ; 614: 322-336, 2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35104706

RESUMEN

HYPOTHESIS: Precise modulation of immuno-inflammatory response is crucial to control periodontal diseases and related systemic comorbidities. The present nanosystem with the controlled-release and cell-penetrating manner enhances the inflammation modulation effects of baicalein in human gingival epithelial cells (hGECs) for better oral healthcare. EXPERIMENTS: We constructed a red-emissive mesoporous silica nanoparticle-based nanosystem with cell-penetrating poly(disulfide) (CPD) capping, through a facile in-situ polymerization approach. It was featured with a glutathione-responsive manner and instant cellular internalization capacity for precisely delivering baicalein intracellularly. Laboratory experiments assessed whether and how the nanosystem per se with the delivered baicalein could modulate immuno-inflammatory responses in hGECs. FINDINGS: The in-situ polymerized CPD layer capped the nanoparticles and yet controlled the release of baicalein in a glutathione-responsive manner. The CPD coating could facilitate cellular internalization of the nanosystem via endocytosis and thiol-mediated approaches. Notably, the intracellularly released baicalein effectively downregulated the expression of pro-inflammatory cytokines through inhibiting the NF-κB signaling pathway. The nanosystem per se could modulate immuno-inflammatory responses by passivating the cellular response to interlukin-1ß. This study highlights that the as-synthesized nanosystem may serve as a novel multi-functional vehicle to modulate innate host response via targeting the NF-κB pathway for precision healthcare.


Asunto(s)
Disulfuros , Glutatión , Inmunomodulación , Nanopartículas , Dióxido de Silicio , Disulfuros/química , Sistemas de Liberación de Medicamentos , Flavanonas/administración & dosificación , Glutatión/química , Humanos , Nanopartículas/química , Nanopartículas/uso terapéutico , Enfermedades Periodontales/tratamiento farmacológico , Polimerizacion , Porosidad , Dióxido de Silicio/química
13.
Cell Surf ; 8: 100072, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35118215

RESUMEN

Humoral immunity plays a defensive role against invading microbes. However, it has been largely overlooked with respect to Aspergillus fumigatus, an airborne fungal pathogen. Previously, we have demonstrated that surfactant protein D (SP-D), a major humoral component in human lung-alveoli, recognizes A. fumigatus conidial surface exposed melanin pigment. Through binding to melanin, SP-D opsonizes conidia, facilitates conidial phagocytosis, and induces the expression of protective pro-inflammatory cytokines in the phagocytic cells. In addition to melanin, SP-D also interacts with galactomannan (GM) and galactosaminogalactan (GAG), the cell wall polysaccharides exposed on germinating conidial surfaces. Therefore, we aimed at unravelling the biological significance of SP-D during the germination process. Here, we demonstrate that SP-D exerts direct fungistatic activity by restricting A. fumigatus hyphal growth. Conidial germination in the presence of SP-D significantly increased the exposure of cell wall polysaccharides chitin, α-1,3-glucan and GAG, and decreased ß-1,3-glucan exposure on hyphae, but that of GM was unaltered. Hyphae grown in presence of SP-D showed positive immunolabelling for SP-D. Additionally, SP-D treated hyphae induced lower levels of pro-inflammatory cytokine, but increased IL-10 (anti-inflammatory cytokine) and IL-8 (a chemokine) secretion by human peripheral blood mononuclear cells (PBMCs), compared to control hyphae. Moreover, germ tube surface modifications due to SP-D treatment resulted in an increased hyphal susceptibility to voriconazole, an antifungal drug. It appears that SP-D exerts its anti-A. fumigatus functions via a range of mechanisms including hyphal growth-restriction, hyphal surface modification, masking of hyphal surface polysaccharides and thus altering hyphal immunostimulatory properties.

14.
mBio ; 12(6): e0282421, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34781737

RESUMEN

The signaling pathways activated following interaction between dendritic cells (DCs) and a pathogen determine the polarization of effector T-cell and regulatory T-cell (Treg) responses to the infection. Several recent studies, mostly in the context of bacterial infections, have shown that the Wnt/ß-catenin pathway plays a major role in imparting tolerogenic features in DCs and in promotion of Treg responses. However, the significance of the Wnt/ß-catenin pathway's involvement in regulating the immune response to the fungal species is not known. Using Aspergillus fumigatus, a ubiquitous airborne opportunistic fungal species, we show here that fungi activate the Wnt/ß-catenin pathway in human DCs and are critical for mediating the immunosuppressive Treg responses. Pharmacological inhibition of this pathway in DCs led to inhibition of maturation-associated molecules and interleukin 10 (IL-10) secretion without affecting the majority of the inflammatory cytokines. Furthermore, blockade of Wnt signaling in DCs suppressed DC-mediated Treg responses in CD4+ T cells and downregulated both tumor necrosis factor alpha (TNF-α) and IL-10 responses in CD8+ T cells. Mechanistically, induction of ß-catenin pathway by A. fumigatus required C-type lectin receptors and promoted Treg polarization via the induction of programmed death-ligand 1 on DCs. Further investigation on the identity of fungal molecular patterns has revealed that the cell wall polysaccharides ß-(1, 3)-glucan and α-(1, 3)-glucan, but not chitin, possess the capacity to activate the ß-catenin pathway. Our data suggest that the Wnt/ß-catenin pathway is a potential therapeutic target to selectively suppress the Treg response and to sustain the protective Th1 response in the context of invasive aspergillosis caused by A. fumigatus. IMPORTANCE The balance between effector CD4+ T-cell and immunosuppressive regulatory T-cell (Treg) responses determines the outcome of an infectious disease. The signaling pathways that regulate human CD4+ T-effector versus Treg responses to the fungi are not completely understood. By using Aspergillus fumigatus, a ubiquitous opportunistic fungal species, we show that fungi activate the Wnt/ß-catenin pathway in human dendritic cells (DCs) that promotes Treg responses via induction of immune checkpoint molecule programmed death ligand 1 on DCs. Blockade of the Wnt/ß-catenin pathway in DCs led to the selective inhibition of Treg without affecting the Th1 response. Dissection of the identity of A. fumigatus pathogen-associated molecular patterns (PAMPs) revealed that cell wall polysaccharides exhibit selectivity in their capacity to activate the ß-catenin pathway in DCs. Our data thus provide a pointer that Wnt/ß-catenin pathway represents potential therapeutic target to selectively suppress Treg responses and to sustain protective a Th1 response against invasive fungal diseases.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/fisiología , Antígeno B7-H1/inmunología , Células Dendríticas/inmunología , Linfocitos T Reguladores/inmunología , beta Catenina/inmunología , Aspergilosis/genética , Aspergilosis/microbiología , Antígeno B7-H1/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Vía de Señalización Wnt , beta Catenina/genética
15.
Front Immunol ; 12: 677798, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34122441

RESUMEN

Humoral immune components have been individually studied in the context of interaction of host with Aspergillus fumigatus, a major airborne fungal pathogen. However, a global view of the multitude and complex nature of humoral immune components is needed to bring new insight into host-Aspergillus interaction. Therefore, we undertook comparative proteomic analysis of the bronchoalveolar lavage fluid collected from individuals infected or colonized with A. fumigatus versus controls, to identify those alveolar humoral components affected upon A. fumigatus infection. Complement proteins C1q, C8 beta-chain, factor-H, ficolin-1, ficolin-2, mannan binding lectin serine peptidase 2, pentraxin-3 and the surfactant protein-D were identified as the major humoral immune components affected by A. fumigatus infection and colonization. Based on this observation, we hypothesize that crosstalk between these humoral components is essential during host-Aspergillus interaction giving new specific leads to study for better understanding the pathogenesis. Furthermore, the affected humoral components could be potential diagnostic markers of A. fumigatus infection or colonization.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/genética , Líquido del Lavado Bronquioalveolar/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Humoral , Proteoma/inmunología , Proteómica/métodos , Anciano , Aspergilosis/microbiología , Líquido del Lavado Bronquioalveolar/microbiología , Estudios de Casos y Controles , Proteínas del Sistema Complemento/inmunología , Citocinas/inmunología , Femenino , Humanos , Inmunidad Innata , Masculino , Persona de Mediana Edad , Mapas de Interacción de Proteínas , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/microbiología , ARN de Hongos/genética , ARN Ribosómico 28S/genética
16.
Front Immunol ; 12: 749074, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867977

RESUMEN

In this study, the human immune response mechanisms against Sporothrix brasiliensis and Sporothrix schenckii, two causative agents of human and animal sporotrichosis, were investigated. The interaction of S. brasiliensis and S. schenckii with human monocyte-derived macrophages (hMDMs) was shown to be dependent on the thermolabile serum complement protein C3, which facilitated the phagocytosis of Sporothrix yeast cells through opsonization. The peptidorhamnomannan (PRM) component of the cell walls of these two Sporothrix yeasts was found to be one of their surfaces exposed pathogen-associated molecular pattern (PAMP), leading to activation of the complement system and deposition of C3b on the Sporothrix yeast surfaces. PRM also showed direct interaction with CD11b, the specific component of the complement receptor-3 (CR3). Furthermore, the blockade of CR3 specifically impacted the interleukin (IL)-1ß secretion by hMDM in response to both S. brasiliensis and S. schenckii, suggesting that the host complement system plays an essential role in the inflammatory immune response against these Sporothrix species. Nevertheless, the structural differences in the PRMs of the two Sporothrix species, as revealed by NMR, were related to the differences observed in the host complement activation pathways. Together, this work reports a new PAMP of the cell surface of pathogenic fungi playing a role through the activation of complement system and via CR3 receptor mediating an inflammatory response to Sporothrix species.


Asunto(s)
Antígenos Fúngicos/inmunología , Proteínas del Sistema Complemento/inmunología , Glicoproteínas/inmunología , Macrófagos/inmunología , Sporothrix , Pared Celular/inmunología , Activación de Complemento , Citocinas/inmunología , Humanos , L-Lactato Deshidrogenasa/inmunología , Antígeno de Macrófago-1/inmunología , Macrófagos/microbiología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Fagocitosis
17.
Front Cell Infect Microbiol ; 11: 643312, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33718288

RESUMEN

Although belong to the same genus, Aspergillus fumigatus is primarily involved in invasive pulmonary infection, whereas Aspergillus flavus is a common cause of superficial infection. In this study, we compared conidia (the infective propagules) of these two Aspergillus species. In immunocompetent mice, intranasal inoculation with conidia of A. flavus resulted in significantly higher inflammatory responses in the lungs compared to mice inoculated with A. fumigatus conidia. In vitro assays revealed that the dormant conidia of A. flavus, unlike A. fumigatus dormant conidia, are immunostimulatory. The conidial surface of A. fumigatus was covered by a rodlet-layer, while that of A. flavus were presented with exposed polysaccharides. A. flavus harbored significantly higher number of proteins in its conidial cell wall compared to A. fumigatus conidia. Notably, ß-1,3-glucan in the A. flavus conidial cell-wall showed significantly higher percentage of branching compared to that of A. fumigatus. The polysaccharides ensemble of A. flavus conidial cell wall stimulated the secretion of proinflammatory cytokines, and conidial cell wall associated proteins specifically stimulated IL-8 secretion from the host immune cells. Furthermore, the two species exhibited different sensitivities to antifungal drugs targeting cell wall polysaccharides, proposing the efficacy of species-specific treatment strategies. Overall, the species-specific organization of the conidial cell wall could be important in establishing infection by the two Aspergillus species.


Asunto(s)
Aspergillus fumigatus , Aspergillus , Animales , Aspergillus flavus , Pared Celular , Ratones , Esporas Fúngicas
18.
J Extracell Vesicles ; 10(10): e12129, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34377375

RESUMEN

Whereas extracellular vesicle (EV) research has become commonplace in different biomedical fields, this field of research is still in its infancy in mycology. Here we provide a robust set of data regarding the structural and compositional aspects of EVs isolated from the fungal pathogenic species Cryptococcus neoformans, C. deneoformans and C. deuterogattii. Using cutting-edge methodological approaches including cryogenic electron microscopy and cryogenic electron tomography, proteomics, and flow cytometry, we revisited cryptococcal EV features and suggest a new EV structural model, in which the vesicular lipid bilayer is covered by mannoprotein-based fibrillar decoration, bearing the capsule polysaccharide as its outer layer. About 10% of the EV population is devoid of fibrillar decoration, adding another aspect to EV diversity. By analysing EV protein cargo from the three species, we characterized the typical Cryptococcus EV proteome. It contains several membrane-bound protein families, including some Tsh proteins bearing a SUR7/PalI motif. The presence of known protective antigens on the surface of Cryptococcus EVs, resembling the morphology of encapsulated virus structures, suggested their potential as a vaccine. Indeed, mice immunized with EVs obtained from an acapsular C. neoformans mutant strain rendered a strong antibody response in mice and significantly prolonged their survival upon C. neoformans infection.


Asunto(s)
Cryptococcus neoformans/inmunología , Cryptococcus neoformans/metabolismo , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Vacunas/inmunología , Secuencias de Aminoácidos , Animales , Antígenos Fúngicos/inmunología , Antígenos Fúngicos/metabolismo , Microscopía por Crioelectrón , Criptococosis/inmunología , Vesículas Extracelulares/microbiología , Femenino , Proteínas Fúngicas/inmunología , Proteínas Fúngicas/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteoma , Proteómica/métodos
19.
Microorganisms ; 8(10)2020 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-33036329

RESUMEN

Candida albicans is a commensal polymorphic and opportunistic fungus, which usually resides as a small community in the oral cavities of a majority of humans. The latter eco-system presents this yeast varied opportunities for mutualistic interactions with other cohabitant oral bacteria, that synergizes its persistence and pathogenicity. Collectively, these communities live within complex plaque biofilms which may adversely affect the oral health and increase the proclivity for oral candidiasis. The proteome of such oral biofilms with myriad interkingdom interactions are largely underexplored. Herein, we employed limma differential expression analysis, and cluster analysis to explore the proteomic interactions of C. albicans biofilms with nine different common oral bacterial species, Aggregatibacter actinomycetemcomitans, Actinomyces naeslundii, Fusobacterium nucleatum, Enterococcus faecalis, Porphyromonas gingivalis, Streptococcus mutants, Streptococcus sanguinis, Streptococcus mitis, and Streptococcus sobrinus. Interestingly, upon exposure of C. albicans biofilms to the foregoing heat-killed bacteria, the proteomes of the fungus associated with cellular respiration, translation, oxidoreductase activity, and ligase activity were significantly altered. Subsequent differential expression and cluster analysis revealed the subtle, yet significant alterations in the C. albicans proteome, particularly on exposure to bacteria with dissimilar cell morphologies, and Gram staining characteristics.

20.
mSphere ; 5(1)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31915215

RESUMEN

Methodologies to identify epitopes or ligands of the fungal cell wall polysaccharides influencing the immune response of human pathogens have to date been imperfect. Using the galactomannan (GM) of Aspergillus fumigatus as a model, we have shown that synthetic oligosaccharides of distinct structures representing key fragments of cell wall polysaccharides are the most precise tools to study the serological and immunomodulatory properties of a fungal polysaccharide.


Asunto(s)
Antígenos Fúngicos/inmunología , Aspergillus fumigatus/química , Pared Celular/inmunología , Mananos/inmunología , Oligosacáridos/síntesis química , Oligosacáridos/inmunología , Antígenos Fúngicos/química , Aspergilosis/microbiología , Epítopos/química , Epítopos/inmunología , Galactosa/análogos & derivados , Humanos , Inmunomodulación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA