Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 517
Filtrar
1.
Exp Eye Res ; 240: 109830, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38364932

RESUMO

Fungal keratitis (FK) is a refractory keratitis caused by excessive inflammation and fungal damage. Excessive inflammation can lead to tissue damage and corneal opacity, resulting in a poor prognosis for FK. Oxymatrine (OMT) is a natural alkaloid, which has rich pharmacological effects, such as antioxidant and anti-inflammation. However, its antifungal activity and the mechanism of action in FK have not been elucidated. This study confirmed that OMT suppressed Aspergillus fumigatus growth, biofilm formation, the integrity of fungal cell and conidial adherence. OMT not only effectively reduced corneal fungal load but also inflammation responses. OMT lessened the recruitment of neutrophils and macrophages in FK. In addition, OMT up-regulated the expression of Nrf2 and down-regulated the expression of IL-18, IL-1ß, caspase-1, NLRP3 and GSDMD. Pre-treatment with Nrf2 inhibitor up-regulated the expression of IL-1ß, IL-18, caspase-1, NLRP3 and GSDMD supressed by OMT. In conclusion, OMT has efficient anti-inflammatory and antifungal effects by suppressing fungal activity and restricting pyroptosis via Nrf2 pathway. OMT is considered as a potential option for the treatment of FK.


Assuntos
Aspergilose , Úlcera da Córnea , Infecções Oculares Fúngicas , Ceratite , Matrinas , Animais , Camundongos , Aspergillus fumigatus/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Interleucina-18 , Aspergilose/tratamento farmacológico , Aspergilose/metabolismo , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Piroptose , Fator 2 Relacionado a NF-E2 , Ceratite/microbiologia , Inflamação , Infecções Oculares Fúngicas/tratamento farmacológico , Infecções Oculares Fúngicas/metabolismo , Caspase 1/metabolismo , Camundongos Endogâmicos C57BL
2.
J Ocul Pharmacol Ther ; 40(1): 89-99, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38346287

RESUMO

Purpose: To characterize the efficiency of glabridin alone and in combination with clinical antifungals in Aspergillus fumigatus keratitis. Methods: The broth microdilution method was performed to investigate whether glabridin exerted an antifungal role on planktonic cells and immature and mature biofilm. Antifungal mechanism was evaluated by Sorbitol and Ergosterol Assays. The synergistic effect of glabridin and antifungals was assessed through the checkerboard microdilution method and time-killing test. Regarding anti-inflammatory role, inflammatory substances induced by A. fumigatus were assessed by real-time quantitative polymerase chain reaction, western blot, and enzyme-linked immunosorbent assay. Drug toxicity was assessed by Draize test in vivo. Macrophage phenotypes were examined by flow cytometry. Results: Regarding antifungal activity, glabridin destroyed fungal cell wall and membrane on planktonic cells and suppressed immature and mature biofilm formation. After combining with natamycin or amphotericin B, glabridin possessed a potent synergistic effect against A. fumigatus. Regarding anti-inflammatory aspects, Dectin-1, toll­like receptor (TLR)-2 and TLR-4 expression of human corneal epithelial cells were significantly elevated after A. fumigatus challenge and reduced by glabridin. The elevated expression of interleukin-1ß and tumor necrosis factor-alpha induced by A. fumigatus or corresponding agonists were reversed by glabridin, equivalent to the effect of corresponding inhibitors. Glabridin could also contribute to anti-inflammation by downregulating inflammatory mediator expression to suppress macrophage infiltration. Conclusions: Glabridin contributed to fungal clearance by destroying fungal cell wall and membrane, and disrupting biofilm. Combining glabridin with clinical antifungals was superior in reducing A. fumigatus growth. Glabridin exerted an anti-inflammatory effect by downregulating proinflammatory substance expression and inhibiting macrophage infiltration, which provide a potential agent and treatment strategies for fungal keratitis.


Assuntos
Aspergilose , Infecções Oculares Fúngicas , Isoflavonas , Ceratite , Fenóis , Humanos , Animais , Camundongos , Aspergillus fumigatus/fisiologia , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Aspergilose/tratamento farmacológico , Ceratite/tratamento farmacológico , Ceratite/microbiologia , Infecções Oculares Fúngicas/tratamento farmacológico , Infecções Oculares Fúngicas/microbiologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Camundongos Endogâmicos C57BL
3.
Infect Immun ; 92(2): e0038023, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38168666

RESUMO

Macrophages act as a first line of defense against pathogens. Against Aspergillus fumigatus, a fungus with pathogenic potential in immunocompromised patients, macrophages can phagocytose fungal spores and inhibit spore germination to prevent the development of tissue-invasive hyphae. However, the cellular pathways that macrophages use to accomplish these tasks and any roles macrophages have later in infection against invasive forms of fungi are still not fully known. Rac-family Rho GTPases are signaling hubs for multiple cellular functions in leukocytes, including cell migration, phagocytosis, reactive oxygen species (ROS) generation, and transcriptional activation. We therefore aimed to further characterize the function of macrophages against A. fumigatus in an in vivo vertebrate infection model by live imaging of the macrophage behavior in A. fumigatus-infected rac2 mutant zebrafish larvae. While Rac2-deficient zebrafish larvae are susceptible to A. fumigatus infection, Rac2 deficiency does not impair macrophage migration to the infection site, interaction with and phagocytosis of spores, spore trafficking to acidified compartments, or spore killing. However, we reveal a role for Rac2 in macrophage-mediated inhibition of spore germination and control of invasive hyphae. Re-expression of Rac2 under a macrophage-specific promoter rescues the survival of A. fumigatus-infected rac2 mutant larvae through increased control of germination and hyphal growth. Altogether, we describe a new role for macrophages against extracellular hyphal growth of A. fumigatus and report that the function of the Rac2 Rho GTPase in macrophages is required for this function.


Assuntos
Aspergilose , Peixe-Zebra , Animais , Humanos , Peixe-Zebra/microbiologia , GTP Fosfo-Hidrolases , Macrófagos/microbiologia , Fagocitose , Aspergilose/microbiologia , Aspergillus fumigatus/fisiologia , Esporos Fúngicos , Proteínas rac de Ligação ao GTP/genética , Proteínas de Peixe-Zebra/genética
4.
ACS Infect Dis ; 10(2): 500-512, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38175918

RESUMO

Fungal keratitis (FK) is a refractory global disease characterized by a high incidence of blindness and a lack of effective therapeutic options, and Aspergillus fumigatus (A. fumigatus, AF) is one of the most common causative fungi. This study aimed to investigate the role of extracellular vesicles (EVs) from A. fumigatus in the immune cell function and their protective role in A. fumigatus keratitis in order to explore their therapeutic potential. First, we isolated and characterized the EVs (AF-derived EVs). In vitro, we stimulated RAW264.7 cells and polymorphonuclear cells with AF-derived EVs. The expression levels of inflammatory factors increased in both immune cells along with an M1 polarization variation of RAW264.7 cells. After being incubated with AF-derived EVs, both immune cells exhibited an increased conidia-phagocytic index and a decreased conidia survival rate. In vivo, we injected EVs subconjunctivally on mice resulting in a heightened production of secretory immunoglobulin A (sIgA) in tear fluid. By the injection of EVs on mice in advance, a significant reduction in severity of A. fumigatus FK was witnessed by lower clinical scores, inflammatory appearances, and mitigated fungal load. Collectively, these results positioned AF-derived EVs as a promising and innovative immune therapy for combating FK, while also providing a platform for further investigation into developing an optimal formulation for modulating inflammation in the context of FK.


Assuntos
Aspergilose , Vesículas Extracelulares , Infecções Oculares Fúngicas , Ceratite , Animais , Camundongos , Aspergillus fumigatus/fisiologia , Aspergilose/tratamento farmacológico , Aspergilose/metabolismo , Ceratite/microbiologia , Inflamação , Infecções Oculares Fúngicas/tratamento farmacológico
5.
Transl Vis Sci Technol ; 12(8): 3, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37531113

RESUMO

Purpose: To investigate the antifungal and anti-inflammatory effects of 0.01% hypochlorous acid (HCLO) on rats with Aspergillus fumigatus keratitis. Methods: The time-kill assay and broth microdilution procedures were used in vitro to demonstrate that 0.01% HCLO was fungicidal and fungistatic. The severity of the disease was evaluated in vivo using a clinical score and slit-lamp photographs. Fungal load, polymorphonuclear neutrophil infiltration, and the production of related proteins were determined using colony plate counting, in vivo confocal microscopy, periodic acid-Schiff staining, fungal fluorescence staining, immunofluorescence staining, myeloperoxidase assay, and Western blotting. Result: In vitro, 0.01% HCLO can destroy A. fumigatus spores in 1 minute. The optical density of the 0.01% HCLO group was significantly lower than that of the phosphate-buffered saline control group (P < 0.01), and no visible mycelium was observed using a fluorescence microscope. 0.01% HCLO reduced the severity of A. fumigatus keratitis in rats by decreasing the clinical score, fungal loading (periodic acid-Schiff, plate count, and fungal fluorescence staining), and inhibiting neutrophil infiltration and activity (immunofluorescence staining and myeloperoxidase). Furthermore, the Western blot analysis revealed that 0.01% HCO decreased protein expression levels of tumor necrosis factor-α and IL-1ß. Conclusions: According to our findings, 0.01% HCLO can kill A. fumigatus spores in vitro. It has antifungal and anti-inflammatory effects on A. fumigatus keratitis in rats. It also inhibited A. fumigatus growth; decreased neutrophil infiltration, tumor necrosis factor-α, and IL-1ß expression; and provided a potential treatment for fungal keratitis. Translational Relevance: This study provides a potential treatment for fungal keratitis in the clinic.


Assuntos
Aspergilose , Infecções Oculares Fúngicas , Ceratite , Ratos , Animais , Aspergillus fumigatus/fisiologia , Peroxidase/uso terapêutico , Ácido Hipocloroso/uso terapêutico , Aspergilose/tratamento farmacológico , Aspergilose/metabolismo , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Fator de Necrose Tumoral alfa , Ácido Periódico/uso terapêutico , Ceratite/tratamento farmacológico , Ceratite/microbiologia , Infecções Oculares Fúngicas/tratamento farmacológico , Infecções Oculares Fúngicas/metabolismo , Infecções Oculares Fúngicas/microbiologia , Anti-Inflamatórios/uso terapêutico
6.
Viruses ; 15(3)2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36992427

RESUMO

Infection with Aspergillus fumigatus polymycovirus 1 (AfuPmV-1) weakens the resistance of biofilms of common A. fumigatus reference strain Af293 in intermicrobial competition with Pseudomonas aeruginosa, and sensitizes A. fumigatus for antifungal effects of nikkomycin Z. We compared the sensitivity of two virus-infected (VI) and one virus-free (VF) Af293 strains to hypertonic salt. Salt stress impairs the growth of VI and VF at all times; VF control growth always exceeds VI, and VF growth in salt always exceeds VI. Since VF growth exceeds VI in the presence and absence of salt, we also examined growth in salt as a percentage of control growth. Initially, as a percentage of control, VI exceeded VF, but at 120 h VF began to exceed VI consistently even by this measure; thus, at that time the growth of VF in salt surges in relation to control growth, or, alternatively, its growth in salt persists compared to the relative inhibition of VI. In summary, virus infection impairs the response of A. fumigatus to several different stresses, including hypertonic salt.


Assuntos
Aspergillus fumigatus , Vírus de RNA , Cloreto de Sódio , Aspergillus fumigatus/efeitos dos fármacos , Aspergillus fumigatus/fisiologia , Aspergillus fumigatus/virologia , Biofilmes , Vírus de RNA/fisiologia , Estresse Salino , Cloreto de Sódio/farmacologia
7.
Curr Eye Res ; 48(4): 348-356, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36785524

RESUMO

PURPOSE: The research was used to uncover the mechanism of glabridin in Aspergillus fumigatus keratitis in anti-fungus and anti-inflammation. METHODS: In vitro, RAW 264.7 cells were infected with A. fumigatus with incubation of glabridin in different concentrations. Real-time quantitative polymerase chain reaction (RT­qPCR), Western blot, and enzyme-linked immunosorbent assay (ELISA) were used to assess the inflammatory severe and alternation with the intervention of Dectin-2 siRNA and glabridin. In vivo, A. fumigatus keratitis mouse models were established by spore intra-stromal injection and treated with glabridin or PBS. And disease scores, inflammatory mediators, and periodic acid-schiff (PAS) staining were exhibited to demonstrate the therapeutic efficiency of glabridin in vivo. Morphological interference assay monitored fungal germination. Scanning and transmission electron microscopy were used to observe the growth of fungi. RESULTS: In RAW 264.7 cells and mouse keratitis models, noncytotoxic 16 µg/mL glabridin showed significant inhibition in the expression of Dectin-2, NLRP3, Caspase-1, IL-1ß, and TNF-α after A. fumigatus infection, almost similar to the intervention of Dectin-2 siRNA. PAS staining illustrated the reduced hyphal distribution in cornea stroma with glabridin treatment. Glabridin remarkably inhibited A. fumigatus growth through delaying germination and disrupting the integrity of the hyphae membrane. CONCLUSION: Glabridin plays an anti-inflammatory role in A. fumigatus challenge via suppression of the Dectin-2 and NLRP3 inflammasome, and plays an anti-fungal role through delaying germination and changing the hyphal integrity.KEY MESSAGESGlabridin plays an anti-inflammatory role in A. fumigatus infection of RAW264.7 cells in a concentration-dependent manner and through Dectin-2 mediation.Glabridin decreases fungal distribution and inflammation in mouse A. fumigatus keratitis.Glabridin inhibits A. fumigatus growth by delaying germination and disrupting cellular structure in vitro.


Assuntos
Aspergilose , Ceratite , Animais , Camundongos , Aspergillus fumigatus/fisiologia , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Ceratite/tratamento farmacológico , Ceratite/microbiologia , Aspergilose/tratamento farmacológico , Aspergilose/metabolismo , Inflamação/tratamento farmacológico , RNA Interferente Pequeno , Anti-Inflamatórios/uso terapêutico , Camundongos Endogâmicos C57BL
8.
Viruses ; 15(1)2023 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-36680240

RESUMO

Infection with Aspergillus fumigatus polymycovirus 1 (AfuPmV-1) weakens resistance of Aspergillus fumigatus common reference strain Af293 biofilms in intermicrobial competition with Pseudomonas aeruginosa. We compared the sensitivity of two infected and one virus-free Af293 strains to antifungal drugs. All three were comparably sensitive to drugs affecting fungal membranes (voriconazole, amphotericin) or cell wall glucan synthesis (micafungin, caspofungin). In contrast, forming biofilms of virus-free Af293 were much more resistant than AfuPmV-1-infected Af293 to nikkomycin Z (NikZ), a drug inhibiting chitin synthase. The IC50 for NikZ on biofilms was between 3.8 and 7.5 µg/mL for virus-free Af293 and 0.94-1.88 µg/mL for infected strains. The IC50 for the virus-free A. fumigatus strain 10AF was ~2 µg/mL in most experiments. NikZ also modestly affected the planktonic growth of infected Af293 more than the virus-free strain (MIC 50%, 2 and 4 µg/mL, respectively). Virus-free Af293 biofilm showed increased metabolism, and fungus growing as biofilm or planktonically showed increased growth compared to infected; these differences do not explain the resistance of the virus-free fungus to NikZ. In summary, AfuPmV-1 infection sensitized A. fumigatus to NikZ, but did not affect response to drugs commonly used against A. fumigatus infection. Virus infection had a greater effect on NikZ inhibition of biofilm than planktonic growth.


Assuntos
Antifúngicos , Vírus de RNA , Antifúngicos/farmacologia , Antifúngicos/metabolismo , Aspergillus fumigatus/fisiologia , Aminoglicosídeos/farmacologia , Aminoglicosídeos/metabolismo , Anfotericina B/metabolismo , Anfotericina B/farmacologia
9.
Int Immunopharmacol ; 115: 109721, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36641891

RESUMO

PURPOSE: We aimed to investigate the therapeutic role of dimethyl fumarate (DMF) in fungal keratitis. METHODS: Human corneal epithelial cells (HCECs) and mouse models of fungal keratitis were used in this study. The antifungal effect of DMF on Aspergillus fumigatus (A. fumigatus) was confirmed by examining the minimum inhibitory concentration (MIC), biofilm formation, conidial adherence and corneal fungal loads. Slit-lamp photography, haematoxylin and eosin staining and immunostaining were used to assess the severity of corneal impairment. RT-PCR, western blot, ELISA, immunohistochemistry and immunostaining were performed to examine the effects of DMF on the expression of the inflammatory mediators during fungal infection. RESULTS: In vitro, DMF limited A. fumigatus growth, biofilm formation, and conidial adherence and reduced the mRNA levels of AldA, GlkA, GAPDH, HxkA, PgkA, Sdh2, GelA and ChsF in A. fumigatus. In vivo, DMF effectively decreased corneal fungal loads. DMF attenuated corneal inflammatory impairment by suppressing inflammatory cell accumulation and downregulating cytokine expression. DMF notably downregulated the high expression of NLRP3, cleaved GSDMD, cleaved caspase-1, mature IL-1ß and mature IL-18 induced by fungi. The production of Nrf2 and HO-1 could be further increased by DMF in infected HCECs. Nrf2 siRNA pretreatment counteracted DMF-mediated downregulation of the expression of the active forms of IL-18, IL-1ß, caspase-1 and GSDMD. CONCLUSION: DMF limits fungal growth by suppressing biofilm formation, conidial adherence and respiratory metabolism. It also exerts an anti-inflammatory effect on fungal keratitis by inhibiting pyroptosis, which could be regulated by Nrf2. Our results suggest that DMF plays a therapeutic role in fungal keratitis.


Assuntos
Aspergilose , Infecções Oculares Fúngicas , Ceratite , Camundongos , Animais , Humanos , Fumarato de Dimetilo/farmacologia , Fumarato de Dimetilo/uso terapêutico , Interleucina-18 , Aspergilose/tratamento farmacológico , Aspergilose/metabolismo , Piroptose , Fator 2 Relacionado a NF-E2 , Ceratite/microbiologia , Aspergillus fumigatus/fisiologia , Infecções Oculares Fúngicas/tratamento farmacológico , Caspase 1 , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
10.
Int Immunopharmacol ; 110: 108992, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35810488

RESUMO

PURPOSE: To investigate the anti-inflammatory and antifungal role of ß-ionone (BI) in fungal keratitis (FK). METHODS: In vitro antifungal activity of BI against Aspergillus fumigatus (A. fumigatus) was evaluated by using minimum inhibitory concentration (MIC), crystal violet staining, biofilm biomass measurement, propidium iodide uptake test, and adherence assay. And RT-PCR was carried out to measure the levels of RodA, RodB, Rho, FKs, CshA-D, RlmA, Cyp51A-B and Cdr1B. Network pharmacology analysis was applied to predict the relationship between BI and FK. Cell Count Kit-8 (CCK8) assay was utilized to detect the cytotoxicity of BI to RAW264.7 and immortalized human corneal epithelial cells (HCECs). The underlying mechanism of BI at regulating the level of inflammatory factors in FK was assessed by RT-PCR, ELISA and Western blot in vitro and in vivo. The therapeutic effect of BI has investigated in A. fumigatus keratitis by employing the clinical score, pathological examination, plate count, immunofluorescence and myeloperoxidase (MPO) assay. We also used the slit-lamp microscopy, clinical scores, and HE staining to assess the effect of natamycin compared with BI treatment in vivo. RESULTS: BI suppressed the growth of A. fumigatus and had a significant effect on A. fumigatus biofilms and membrane permeability. RT-PCR demonstrated that exposure of A. fumigatus to BI inhibited the expression of genes that function in hydrophobin (RodA, RodB), cell wall integrity (Rho, FKs, CshA-D, RlmA), azole susceptibility (Cyp51A-B, Cdr1B). Network pharmacology showed that the effects of BI in FK implicate with C-type lectin receptor signaling pathway. In vivo, after A. fumigatus infection, BI treatment markedly reduced the severity of FK by decreasing clinical score, neutrophil recruitment, and fungal load. And BI treatment also obviously reduced the expression of inflammatory cytokines, Lectin-like oxidized LDL receptor (LOX-1), phosphorylation of p38MAPK and p-JNK versus the DMSO-treated group. BI and natamycin both significantly increased corneal transparency and decreased inflammatory cell recruitment in the FK in the mice model. CONCLUSION: These results indicated that BI had fungicidal activities against A. fumigatus. It also ameliorated FK in mice by reducing inflammation, which was regulated by LOX-1, p-p38MAPK and p-JNK.


Assuntos
Infecções Oculares Fúngicas , Ceratite , Animais , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Aspergillus fumigatus/fisiologia , Infecções Oculares Fúngicas/tratamento farmacológico , Humanos , Ceratite/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Natamicina/uso terapêutico , Norisoprenoides , Receptores Depuradores Classe E/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Eur J Pharmacol ; 924: 174955, 2022 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-35436473

RESUMO

Fungal keratitis is a corneal infection, which severely impairs vision. The fungal pathogen provokes host immune response, but the excessive inflammatory response causes significant collateral damage to the cornea. Eugenol, the main component of clove oil, has been found to have a broad range of pharmacological activities including anti-microbial, antioxidation and anti-inflammation. However, the role of eugenol in Aspergillus fumigatus (A. fumigatus) keratitis is unknown. In this study, we demonstrated that eugenol reduced mice keratitis severity, inflammatory cells infiltration, pro-inflammatory cytokine expression, and the fungal load. Eugenol also decreased the expressions of pro-inflammatory cytokines in human corneal epithelial cells (HCECs). We confirmed that the anti-inflammatory effects of eugenol were related to activating nuclear factor erythroid 2-related factor 2/Heme Oxygenase-1 (Nrf2/HO-1) signaling pathway. Moreover, we demonstrated that eugenol could inhibit the A. fumigatus growth and adhesion to host cells, as well as damage the fungal biofilm. The antifungal mechanism seemed to be disrupting the integrity of the fungal membrane and reducing the biosynthesis of ergosterol. Taken together, our research suggested that eugenol exerted protective effects on mouse A. fumigatus keratitis, due to its anti-inflammatory and antifungal activity.


Assuntos
Aspergilose , Ceratite , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Aspergilose/tratamento farmacológico , Aspergilose/metabolismo , Aspergilose/microbiologia , Aspergillus fumigatus/fisiologia , Citocinas/metabolismo , Eugenol/farmacologia , Eugenol/uso terapêutico , Ceratite/tratamento farmacológico , Ceratite/metabolismo , Ceratite/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
12.
mSphere ; 7(1): e0094021, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-34986319

RESUMO

Fungal infections remain a major global concern. Emerging fungal pathogens and increasing rates of resistance mean that additional research efforts and resources must be allocated to advancing our understanding of fungal pathogenesis and developing new therapeutic interventions. Neutrophilic granulocytes are a major cell type involved in protection against the important fungal pathogen Aspergillus fumigatus, where they employ numerous defense mechanisms, including production of antimicrobial extracellular vesicles. A major drawback to work with neutrophils is the lack of a suitable cell line system for the study of fungal pathogenesis. To address this problem, we assessed the feasibility of using differentiated PLB-985 neutrophil-like cells as an in vitro model to study A. fumigatus infection. We find that dimethylformamide-differentiated PLB-985 cells provide a useful recapitulation of many aspects of A. fumigatus interactions with primary human polymorphonuclear leukocytes. We show that differentiated PLB-985 cells phagocytose fungal conidia and acidify conidia-containing phagolysosomes similar to primary neutrophils, release neutrophil extracellular traps, and also produce antifungal extracellular vesicles in response to infection. In addition, we provide an improved method for the isolation of extracellular vesicles produced during infection by employing a size exclusion chromatography-based approach. Advanced liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomics revealed an enrichment of extracellular vesicle marker proteins and a decrease of cytoplasmic proteins in extracellular vesicles isolated using this improved method. Ultimately, we find that differentiated PLB-985 cells can serve as a genetically tractable model to study many aspects of A. fumigatus pathogenesis. IMPORTANCE Polymorphonuclear leukocytes are an important defense against human fungal pathogens, yet our model systems to study this group of cells remain very limited in scope. In this study, we established that differentiated PLB-985 cells can serve as a model to recapitulate several important aspects of human polymorphonuclear leukocyte interactions with the important human fungal pathogen Aspergillus fumigatus. The proposed addition of a cultured neutrophil-like cell line to the experimental toolbox to study fungal pathogenesis will allow for a more mechanistic description of neutrophil antifungal biology. In addition, the easier handling of the cell line compared to primary human neutrophils allowed us to use PLB-985 cells to provide an improved method for isolation of neutrophil-derived extracellular vesicles using size exclusion chromatography. Together, these results provide significant tools and a baseline knowledge for the future study of neutrophil-derived extracellular vesicles in the laboratory.


Assuntos
Aspergillus fumigatus , Neutrófilos , Antifúngicos , Aspergillus fumigatus/fisiologia , Cromatografia Líquida , Humanos , Neutrófilos/microbiologia , Espectrometria de Massas em Tandem
13.
Exp Eye Res ; 216: 108960, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35085580

RESUMO

Fungal keratitis is one of leading reasons for blindness in the world, which causes corneal blindness mainly due to excessive inflammatory responses. Kaempferol (KAE) is a natural flavonoid which has potent anti-inflammatory effects. However, whether KAE plays protective roles in fungal keratitis and the potentially protective mechanisms are unrevealed. Here we first investigated the anti-inflammatory and antifungal effects of KAE on Aspergillus fumigatus (A. fumigatus) keratitis in C57BL/6 mice. We found that treatment of KAE ameliorated the severity of keratitis, inhibited macrophages and neutrophils recruitment, depressed corneal fungal load, and declined the expression of TLR4 and Dectin-1 in A. fumigatus infected mice corneas. And in activated hyphae or Curdlan stimulated macrophages, pretreatment of KAE also significantly decreased the mRNA and protein expression of IL-1ß, TNF-α, MIP-2 and the phosphorylated-p38 (p-p38)/p38 MAPK ratio. In summary, KAE ameliorated the prognosis of fungal keratitis in C57BL/6 mice by reducing corneal fungal load, depressing the inflammatory cells recruitment, and downregulating the expression of inflammatory factors, and those effects depended on the inhibition of Dectin-1 and p38 MAPK pathway.


Assuntos
Aspergilose/tratamento farmacológico , Aspergillus fumigatus/efeitos dos fármacos , Úlcera da Córnea/tratamento farmacológico , Infecções Oculares Fúngicas/tratamento farmacológico , Quempferóis/uso terapêutico , Lectinas Tipo C/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Aspergilose/metabolismo , Aspergilose/microbiologia , Aspergillus fumigatus/fisiologia , Contagem de Colônia Microbiana , Úlcera da Córnea/metabolismo , Úlcera da Córnea/microbiologia , Modelos Animais de Doenças , Infecções Oculares Fúngicas/metabolismo , Infecções Oculares Fúngicas/microbiologia , Feminino , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Prognóstico
14.
Exp Eye Res ; 216: 108941, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35077754

RESUMO

Fungal keratitis (FK) is one of the main causes of blindness in China. People with diabetes are susceptible to corneal epithelial disease, even fungal keratitis. At present, there are few studies on this disease. Resolvins (Rv) has been reported as a mediators that exert crucial anti-inflammatory and immune regulation roles in serval diseases. In order to investigate the roles and underlying mechanism of Resolvins D1 (RvD1) on the Aspergillus fumigatus (A. fumigatus) keratitis in diabetes, we established in vivo and in vitro models of A. fumigatus keratitis, which were then exposed to high glucose. The expression levels of RvD1, 5-lipoxygenase (5-LOX), and 15-lipoxygenase (15-LOX) in A. fumigatus keratitis patients with diabetes were determined through Enzyme Linked Immunosorbent Assay (ELISA), Western blot and immunohistochemistry. Reactive Oxygen Species (ROS) production, ELISA, flow cytometry, Hematoxylin-Eosin (HE) staining and fungal loading determination were conducted to evaluate the severity of A. fumigatus infection. Lymphangiogenesis and angiogenesis were examined by immunofluorescence assay. Western blot was applied to detect the proteins of the MAPK-NF-κB pathway. The results showed that RvD1 diminished the high glucose-induced oxidative stress and inflammatory response, as evidenced by the reduction of ROS production, Interleukin-6 (IL-6), Interleukin-8 (IL-8), Heme Oxygenase-1 (HMOX-1), and the elevation of Cyclooxygenase-2 (COX2), Superoxide Dismutase (SOD-1), and Glutathione Peroxidase-2 (GPX2) levels in A. fumigatus-infected Human Corneal Endothelial Cells (HCECs). Additionally, lymphangiogenesis and angiogenesis prominently decreased after intervention with RvD1. Furthermore, RvD1 significantly reduced the levels of p-MEK1/2 and p-ERK1/2, and restrained the NF-κB and GPR32 activation. The above results showed that RvD1 protects against A. fumigatus keratitis in diabetes by suppressing oxidative stress, inflammatory response, fungal growth, and immunoreaction via modulating MAPK-NF-κB pathway. RvD1 provides clues for the therapeutic targets of Fungal keratitis complicated with diabetes.


Assuntos
Aspergilose/prevenção & controle , Úlcera da Córnea/prevenção & controle , Complicações do Diabetes/microbiologia , Ácidos Docosa-Hexaenoicos/fisiologia , Infecções Oculares Fúngicas/prevenção & controle , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Animais , Araquidonato 15-Lipoxigenase/metabolismo , Araquidonato 5-Lipoxigenase/metabolismo , Aspergilose/metabolismo , Aspergilose/microbiologia , Aspergillus fumigatus/fisiologia , Western Blotting , Células Cultivadas , Úlcera da Córnea/metabolismo , Úlcera da Córnea/microbiologia , Complicações do Diabetes/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Epitélio Corneano/efeitos dos fármacos , Epitélio Corneano/microbiologia , Infecções Oculares Fúngicas/metabolismo , Infecções Oculares Fúngicas/microbiologia , Citometria de Fluxo , Glucose/farmacologia , Humanos , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Oxirredutases/metabolismo , Espécies Reativas de Oxigênio/metabolismo
15.
Exp Eye Res ; 214: 108883, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34896107

RESUMO

PURPOSE: To investigate the effect of Glabridin (GLD) in Aspergillus fumigatus keratitis and its associated mechanisms. METHODS: Aspergillus fumigatus (A. fumigatus) conidia was inoculated in 96-well plate, and minimal inhibitory concentration (MIC) and biofilm formation ability were evaluated after GLD treatment. Spore adhesion ability was evaluated in conidia infected human corneal epithelial cells (HCECs). Keratitis mouse model was created by corneal intrastromal injection with A. fumigatus conidia, and GLD treatment started at the day after infection. The number of fungal colonies was calculated by plate count, and degree of corneal inflammation was assessed by clinical score. Flow cytometry, myeloperoxidase (MPO), and immunofluorescence staining (IFS) experiments were used to assess neutrophil infiltrations. PCR, ELISA and Western blot were conducted to determine levels of TLR4, Dectin-1 as well as downstream inflammatory factors. RESULTS: GLD treatment suppressed the proliferation, biofilm formation abilities and adhesive capability of A. fumigatus. In mice upon A. fumigatus infection, treatment of GLD showed significantly decreased severity of corneal inflammation, reduced number of A. fumigatus in cornea, and suppressed neutrophil infiltration in cornea. GLD treatment obviously inhibited mRNA and protein levels of Dectin-1, TLR4 and proinflammatory mediators such as IL-1ß, HMGB1, and TNF-α in mice corneas compared to the control group. CONCLUSION: GLD has antifungal and anti-inflammatory effects in fungal keratitis through suppressing A. fumigatus proliferation and alleviating neutrophil infiltration, and repressing the expression of TLR4, Dectin-1 and proinflammatory mediators.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antifúngicos/uso terapêutico , Aspergilose/tratamento farmacológico , Aspergillus fumigatus/fisiologia , Úlcera da Córnea/tratamento farmacológico , Infecções Oculares Fúngicas/tratamento farmacológico , Isoflavonas/uso terapêutico , Fenóis/uso terapêutico , Animais , Aspergilose/microbiologia , Aspergillus fumigatus/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Western Blotting , Úlcera da Córnea/microbiologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Infecções Oculares Fúngicas/microbiologia , Feminino , Citometria de Fluxo , Lectinas Tipo C/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Infiltração de Neutrófilos , Reação em Cadeia da Polimerase , Receptor 4 Toll-Like/metabolismo
16.
Curr Eye Res ; 47(3): 343-351, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34766863

RESUMO

PURPOSE: To investigate the anti-inflammatory and antifungal role of α-melanocyte stimulating hormone (α-MSH) in Aspergillus Fumigatus (A. fumigatus) keratitis. METHOD: Corneas of C57BL/6 mice were infected with A. Fumigatus. α-MSH (5 ul, 1×10-4 mmol/ml) was given by subconjunctival injection from day 1 to day 3 post infection (p.i.). After 3 days p.i., clinical score was recorded and HE staining was tested. Fungal load in mice corneas was observed by plate counting. Proinflammatory mediators and pattern recognition receptors (PRRs) were detected. The number of neutrophils and macrophages was tested by immunofluorescence staining. The role of α-MSH in RAW264.7 cells after A. fumigatus stimulation were evaluated by PCR and Western blot, and MPKA protein levels including total-JNK (T-JNK), phosphorylated-JNK (P-JNK), total-ERK (T-ERK), and phosphorylated-ERK (P-ERK) were tested via Western blot with or without α-MSH treatment. RESULTS: Compared with PBS control group, α-MSH treatment alleviated disease response and decreased clinical score at 3 days p.i. HE staining showed less infiltration in corneal tissue after α-MSH treatment. Plate counting experiment showed that number of viable fungus in corneas of α-MSH treated group was less than control group. mRNA levels of IL-1ß, TNF-α, IL-6, MIP-2, LOX-1, Dectin-1, and iNOS were decreased. Protein levels of IL-1ß, TNF-α, IL-6, and Dectin-1 were decreased. α-MSH treatment also decreased the infiltrating neutrophils and macrophages. The levels of proinflammatory cytokines, Dectin-1 and LOX-1 stimulated by A. fumigatus, were also suppressed by pretreatment of α-MSH in RAW264.7 cells. The ratio of P-JNK/T-JNK and P-ERK/T-ERK was downregulated in α-MSH group compared with PBS control group. CONCLUSION: α-MSH alleviates the severity and decreases fungal load of A. fumigatus keratitis in mice. Migration of neutrophils and macrophages are restrained. α-MSH downregulates the expression of dectin-1 and the ratio of P-JNK/T-JNK and P-ERK/T-ERK in A. fumigatus infection.


Assuntos
Infecções Oculares Fúngicas , Ceratite , Animais , Anti-Inflamatórios/farmacologia , Aspergillus fumigatus/fisiologia , Modelos Animais de Doenças , Infecções Oculares Fúngicas/microbiologia , Interleucina-6 , Ceratite/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Depuradores Classe E/uso terapêutico , Fator de Necrose Tumoral alfa , alfa-MSH/uso terapêutico
17.
Curr Eye Res ; 47(4): 517-524, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34797193

RESUMO

PURPOSE: This study aimed to investigate the anti-inflammatory effect and antifungal effect of punicalagin in murine fungal keratitis. METHODS: We used in vitro and in vivo protocols to assess the anti-inflammatory effect and antifungal effect of punicalagin. In vitro, time kill and mycelial stain were done. In vivo, murine fungal keratitis was established and treated with PBS or PUN. Clinical scores were taken on days 1, 3, and 5 post infection. The mRNA and protein levels of inflammatory factors were detected by RT-PCR and Western blot, and the number and location of macrophages were analyzed by flow cytometry and immunofluorescence. Also, fungal plate counting was used to assess the antifungal effect. The DCFH-DA fluorescence probe detected the ROS level. RESULTS: In vitro, PUN showed activity against A.fumigatus. (A.F.), with MIC90 values of 250 µg/ml, and significantly reduced A.F. biofilm formation (p < .001). In vivo, the mouse fungal keratitis model after punicalagin treatment exhibited less disease, lower clinical scores (p < .05), lower reduced macrophage infiltrate (p < .001), and fungal load (p < .001) than those treated with PBS. Treatment with punicalagin also reduced the mRNA expression and protein level of pro-inflammatory factors. At the cellular level, PUN significantly reduced the mRNA expression of inflammatory factors and ROS production caused by the stimulation of mycelia in RAW264.7 (p < .001). CONCLUSIONS: The results show that punicalagin is beneficial in the treatment of murine fungal keratitis. The mechanism of its anti-inflammatory effect was synthetical, including antifungal activity, an inhibitory effect of proinflammatory factor and macrophages, and anti-oxidation.


Assuntos
Aspergilose , Úlcera da Córnea , Infecções Oculares Fúngicas , Ceratite , Animais , Anti-Inflamatórios/farmacologia , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Aspergilose/metabolismo , Aspergillus fumigatus/fisiologia , Úlcera da Córnea/tratamento farmacológico , Modelos Animais de Doenças , Infecções Oculares Fúngicas/microbiologia , Taninos Hidrolisáveis , Ceratite/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Espécies Reativas de Oxigênio
18.
Immunohorizons ; 5(12): 983-993, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34937773

RESUMO

Aspergillus fumigatus is a ubiquitous mold associated with the development of pulmonary diseases that include invasive pulmonary aspergillosis (IPA), an often fatal opportunistic infection. FIBCD1 is a transmembrane endocytic membrane receptor widely expressed on human epithelium. Although FIBCD1 was previously shown to bind chitin, modulate fungal colonization of the gut, and inhibit intestinal inflammation, the role of FIBCD1 in the context of lung fungal infection remains unknown. In this study, we observed that mortality, fungal burden, and tissue histopathology were decreased in the absence of FIBCD1 in murine IPA. Quantitative RT-PCR analyses demonstrated decreased inflammatory cytokines in the lungs of neutrophil-depleted FIBCD1-/- mice with IPA, when compared with wild-type controls. In contrast, inflammatory cytokines were increased in immune-competent FIBCD1-/- mice after fungal aspiration, suggesting that the presence of neutrophils is associated with cytokine modulation. In contrast to the clear IPA phenotype, FIBCD1-/- mice with systemic infection or bleomycin-induced lung injury exhibited similar morbidity and mortality when compared with their wild-type counterparts. Thus, our study identifies a detrimental role of FIBCD1 in IPA.


Assuntos
Aspergillus fumigatus/fisiologia , Aspergilose Pulmonar Invasiva/metabolismo , Pulmão/patologia , Receptores de Superfície Celular/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Aspergilose Pulmonar Invasiva/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Receptores de Superfície Celular/genética , Índice de Gravidade de Doença
19.
Nat Microbiol ; 6(12): 1526-1536, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34819642

RESUMO

Aspergillus fumigatus is an environmental saprobe and opportunistic human fungal pathogen. Despite an estimated annual occurrence of more than 300,000 cases of invasive disease worldwide, a comprehensive survey of the genomic diversity present in A. fumigatus-including the relationship between clinical and environmental isolates and how this genetic diversity contributes to virulence and antifungal drug resistance-has been lacking. In this study we define the pan-genome of A. fumigatus using a collection of 300 globally sampled genomes (83 clinical and 217 environmental isolates). We found that 7,563 of the 10,907 unique orthogroups (69%) are core and present in all isolates and the remaining 3,344 show presence/absence of variation, representing 16-22% of the genome of each isolate. Using this large genomic dataset of environmental and clinical samples, we found an enrichment for clinical isolates in a genetic cluster whose genomes also contain more accessory genes, including genes coding for transmembrane transporters and proteins with iron-binding activity, and genes involved in both carbohydrate and amino-acid metabolism. Finally, we leverage the power of genome-wide association studies to identify genomic variation associated with clinical isolates and triazole resistance as well as characterize genetic variation in known virulence factors. This characterization of the genomic diversity of A. fumigatus allows us to move away from a single reference genome that does not necessarily represent the species as a whole and better understand its pathogenic versatility, ultimately leading to better management of these infections.


Assuntos
Aspergilose/microbiologia , Aspergillus fumigatus/genética , Genoma Fúngico , Aspergillus fumigatus/isolamento & purificação , Aspergillus fumigatus/fisiologia , Proteínas Fúngicas/genética , Variação Genética , Estudo de Associação Genômica Ampla , Humanos , Fatores de Virulência/genética
20.
mBio ; 12(6): e0282421, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34781737

RESUMO

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.


Assuntos
Aspergilose/imunologia , Aspergillus fumigatus/fisiologia , Antígeno B7-H1/imunologia , Células Dendríticas/imunologia , Linfócitos T Reguladores/imunologia , beta Catenina/imunologia , Aspergilose/genética , Aspergilose/microbiologia , Antígeno B7-H1/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Humanos , Interleucina-10/genética , Interleucina-10/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Via de Sinalização Wnt , beta Catenina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...