Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Immunol ; 193(7): 3332-43, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25172488

RESUMEN

CD4(+) T cells orchestrate immune responses against fungi, such as Aspergillus fumigatus, a major fungal pathogen in humans. The complexity of the fungal genome and lifestyle questions the existence of one or a few immune-dominant Ags and complicates systematic screening for immunogenic Ags useful for immunotherapy or diagnostics. In this study, we used a recently developed flow cytometric assay for the direct ex vivo characterization of A. fumigatus-specific CD4(+) T cells for rapid identification of physiological T cell targets in healthy donors. We show that the T cell response is primarily directed against metabolically active A. fumigatus morphotypes and is stronger against membrane protein fractions compared with cell wall or cytosolic proteins. Further analysis of 15 selected single A. fumigatus proteins revealed a highly diverse reactivity pattern that was donor and protein dependent. Importantly, the parallel assessment of T cell frequency, phenotype, and function allowed us to differentiate between proteins that elicit strong memory T cell responses in vivo versus Ags that induce T cell exhaustion or no reactivity in vivo. The regulatory T cell (Treg) response mirrors the conventional T cell response in terms of numbers and target specificity. Thus, our data reveal that the fungal T cell immunome is complex, but the ex vivo characterization of reactive T cells allows us to classify Ags and to predict potential immunogenic targets. A. fumigatus-specific conventional T cell responses are counterbalanced by a strong Treg response, suggesting that Treg-depletion strategies may be helpful in improving antifungal immunity.


Asunto(s)
Antígenos Fúngicos/inmunología , Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Memoria Inmunológica , Linfocitos T Reguladores/inmunología , Aspergilosis/patología , Aspergilosis/terapia , Femenino , Humanos , Masculino , Linfocitos T Reguladores/patología
2.
Mol Microbiol ; 76(5): 1205-21, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20398215

RESUMEN

O-mannosylation is an essential protein modification in eukaryotes. It is initiated at the endoplasmic reticulum by O-mannosyltransferases (PMT) that are evolutionary conserved from yeast to humans. The PMT family is phylogenetically classified into PMT1, PMT2 and PMT4 subfamilies, which differ in protein substrate specificity and number of genes per subfamily. In this study, we characterized for the first time the whole PMT family of a pathogenic filamentous fungus, Aspergillus fumigatus. Genome analysis showed that only one member of each subfamily is present in A. fumigatus, PMT1, PMT2 and PMT4. Despite the fact that all PMTs are transmembrane proteins with conserved peptide motifs, the phenotype of each PMT deletion mutant was very different in A. fumigatus. If disruption of PMT1 did not reveal any phenotype, deletion of PMT2 was lethal. Disruption of PMT4 resulted in abnormal mycelial growth and highly reduced conidiation associated to significant proteomic changes. The double pmt1pmt4 mutant was lethal. The single pmt4 mutant exhibited an exquisite sensitivity to echinocandins that is associated to major changes in the expression of signal transduction cascade genes. These results indicate that the PMT family members play a major role in growth, morphogenesis and viability of A. fumigatus.


Asunto(s)
Aspergillus fumigatus/enzimología , Aspergillus fumigatus/fisiología , Supervivencia Celular/fisiología , Proteínas Fúngicas/metabolismo , Isoenzimas/metabolismo , Manosiltransferasas/metabolismo , Morfogénesis/fisiología , Animales , Antifúngicos/farmacología , Aspergilosis/microbiología , Aspergillus fumigatus/citología , Aspergillus fumigatus/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Equinocandinas/farmacología , Proteínas Fúngicas/clasificación , Proteínas Fúngicas/genética , Eliminación de Gen , Genes Fúngicos , Prueba de Complementación Genética , Humanos , Isoenzimas/clasificación , Isoenzimas/genética , Masculino , Manosiltransferasas/clasificación , Manosiltransferasas/genética , Ratones , Mutación , Micelio/metabolismo , Micelio/ultraestructura , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
3.
Int J Med Microbiol ; 301(5): 368-77, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21565549

RESUMEN

Both fungi Candida albicans and Aspergillus fumigatus can cause a number of life-threatening systemic infections in humans. The commensal yeast C. albicans is one of the main causes of nosocomial fungal infectious diseases, whereas the filamentous fungus A. fumigatus has become one of the most prevalent airborne fungal pathogens. Early diagnosis of these fungal infections is challenging, only a limited number of antifungals for treatment are available, and the molecular details of pathogenicity are hardly understood. The completion of both the A. fumigatus and C. albicans genome sequence provides the opportunity to improve diagnosis, to define new drug targets, to understand the functions of many uncharacterised proteins, and to study protein regulation on a global scale. With the application of proteomic tools, particularly two-dimensional gel electrophoresis and LC/MS-based methods, a comprehensive overview about the proteins of A. fumigatus and C. albicans present or induced during environmental changes and stress conditions has been obtained in the past 5 years. However, for the discovery of further putative virulence determinants, more sensitive and targeted proteomic methods have to be applied. Here, we review the recent proteome data generated for A. fumigatus and C. albicans that are related to factors required for pathogenicity.


Asunto(s)
Aspergillus fumigatus/patogenicidad , Candida albicans/patogenicidad , Proteómica/métodos , Factores de Virulencia/análisis , Cromatografía Liquida/métodos , Electroforesis en Gel Bidimensional/métodos , Humanos , Espectrometría de Masas/métodos , Virulencia
4.
Int J Med Microbiol ; 301(7): 602-11, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21658997

RESUMEN

Surface-associated and secreted proteins represent primarily exposed components of Aspergillus fumigatus during host infection. Several secreted proteins are known to be involved in defense mechanisms or immune evasion, thus, probably contributing to pathogenicity. Furthermore, several secreted antigens were identified as possible biomarkers for the verification of diseases caused by Aspergillus species. Nevertheless, there is only limited knowledge about the composition of the secretome and about molecular functions of particular proteins. To identify secreted proteins potentially essential for virulence, the core secretome of A. fumigatus grown in minimal medium was determined. Two-dimensional gel electrophoretic separation and subsequent MALDI-TOF-MS/MS analyses resulted in the identification of 64 different proteins. Additionally, secretome analyses of A. fumigatus utilizing elastin, collagen or keratin as main carbon and nitrogen source were performed. Thereby, the alkaline serine protease Alp1 was identified as the most abundant protein and hence presumably represents an important protease during host infection. Interestingly, the Asp-hemolysin (Asp-HS), which belongs to the protein family of aegerolysins and which was often suggested to be involved in fungal virulence, was present in the secretome under all growth conditions tested. In addition, a second, non-secreted protein with an aegerolysin domain annotated as Asp-hemolysin-like (HS-like) protein can be found to be encoded in the genome of A. fumigatus. Generation and analysis of Asp-HS and HS-like deletion strains revealed no differences in phenotype compared to the corresponding wild-type strain. Furthermore, hemolysis and cytotoxicity was not altered in both single-deletion and double-deletion mutants lacking both aegerolysin genes. All mutant strains showed no attenuation in virulence in a mouse infection model for invasive pulmonary aspergillosis. Overall, this study provides a comprehensive analysis of secreted proteins of A. fumigatus and a detailed characterization of hemolysin mutants.


Asunto(s)
Aspergillus fumigatus/metabolismo , Medios de Cultivo/química , Proteínas Fúngicas/metabolismo , Proteínas Hemolisinas/metabolismo , Proteoma/análisis , Animales , Aspergillus fumigatus/crecimiento & desarrollo , Carbono/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Elastina/metabolismo , Electroforesis en Gel Bidimensional , Femenino , Proteínas Fúngicas/genética , Eliminación de Gen , Proteínas Hemolisinas/genética , Aspergilosis Pulmonar Invasiva/microbiología , Aspergilosis Pulmonar Invasiva/patología , Queratinas/metabolismo , Ratones , Nitrógeno/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
5.
Infect Immun ; 78(8): 3585-94, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20498262

RESUMEN

The opportunistic human pathogenic fungus Aspergillus fumigatus is a major cause of fungal infections in immunocompromised patients. Innate immunity plays an important role in the defense against infections. The complement system represents an essential part of the innate immune system. This cascade system is activated on the surface of A. fumigatus conidia and hyphae and enhances phagocytosis of conidia. A. fumigatus conidia but not hyphae bind to their surface host complement regulators factor H, FHL-1, and CFHR1, which control complement activation. Here, we show that A. fumigatus hyphae possess an additional endogenous activity to control complement activation. A. fumigatus culture supernatant efficiently cleaved complement components C3, C4, C5, and C1q as well as immunoglobulin G. Secretome analysis and protease inhibitor studies identified the secreted alkaline protease Alp1, which is present in large amounts in the culture supernatant, as the central molecule responsible for this cleavage. An alp1 deletion strain was generated, and the culture supernatant possessed minimal complement-degrading activity. Moreover, protein extract derived from an Escherichia coli strain overproducing Alp1 cleaved C3b, C4b, and C5. Thus, the protease Alp1 is responsible for the observed cleavage and degrades a broad range of different substrates. In summary, we identified a novel mechanism in A. fumigatus that contributes to evasion from the host complement attack.


Asunto(s)
Aspergillus fumigatus/enzimología , Aspergillus fumigatus/inmunología , Complemento C3/metabolismo , Complemento C4/metabolismo , Complemento C5/metabolismo , Proteínas Fúngicas/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Aspergillus fumigatus/genética , Complemento C1q/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Eliminación de Gen , Humanos , Hifa/enzimología , Hifa/inmunología , Evasión Inmune , Inmunoglobulina G/metabolismo , Ratones , Serina Endopeptidasas/genética
6.
ACS Synth Biol ; 9(10): 2784-2796, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-32966739

RESUMEN

Human milk oligosaccharides (HMOs) are unique components of human breast milk. Their large-scale production by fermentation allows infant formulas to be fortified with HMOs, but current fermentation processes require lactose as a starting material, increasing the costs, bioburden, and environmental impact of manufacturing. Here we report the development of an Escherichia coli strain that produces 2'-fucosyllactose (2'-FL), the most abundant HMO, de novo using sucrose as the sole carbon source. Strain engineering required the expression of a novel glucose-accepting galactosyltransferase, overexpression of the de novo UDP-d-galactose and GDP-l-fucose pathways, the engineering of an intracellular pool of free glucose, and overexpression of a suitable α(1,2)-fucosyltransferase. The export of 2'-FL was facilitated using a sugar efflux transporter. The final production strain achieved 2'-FL yields exceeding 60 g/L after fermentation for 84 h. This efficient strategy facilitates the lactose-independent production of HMOs by fermentation, which will improve product quality and reduce the costs of manufacturing.


Asunto(s)
Escherichia coli/genética , Escherichia coli/metabolismo , Ingeniería Metabólica/métodos , Leche Humana/química , Sacarosa/metabolismo , Trisacáridos/biosíntesis , Técnicas de Cultivo Celular por Lotes , Carbono/metabolismo , Fermentación , Calidad de los Alimentos , Fucosa/metabolismo , Fucosiltransferasas/metabolismo , Galactosa/metabolismo , Galactosiltransferasas/metabolismo , Humanos , Fórmulas Infantiles/química , Lactosa/metabolismo
7.
Front Immunol ; 10: 2573, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31824478

RESUMEN

The opportunistic fungal pathogen Aspergillus fumigatus can cause severe infections, particularly in immunocompromised individuals. Upon infection, A. fumigatus faces the powerful and directly acting immune defense of the human host. The mechanisms on how A. fumigatus evades innate immune attack and complement are still poorly understood. Here, we identify A. fumigatus enolase, AfEno1, which was also characterized as fungal allergen, as a surface ligand for human plasma complement regulators. AfEno1 binds factor H, factor-H-like protein 1 (FHL-1), C4b binding protein (C4BP), and plasminogen. Factor H attaches to AfEno1 via two regions, via short conserved repeats (SCRs) 6-7 and 19-20, and FHL-1 contacts AfEno1 via SCRs 6-7. Both regulators when bound to AfEno1 retain cofactor activity and assist in C3b inactivation. Similarly, the classical pathway regulator C4BP binds to AfEno1 and bound to AfEno1; C4BP assists in C4b inactivation. Plasminogen which binds to AfEno1 via lysine residues is accessible for the tissue-type plasminogen activator (tPA), and active plasmin cleaves the chromogenic substrate S2251, degrades fibrinogen, and inactivates C3 and C3b. Plasmin attached to swollen A. fumigatus conidia damages human A549 lung epithelial cells, reduces the cellular metabolic activity, and induces cell retraction, which results in exposure of the extracellular matrix. Thus, A. fumigatus AfEno1 is a moonlighting protein and virulence factor which recruits several human regulators. The attached human regulators allow the fungal pathogen to control complement at the level of C3 and to damage endothelial cell layers and tissue components.


Asunto(s)
Aspergillus fumigatus/enzimología , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/metabolismo , Proteínas Fúngicas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Plasminógeno/metabolismo , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/microbiología , Aspergilosis/inmunología , Aspergilosis/metabolismo , Aspergilosis/microbiología , Aspergillus fumigatus/inmunología , Línea Celular , Factor H de Complemento/inmunología , Proteínas Fúngicas/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune , Cinética , Fosfopiruvato Hidratasa/inmunología , Unión Proteica
8.
Front Immunol ; 9: 1635, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30166981

RESUMEN

The opportunistic fungal pathogen Aspergillus fumigatus can cause life-threatening infections, particularly in immunocompromised patients. Most pathogenic microbes control host innate immune responses at the earliest time, already before infiltrating host immune cells arrive at the site of infection. Here, we identify Aspf2 as the first A. fumigatus Factor H-binding protein. Aspf2 recruits several human plasma regulators, Factor H, factor-H-like protein 1 (FHL-1), FHR1, and plasminogen. Factor H contacts Aspf2 via two regions located in SCRs6-7 and SCR20. FHL-1 binds via SCRs6-7, and FHR1 via SCRs3-5. Factor H and FHL-1 attached to Aspf2-maintained cofactor activity and assisted in C3b inactivation. A Δaspf2 knockout strain was generated which bound Factor H with 28% and FHL-1 with 42% lower intensity. In agreement with less immune regulator acquisition, when challenged with complement-active normal human serum, Δaspf2 conidia had substantially more C3b (>57%) deposited on their surface. Consequently, Δaspf2 conidia were more efficiently phagocytosed (>20%) and killed (44%) by human neutrophils as wild-type conidia. Furthermore, Aspf2 recruited human plasminogen and, when activated by tissue-type plasminogen activator, newly generated plasmin cleaved the chromogenic substrate S2251 and degraded fibrinogen. Furthermore, plasmin attached to conidia damaged human lung epithelial cells, induced cell retraction, and caused matrix exposure. Thus, Aspf2 is a central immune evasion protein and plasminogen ligand of A. fumigatus. By blocking host innate immune attack and by disrupting human lung epithelial cell layers, Aspf2 assists in early steps of fungal infection and likely allows tissue penetration.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Proteínas Fúngicas/inmunología , Aspergilosis/microbiología , Proteínas Inactivadoras del Complemento C3b/inmunología , Proteínas Inactivadoras del Complemento C3b/metabolismo , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/aislamiento & purificación , Proteínas Fúngicas/metabolismo , Humanos , Evasión Inmune , Inmunidad Innata , Plasminógeno/inmunología , Plasminógeno/metabolismo , Unión Proteica
9.
J Proteomics ; 75(13): 4038-49, 2012 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-22634043

RESUMEN

The isoprenoid alcohol farnesol represents a quorum-sensing molecule in pathogenic yeasts, but was also shown to inhibit the growth of many filamentous fungi. In order to gain a deeper insight into the antifungal activity of farnesol, we performed 2D-differential gel electrophoretic analysis (2D-DIGE) of Aspergillus nidulans exposed to farnesol. We observed an increased abundance of antioxidative enzymes and proteins involved in protein folding and the ubiquitin-mediated protein degradation. A striking finding was the strong up-regulation of a dehydrin-like protein (DlpA). Expression analyses suggested the involvement of DlpA in the cellular response to oxidative, osmotic and cold stress. In line with these data, we demonstrated that dlpA expression was regulated by the MAP kinase SakA/HogA. The generation of both a dlpA Tet(on) antisense RNA-producing A. nidulans strain (dlpA-inv) and a ΔdlpA deletion mutant indicated a role of DlpA in conidiation and stress resistance of dormant conidia against heat and ROS. Furthermore, the production of the secondary metabolite sterigmatocystin was absent in both strains dlpA-inv and ΔdlpA. Our results demonstrate the complexity of the farnesol-mediated stress response in A. nidulans and describe a farnesol-inducible dehydrin-like protein that contributes to the high tolerance of resting conidia against oxidative and heat stress.


Asunto(s)
Aspergillus nidulans/efectos de los fármacos , Farnesol/farmacología , Estrés Oxidativo/efectos de los fármacos , Aspergillus nidulans/genética , Proteínas Fúngicas/biosíntesis , Proteínas Fúngicas/metabolismo , Proteínas de Choque Térmico/biosíntesis , Proteínas de Choque Térmico/fisiología , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/metabolismo , Proteolisis , Proteoma/metabolismo , Esporas Fúngicas/efectos de los fármacos , Esporas Fúngicas/metabolismo , Esterigmatocistina/biosíntesis , Electroforesis Bidimensional Diferencial en Gel , Respuesta de Proteína Desplegada/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA