Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
J Fungi (Basel) ; 6(3)2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32859091

RESUMEN

Immune inertness of Aspergillus fumigatus conidia is attributed to its surface rodlet-layer made up of RodAp, characterized by eight conserved cysteine residues forming four disulfide bonds. Earlier, we showed that the conserved cysteine residue point (ccrp) mutations result in conidia devoid of the rodlet layer. Here, we extended our study comparing the surface organization and immunoreactivity of conidia carrying ccrp-mutations with the RODA deletion mutant (∆rodA). Western blot analysis using anti-RodAp antibodies indicated the absence of RodAp in the cytoplasm of ccrp-mutant conidia. Immunolabeling revealed differential reactivity to conidial surface glucans, the ccrp-mutant conidia preferentially binding to α-(1,3)-glucan, ∆rodA conidia selectively bound to ß-(1,3)-glucan; the parental strain conidia showed negative labeling. However, permeability of ccrp-mutants and ∆rodA was similar to the parental strain conidia. Proteomic analyses of the conidial surface exposed proteins of the ccrp-mutants showed more similarities with the parental strain, but were significantly different from the ∆rodA. Ccrp-mutant conidia were less immunostimulatory compared to ∆rodA conidia. Our data suggest that (i) the conserved cysteine residues are essential for the trafficking of RodAp and the organization of the rodlet layer on the conidial surface, and (ii) targeted point mutation could be an alternative approach to study the role of fungal cell-wall genes in host-fungal interaction.

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.
ACS Appl Mater Interfaces ; 11(50): 46591-46603, 2019 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-31742377

RESUMEN

Oral candidiasis as a highly prevalent and recurrent infection in medically compromised individuals is mainly caused by the opportunistic fungal pathogen Candida albicans. This epithelial infection, if not controlled effectively, can progress to life-threatening systemic conditions and complications. The efficacy of current frontline antifungals is limited due to their poor bioavailability and systemic toxicity. As such, an efficient intervention is essential for controlling disease progression and recurrence. Herein, a theranostic nanoplatform (CD-Gu+-AmB) was developed to track the penetration of antifungals and perturb the invasion of C. albicans at oral epithelial tissues, via decorating the homemade red-emissive carbon dots (CD) with positively charged guanidine groups (Gu+) followed by conjugation with antifungal polyene (amphotericin B, AmB) in a reacting site-controllable manner. The generated CD-Gu+-AmB favorably gathered within the Candida cells and exhibited potent antifungal effects in both planktonic and biofilm forms. It selectively accumulated in the nuclei of human oral keratinocytes and exhibited undetectable toxicity to the host cells. Moreover, we reported for the first time the penetration and exfoliation profiles of CD in a three-dimensional organotypic model of human oral epithelial tissues, demonstrating that the extra- and intracellular accumulation of CD-Gu+-AmB effectively resisted the invasion of C. albicans by forming a "shielding" layer throughout the entire tissue. This study establishes a multifunctional CD-based theranostic nanoplatform functioning as a traceable and topically applied antifungal to arm oral epithelia, thereby shedding light on early intervention of mucosal candidiasis for oral and general health.


Asunto(s)
Anfotericina B/farmacología , Antifúngicos/farmacología , Candidiasis/tratamiento farmacológico , Infecciones Fúngicas Invasoras/tratamiento farmacológico , Anfotericina B/química , Antifúngicos/química , Biopelículas/efectos de los fármacos , Disponibilidad Biológica , Candida albicans/efectos de los fármacos , Candida albicans/patogenicidad , Candidiasis/microbiología , Carbono/química , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Guanosina Monofosfato/química , Humanos , Infecciones Fúngicas Invasoras/microbiología , Queratinocitos/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/microbiología , Polienos/química , Polienos/farmacología , Puntos Cuánticos/química
8.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA