Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 5817, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987270

RESUMEN

Respiratory infections caused by the human fungal pathogen Aspergillus fumigatus are a major cause of mortality for immunocompromised patients. Exposure to these pathogens occurs through inhalation, although the role of the respiratory epithelium in disease pathogenesis has not been fully defined. Employing a primary human airway epithelial model, we demonstrate that fungal melanins potently block the post-translational secretion of the chemokines CXCL1 and CXCL8 independent of transcription or the requirement of melanin to be phagocytosed, leading to a significant reduction in neutrophil recruitment to the apical airway both in vitro and in vivo. Aspergillus-derived melanin, a major constituent of the fungal cell wall, dampened airway epithelial chemokine secretion in response to fungi, bacteria, and exogenous cytokines. Furthermore, melanin muted pathogen-mediated calcium fluxing and hindered actin filamentation. Taken together, our results reveal a critical role for melanin interaction with airway epithelium in shaping the host response to fungal and bacterial pathogens.


Asunto(s)
Aspergillus fumigatus , Calcio , Quimiocina CXCL1 , Interleucina-8 , Melaninas , Melaninas/metabolismo , Humanos , Interleucina-8/metabolismo , Calcio/metabolismo , Quimiocina CXCL1/metabolismo , Animales , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Ratones , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Quimiocinas/metabolismo , Ratones Endogámicos C57BL
2.
Front Cell Infect Microbiol ; 13: 1241770, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37724291

RESUMEN

Introduction: Invasive aspergillosis (IA) is the most prevalent infectious complication in patients with chronic granulomatous disease (CGD). Yet, understanding of fungal pathogenesis in the CGD host remains limited, particularly with regards to A. nidulans infection. Methods: We have used a murine model of X-linked CGD to investigate how the pathogenesis of IA varies between A. fumigatus and A. nidulans, comparing infection in both X-linked CGD (gp91-/-) mice and their parent C57BL/6 (WT) mice. A 14-colour flow cytometry panel was used to assess the cell dynamics over the course of infection, with parallel assessment of pulmonary cytokine production and lung histology. Results: We observed a lack of association between pulmonary pathology and infection outcome in gp91-/- mice, with no significant mortality in A. nidulans infected mice. An overwhelming and persistent neutrophil recruitment and IL-1 release in gp91-/- mice following both A. fumigatus and A. nidulans infection was observed, with divergent macrophage, dendritic cell and eosinophil responses and distinct cytokine profiles between the two infections. Conclusion: We have provided an in-depth characterisation of the immune response to pulmonary aspergillosis in an X-linked CGD murine model. This provides the first description of distinct pulmonary inflammatory environments in A. fumigatus and A. nidulans infection in X-linked CGD and identifies several new avenues for further research.


Asunto(s)
Aspergilosis , Aspergillus nidulans , Enfermedad Granulomatosa Crónica , Infecciones Fúngicas Invasoras , Animales , Ratones , Ratones Endogámicos C57BL , Aspergillus fumigatus/genética , Aspergillus nidulans/genética , Enfermedad Granulomatosa Crónica/complicaciones , Modelos Animales de Enfermedad , Citocinas
3.
Sci Adv ; 7(3)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33523895

RESUMEN

Macrophages provide a first line of defense against microorganisms, and while some mechanisms to kill pathogens such as the oxidative burst are well described, others are still undefined or unknown. Here, we report that the Rab32 guanosine triphosphatase and its guanine nucleotide exchange factor BLOC-3 (biogenesis of lysosome-related organelles complex-3) are central components of a trafficking pathway that controls both bacterial and fungal intracellular pathogens. This host-defense mechanism is active in both human and murine macrophages and is independent of well-known antimicrobial mechanisms such as the NADPH (reduced form of nicotinamide adenine dinucleotide phosphate)-dependent oxidative burst, production of nitric oxide, and antimicrobial peptides. To survive in human macrophages, Salmonella Typhi actively counteracts the Rab32/BLOC-3 pathway through its Salmonella pathogenicity island-1-encoded type III secretion system. These findings demonstrate that the Rab32/BLOC-3 pathway is a novel and universal host-defense pathway and protects mammalian species from various pathogens.


Asunto(s)
Salmonella typhi , Proteínas de Unión al GTP rab , Animales , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Lisosomas/metabolismo , Macrófagos/metabolismo , Mamíferos/metabolismo , Ratones , Proteínas de Unión al GTP rab/metabolismo
4.
Front Immunol ; 11: 2071, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33013877

RESUMEN

We specify the clinical features of a spontaneous experimental autoimmune uveitis (EAU) model, in which foreign hen-egg lysozyme (HEL) is expressed in the retina, controlled by the promoter for interphotoreceptor retinol binding protein (IRBP). We previously reported 100% P21 (post-partum day) IRBP:HEL single transgenic (sTg) mice, when crossed to transgenic T cell receptor mice (3A9) generating the double transgenic (dTg) genotype, develop EAU despite profound lymphopenia (thymic HEL-specific T cell deletion). In this work, we characterized the immune component of this model and found conventional dTg CD4+ T cells were less anergic than those from 3A9 controls. Furthermore, prior in vitro HEL-activation of 3A9 anergic T cells (Tan) rendered them uveitogenic upon adoptive transfer (Tx) to sTg mice, while antigen-experienced (AgX, dTg), but not naïve (3A9) T cells halted disease in P21 dTg mice. Flow cytometric analysis of the AgX cells elucidated the underlying pathology: FoxP3+CD25hiCD4+ T regulatory cells (Treg) comprised ∼18%, while FR4+CD73+FoxP3-CD25lo/-CD4+ Tan comprised ∼1.2% of total cells. Further Treg-enrichment (∼80%) of the AgX population indicated FoxP3+CD25hiCD4+ Treg played a key role in EAU-suppression while FoxP3-CD25lo/-CD4+ T cells did not. Here we present the novel concept of dual immunological tolerance where spontaneous EAU is due to escape from anergy with consequent failure of Treg induction and subsequent imbalance in the [Treg:Teffector] cell ratio. The reduced numbers of Tan, normally sustaining Treg to prevent autoimmunity, are the trigger for disease, while immune homeostasis can be restored by supplementation with AgX, but not naïve, antigen-specific Treg.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Inmunoterapia Adoptiva/métodos , Retina/patología , Linfocitos T Reguladores/inmunología , Uveítis/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas del Ojo/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Tolerancia Inmunológica , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Unión al Retinol/inmunología , Linfocitos T Reguladores/trasplante
5.
PLoS One ; 14(8): e0220867, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31393930

RESUMEN

Phagocytosis is a receptor-mediated process critical to innate immune clearance of pathogens. It proceeds in a regulated sequence of stages: (a) migration of phagocytes towards pathogens, (b) recognition of PAMPs and binding through PRRs, (c) engulfment and internalisation into phagosomes, (d) phagosome maturation, and (e) killing of pathogen or host cells. However, little is known about the role that individual receptors play in these discrete stages in the recognition of fungal cells. In a previous study, we found that dectin-2 deficiency impacted some but not all stages of macrophage-mediated phagocytosis of Candida glabrata. Because the C-type lectin receptor dectin-2 critically requires coupling to the FcRγ chain for signalling, we hypothesised that this coupling may be important for regulating phagocytosis of fungal cargo. We therefore examined how deficiency in FcRγ itself or two receptors to which it couples (dectin-2 and mincle) impacts phagocytosis of six fungal organisms representing three different fungal taxa. Our data show that deficiency in these proteins impairs murine bone marrow-derived macrophage migration, engulfment, and phagosome maturation, but not macrophage survival. Therefore, FcRγ engagement with selective C-type lectin receptors (CLRs) critically affects the spatio-temporal dynamics of fungal phagocytosis.


Asunto(s)
Hongos/inmunología , Fagocitosis , Receptores de Reconocimiento de Patrones/inmunología , Animales , Candida/inmunología , Movimiento Celular , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Macrófagos/citología , Malassezia/inmunología , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Mucor/inmunología , Unión Proteica , Receptores Fc/inmunología , Receptores Fc/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Saccharomyces/inmunología
6.
Immunity ; 50(2): 446-461.e9, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30709742

RESUMEN

Production of interleukin-17 (IL-17) and IL-22 by T helper 17 (Th17) cells and group 3 innate lymphoid cells (ILC3s) in response to the gut microbiota ensures maintenance of intestinal barrier function. Here, we examined the mechanisms whereby the immune system detects microbiota in the steady state. A Syk-kinase-coupled signaling pathway in dendritic cells (DCs) was critical for commensal-dependent production of IL-17 and IL-22 by CD4+ T cells. The Syk-coupled C-type lectin receptor Mincle detected mucosal-resident commensals in the Peyer's patches (PPs), triggered IL-6 and IL-23p19 expression, and thereby regulated function of intestinal Th17- and IL-17-secreting ILCs. Mice deficient in Mincle or with selective depletion of Syk in CD11c+ cells had impaired production of intestinal RegIIIγ and IgA and increased systemic translocation of gut microbiota. Consequently, Mincle deficiency led to liver inflammation and deregulated lipid metabolism. Thus, sensing of commensals by Mincle and Syk signaling in CD11c+ cells reinforces intestinal immune barrier and promotes host-microbiota mutualism, preventing systemic inflammation.


Asunto(s)
Células Dendríticas/inmunología , Microbioma Gastrointestinal/inmunología , Interleucina-17/inmunología , Interleucinas/inmunología , Lectinas Tipo C/inmunología , Proteínas de la Membrana/inmunología , Quinasa Syk/inmunología , Animales , Células Dendríticas/metabolismo , Microbioma Gastrointestinal/fisiología , Humanos , Interleucina-17/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/microbiología , Transducción de Señal/inmunología , Quinasa Syk/genética , Quinasa Syk/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Interleucina-22
8.
Nature ; 555(7696): 382-386, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29489751

RESUMEN

Resistance to infection is critically dependent on the ability of pattern recognition receptors to recognize microbial invasion and induce protective immune responses. One such family of receptors are the C-type lectins, which are central to antifungal immunity. These receptors activate key effector mechanisms upon recognition of conserved fungal cell-wall carbohydrates. However, several other immunologically active fungal ligands have been described; these include melanin, for which the mechanism of recognition is hitherto undefined. Here we identify a C-type lectin receptor, melanin-sensing C-type lectin receptor (MelLec), that has an essential role in antifungal immunity through recognition of the naphthalene-diol unit of 1,8-dihydroxynaphthalene (DHN)-melanin. MelLec recognizes melanin in conidial spores of Aspergillus fumigatus as well as in other DHN-melanized fungi. MelLec is ubiquitously expressed by CD31+ endothelial cells in mice, and is also expressed by a sub-population of these cells that co-express epithelial cell adhesion molecule and are detected only in the lung and the liver. In mouse models, MelLec was required for protection against disseminated infection with A. fumigatus. In humans, MelLec is also expressed by myeloid cells, and we identified a single nucleotide polymorphism of this receptor that negatively affected myeloid inflammatory responses and significantly increased the susceptibility of stem-cell transplant recipients to disseminated Aspergillus infections. MelLec therefore recognizes an immunologically active component commonly found on fungi and has an essential role in protective antifungal immunity in both mice and humans.


Asunto(s)
Aspergillus fumigatus/inmunología , Lectinas Tipo C/inmunología , Melaninas/inmunología , Naftoles/inmunología , Animales , Aspergilosis/inmunología , Aspergilosis/microbiología , Aspergilosis/prevención & control , Aspergillus fumigatus/química , Aspergillus fumigatus/patogenicidad , Pared Celular/química , Pared Celular/inmunología , Femenino , Humanos , Macrófagos/inmunología , Melaninas/química , Ratones , Ratones Endogámicos C57BL , Naftoles/química , Ratas , Ratas Sprague-Dawley , Esporas Fúngicas/química , Esporas Fúngicas/inmunología , Especificidad por Sustrato
9.
Microbes Infect ; 18(7-8): 505-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27005451

RESUMEN

The heterodimeric mycobacterial receptors, macrophage C-type lectin (MCL) and macrophage inducible C-type lectin (Mincle), are upregulated at the cell surface following microbial challenge, but the mechanisms underlying this response are unclear. Here we report that microbial stimulation triggers Mincle expression through the myeloid differentiation primary response gene 88 (MyD88) pathway; a process that does not require MCL. Conversely, we show that MCL is constitutively expressed but retained intracellularly until Mincle is induced, whereupon the receptors form heterodimers which are translocated to the cell surface. Thus this "two-step" model for induction of these key receptors provides new insights into the underlying mechanisms of anti-mycobacterial immunity.


Asunto(s)
Interacciones Huésped-Patógeno , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Mycobacterium/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Células Cultivadas , Expresión Génica , Lectinas Tipo C/genética , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Receptores de Superficie Celular/genética , Receptores Inmunológicos/genética
10.
Eur J Immunol ; 46(2): 381-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26558717

RESUMEN

The C-type lectin receptor (CTLR), Clec4d (MCL, CLECSF8), is a member of the Dectin-2 cluster of CTLRs, which also includes the related receptors Mincle and Dectin-2. Like Mincle, Clec4d recognizes mycobacterial cord factor, trehalose dimycolate, and we recently demonstrated its key role in anti-mycobacterial immunity in mouse and man. Here, we characterized receptor expression in naïve mice, under inflammatory conditions, and during Mycobacterium bovis BCG infection using newly generated monoclonal antibodies. In naïve mice, Clec4d was predominantly expressed on myeloid cells within the peritoneal cavity, blood, and bone marrow. Unexpectedly, basal expression of Clec4d was very low on leukocytes in the lung. However, receptor expression was significantly upregulated on pulmonary myeloid cells during M. bovis BCG infection. Moreover, Clec4d expression could be strongly induced in vitro and in vivo by various microbial stimuli, including TLR agonists, but not exogenous cytokines. Notably, we show that Clec4d requires association with the signaling adaptor FcRγ and Mincle, but not Dectin-2, for surface expression. In addition, we provide evidence that Clec4d and Mincle, but not Dectin-2, are interdependently coregulated during inflammation and infection. These data show that Clec4d is an inducible myeloid-expressed CTLR in mice, whose expression is tightly linked to that of Mincle.


Asunto(s)
Factores Cordón/metabolismo , Lectinas Tipo C/metabolismo , Leucocitos/inmunología , Mycobacterium bovis/inmunología , Células Mieloides/inmunología , Receptores de IgG/metabolismo , Receptores Inmunológicos/metabolismo , Tuberculosis/inmunología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Inmunidad Innata , Lectinas Tipo C/genética , Leucocitos/microbiología , Pulmón/microbiología , Pulmón/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium bovis/metabolismo , Células Mieloides/microbiología , Cavidad Peritoneal/microbiología , Cavidad Peritoneal/patología , Receptores Inmunológicos/genética , Transducción de Señal , Tuberculosis/veterinaria
11.
Ann Rheum Dis ; 75(7): 1386-91, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26275430

RESUMEN

BACKGROUND: Myeloid inhibitory C-type lectin-like receptor (MICL, Clec12A) is a C-type lectin receptor (CLR) expressed predominantly by myeloid cells. Previous studies have suggested that MICL is involved in controlling inflammation. OBJECTIVE: To determine the role of this CLR in inflammatory pathology using Clec12A(-/-) mice. METHODS: Clec12A(-/-) mice were generated commercially and primarily characterised using the collagen antibody-induced arthritis (CAIA) model. Mechanisms and progress of disease were characterised by clinical scoring, histology, flow cytometry, irradiation bone-marrow chimera generation, administration of blocking antibodies and in vivo imaging. Characterisation of MICL in patients with rheumatoid arthritis (RA) was determined by immunohistochemistry and single nucleotide polymorphism analysis. Anti-MICL antibodies were detected in patient serum by ELISA and dot-blot analysis. RESULTS: MICL-deficient animals did not present with pan-immune dysfunction, but exhibited markedly exacerbated inflammation during CAIA, owing to the inappropriate activation of myeloid cells. Polymorphisms of MICL were not associated with disease in patients with RA, but this CLR was the target of autoantibodies in a subset of patients with RA. In wild-type mice the administration of such antibodies recapitulated the Clec12A(-/-) phenotype. CONCLUSIONS: MICL plays an essential role in regulating inflammation during arthritis and is an autoantigen in a subset of patients with RA. These data suggest an entirely new mechanism underlying RA pathogenesis, whereby the threshold of myeloid cell activation can be modulated by autoantibodies that bind to cell membrane-expressed inhibitory receptors.


Asunto(s)
Artritis Experimental/genética , Artritis Reumatoide/genética , Lectinas Tipo C/fisiología , Receptores Mitogénicos/fisiología , Animales , Artritis Reumatoide/sangre , Artritis Reumatoide/etiología , Artritis Reumatoide/patología , Autoanticuerpos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Lectinas Tipo C/deficiencia , Lectinas Tipo C/inmunología , Ratones , Células Mieloides/metabolismo , Polimorfismo Genético , Receptores Mitogénicos/deficiencia , Receptores Mitogénicos/inmunología , Membrana Sinovial/patología
12.
J Immunol ; 195(8): 3781-92, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26336150

RESUMEN

Candida is the third most common cause of bloodstream infections in hospitalized patients. Immunity to C. albicans, the most frequent species to be isolated in candidiasis, involves a well-characterized Dectin-1/caspase-associated recruitment domain adaptor 9 (CARD9)/IL-17 signaling axis. Infections caused by non-albicans Candida species are on the rise, but surprisingly little is known about immunity to these pathogens. In this study, we evaluated a systemic infection model of C. tropicalis, a clinically relevant, but poorly understood, non-albicans Candida. Mice lacking CARD9 were profoundly susceptible to C. tropicalis, displaying elevated fungal burdens in visceral organs and increased mortality compared with wild-type (WT) controls. Unlike C. albicans, IL-17 responses were induced normally in CARD9(-/-) mice following C. tropicalis infection. Moreover, there was no difference in susceptibility to C. tropicalis infection between WT and IL-23p19(-/-), IL-17RA(-/-), or Act1(-/-) mice. However, TNF-α expression was markedly impaired in CARD9(-/-) mice. Consistently, WT mice depleted of TNF-α were more susceptible to C. tropicalis, and CARD9-deficient neutrophils and monocytes failed to produce TNF-α following stimulation with C. tropicalis Ags. Both neutrophils and monocytes were necessary for defense against C. tropicalis, because their depletion in WT mice enhanced susceptibility to C. tropicalis. Disease in CARD9(-/-) mice was not due to defective neutrophil or monocyte recruitment to infected kidneys. However, TNF-α treatment of neutrophils in vitro enhanced their ability to kill C. tropicalis. Thus, protection against systemic C. tropicalis infection requires CARD9 and TNF-α, but not IL-17, signaling. Moreover, CARD9-dependent production of TNF-α enhances the candidacidal capacity of neutrophils, limiting fungal disease during disseminated C. tropicalis infection.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/inmunología , Candida tropicalis/inmunología , Candidiasis/inmunología , Interleucina-17/inmunología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Candidiasis/genética , Candidiasis/patología , Interleucina-17/genética , Ratones , Ratones Noqueados , Monocitos/inmunología , Monocitos/patología , Neutrófilos/inmunología , Neutrófilos/patología , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/genética
13.
Mol Immunol ; 67(2 Pt B): 398-406, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26216045

RESUMEN

Mycobacteria in complete Freund's adjuvant (CFA) are an essential component of immunization protocols in a number of autoimmune disease animal models including experimental autoimmune encephalomyelitis and uveoretinitis (EAE and EAU, respectively). We determined the role in EAU of two C-type lectin receptors on myeloid cells that recognize and respond to mycobacteria. Using receptor-specific antibodies and knockout mice, we demonstrated for the first time that the macrophage mannose receptor delays disease development but does not affect severity. In contrast, dectin-1 is critically involved in the development of CFA-mediated EAU. Disease severity is reduced in dectin-1 knockout mice and antibody blockade of dectin-1 during the induction, but not the effector phase, prevents EAU development. Significantly, similar blockade of dectin-1 in vivo has no effect in non-CFA-mediated, spontaneously induced or adoptive transfer models of EAU. Thus dectin-1 plays a critical role in the ability of complete Freund's adjuvant to induce EAU in mice.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Lectinas Tipo C/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Retinitis/metabolismo , Uveítis/metabolismo , Animales , Anticuerpos Bloqueadores/farmacología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Adyuvante de Freund/inmunología , Humanos , Inmunización , Mediadores de Inflamación/metabolismo , Lectinas Tipo C/deficiencia , Lectinas Tipo C/inmunología , Ganglios Linfáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Reconocimiento de Patrones/deficiencia , Receptores de Reconocimiento de Patrones/inmunología , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Retinitis/inmunología , Retinitis/patología , Proteínas de Unión al Retinol/metabolismo , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Factores de Tiempo , Uveítis/inmunología , Uveítis/patología
14.
Cell Host Microbe ; 17(2): 252-9, 2015 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-25674984

RESUMEN

The interaction of microbes with pattern recognition receptors (PRRs) is essential for protective immunity. While many PRRs that recognize mycobacteria have been identified, none is essentially required for host defense in vivo. Here, we have identified the C-type lectin receptor CLECSF8 (CLEC4D, MCL) as a key molecule in anti-mycobacterial host defense. Clecsf8-/- mice exhibit higher bacterial burdens and increased mortality upon M. tuberculosis infection. Additionally, Clecsf8 deficiency is associated with exacerbated pulmonary inflammation, characterized by enhanced neutrophil recruitment. Clecsf8-/- mice show reduced mycobacterial uptake by pulmonary leukocytes, but infection with opsonized bacteria can restore this phagocytic defect as well as decrease bacterial burdens. Notably, a CLECSF8 polymorphism identified in humans is associated with an increased susceptibility to pulmonary tuberculosis. We conclude that CLECSF8 plays a non-redundant role in anti-mycobacterial immunity in mouse and in man.


Asunto(s)
Lectinas Tipo C/metabolismo , Proteínas de la Membrana/metabolismo , Mycobacterium tuberculosis/inmunología , Animales , Carga Bacteriana , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Pulmón/patología , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Fagocitosis , Polimorfismo Genético , Receptores Inmunológicos/metabolismo , Análisis de Supervivencia , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
15.
mBio ; 5(6): e01874, 2014 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-25467440

RESUMEN

UNLABELLED: Candida albicans is a major life-threatening human fungal pathogen in the immunocompromised host. Host defense against systemic Candida infection relies heavily on the capacity of professional phagocytes of the innate immune system to ingest and destroy fungal cells. A number of pathogens, including C. albicans, have evolved mechanisms that attenuate the efficiency of phagosome-mediated inactivation, promoting their survival and replication within the host. Here we visualize host-pathogen interactions using live-cell imaging and show that viable, but not heat- or UV-killed C. albicans cells profoundly delay phagosome maturation in macrophage cell lines and primary macrophages. The ability of C. albicans to delay phagosome maturation is dependent on cell wall composition and fungal morphology. Loss of cell wall O-mannan is associated with enhanced acquisition of phagosome maturation markers, distinct changes in Rab GTPase acquisition by the maturing phagosome, impaired hyphal growth within macrophage phagosomes, profound changes in macrophage actin dynamics, and ultimately a reduced ability of fungal cells to escape from macrophage phagosomes. The loss of cell wall O-mannan leads to exposure of ß-glucan in the inner cell wall, facilitating recognition by Dectin-1, which is associated with enhanced phagosome maturation. IMPORTANCE: Innate cells engulf and destroy invading organisms by phagocytosis, which is essential for the elimination of fungal cells to protect against systemic life-threatening infections. Yet comparatively little is known about what controls the maturation of phagosomes following ingestion of fungal cells. We used live-cell microscopy and fluorescent protein reporter macrophages to understand how C. albicans viability, filamentous growth, and cell wall composition affect phagosome maturation and the survival of the pathogen within host macrophages. We have demonstrated that cell wall glycosylation and yeast-hypha morphogenesis are required for disruption of host processes that function to inactivate pathogens, leading to survival and escape of this fungal pathogen from within host phagocytes. The methods employed here are applicable to study interactions of other pathogens with phagocytic cells to dissect how specific microbial features impact different stages of phagosome maturation and the survival of the pathogen or host.


Asunto(s)
Candida albicans/inmunología , Pared Celular/química , Hifa/inmunología , Macrófagos/inmunología , Mananos/metabolismo , Fagosomas/inmunología , beta-Glucanos/metabolismo , Candida albicans/química , Candida albicans/metabolismo , Células Cultivadas , Interacciones Huésped-Patógeno , Humanos , Hifa/química , Hifa/metabolismo , Evasión Inmune , Macrófagos/microbiología , Fagosomas/microbiología
16.
J Immunol ; 193(11): 5381-5, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25344471

RESUMEN

Candida albicans is the leading cause of systemic candidiasis, a fungal disease associated with high mortality and poor treatment options. The kidney is the target organ during infection and whose control is largely dependent on innate immunity, because lymphocytes appear redundant for protection. In this article, we show that this apparent redundancy stems from a failure of Ag-specific CD4(+) T cells to migrate into infected kidneys. In contrast, Ag-specific CD8(+) T cells are recruited normally. Using Ag-loaded immunoliposomes to artificially reverse this defective migration, we show that recruited Ag-specific CD4(+) T cells polarize toward a Th17 phenotype in the kidney and are protective during fungal infection. Therefore, our data explain the redundancy of CD4(+) T cells for defense against systemic infection with C. albicans and have important implications for our understanding of antifungal immunity and the control of renal infections.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Candida albicans/inmunología , Candidiasis/inmunología , Riñón/inmunología , Células Th17/inmunología , Animales , Antígenos/inmunología , Antígenos/metabolismo , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/microbiología , Movimiento Celular , Células Cultivadas , Citoprotección , Femenino , Inmunidad Innata , Riñón/microbiología , Liposomas/inmunología , Liposomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Células Th17/microbiología
17.
Infect Immun ; 82(3): 1064-73, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24343653

RESUMEN

Although Candida glabrata is an important pathogenic Candida species, relatively little is known about its innate immune recognition. Here, we explore the potential role of Dectin-2 for host defense against C. glabrata. Dectin-2-deficient (Dectin-2(-/-)) mice were found to be more susceptible to C. glabrata infections, showing a defective fungal clearance in kidneys but not in the liver. The increased susceptibility to infection was accompanied by lower production of T helper 1 (Th1) and Th17-derived cytokines by splenocytes of Dectin-2(-/-) mice, while macrophage-derived cytokines were less affected. These defects were associated with a moderate yet significant decrease in phagocytosis of the fungus by the Dectin-2(-/-) macrophages and neutrophils. Neutrophils of Dectin-2(-/-) mice also displayed lower production of reactive oxygen species (ROS) upon challenge with opsonized C. glabrata or C. albicans. This study suggests that Dectin-2 is important in host defense against C. glabrata and provides new insights into the host defense mechanisms against this important fungal pathogen.


Asunto(s)
Candida glabrata/inmunología , Candidiasis/inmunología , Lectinas Tipo C/inmunología , Animales , Candida albicans/inmunología , Candidiasis/microbiología , Citocinas/inmunología , Femenino , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neutrófilos/microbiología , Fagocitosis/inmunología , Especies Reactivas de Oxígeno/inmunología , Células TH1/inmunología , Células TH1/microbiología
18.
Nat Med ; 18(9): 1401-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22922409

RESUMEN

Complement is an ancient danger-sensing system that contributes to host defense, immune surveillance and homeostasis. C5a and its G protein­coupled receptor mediate many of the proinflammatory properties of complement. Despite the key role of C5a in allergic asthma, autoimmune arthritis, sepsis and cancer, knowledge about its regulation is limited. Here we demonstrate that IgG1 immune complexes (ICs), the inhibitory IgG receptor FcγRIIB and the C-type lectin­like receptor dectin-1 suppress C5a receptor (C5aR) functions. IgG1 ICs promote the association of FcγRIIB with dectin-1, resulting in phosphorylation of Src homology 2 domain­containing inositol phosphatase (SHIP) downstream of FcγRIIB and spleen tyrosine kinase downstream of dectin-1. This pathway blocks C5aR-mediated ERK1/2 phosphorylation, C5a effector functions in vitro and C5a-dependent inflammatory responses in vivo, including peritonitis and skin blisters in experimental epidermolysis bullosa acquisita. Notably, high galactosylation of IgG N-glycans is crucial for this inhibitory property of IgG1 ICs, as it promotes the association between FcγRIIB and dectin-1. Thus, galactosylated IgG1 and FcγRIIB exert anti-inflammatory properties beyond their impact on activating FcγRs.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Complemento C5a/inmunología , Inmunoglobulina G/inmunología , Lectinas Tipo C/metabolismo , Receptores de Complemento/metabolismo , Receptores de IgG/metabolismo , Análisis de Varianza , Animales , Anticuerpos Monoclonales , Western Blotting , Calcio/metabolismo , Adhesión Celular/inmunología , Complemento C5a/administración & dosificación , Femenino , Inositol Polifosfato 5-Fosfatasas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lectinas Tipo C/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Receptor de Anafilatoxina C5a , Receptores de IgG/genética , Receptores de IgG/inmunología , Resonancia por Plasmón de Superficie , Quinasa Syk
19.
J Biol Chem ; 287(31): 25964-74, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22689578

RESUMEN

CLECSF8 is a poorly characterized member of the "Dectin-2 cluster" of C-type lectin receptors and was originally thought to be expressed exclusively by macrophages. We show here that CLECSF8 is primarily expressed by peripheral blood neutrophils and monocytes and weakly by several subsets of peripheral blood dendritic cells. However, expression of this receptor is lost upon in vitro differentiation of monocytes into dendritic cells or macrophages. Like the other members of the Dectin-2 family, which require association of their transmembrane domains with signaling adaptors for surface expression, CLECSF8 is retained intracellularly when expressed in non-myeloid cells. However, we demonstrate that CLECSF8 does not associate with any known signaling adaptor molecule, including DAP10, DAP12, or the FcRγ chain, and we found that the C-type lectin domain of CLECSF8 was responsible for its intracellular retention. Although CLECSF8 does not contain a signaling motif in its cytoplasmic domain, we show that this receptor is capable of inducing signaling via Syk kinase in myeloid cells and that it can induce phagocytosis, proinflammatory cytokine production, and the respiratory burst. These data therefore indicate that CLECSF8 functions as an activation receptor on myeloid cells and associates with a novel adaptor molecule. Characterization of the CLECSF8-deficient mice and screening for ligands using oligosaccharide microarrays did not provide further insights into the physiological function of this receptor.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lectinas Tipo C/metabolismo , Células Mieloides/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Expresión Génica , Regulación de la Expresión Génica , Humanos , Lectinas Tipo C/química , Ratones , Células Mieloides/enzimología , Células Mieloides/fisiología , Especificidad de Órganos , Fagocitosis , Cultivo Primario de Células , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores Inmunológicos/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Estallido Respiratorio , Transducción de Señal , Quinasa Syk , Factor de Necrosis Tumoral alfa/metabolismo
20.
PLoS One ; 7(4): e35675, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22536422

RESUMEN

The innate recognition of fungi by leukocytes is mediated by pattern recognition receptors (PRR), such as Dectin-1, and is thought to occur at the cell surface triggering intracellular signalling cascades which lead to the induction of protective host responses. In the lung, this recognition is aided by surfactant which also serves to maintain the balance between inflammation and pulmonary function, although the underlying mechanisms are unknown. Here we have explored pulmonary innate recognition of a variety of fungal particles, including zymosan, Candida albicans and Aspergillus fumigatus, and demonstrate that opsonisation with surfactant components can limit inflammation by reducing host-cell fungal interactions. However, we found that this opsonisation does not contribute directly to innate fungal recognition and that this process is mediated through non-opsonic PRRs, including Dectin-1. Moreover, we found that pulmonary inflammatory responses to resting Aspergillus conidia were initiated by these PRRs in acidified phagolysosomes, following the uptake of fungal particles by leukocytes. Our data therefore provides crucial new insights into the mechanisms by which surfactant can maintain pulmonary function in the face of microbial challenge, and defines the phagolysosome as a novel intracellular compartment involved in the innate sensing of extracellular pathogens in the lung.


Asunto(s)
Aspergillus fumigatus/inmunología , Candida albicans/inmunología , Candidiasis/inmunología , Inmunidad Innata , Aspergilosis Pulmonar/inmunología , Animales , Antígenos Fúngicos/inmunología , Aspergillus fumigatus/fisiología , Líquido del Lavado Bronquioalveolar , Femenino , Interacciones Huésped-Patógeno , Lectinas Tipo C/metabolismo , Enfermedades Pulmonares Fúngicas/inmunología , Lisosomas/inmunología , Lisosomas/microbiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Fagocitosis/inmunología , Fagosomas/inmunología , Fagosomas/microbiología , Neumonía/inmunología , Neumonía/metabolismo , Neumonía/microbiología , Unión Proteica , Surfactantes Pulmonares/metabolismo , Receptores Inmunológicos/metabolismo , Esporas Fúngicas/inmunología , Zimosan/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...