Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nat Immunol ; 20(5): 559-570, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30996332

RESUMEN

The C-type lectin receptor-Syk (spleen tyrosine kinase) adaptor CARD9 facilitates protective antifungal immunity within the central nervous system (CNS), as human deficiency in CARD9 causes susceptibility to fungus-specific, CNS-targeted infection. CARD9 promotes the recruitment of neutrophils to the fungus-infected CNS, which mediates fungal clearance. In the present study we investigated host and pathogen factors that promote protective neutrophil recruitment during invasion of the CNS by Candida albicans. The cytokine IL-1ß served an essential function in CNS antifungal immunity by driving production of the chemokine CXCL1, which recruited neutrophils expressing the chemokine receptor CXCR2. Neutrophil-recruiting production of IL-1ß and CXCL1 was induced in microglia by the fungus-secreted toxin Candidalysin, in a manner dependent on the kinase p38 and the transcription factor c-Fos. Notably, microglia relied on CARD9 for production of IL-1ß, via both transcriptional regulation of Il1b and inflammasome activation, and of CXCL1 in the fungus-infected CNS. Microglia-specific Card9 deletion impaired the production of IL-1ß and CXCL1 and neutrophil recruitment, and increased fungal proliferation in the CNS. Thus, an intricate network of host-pathogen interactions promotes antifungal immunity in the CNS; this is impaired in human deficiency in CARD9, which leads to fungal disease of the CNS.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/inmunología , Candidiasis/inmunología , Quimiocina CXCL1/inmunología , Interleucina-1beta/inmunología , Microglía/inmunología , Neutrófilos/inmunología , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/microbiología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Candida albicans/inmunología , Candida albicans/fisiología , Candidiasis/genética , Candidiasis/microbiología , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inflamasomas/genética , Inflamasomas/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Microglía/microbiología , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología
2.
PLoS Pathog ; 19(5): e1011355, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37200244

RESUMEN

Immune checkpoint inhibitor (ICI) therapy represents a breakthrough cancer treatment by stimulating dysfunctional T cells in the tumour environment to kill cancer cells. Beyond effects on anticancer immunity, ICI therapy may be associated with increased susceptibility to or more rapid resolution of chronic infections, particularly those caused by human fungal pathogens. In this concise review, we summarise recent observations and findings that implicate immune checkpoint blockade in fungal infection outcomes.


Asunto(s)
Neoplasias , Humanos , Neoplasias/patología , Inmunoterapia , Antígeno CTLA-4
3.
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
4.
Microb Pathog ; 185: 106437, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37913825

RESUMEN

BACKGROUND: Our previous proteomics data obtained from Candida albicans recovered after serial passage in a murine model of systemic infection revealed that Orf19.36.1 expression correlates with the virulence of the fungus. Therefore, the impact of ORF19.36.1 upon virulence was tested in this study. MATERIALS & METHODS: CRISPR-Cas9 technology was used to construct homozygous C. albicans orf19.36.1 null mutants and the phenotypes of these mutants examined in vitro (filamentation, invasion, adhesion, biofilm formation, hydrolase activities) and in vivo assays. RESULTS: The deletion of ORF19.36.1 did not significantly impact the phenotypes examined or the virulence of C. albicans in two infection models. CONCLUSION: These results suggest that, although Orf19.36.1 expression correlates with virulence, this protein is not essential for C. albicans pathobiology.


Asunto(s)
Candida albicans , Candidiasis , Proteínas Fúngicas , Animales , Ratones , Candidiasis/microbiología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Virulencia/genética
5.
Trends Immunol ; 41(1): 61-76, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31813764

RESUMEN

Fungi can cause disease in humans, from mucocutaneous to life-threatening systemic infections. Initiation of antifungal immunity involves fungal recognition by pattern recognition receptors such as C-type lectin receptors (CLRs). These germline-encoded receptors trigger a multitude of innate responses including phagocytosis, fungal killing, and antigen presentation which can also shape the development of adaptive immunity. Recently, studies have shed light on how CLRs directly or indirectly modulate lymphocyte function. Moreover, CLR-mediated recognition of commensal fungi maintains homeostasis and prevents invasion from opportunistic commensals. We present an overview of current knowledge of antifungal T cell immune responses, with emphasis on the role of C-type lectins, and discuss how these receptors modulate these responses at different levels.


Asunto(s)
Inmunidad Innata , Lectinas Tipo C , Micosis , Linfocitos T , Hongos/inmunología , Humanos , Inmunidad Innata/inmunología , Lectinas Tipo C/inmunología , Micosis/inmunología , Linfocitos T/inmunología
6.
PLoS Pathog ; 14(5): e1006978, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29775474

RESUMEN

Fungal cells change shape in response to environmental stimuli, and these morphogenic transitions drive pathogenesis and niche adaptation. For example, dimorphic fungi switch between yeast and hyphae in response to changing temperature. The basidiomycete Cryptococcus neoformans undergoes an unusual morphogenetic transition in the host lung from haploid yeast to large, highly polyploid cells termed Titan cells. Titan cells influence fungal interaction with host cells, including through increased drug resistance, altered cell size, and altered Pathogen Associated Molecular Pattern exposure. Despite the important role these cells play in pathogenesis, understanding the environmental stimuli that drive the morphological transition, and the molecular mechanisms underlying their unique biology, has been hampered by the lack of a reproducible in vitro induction system. Here we demonstrate reproducible in vitro Titan cell induction in response to environmental stimuli consistent with the host lung. In vitro Titan cells exhibit all the properties of in vivo generated Titan cells, the current gold standard, including altered capsule, cell wall, size, high mother cell ploidy, and aneuploid progeny. We identify the bacterial peptidoglycan subunit Muramyl Dipeptide as a serum compound associated with shift in cell size and ploidy, and demonstrate the capacity of bronchial lavage fluid and bacterial co-culture to induce Titanisation. Additionally, we demonstrate the capacity of our assay to identify established (cAMP/PKA) and previously undescribed (USV101) regulators of Titanisation in vitro. Finally, we investigate the Titanisation capacity of clinical isolates and their impact on disease outcome. Together, these findings provide new insight into the environmental stimuli and molecular mechanisms underlying the yeast-to-Titan transition and establish an essential in vitro model for the future characterization of this important morphotype.


Asunto(s)
Cryptococcus neoformans/citología , Cryptococcus neoformans/patogenicidad , Animales , Criptococosis/microbiología , Cryptococcus neoformans/genética , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas Fúngicas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Hifa/citología , Hifa/crecimiento & desarrollo , Hifa/patogenicidad , Pulmón/microbiología , Enfermedades Pulmonares Fúngicas/microbiología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Modelos Biológicos , Morfogénesis , Poliploidía , Factores de Transcripción/metabolismo , Virulencia
7.
Nature ; 574(7777): 184-185, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31591542
8.
Adv Exp Med Biol ; 1185: 353-358, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31884637

RESUMEN

Neurotrophic factors can promote the survival of degenerating retinal cells through the activation of STAT3 pathway. Thus, augmenting STAT3 activation in the retina has been proposed as potential therapy for retinal dystrophies. On the other hand, aberrant activation of STAT3 pathway is oncogenic and implicated in diverse human diseases. Furthermore, the STAT3/SOCS3 axis has been shown to induce the degradation of rhodopsin during retinal inflammation. In this study, we generated and used mice with constitutive activation of STAT3 pathway in the retina to evaluate the safety and consequences of enhancing STAT3 activities in the retina as a potential treatment for retinal degenerative diseases. We show that long-term activation of the STAT3 pathway can induce retinal degenerative changes and also exacerbate uveitis and other intraocular inflammatory diseases. Mechanisms underlying the development of vision impairment in the STAT3c-Tg mice derived in part from STAT3-mediated inhibition of rhodopsin and overexpression of SOCS3 in the retina. These results suggest that much caution should be exercised in the use of STAT3 augmentation therapy for retinal dystrophies.


Asunto(s)
Envejecimiento , Retina/patología , Degeneración Retiniana/patología , Factor de Transcripción STAT3/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas , Uveítis/patología
9.
mSphere ; 9(6): e0028124, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38771036

RESUMEN

The fungal pathogen Cryptococcus neoformans (C. neoformans) forms yeast cells of different sizes and morphological characteristics during infection. These features are usually not seen in standard laboratory in vitro conditions. Here, we describe in vivo cell morphologies when C. neoformans is grown in human plasma-like medium at 37°C, 5% CO2. We observed mixed-size populations of cells less than 1 µm up to 16.8 µm in cell diameter, increased capsule size, high chitin, and DNA content in larger cells. Our findings show that serum is not required for human plasma-like medium (HPLM)-induced C. neoformans cellular heterogeneity. Thus, this new method offers an opportunity to investigate factors of C. neoformans that mediate pathogenesis or host-pathogen interactions in a physiologically relevant setting.IMPORTANCEWe provide a description of new in vitro culture condition using the human plasma-like medium that supports the formation of the full range of in vivo cell morphologies of C. neoformans.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Medios de Cultivo , Cryptococcus neoformans/citología , Humanos , Medios de Cultivo/química , Criptococosis/microbiología , Animales , Ratones , Plasma/microbiología , Interacciones Huésped-Patógeno
10.
Cell Surf ; 11: 100128, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38938582

RESUMEN

Host recognition of the pathogen-associated molecular pattern (PAMP), ß-1,3-glucan, plays a major role in antifungal immunity. ß-1,3-glucan is an essential component of the inner cell wall of the opportunistic pathogen Candida albicans. Most ß-1,3-glucan is shielded by the outer cell wall layer of mannan fibrils, but some can become exposed at the cell surface. In response to host signals such as lactate, C. albicans shaves the exposed ß-1,3-glucan from its cell surface, thereby reducing the ability of innate immune cells to recognise and kill the fungus. We have used sets of barcoded xog1 and eng1 mutants to compare the impacts of the secreted ß-glucanases Xog1 and Eng1 upon C. albicans in vitro and in vivo. Flow cytometry of Fc-dectin-1-stained strains revealed that Eng1 plays the greater role in lactate-induced ß-1,3-glucan masking. Transmission electron microscopy and stress assays showed that neither Eng1 nor Xog1 are essential for cell wall maintenance, but the inactivation of either enzyme compromised fungal adhesion to gut and vaginal epithelial cells. Competitive barcode sequencing suggested that neither Eng1 nor Xog1 strongly influence C. albicans fitness during systemic infection or vaginal colonisation in mice. However, the deletion of XOG1 enhanced C. albicans fitness during gut colonisation. We conclude that both Eng1 and Xog1 exert subtle effects on the C. albicans cell surface that influence fungal adhesion to host cells and that affect fungal colonisation in certain host niches.

11.
J Biol Chem ; 287(36): 30436-43, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22761423

RESUMEN

An important feature of the adaptive immune response is its remarkable capacity to regulate the duration of inflammatory responses, and effector T cells have been shown to limit excessive immune responses by producing anti-inflammatory cytokines such as IL-10 and IL-27. However, how anti-inflammatory cytokines mediate their suppressive activities is not well understood. In this study, we show that STAT3 contributes to mechanisms that control the duration of T cell proliferation by regulating the subcellular location of FoxO1 and FoxO3a, two Class O Forkhead transcription factors that mediate lymphocyte quiescence and inhibit T cell activation. We show that active FoxO1 and FoxO3a reside exclusively in the nucleus of naïve T cells whereas inactive pFoxO1 and pFoxO3a were most abundant in activated T cells and sequestered in their cytoplasm in association with unphosphorylated STAT3 (U-STAT3) and 14-3-3. We further show that FoxO1/FoxO3a rapidly relocalized into the nucleus in response to pSTAT3 activation by IL-6 or IL-10, and the accumulation of FoxO1/FoxO3a in their nuclei coincided with increased expression of p27(Kip1) and p21(WAF1). STAT3 inhibitors completely abrogated cytokine-induced translocation of FoxO1/FoxO3a into the nucleus. In naïve or resting STAT3-deficient T cells, expression of pFoxO1/pFoxO3a was predominantly in the cytoplasm and correlated with defects in p27(Kip1) and p21(WAF1) expression, suggesting requirement of STAT3 for importation or retention of FoxO in the nucleus and attenuation of lymphocyte proliferation. Taken together, these results suggest that U-STAT3 collaborates with 14-3-3 to sequester pFoxO1/pFoxO3a in cytoplasm and thus prolong T cell activation, whereas pSTAT3 activation by anti-inflammatory cytokines would curtail the duration of TCR activation and re-establish lymphocyte quiescence by inducing nuclear localization of FoxO1/FoxO3a and FoxO-mediated expression of growth-inhibitory proteins.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/inmunología , Proteínas 14-3-3/metabolismo , Transporte Activo de Núcleo Celular/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Núcleo Celular/genética , Núcleo Celular/inmunología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/inmunología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/inmunología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Citoplasma/genética , Citoplasma/inmunología , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Células Jurkat , Activación de Linfocitos/fisiología , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología
12.
Mediators Inflamm ; 2013: 359674, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204098

RESUMEN

STAT3 regulates CD4+ T cell survival and differentiation. However, its effects on CD8+ T cells are not well understood. Here, we show that in comparison to WT CD8+ T cells, STAT3-deficient CD8+ T cells exhibit a preactivated memory-like phenotype, produce more IL-2, proliferate faster, and are more sensitive to activation-induced cell death (AICD). The enhanced proliferation and sensitivity to AICD correlated with downregulation of class-O forkhead transcription factors (FoxO1, FoxO3A), p21(waf1), p27(KIP1), Bcl-2, OX-40, and upregulation of FasL, Bax, and Bad. We examined whether STAT3-deficient CD8+ T cells can mount effective response during herpes simplex virus (HSV-1) infection and experimental autoimmune uveitis (EAU). Compared to WT mice, HSV-1-infected STAT3-deficient mice (STAT3KO) produced less IFN-γ and virus-specific KLRG-1+ CD8+ T cells. STAT3KO mice are also resistant to EAU and produced less IL-17-producing Tc17 cells. Resistance of STAT3KO to EAU correlated with marked expansion of IL-10-producing regulatory CD8+ T cells (CD8-Treg) implicated in recovery from autoimmune encephalomyelitis. Thus, increases of IL-6-induced STAT3 activation observed during inflammation may inhibit expansion of CD8-Tregs, thereby impeding recovery from uveitis. These results suggest that STAT3 is a potential therapeutic target for upregulating CD8+ T cell-mediated responses to viruses and suggest the successful therapeutic targeting of STAT3 as treatment for uveitis, derived, in part, from promoting CD8-Treg expansion.


Asunto(s)
Linfocitos T CD8-positivos/citología , Herpes Simple/metabolismo , Interleucina-10/metabolismo , Factor de Transcripción STAT3/genética , Uveítis/inmunología , Animales , Apoptosis , Enfermedades Autoinmunes/inmunología , Proliferación Celular , Separación Celular , Citometría de Flujo , Regulación de la Expresión Génica , Herpes Simple/virología , Herpesvirus Humano 1 , Inflamación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba , Uveítis/virología
13.
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
14.
Cell Surf ; 8: 100084, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36299406

RESUMEN

The immunogenicity of Candida albicans cells is influenced by changes in the exposure of microbe-associated molecular patterns (MAMPs) on the fungal cell surface. Previously, the degree of exposure on the C. albicans cell surface of the immunoinflammatory MAMP ß-(1,3)-glucan was shown to correlate inversely with colonisation levels in the gastrointestinal (GI) tract. This is important because life-threatening systemic candidiasis in critically ill patients often arises from translocation of C. albicans strains present in the patient's GI tract. Therefore, using a murine model, we have examined the impact of gut-related factors upon ß-glucan exposure and colonisation levels in the GI tract. The degree of ß-glucan exposure was examined by imaging flow cytometry of C. albicans cells taken directly from GI compartments, and compared with colonisation levels. Fungal ß-glucan exposure was lower in the cecum than the small intestine, and fungal burdens were correspondingly higher in the cecum. This inverse correlation did not hold for the large intestine. The gut fermentation acid, lactate, triggers ß-glucan masking in vitro, leading to attenuated anti-Candida immune responses. Additional fermentation acids are present in the GI tract, including acetate, propionate, and butyrate. We show that these acids also influence ß-glucan exposure on C. albicans cells in vitro and, like lactate, they influence ß-glucan exposure via Gpr1/Gpa2-mediated signalling. Significantly, C. albicans gpr1Δ gpa2Δ cells displayed elevated ß-glucan exposure in the large intestine and a corresponding decrease in fungal burden, consistent with the idea that Gpr1/Gpa2-mediated ß-glucan masking influences colonisation of this GI compartment. Finally, extracts from the murine gut and culture supernatants from the mannan grazing gut anaerobe Bacteroides thetaiotaomicron promote ß-glucan exposure at the C. albicans cell surface. Therefore, the local microbiota influences ß-glucan exposure levels directly (via mannan grazing) and indirectly (via fermentation acids), whilst ß-glucan masking appears to promote C. albicans colonisation of the murine large intestine.

15.
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
16.
Front Immunol ; 12: 675702, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34122436

RESUMEN

Environmental factors, particularly fungi, influence the pathogenesis of allergic airway inflammation, but the mechanisms underlying these effects are still unclear. Melanin is one fungal component which is thought to modulate pulmonary inflammation. We recently identified a novel C-type lectin receptor, MelLec (Clec1a), which recognizes fungal 1,8-dihydroxynaphthalene (DHN)-melanin and is able to regulate inflammatory responses. Here we show that MelLec promotes pulmonary allergic inflammation and drives the development of Th17 T-cells in response to spores of Aspergillus fumigatus. Unexpectedly, we found that MelLec deficiency was protective, with MelLec-/- animals showing normal weight gain and significantly reduced pulmonary inflammation in our allergic model. The lungs of treated MelLec-/- mice displayed significantly reduced inflammatory foci and reduced bronchial wall thickening, which correlated with a reduced cellular influx (particularly neutrophils and inflammatory monocytes) and levels of inflammatory cytokines and chemokines. Notably, fungal burdens were increased in MelLec-/- animals, without apparent adverse effects, and there were no alterations in the survival of these mice. Characterization of the pulmonary T-cell populations, revealed a significant reduction in Th17 cells, and no alterations in Th2, Th1 or Treg cells. Thus, our data reveal that while MelLec is required to control pulmonary fungal burden, the inflammatory responses mediated by this receptor negatively impact the animal welfare in this allergic model.


Asunto(s)
Aspergillus fumigatus/patogenicidad , Asma/etiología , Lectinas Tipo C/fisiología , Animales , Asma/inmunología , Bronquios/patología , Citocinas/biosíntesis , Melaninas/fisiología , Ratones , Ratones Endogámicos C57BL , Células Th17/inmunología
17.
Microorganisms ; 8(7)2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32708909

RESUMEN

The discovery of liquid water at several locations in the solar system raises the possibility that microbial life may have evolved outside Earth and as such could be accidently introduced into the Earth's ecosystem. Unusual sugars or amino acids, like non-proteinogenic isovaline and α-aminoisobutyric acid that are vanishingly rare or absent from life forms on Earth, have been found in high abundance on non-terrestrial carbonaceous meteorites. It is therefore conceivable that exo-microorganisms might contain proteins that include these rare amino acids. We therefore asked whether the mammalian immune system would be able to recognize and induce appropriate immune responses to putative proteinaceous antigens that include these rare amino acids. To address this, we synthesised peptide antigens based on a backbone of ovalbumin and introduced isovaline and α-aminoisobutyric acid residues and demonstrated that these peptides can promote naïve OT-I cell activation and proliferation, but did so less efficiently than the canonical peptides. This is relevant to the biosecurity of missions that may retrieve samples from exoplanets and moons that have conditions that may be permissive for life, suggesting that accidental contamination and exposure to exo-microorganisms with such distinct proteomes might pose an immunological challenge.

18.
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
20.
Nat Commun ; 8(1): 719, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28959012

RESUMEN

Interleukin 35 (IL-35) is a heterodimeric cytokine composed of IL-12p35 and Ebi3 subunits. IL-35 suppresses autoimmune diseases while preventing host defense to infection and promoting tumor growth and metastasis by converting resting B and T cells into IL-10-producing and IL-35-producing regulatory B (Breg) and T (Treg) cells. Despite sharing the IL-12p35 subunit, IL-12 (IL-12p35/IL-12p40) promotes inflammatory responses whereas IL-35 (IL-12p35/Ebi3) induces regulatory responses, suggesting that IL-12p35 may have unknown intrinsic immune-regulatory functions regulated by its heterodimeric partner. Here we show that the IL-12p35 subunit has immunoregulatory functions hitherto attributed to IL-35. IL-12p35 suppresses lymphocyte proliferation, induces expansion of IL-10-expressing and IL-35-expressing B cells and ameliorates autoimmune uveitis in mice by antagonizing pathogenic Th17 responses. Recapitulation of essential immunosuppressive activities of IL-35 indicates that IL-12p35 may be utilized for in vivo expansion of Breg cells and autologous Breg cell immunotherapy. Furthermore, our uveitis data suggest that intrinsic immunoregulatory activities of other single chain IL-12 subunits might be exploited to treat other autoimmune diseases.IL-12p35 is common to IL-35 and IL-12, which have opposing effects on inflammation. Here the authors show that the IL-12p35 subunit induces regulatory B cells and can be used therapeutically to limit autoimmune uveitis in mice.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Linfocitos B Reguladores/metabolismo , Interleucina-10/metabolismo , Subunidad p35 de la Interleucina-12/metabolismo , Animales , Proliferación Celular , Terapia de Inmunosupresión , Subunidad beta 2 del Receptor de Interleucina-12/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Multimerización de Proteína , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Uveítis/inmunología , Uveítis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA