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











Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 34(4): 108673, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33503414

RESUMEN

Indoleamine 2,3-dioxygenases (IDOs) degrade l-tryptophan to kynurenines and drive the de novo synthesis of nicotinamide adenine dinucleotide. Unsurprisingly, various invertebrates, vertebrates, and even fungi produce IDO. In mammals, IDO1 also serves as a homeostatic regulator, modulating immune response to infection via local tryptophan deprivation, active catabolite production, and non-enzymatic cell signaling. Whether fungal Idos have pleiotropic functions that impact on host-fungal physiology is unclear. Here, we show that Aspergillus fumigatus possesses three ido genes that are expressed under conditions of hypoxia or tryptophan abundance. Loss of these genes results in increased fungal pathogenicity and inflammation in a mouse model of aspergillosis, driven by an alternative tryptophan degradation pathway to indole derivatives and the host aryl hydrocarbon receptor. Fungal tryptophan metabolic pathways thus cooperate with the host xenobiotic response to shape host-microbe interactions in local tissue microenvironments.


Asunto(s)
Aspergilosis/fisiopatología , Aspergillus fumigatus/patogenicidad , Triptófano/metabolismo , Animales , Humanos , Ratones
2.
Front Immunol ; 10: 890, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31134053

RESUMEN

Aspergillus is the causative agent of human diseases ranging from asthma to invasive infection. Genetic and environmental factors are crucial in regulating the interaction between the host and Aspergillus. The role played by the enzyme indoleamine 2,3-dioxygenase 1 (IDO1), which catalyzes the first and rate-limiting step of tryptophan catabolism along the kynurenine pathway, is increasingly being recognized, but whether and how genetic variation of IDO1 influences the risk of aspergillosis in susceptible patients is incompletely understood. In addition, whether the closely related protein IDO2 plays a similar role remains unexplored. In the present study, we performed genetic association studies in two different cohorts of susceptible patients [cystic fibrosis (CF) patients and recipients of hematopoietic stem cell transplantation (HSCT)], and identified IDO1 polymorphisms that associate with the risk of infection in both cohorts. By using human bronchial epithelial cells and PBMC from CF and HSCT patients, respectively, we could show that the IDO1 polymorphisms appeared to down-modulate IDO1 expression and function in response to IFNγ or Aspergillus conidia, and to associate with an increased inflammatory response. In contrast, IDO2 polymorphisms, including variants known to profoundly affect protein expression and function, were differently associated with the risk of aspergillosis in the two cohorts of patients as no association was found in CF patients as opposed to recipients of HSCT. By resorting to a murine model of bone marrow transplantation, we could show that the absence of IDO2 more severely affected fungal burden and lung pathology upon infection with Aspergillus as compared to IDO1, and this effect appeared to be linked to a deficit in the antifungal effector phagocytic activity. Thus, our study confirms and extends the role of IDO1 in the response to Aspergillus, and shed light on the possible involvement of IDO2 in specific clinical settings.


Asunto(s)
Aspergilosis/genética , Predisposición Genética a la Enfermedad/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Polimorfismo Genético , Adolescente , Adulto , Animales , Aspergilosis/enzimología , Aspergilosis/microbiología , Aspergillus/fisiología , Niño , Preescolar , Fibrosis Quística/enzimología , Fibrosis Quística/genética , Fibrosis Quística/microbiología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Femenino , Trasplante de Células Madre Hematopoyéticas/estadística & datos numéricos , Interacciones Huésped-Patógeno , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/microbiología , Masculino , Ratones , Adulto Joven
3.
Cell Rep ; 20(7): 1667-1680, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813677

RESUMEN

The interleukin 17 (IL-17) cytokine and receptor family is central to antimicrobial resistance and inflammation in the lung. Mice lacking IL-17A, IL-17F, or the IL-17RA subunit were compared with wild-type mice for susceptibility to airway inflammation in models of infection and allergy. Signaling through IL-17RA was required for efficient microbial clearance and prevention of allergy; in the absence of IL-17RA, signaling through IL-17RC on epithelial cells, predominantly by IL-17F, significantly exacerbated lower airway Aspergillus or Pseudomonas infection and allergic airway inflammation. In contrast, following infection with the upper respiratory pathogen Staphylococcus aureus, the IL-17F/IL-17RC axis mediated protection. Thus, IL-17A and IL-17F exert distinct biological effects during pulmonary infection; the IL-17F/IL-17RC signaling axis has the potential to significantly worsen pathogen-associated inflammation of the lower respiratory tract in particular, and should be investigated further as a therapeutic target for treating pathological inflammation in the lung.


Asunto(s)
Aspergilosis/inmunología , Hipersensibilidad/inmunología , Interleucina-17/inmunología , Infecciones por Pseudomonas/inmunología , Receptores de Interleucina-17/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Aspergilosis/genética , Aspergilosis/microbiología , Aspergilosis/patología , Aspergillus/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Hipersensibilidad/genética , Hipersensibilidad/microbiología , Hipersensibilidad/patología , Interleucina-17/deficiencia , Interleucina-17/genética , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Pseudomonas/inmunología , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Receptores de Interleucina-17/deficiencia , Receptores de Interleucina-17/genética , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Transducción de Señal , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/inmunología
4.
Nat Commun ; 8: 14017, 2017 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-28090087

RESUMEN

T helper 9 (Th9) cells contribute to lung inflammation and allergy as sources of interleukin-9 (IL-9). However, the mechanisms by which IL-9/Th9 mediate immunopathology in the lung are unknown. Here we report an IL-9-driven positive feedback loop that reinforces allergic inflammation. We show that IL-9 increases IL-2 production by mast cells, which leads to expansion of CD25+ type 2 innate lymphoid cells (ILC2) and subsequent activation of Th9 cells. Blocking IL-9 or inhibiting CD117 (c-Kit) signalling counteracts the pathogenic effect of the described IL-9-mast cell-IL-2 signalling axis. Overproduction of IL-9 is observed in expectorates from cystic fibrosis (CF) patients, and a sex-specific variant of IL-9 is predictive of allergic reactions in female patients. Our results suggest that blocking IL-9 may be a therapeutic strategy to ameliorate inflammation associated with microbial colonization in the lung, and offers a plausible explanation for gender differences in clinical outcomes of patients with CF.


Asunto(s)
Fibrosis Quística/inmunología , Linfocitos/inmunología , Mastocitos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Adolescente , Adulto , Animales , Niño , Preescolar , Fibrosis Quística/genética , Femenino , Humanos , Inmunidad Innata , Lactante , Interleucina-9/inmunología , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/inmunología , Adulto Joven
5.
Cell Host Microbe ; 20(6): 744-757, 2016 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-27889463

RESUMEN

Defects in a form of noncanonical autophagy, known as LC3-associated phagocytosis (LAP), lead to increased inflammatory pathology during fungal infection. Although LAP contributes to fungal degradation, the molecular mechanisms underlying LAP-mediated modulation of inflammation are unknown. We describe a mechanism by which inflammation is regulated during LAP through the death-associated protein kinase 1 (DAPK1). The ATF6/C/EBP-ß/DAPK1 axis activated by IFN-γ not only mediates LAP to Aspergillus fumigatus but also concomitantly inhibits Nod-like receptor protein 3 (NLRP3) activation and restrains pathogenic inflammation. In mouse models and patient samples of chronic granulomatous disease, which exhibit defective autophagy and increased inflammasome activity, IFN-γ restores reduced DAPK1 activity and dampens fungal growth. Additionally, in a cohort of hematopoietic stem cell-transplanted patients, a genetic DAPK1 deficiency is associated with increased inflammation and heightened aspergillosis susceptibility. Thus, DAPK1 is a potential drugable player in regulating the inflammatory response during fungal clearance initiated by IFN-γ.


Asunto(s)
Autofagia/efectos de los fármacos , Autofagia/fisiología , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Hongos/metabolismo , Inflamación/metabolismo , Interferón gamma/metabolismo , Animales , Aspergilosis/microbiología , Aspergillus fumigatus/metabolismo , Línea Celular , Proteínas Quinasas Asociadas a Muerte Celular/efectos de los fármacos , Proteínas Quinasas Asociadas a Muerte Celular/genética , Hongos/patogenicidad , Regulación Fúngica de la Expresión Génica , Enfermedad Granulomatosa Crónica/microbiología , Humanos , Interferón gamma/farmacología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Proteínas NLR/efectos de los fármacos , Fagocitosis , Fagosomas , Células RAW 264.7/efectos de los fármacos , Células RAW 264.7/microbiología , Esporas Fúngicas/metabolismo
6.
Nat Commun ; 7: 10791, 2016 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-26972847

RESUMEN

Dysregulated inflammasome activation contributes to respiratory infections and pathologic airway inflammation. Through basic and translational approaches involving murine models and human genetic epidemiology, we show here the importance of the different inflammasomes in regulating inflammatory responses in mice and humans with cystic fibrosis (CF), a life-threatening disorder of the lungs and digestive system. While both contributing to pathogen clearance, NLRP3 more than NLRC4 contributes to deleterious inflammatory responses in CF and correlates with defective NLRC4-dependent IL-1Ra production. Disease susceptibility in mice and microbial colonization in humans occurs in conditions of genetic deficiency of NLRC4 or IL-1Ra and can be rescued by administration of the recombinant IL-1Ra, anakinra. These results indicate that pathogenic NLRP3 activity in CF could be negatively regulated by IL-1Ra and provide a proof-of-concept evidence that inflammasomes are potential targets to limit the pathological consequences of microbial colonization in CF.


Asunto(s)
Aspergilosis/inmunología , Fibrosis Quística/inmunología , Citocinas/genética , Células Epiteliales/inmunología , Inflamasomas/genética , Proteína Antagonista del Receptor de Interleucina 1/genética , Pulmón/metabolismo , Infecciones por Pseudomonas/inmunología , Adolescente , Adulto , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Aspergillus fumigatus , Autofagia/genética , Autofagia/inmunología , Western Blotting , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular , Niño , Preescolar , Fibrosis Quística/genética , Fibrosis Quística/microbiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Lactante , Inflamasomas/inmunología , Inflamación , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR , Polimorfismo de Nucleótido Simple , Pseudomonas aeruginosa , Mucosa Respiratoria/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
7.
Front Immunol ; 5: 506, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25360137

RESUMEN

Fungal diseases represent an important paradigm in immunology since they can result from either the lack of recognition or over-activation of the inflammatory response. Current understanding of the pathophysiology underlying fungal infections and diseases highlights the multiple cell populations and cell-signaling pathways involved in these conditions. A systems biology approach that integrates investigations of immunity at the systems-level is required to generate novel insights into this complexity and to decipher the dynamics of the host-fungus interaction. It is becoming clear that a three-way interaction between the host, microbiota, and fungi dictates the types of host-fungus relationship. Tryptophan metabolism helps support this interaction, being exploited by the mammalian host and commensals to increase fitness in response to fungi via resistance and tolerance mechanisms of antifungal immunity. The cellular and molecular mechanisms that provide immune homeostasis with the fungal biota and its possible rupture in fungal infections and diseases will be discussed within the expanding role of antifungal Th cell responses.

8.
Am J Respir Crit Care Med ; 188(11): 1338-50, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24127697

RESUMEN

RATIONALE: Hypoxia regulates the inflammatory-antiinflammatory balance by the receptor for advanced glycation end products (RAGE), a versatile sensor of damage-associated molecular patterns. The multiligand nature of RAGE places this receptor in the midst of chronic inflammatory diseases. OBJECTIVES: To characterize the impact of the hypoxia-RAGE pathway on pathogenic airway inflammation preventing effective pathogen clearance in cystic fibrosis (CF) and elucidate the potential role of this danger signal in pathogenesis and therapy of lung inflammation. METHODS: We used in vivo and in vitro models to study the impact of hypoxia on RAGE expression and activity in human and murine CF, the nature of the RAGE ligand, and the impact of RAGE on lung inflammation and antimicrobial resistance in fungal and bacterial pneumonia. MEASUREMENTS AND MAIN RESULTS: Sustained expression of RAGE and its ligand S100B was observed in murine lung and human epithelial cells and exerted a proximal role in promoting inflammation in murine and human CF, as revealed by functional studies and analysis of the genetic variability of AGER in patients with CF. Both hypoxia and infections contributed to the sustained activation of the S100B-RAGE pathway, being RAGE up-regulated by hypoxia and S100B by infection by Toll-like receptors. Inhibiting the RAGE pathway in vivo with soluble (s) RAGE reduced pathogen load and inflammation in experimental CF, whereas sRAGE production was defective in patients with CF. CONCLUSIONS: A causal link between hyperactivation of RAGE and inflammation in CF has been observed, such that targeting pathogenic inflammation alleviated inflammation in CF and measurement of sRAGE levels could be a useful biomarker for RAGE-dependent inflammation in patients with CF.


Asunto(s)
Fibrosis Quística/patología , Hipoxia/patología , Mediadores de Inflamación/fisiología , Neumonía/etiología , Receptores Inmunológicos/inmunología , Animales , Aspergilosis/microbiología , Biomarcadores , Western Blotting , Fibrosis Quística/complicaciones , Fibrosis Quística/microbiología , Farmacorresistencia Microbiana , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Hipoxia/complicaciones , Hipoxia/etiología , Italia , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neumonía/tratamiento farmacológico , Neumonía/microbiología , Infecciones por Pseudomonas/microbiología , Receptor para Productos Finales de Glicación Avanzada , Mucosa Respiratoria , Técnicas de Cultivo de Tejidos , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA