Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34168077

RESUMEN

Dual oxidase 1 (DUOX1) is an NADPH oxidase that is highly expre-ssed in respiratory epithelial cells and produces H2O2 in the airway lumen. While a line of prior in vitro observations suggested that DUOX1 works in partnership with an airway peroxidase, lactoperoxidase (LPO), to produce antimicrobial hypothiocyanite (OSCN-) in the airways, the in vivo role of DUOX1 in mammalian organisms has remained unproven to date. Here, we show that Duox1 promotes antiviral innate immunity in vivo. Upon influenza airway challenge, Duox1-/- mice have enhanced mortality, morbidity, and impaired lung viral clearance. Duox1 increases the airway levels of several cytokines (IL-1ß, IL-2, CCL1, CCL3, CCL11, CCL19, CCL20, CCL27, CXCL5, and CXCL11), contributes to innate immune cell recruitment, and affects epithelial apoptosis in the airways. In primary human tracheobronchial epithelial cells, OSCN- is generated by LPO using DUOX1-derived H2O2 and inactivates several influenza strains in vitro. We also show that OSCN- diminishes influenza replication and viral RNA synthesis in infected host cells that is inhibited by the H2O2 scavenger catalase. Binding of the influenza virus to host cells and viral entry are both reduced by OSCN- in an H2O2-dependent manner in vitro. OSCN- does not affect the neuraminidase activity or morphology of the influenza virus. Overall, this antiviral function of Duox1 identifies an in vivo role of this gene, defines the steps in the infection cycle targeted by OSCN-, and proposes that boosting this mechanism in vivo can have therapeutic potential in treating viral infections.


Asunto(s)
Antivirales/inmunología , Oxidasas Duales/metabolismo , Inmunidad Innata , Animales , Apoptosis , Bronquios/patología , Bronquios/virología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/patología , Humanos , Peróxido de Hidrógeno/metabolismo , Gripe Humana/inmunología , Gripe Humana/patología , Gripe Humana/virología , Lactoperoxidasa/metabolismo , Ratones , Neuraminidasa/química , Neuraminidasa/metabolismo , Orthomyxoviridae/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Proteolisis , ARN Viral/metabolismo , Tiocianatos , Proteínas Virales/química , Proteínas Virales/metabolismo , Inactivación de Virus , Internalización del Virus , Replicación Viral
2.
Proc Natl Acad Sci U S A ; 117(3): 1762-1771, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31900358

RESUMEN

The pathological hallmark of synucleinopathies, including Lewy body dementia and Parkinson's disease (PD), is the presence of Lewy bodies, which are primarily composed of intracellular inclusions of misfolded α-synuclein (α-syn) among other proteins. α-Syn is found in extracellular biological fluids in PD patients and has been implicated in modulating immune responses in the central nervous system (CNS) and the periphery. Natural killer (NK) cells are innate effector lymphocytes that are present in the CNS in homeostatic and pathological conditions. NK cell numbers are increased in the blood of PD patients and their activity is associated with disease severity; however, the role of NK cells in the context of α-synucleinopathies has never been explored. Here, we show that human NK cells can efficiently internalize and degrade α-syn aggregates via the endosomal/lysosomal pathway. We demonstrate that α-syn aggregates attenuate NK cell cytotoxicity in a dose-dependent manner and decrease the release of the proinflammatory cytokine, IFN-γ. To address the role of NK cells in PD pathogenesis, NK cell function was investigated in a preformed fibril α-syn-induced mouse PD model. Our studies demonstrate that in vivo depletion of NK cells in a preclinical mouse PD model resulted in exacerbated motor deficits and increased phosphorylated α-syn deposits. Collectively, our data provide a role of NK cells in modulating synuclein pathology and motor symptoms in a preclinical mouse model of PD, which could be developed into a therapeutic for PD and other synucleinopathies.


Asunto(s)
Células Asesinas Naturales/metabolismo , Sinucleinopatías/metabolismo , Sinucleínas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Sistema Nervioso Central/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Enfermedad de Parkinson/metabolismo , Sinucleinopatías/genética , Sinucleinopatías/patología
3.
Infect Immun ; 90(5): e0068221, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35499339

RESUMEN

Human intelectin-1 (hIntL-1) is a secreted glycoprotein capable of binding exocyclic 1,2-diols within surface glycans of human pathogens such as Streptococcus pneumoniae, Vibrio cholerae, and Helicobacter pylori. For the latter, lectin binding was shown to cause bacterial agglutination and increased phagocytosis, suggesting a role for hIntL-1 in pathogen surveillance. In this study, we investigated the interactions between hIntL-1 and S. pneumoniae, the leading cause of bacterial pneumonia. We show that hIntL-1 also agglutinates S. pneumoniae serotype 43, which displays an exocyclic 1,2-diol moiety in its capsular polysaccharide but is unable to kill in a complement-dependent manner or to promote bacterial killing by peripheral blood mononuclear cells. In contrast, hIntL-1 not only significantly increases serotype-specific S. pneumoniae killing by neutrophils but also enhances the attachment of these bacteria to A549 lung epithelial cells. Taken together, our results suggest that hIntL-1 participates in host surveillance through microbe sequestration and enhanced targeting to neutrophils.


Asunto(s)
Neutrófilos , Streptococcus pneumoniae , Citocinas/metabolismo , Proteínas Ligadas a GPI/metabolismo , Humanos , Lectinas/metabolismo , Leucocitos Mononucleares/metabolismo , Neutrófilos/metabolismo , Fagocitosis , Polisacáridos/metabolismo , Serogrupo , Streptococcus pneumoniae/metabolismo
4.
Emerg Infect Dis ; 27(8): 2107-2116, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34286682

RESUMEN

Conventional pertussis animal models deliver hundreds of thousands of Bordetella pertussis bacteria deep into the lungs, rapidly inducing severe pneumonic pathology and a robust immune response. However, human infections usually begin with colonization and growth in the upper respiratory tract. We inoculated only the nasopharynx of mice to explore the course of infection in a more natural exposure model. Nasopharyngeal colonization resulted in robust growth in the upper respiratory tract but elicited little immune response, enabling prolonged and persistent infection. Immunization with human acellular pertussis vaccine, which prevents severe lung infections in the conventional pneumonic infection model, had little effect on nasopharyngeal colonization. Our infection model revealed that B. pertussis can efficiently colonize the mouse nasopharynx, grow and spread within and between respiratory organs, evade robust host immunity, and persist for months. This experimental approach can measure aspects of the infection processes not observed in the conventional pneumonic infection model.


Asunto(s)
Infecciones por Bordetella , Tos Ferina , Animales , Bordetella pertussis , Evasión Inmune , Ratones , Nasofaringe , Vacuna contra la Tos Ferina , Tos Ferina/prevención & control
5.
BMC Immunol ; 22(1): 7, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33478382

RESUMEN

BACKGROUND: Neutrophils are key components of the exacerbated inflammation and tissue damage in cystic fibrosis (CF) airways. Neutrophil extracellular traps (NETs) trap and kill extracellular pathogens. While NETs are abundant in the airways of CF patients and have been hypothesized to contribute to lung damage in CF, the in vivo role of NETs remains controversial, partially due to lack of appropriate animal models. The goal of this study was to detect NETs and to further characterize neutrophil-mediated inflammation in the airways of mice overexpressing the epithelial sodium channel (ßENaC-Tg mice on C57BL/6 background) in their lung with CF-like airway disease, in the absence of any apparent bacterial infections. METHODS: Histology scoring of lung tissues, flow cytometry, multiplex ELISA, immunohistochemistry and immunofluorescence were used to characterize NETs and the airway environment in uninfected, ßENaC-Tg mice at 6 and 8 weeks of age, the most chronic time points so far studied in this model. RESULTS: Excessive neutrophilic infiltration characterized the lungs of uninfected, ßENaC-Tg mice at 6 and 8 weeks of age. The bronchoalveolar lavage fluid (BALF) of ßENaC-Tg mice contains increased levels of CF-associated cytokines and chemokines: KC, MIP-1α/ß, MCP-1, G-CSF, IL-5, and IL-6. The BALF of ßENaC-Tg mice contain MPO-DNA complexes, indicative of the presence of NETs. Immunofluorescence and flow cytometry of BALF neutrophils and lung tissues demonstrated increased histone citrullination, a NET-specific marker, in ßENaC-Tg mice. CONCLUSIONS: NETs are detected in the airways of ßENaC-Tg mice, in the absence of bacterial infections. These data demonstrate the usefulness of the ßENaC-Tg mouse to serve as a model for studying the role of NETs in chronic CF airway inflammation.


Asunto(s)
Fibrosis Quística/inmunología , Canales Epiteliales de Sodio/inmunología , Trampas Extracelulares/inmunología , Pulmón/inmunología , Neutrófilos/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Citrulinación , Fibrosis Quística/genética , Fibrosis Quística/patología , Citocinas/metabolismo , Canales Epiteliales de Sodio/genética , Histonas/metabolismo , Inflamación , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infiltración Neutrófila
6.
J Immunol ; 198(1): 428-442, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27903742

RESUMEN

Human neutrophils (polymorphonuclear leukocytes [PMNs]) generate inflammatory responses within the joints of gout patients upon encountering monosodium urate (MSU) crystals. Neutrophil extracellular traps (NETs) are found abundantly in the synovial fluid of gout patients. The detailed mechanism of MSU crystal-induced NET formation remains unknown. Our goal was to shed light on possible roles of purinergic signaling and neutrophil migration in mediating NET formation induced by MSU crystals. Interaction of human neutrophils with MSU crystals was evaluated by high-throughput live imaging using confocal microscopy. We quantitated NET levels in gout synovial fluid supernatants and detected enzymatically active neutrophil primary granule enzymes, myeloperoxidase, and human neutrophil elastase. Suramin and PPADS, general P2Y receptor blockers, and MRS2578, an inhibitor of the purinergic P2Y6 receptor, blocked NET formation triggered by MSU crystals. AR-C25118925XX (P2Y2 antagonist) did not inhibit MSU crystal-stimulated NET release. Live imaging of PMNs showed that MRS2578 represses neutrophil migration and blocked characteristic formation of MSU crystal-NET aggregates called aggregated NETs. Interestingly, the store-operated calcium entry channel inhibitor (SK&F96365) also reduced MSU crystal-induced NET release. Our results indicate that the P2Y6/store-operated calcium entry/IL-8 axis is involved in MSU crystal-induced aggregated NET formation, but MRS2578 could have additional effects affecting PMN migration. The work presented in the present study could lead to a better understanding of gouty joint inflammation and help improve the treatment and care of gout patients.


Asunto(s)
Trampas Extracelulares/efectos de los fármacos , Isotiocianatos/farmacología , Activación Neutrófila/efectos de los fármacos , Receptores Purinérgicos P2/metabolismo , Tiourea/análogos & derivados , Ácido Úrico/inmunología , Quimiotaxis de Leucocito/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Trampas Extracelulares/inmunología , Técnica del Anticuerpo Fluorescente , Gota/inmunología , Gota/metabolismo , Gota/patología , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas In Vitro , Microscopía Confocal , Activación Neutrófila/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Tiourea/farmacología
7.
PLoS Pathog ; 12(11): e1005987, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27855208

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen causing severe infections often characterized by robust neutrophilic infiltration. Neutrophils provide the first line of defense against P. aeruginosa. Aside from their defense conferred by phagocytic activity, neutrophils also release neutrophil extracellular traps (NETs) to immobilize bacteria. Although NET formation is an important antimicrobial process, the details of its mechanism are largely unknown. The identity of the main components of P. aeruginosa responsible for triggering NET formation is unclear. In this study, our focus was to identify the main bacterial factors mediating NET formation and to gain insight into the underlying mechanism. We found that P. aeruginosa in its exponential growth phase promoted strong NET formation in human neutrophils while its NET-inducing ability dramatically decreased at later stages of bacterial growth. We identified the flagellum as the primary component of P. aeruginosa responsible for inducing NET extrusion as flagellum-deficient bacteria remained seriously impaired in triggering NET formation. Purified P. aeruginosa flagellin, the monomeric component of the flagellum, does not stimulate NET formation in human neutrophils. P. aeruginosa-induced NET formation is independent of the flagellum-sensing receptors TLR5 and NLRC4 in both human and mouse neutrophils. Interestingly, we found that flagellar motility, not flagellum binding to neutrophils per se, mediates NET release induced by flagellated bacteria. Immotile, flagellar motor-deficient bacterial strains producing paralyzed flagella did not induce NET formation. Forced contact between immotile P. aeruginosa and neutrophils restored their NET-inducing ability. Both the motAB and motCD genetic loci encoding flagellar motor genes contribute to maximal NET release; however the motCD genes play a more important role. Phagocytosis of P. aeruginosa and superoxide production by neutrophils were also largely dependent upon a functional flagellum. Taken together, the flagellum is herein presented for the first time as the main organelle of planktonic bacteria responsible for mediating NET release. Furthermore, flagellar motility, rather than binding of the flagellum to flagellum-sensing receptors on host cells, is required for P. aeruginosa to induce NET release.


Asunto(s)
Movimiento Celular/inmunología , Trampas Extracelulares/inmunología , Flagelos/inmunología , Neutrófilos/inmunología , Infecciones por Pseudomonas/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Pseudomonas aeruginosa/inmunología
8.
Rheumatology (Oxford) ; 56(4): 644-653, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28013195

RESUMEN

Objectives: The aim was to investigate the effects of nicotine on neutrophil extracellular traps (NETs) formation in current and non-smokers and on a murine model of RA. Methods: We compared spontaneous and phorbol 12-myristate 13-acetate-induced NETosis between current and non-smokers by DNA release binding. Nicotine-induced NETosis from non-smokers was assessed by DNA release binding, NET-specific (myeloperoxidase (MPO)-DNA complex) ELISA and real-time fluorescence microscopy. We also used immunofluorescent staining to detect nicotinic acetylcholine receptors (nAChRs) on neutrophils and performed a functional analysis to assess the role of nAChRs in nicotine-induced NETosis. Finally, we investigated the effects of systemic nicotine exposure on arthritis severity and NETosis in the CIA mouse model. Results: Neutrophils derived from current smokers displayed elevated levels of spontaneous and phorbol 12-myristate 13-acetate-induced NETosis. Nicotine induced dose-dependent NETosis in ex vivo neutrophils from healthy non-smokers, and co-incubation with ACPA-immune complexes or TNF-α facilitated a synergistic effect on NETosis. Real-time fluorescence microscopy revealed robust formation of NET-like structures in nicotine-exposed neutrophils. Immunofluorescent staining demonstrated the presence of the α7 subunit of the nAChR on neutrophils. Stimulation of neutrophils with an α7-specific nAChR agonist induced NETosis, whereas pretreatment with an nAChR antagonist attenuated nicotine-induced NETosis. Nicotine administration to mice with CIA exacerbated inflammatory arthritis, with higher plasma levels of NET-associated MPO-DNA complex. Conclusion: We demonstrate that nicotine is a potent inducer of NETosis, which may play an important role in accelerating arthritis in the CIA model. This study generates awareness of and the mechanisms by which nicotine-containing products, including e-cigarettes, may have deleterious effects on patients with RA.


Asunto(s)
Artritis Reumatoide/etiología , Trampas Extracelulares/efectos de los fármacos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Animales , Artritis Experimental/etiología , Cartílago/fisiología , Relación Dosis-Respuesta a Droga , Sistemas Electrónicos de Liberación de Nicotina , Ensayo de Inmunoadsorción Enzimática , Humanos , Infusiones Subcutáneas , Masculino , Ratones Endogámicos DBA , Neutrófilos/efectos de los fármacos , Nicotina/administración & dosificación , Agonistas Nicotínicos/administración & dosificación , Peroxidasa/metabolismo , Fumar/efectos adversos , Acetato de Tetradecanoilforbol/farmacología
9.
Inflamm Res ; 66(3): 227-237, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27853847

RESUMEN

OBJECTIVE AND DESIGN: Arthritic gout is caused by joint inflammation triggered by the damaging effects of monosodium uric acid (MSU) crystal accumulation in the synovial space. Neutrophils play a major role in mediating joint inflammation in gout. Along with neutrophils, other immune cells, such as macrophages, are present in inflamed joints and contribute to gout pathogenesis. Neutrophils form neutrophil extracellular traps (NETs) in response to MSU crystals. In the presence of MSU crystals, macrophages release IL-1ß, a cytokine crucial to initiate gout pathogenesis and neutrophil recruitment. Our research investigated interactions between human macrophages and neutrophils in an in vitro model system and asked how macrophages affect NET formation stimulated by MSU crystals. MATERIALS OR SUBJECTS: Human neutrophils and PBMCs were isolated from peripheral blood of healthy volunteers. PBMCs were differentiated into macrophages in vitro using human M-CSF. TREATMENT: Human neutrophils were pretreated with macrophage-conditioned media, neutrophil-conditioned media, recombinant human IL-1ß or anakinra prior to stimulation by MSU crystals. METHOD: Interaction of neutrophils with MSU crystals was evaluated by live imaging using confocal microscopy. The presence of myeloperoxidase (MPO) and neutrophil elastase (NE) was measured by ELISA. NET formation was quantitated by Sytox Orange-based extracellular DNA release assay and NE-DNA ELISA. AggNET formation was assessed by macroscopic evaluation. RESULTS: We found that crystal- and cell-free supernatants of macrophages stimulated with MSU crystals promote MSU crystal-stimulated NET formation in human neutrophils. This observation was confirmed by additional assays measuring the release of MPO, NE, and the enzymatic activity of NE. MSU crystal-induced NET formation remained unchanged when neutrophil supernatants were tested. IL-1ß is a crucial cytokine orchestrating the onset of inflammation in gout and is known to be released in large amounts from macrophages following MSU crystal stimulation. We found that recombinant IL-1ß strongly promoted MSU crystal-induced NET formation in human neutrophils. Interestingly, IL-1ß alone did not induce any NET release. We also found that clinical grade anakinra, an IL-1 receptor blocker, strongly reduced the NETosis-enhancing effect of macrophage supernatants indicating that IL-1ß is mainly responsible for this effect. CONCLUSIONS: Macrophage-derived IL-1ß enhances MSU crystal-induced NET release in neutrophils. We identified a new mechanism by which macrophages and IL-1ß affect neutrophil functions, and could contribute to the inflammatory conditions present in gout. Our results also revealed a new anti-inflammatory mechanism of anakinra.


Asunto(s)
Trampas Extracelulares , Interleucina-1beta/inmunología , Macrófagos/inmunología , Neutrófilos/inmunología , Ácido Úrico/farmacología , Gota/inmunología , Humanos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-8/inmunología , Elastasa de Leucocito/inmunología , Macrófagos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Peroxidasa/inmunología
10.
Inflamm Res ; 65(1): 71-80, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26608498

RESUMEN

OBJECTIVE AND DESIGN: Our aim was to study whether an extracellular, oxidative antimicrobial mechanism inherent to tracheal epithelial cells is capable of inactivating influenza H1N2 virus. MATERIAL OR SUBJECTS: Epithelial cells were isolated from tracheas of male Sprague-Dawley rats. Both primary human and rat tracheobronchial epithelial cells were differentiated in air-liquid interface cultures. TREATMENT: A/swine/Illinois/02860/09 (swH1N2) influenza A virions were added to the apical side of airway cells for 1 h in the presence or absence of lactoperoxidase or thiocyanate. METHODS: Characterization of rat epithelial cells (morphology, Duox expression) occurred via western blotting, PCR, hydrogen peroxide production measurement and histology. The number of viable virions was determined by plaque assays. Statistical difference of the results was analyzed by ANOVA and Tukey's test. RESULTS: Our data show that rat tracheobronchial epithelial cells develop a differentiated, polarized monolayer with high transepithelial electrical resistance, mucin production and expression of dual oxidases. Influenza A virions are inactivated by human and rat epithelial cells via a dual oxidase-, lactoperoxidase- and thiocyanate-dependent mechanism. CONCLUSIONS: Differentiated air-liquid interface cultures of rat tracheal epithelial cells provide a novel model to study airway epithelium-influenza interactions. The dual oxidase/lactoperoxidase/thiocyanate extracellular oxidative system producing hypothiocyanite is a fast and potent anti-influenza mechanism inactivating H1N2 viruses prior to infection of the epithelium.


Asunto(s)
Células Epiteliales/metabolismo , Subtipo H1N2 del Virus de la Influenza A/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Tiocianatos/metabolismo , Animales , Perros , Humanos , Peróxido de Hidrógeno/metabolismo , Lactoperoxidasa/metabolismo , Células de Riñón Canino Madin Darby , Masculino , Mucinas/biosíntesis , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Mucosa Respiratoria/citología
11.
J Immunol ; 192(10): 4728-38, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24740504

RESUMEN

Cystic fibrosis (CF) airways are characterized by bacterial infections, excess mucus production, and robust neutrophil recruitment. The main CF airway pathogen is Pseudomonas aeruginosa. Neutrophils are not capable of clearing the infection. Neutrophil primary granule components, myeloperoxidase (MPO) and human neutrophil elastase (HNE), are inflammatory markers in CF airways, and their increased levels are associated with poor lung function. Identifying the mechanism of MPO and HNE release from neutrophils is of high clinical relevance for CF. In this article, we show that human neutrophils release large amounts of neutrophil extracellular traps (NETs) in the presence of P. aeruginosa. Bacteria are entangled in NETs and colocalize with extracellular DNA. MPO, HNE, and citrullinated histone H4 are all associated with DNA in Pseudomonas-triggered NETs. Both laboratory standard strains and CF isolates of P. aeruginosa induce DNA, MPO, and HNE release from human neutrophils. The increase in peroxidase activity of neutrophil supernatants after Pseudomonas exposure indicates that enzymatically active MPO is released. P. aeruginosa induces a robust respiratory burst in neutrophils that is required for extracellular DNA release. Inhibition of the cytoskeleton prevents Pseudomonas-initiated superoxide production and DNA release. NADPH oxidase inhibition suppresses Pseudomonas-induced release of active MPO and HNE. Blocking MEK/ERK signaling results in only minimal inhibition of DNA release induced by Pseudomonas. Our data describe in vitro details of DNA, MPO, and HNE release from neutrophils activated by P. aeruginosa. We propose that Pseudomonas-induced NET formation is an important mechanism contributing to inflammatory conditions characteristic of CF airways.


Asunto(s)
Fibrosis Quística/inmunología , ADN/inmunología , NADPH Oxidasas/inmunología , Neutrófilos/inmunología , Pseudomonas aeruginosa/inmunología , Superóxidos/inmunología , Adolescente , Adulto , Biomarcadores/metabolismo , Fibrosis Quística/metabolismo , Fibrosis Quística/microbiología , Fibrosis Quística/patología , ADN/metabolismo , Femenino , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Sistema de Señalización de MAP Quinasas/inmunología , Masculino , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patología , Peroxidasa/inmunología , Peroxidasa/metabolismo , Superóxidos/metabolismo
12.
J Biol Chem ; 289(52): 36089-100, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25378393

RESUMEN

Signal transduction via NFκB and MAP kinase cascades is a universal response initiated upon pathogen recognition by Toll-like receptors (TLRs). How activation of these divergent signaling pathways is integrated to dictate distinct immune responses to diverse pathogens is still incompletely understood. Herein, contrary to current perception, we demonstrate that a signaling pathway defined by the inhibitor of κB kinase ß (IKKß), MAP3 kinase tumor progression locus 2 (Tpl2/MAP3K8), and MAP kinase ERK is differentially activated by TLRs. TLRs 2, 4, and 7 directly activate this inflammatory axis, inducing immediate ERK phosphorylation and early TNFα secretion. In addition to TLR adaptor proteins, IKKß-Tpl2-ERK activation by TLR4 is regulated by the TLR4 co-receptor CD14 and the tyrosine kinase Syk. Signals from TLRs 3 and 9 do not initiate early activation of IKKß-Tpl2-ERK pathway but instead induce delayed, NADPH-oxidase-dependent ERK phosphorylation and TNFα secretion via autocrine reactive oxygen species signaling. Unexpectedly, Tpl2 is an essential regulator of ROS production during TLR signaling. Overall, our study reveals distinct mechanisms activating a common inflammatory signaling cascade and delineates differences in MyD88-dependent signaling between endosomal TLRs 7 and 9. These findings further confirm the importance of Tpl2 in innate host defense mechanisms and also enhance our understanding of how the immune system tailors pathogen-specific gene expression patterns.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas Quinasa Quinasa PAM/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/fisiología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 9/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Comunicación Autocrina , Células Cultivadas , Quinasa I-kappa B/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio , Quinasa Syk , Receptor Toll-Like 4/metabolismo , Activación Transcripcional , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Immunol ; 190(12): 6488-500, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23677474

RESUMEN

Pseudogout is an autoinflammatory condition triggered by calcium pyrophosphate dehydrate (CPPD) crystal deposition in the joints. The innate immune system is irritated by and responds to the presence of the crystals with an inflammatory response. The synovial fluid contains activated inflammatory macrophages and neutrophil granulocytes. Several details of crystal-induced macrophage activation were recently uncovered, but very little is known about interactions of CPPD crystals with neutrophils. In this study, we show that human neutrophils engulf CPPD crystals and form large amounts of neutrophil extracellular traps (NETs) in vitro. Released extracellular DNA binds myeloperoxidase and citrullinated histone H4. CPPD crystal-stimulated neutrophils and their nuclear DNA undergo morphological changes characteristic for NET formation. The ERK/MEK signaling pathway, heat shock protein 90, PI3K, and an intact cytoskeleton are required for CPPD-induced NET formation. Blocking crystal-activated respiratory burst has, however, no effect on NETs. Human neutrophils release IL-1ß and IL-8 in response to CPPD crystals, and blocking CXCR2, the main IL-8R, diminishes NET formation. Proinflammatory cytokines, TNF-α, GM-CSF, and IL-1ß, increase NET release by the crystals. Enhanced bacterial killing by CPPD-induced NETs demonstrates their ability to cause cellular damage. Our work documents and provides details about extracellular trap release in human neutrophils activated by CPPD microcrystals. We suggest that crystal-triggered NET formation can be a novel contributor to inflammatory conditions observed in CPPD crystal-driven synovitis.


Asunto(s)
Pirofosfato de Calcio/inmunología , Condrocalcinosis/inmunología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Condrocalcinosis/patología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Fagocitosis/inmunología
14.
Proc Natl Acad Sci U S A ; 109(14): 5429-34, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22431641

RESUMEN

The malaria parasite, Plasmodium falciparum, and the human immune system have coevolved to ensure that the parasite is not eliminated and reinfection is not resisted. This relationship is likely mediated through a myriad of host-parasite interactions, although surprisingly few such interactions have been identified. Here we show that the 33-kDa fragment of P. falciparum merozoite surface protein 1 (MSP1(33)), an abundant protein that is shed during red blood cell invasion, binds to the proinflammatory protein, S100P. MSP1(33) blocks S100P-induced NFκB activation in monocytes and chemotaxis in neutrophils. Remarkably, S100P binds to both dimorphic alleles of MSP1, estimated to have diverged >27 Mya, suggesting an ancient, conserved relationship between these parasite and host proteins that may serve to attenuate potentially damaging inflammatory responses.


Asunto(s)
Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteína 1 de Superficie de Merozoito/fisiología , Proteínas de Neoplasias/antagonistas & inhibidores , Plasmodium falciparum/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al Calcio/química , Cromatografía en Gel , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Humanos , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Homología de Secuencia de Aminoácido , Resonancia por Plasmón de Superficie
15.
Am J Respir Cell Mol Biol ; 50(1): 125-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23962049

RESUMEN

Oxidative stress has been implicated in the pathogenesis of bronchial asthma. Besides granulocytes, the airway epithelium can produce large amounts of reactive oxygen species and can contribute to asthma-related oxidative stress. Histamine is a major inflammatory mediator present in large quantities in asthmatic airways. Whether histamine triggers epithelium-derived oxidative stress is unknown. We therefore aimed at characterizing human airway epithelial H2O2 production stimulated by histamine. We found that air-liquid interface cultures of primary human bronchial epithelial cells (BECs) and an immortalized BEC model (Cdk4/hTERT HBEC) produce H2O2 in response to histamine. The main source of airway epithelial H2O2 is an NADPH dual oxidase, Duox1. Out of the four histamine receptors (H1R-H4R), H1R has the highest expression in BECs and mediates the H2O2-producing effects of histamine. IL-4 induces Duox1 gene and protein expression levels and enhances histamine-induced H2O2 production by epithelial cells. Using HEK-293 cells expressing Duox1 or Duox2 and endogenous H1R, histamine triggers an immediate intracellular calcium signal and H2O2 release. Overexpression of H1R further increases the oxidative output of Duox-expressing HEK-293 cells. Our observations show that BECs respond to histamine with Duox-mediated H2O2 production. These findings reveal a mechanism that could be an important contributor to oxidative stress characteristic of asthmatic airways, suggesting novel therapeutic targets for treating asthmatic airway disease.


Asunto(s)
Bronquios/metabolismo , Células Epiteliales/metabolismo , Histamina/metabolismo , Peróxido de Hidrógeno/metabolismo , NADPH Oxidasas/metabolismo , Receptores Histamínicos H1/metabolismo , Células Cultivadas , Quinasa 4 Dependiente de la Ciclina/metabolismo , Citocinas/metabolismo , Oxidasas Duales , Células HEK293 , Humanos , Interleucina-4/metabolismo , Telomerasa/metabolismo , Células Th2/metabolismo
16.
Inflamm Res ; 63(10): 821-30, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25048991

RESUMEN

OBJECTIVE AND DESIGN: We studied the involvement of calcium and calcium-activated NADPH oxidases in NLRP3 inflammasome activation and IL-1ß release to better understand inflammasome signaling in macrophages. MATERIAL OR SUBJECTS: Human volunteer blood donors were recruited to isolate monocytes to differentiate them into macrophages. Wild-type or DUOX1-deficient C57/B6 mice were used to prepare bone marrow-derived macrophages. TREATMENT: Murine or human macrophages were treated in vitro with NLRP3 inflammasome agonists (ATP, silica crystals) or calcium agonists (thapsigargin, ionomycin) in calcium-containing or calcium-free medium. METHODS: Intracellular calcium changes were followed by measuring FURA2-based fluorescence. Gene expression changes were measured by quantitative real-time PCR. Protein expression was assessed by western blotting. Enzymatic activity was measured by fluorescence caspase-1 activity assay. IL-1ß release was determined by ELISA. ELISA data were analyzed by ANOVA and Tukey's post hoc test. RESULTS: Our data show that calcium is essential for IL-1ß release in human macrophages. Increases in cytosolic calcium alone lead to IL-1ß secretion. Calcium removal blocks caspase-1 activation. Human macrophages express Duox1, a calcium-regulated NADPH oxidase that produces reactive oxygen species. However, Duox1-deficient murine macrophages show normal IL-1ß release. CONCLUSIONS: Human macrophage inflammasome activation and IL-1ß secretion requires calcium but does not involve NADPH oxidases.


Asunto(s)
Calcio/metabolismo , Proteínas Portadoras/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Adenosina Trifosfato/farmacología , Animales , Oxidasas Duales , Células HEK293 , Humanos , Inflamasomas , Ionomicina/farmacología , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Dióxido de Silicio/farmacología , Tapsigargina/farmacología
17.
Free Radic Biol Med ; 219: 104-111, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38608822

RESUMEN

Hypothiocyanous acid (HOSCN) is an endogenous oxidant produced by peroxidase oxidation of thiocyanate (SCN-), an ubiquitous sulfur-containing pseudohalide synthesized from cyanide. HOSCN serves as a potent microbicidal agent against pathogenic bacteria, viruses, and fungi, functioning through thiol-targeting mechanisms, independent of currently approved antimicrobials. Additionally, SCN- reacts with hypochlorous acid (HOCl), a highly reactive oxidant produced by myeloperoxidase (MPO) at sites of inflammation, also producing HOSCN. This imparts both antioxidant and antimicrobial potential to SCN-. In this review, we discuss roles of HOSCN/SCN- in immunity and potential therapeutic implications for combating infections.


Asunto(s)
Antiinfecciosos , Tiocianatos , Animales , Humanos , Antiinfecciosos/farmacología , Antiinfecciosos/uso terapéutico , Antiinfecciosos/química , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Ácido Hipocloroso/metabolismo , Ácido Hipocloroso/uso terapéutico , Ácido Hipocloroso/química , Oxidación-Reducción , Peroxidasa/metabolismo , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/microbiología , Tiocianatos/uso terapéutico , Tiocianatos/química , Tiocianatos/farmacología , Tiocianatos/metabolismo
18.
Sci Rep ; 14(1): 3986, 2024 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-38368463

RESUMEN

Bone marrow stromal cells (BMSCs) have immunomodulatory activities in numerous species and have been used in clinical trials. BMSCs also make antibacterial agents. Since hepcidin is known to have antimicrobial effects in fish, we wondered if it might also be used as an antimicrobial agent by mammalian BMSCs. In the present study, we show hepcidin expression in both mouse (mBMSC) and human BMSCs (hBMSC). We observed a hBMSC hepcidin-dependent degradation of ferroportin in HEK-293 reporter cells in vitro. In human and mouse bone marrows (BM) we detected hepcidin-positive BMSCs in close proximity to hematopoietic progenitors. The conditioned culture medium of hBMSCs significantly reduced bacterial proliferation that was partially blocked by a hepcidin-neutralizing antibody. Similarly, medium in which hepcidin-deficient (Hamp-/-) mouse BMSCs had been grown was significantly less effective in reducing bacterial counts than the medium of wild-type cells. In a zymosan-induced peritonitis mouse model we found that mBMSC-derived hepcidin reduced the number of invading polymorphonuclear (PMN) cells in the peritoneal cavity. Our results show that BMSC-derived hepcidin has antimicrobial properties in vitro and also reduces inflammation in vivo. We conclude that hepcidin should be added to the expanding arsenal of agents available to BMSCs to fight infections and inflammation.


Asunto(s)
Antiinfecciosos , Células Madre Mesenquimatosas , Humanos , Ratones , Animales , Hepcidinas/metabolismo , Células HEK293 , Antiinfecciosos/farmacología , Inflamación/metabolismo , Células de la Médula Ósea , Mamíferos
19.
Stem Cells ; 30(2): 222-31, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22045589

RESUMEN

There are several clinical trials worldwide using bone marrow stromal cells (BMSCs) as a cellular therapy to modulate immune responses in patients suffering from various inflammatory conditions. A deeper understanding of the molecular mechanisms involved in this modulatory effect could help us design better, more effective protocols to treat immune mediated diseases. In this study, we demonstrated that human BMSCs express H1, H2, and H4 histamine receptors and they respond to histamine stimulation with an increased interleukin 6 (IL-6) production both in vitro and in vivo. Using different receptor antagonists, we pinpointed the importance of the H1 histamine receptor, while Western blot analysis and application of various mitogen-activated protein kinase inhibitors highlighted the role of p38, extracellular signal-regulated kinase, and c-Jun N-terminal kinase kinases in the observed effect. When BMSCs were pretreated with either histamine or degranulated human mast cells, they exhibited an enhanced IL-6-dependent antiapoptotic effect on neutrophil granulocytes. Based on these observations, it is likely that introduction of BMSCs into a histamine-rich environment (such as any allergic setting) or pretreatment of these cells with synthetic histamine could have a significant modulatory effect on the therapeutic potential of BMSCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Histamina/fisiología , Receptores Histamínicos/fisiología , Células del Estroma/metabolismo , Animales , Apoptosis , Células de la Médula Ósea/fisiología , Células Cultivadas , Técnicas de Cocultivo , Expresión Génica , Granulocitos/metabolismo , Histamina/farmacología , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Mastocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Histamínicos/genética , Receptores Histamínicos/metabolismo , Células del Estroma/fisiología
20.
J Immunol ; 186(2): 1180-8, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21148036

RESUMEN

TNF, an important mediator of inflammatory and innate immune responses, can be regulated by binding to soluble TNF receptors. The 55-kDa type 1 TNFR (TNFR1), the key receptor for TNF signaling, is released to the extracellular space by two mechanisms, the inducible cleavage and shedding of 34-kDa soluble TNFR1 (sTNFR1) ectodomains and the constitutive release of full-length 55-kDa TNFR1 within exosome-like vesicles. The aim of this study was to identify and characterize TLR signaling pathways that mediate TNFR1 release to the extracellular space. To our knowledge, we demonstrate for the first time that polyinosinic-polycytidylic acid [poly (I:C)], a synthetic dsRNA analogue that signals via TLR3, induces sTNFR1 shedding from human airway epithelial (NCI-H292) cells, whereas ligands for other microbial pattern recognition receptors, including TLR4, TLR7, and nucleotide-binding oligomerization domain containing 2, do not. Furthermore, poly (I:C) selectively induces the cleavage of 34-kDa sTNFR1 ectodomains but does not enhance the release of full-length 55-kDa TNFR1 within exosome-like vesicles. RNA interference experiments demonstrated that poly (I:C)-induced sTNFR1 shedding is mediated via activation of TLR3-TRIF-RIP1 signaling, with subsequent activation of two downstream pathways. One pathway involves the dual oxidase 2-mediated generation of reactive oxygen species, and the other pathway is via the caspase-mediated activation of apoptosis. Thus, the ability of dsRNA to induce the cleavage and shedding of the 34-kDa sTNFR1 from human bronchial epithelial cells represents a novel mechanism by which innate immune responses to viral infections are modulated.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/fisiología , Caspasas/fisiología , NADPH Oxidasas/fisiología , Proteínas de Complejo Poro Nuclear/fisiología , ARN Bicatenario/farmacología , Proteínas de Unión al ARN/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Mucosa Respiratoria/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 3/fisiología , Apoptosis/inmunología , Bronquios/citología , Bronquios/inmunología , Bronquios/metabolismo , Línea Celular , Oxidasas Duales , Espacio Extracelular/inmunología , Espacio Extracelular/metabolismo , Humanos , Peso Molecular , Poli I-C/genética , Poli I-C/farmacología , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Transducción de Señal/genética , Solubilidad , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA