Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(1): 65-75, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26595887

RESUMEN

Viral respiratory tract infections are the main causative agents of the onset of infection-induced asthma and asthma exacerbations that remain mechanistically unexplained. Here we found that deficiency in signaling via type I interferon receptor led to deregulated activation of group 2 innate lymphoid cells (ILC2 cells) and infection-associated type 2 immunopathology. Type I interferons directly and negatively regulated mouse and human ILC2 cells in a manner dependent on the transcriptional activator ISGF3 that led to altered cytokine production, cell proliferation and increased cell death. In addition, interferon-γ (IFN-γ) and interleukin 27 (IL-27) altered ILC2 function dependent on the transcription factor STAT1. These results demonstrate that type I and type II interferons, together with IL-27, regulate ILC2 cells to restrict type 2 immunopathology.


Asunto(s)
Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Linfocitos/inmunología , Infecciones del Sistema Respiratorio/inmunología , Animales , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones del Sistema Respiratorio/patología
2.
Nat Immunol ; 13(6): 543-550, 2012 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-22544393

RESUMEN

Type I interferon is an integral component of the antiviral response, and its production is tightly controlled at the levels of transcription and translation. The eukaryotic translation-initiation factor eIF4E is a rate-limiting factor whose activity is regulated by phosphorylation of Ser209. Here we found that mice and fibroblasts in which eIF4E cannot be phosphorylated were less susceptible to virus infection. More production of type I interferon, resulting from less translation of Nfkbia mRNA (which encodes the inhibitor IκBα), largely explained this phenotype. The lower abundance of IκBα resulted in enhanced activity of the transcription factor NF-κB, which promoted the production of interferon-ß (IFN-ß). Thus, regulated phosphorylation of eIF4E has a key role in antiviral host defense by selectively controlling the translation of an mRNA that encodes a critical suppressor of the innate antiviral response.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Interferón Tipo I/biosíntesis , FN-kappa B/metabolismo , Estomatitis Vesicular/inmunología , Virus de la Estomatitis Vesicular Indiana/fisiología , Animales , Ensayo de Cambio de Movilidad Electroforética , Factor 4E Eucariótico de Iniciación/inmunología , Femenino , Proteínas I-kappa B/biosíntesis , Proteínas I-kappa B/genética , Proteínas I-kappa B/inmunología , Inmunidad Innata/inmunología , Immunoblotting , Interferón Tipo I/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidor NF-kappaB alfa , FN-kappa B/inmunología , Fosforilación , Biosíntesis de Proteínas , ARN Mensajero/química , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Organismos Libres de Patógenos Específicos , Estomatitis Vesicular/genética , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/inmunología , Replicación Viral
3.
Photochem Photobiol Sci ; 23(2): 339-354, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38308169

RESUMEN

Ultraviolet radiation's germicidal efficacy depends on several parameters, including wavelength, radiant exposure, microbial physiology, biological matrices, and surfaces. In this work, several ultraviolet radiation sources (a low-pressure mercury lamp, a KrCl excimer, and four UV LEDs) emitting continuous or pulsed irradiation were compared. The greatest log reductions in E. coli cells and B. subtilis endospores were 4.1 ± 0.2 (18 mJ cm-2) and 4.5 ± 0.1 (42 mJ cm-2) with continuous 222 nm, respectively. The highest MS2 log reduction observed was 2.7 ± 0.1 (277 nm at 3809 mJ cm-2). Log reductions of SARS-CoV-2 with continuous 222 nm and 277 nm were ≥ 3.4 ± 0.7, with 13.3 mJ cm-2 and 60 mJ cm-2, respectively. There was no statistical difference between continuous and pulsed irradiation (0.83-16.7% [222 nm and 277 nm] or 0.83-20% [280 nm] duty rates) on E. coli inactivation. Pulsed 260 nm radiation (0.5% duty rate) at 260 nm yielded significantly greater log reduction for both bacteria than continuous 260 nm radiation. There was no statistical difference in SARS-CoV-2 inactivation between continuous and pulsed 222 nm UV-C radiation and pulsed 277 nm radiation demonstrated greater germicidal efficacy than continuous 277 nm radiation. Greater radiant exposure for all radiation sources was required to inactivate MS2 bacteriophage. Findings demonstrate that pulsed irradiation could be more useful than continuous UV radiation in human-occupied spaces, but threshold limit values should be respected. Pathogen-specific sensitivities, experimental setup, and quantification methods for determining germicidal efficacy remain important factors when optimizing ultraviolet radiation for surface decontamination or other applications.


Asunto(s)
COVID-19 , Rayos Ultravioleta , Humanos , SARS-CoV-2 , Escherichia coli/efectos de la radiación , Desinfección/métodos
4.
Immunity ; 40(4): 554-68, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24726877

RESUMEN

Aspirin gained tremendous popularity during the 1918 Spanish Influenza virus pandemic, 50 years prior to the demonstration of their inhibitory action on prostaglandins. Here, we show that during influenza A virus (IAV) infection, prostaglandin E2 (PGE2) was upregulated, which led to the inhibition of type I interferon (IFN) production and apoptosis in macrophages, thereby causing an increase in virus replication. This inhibitory role of PGE2 was not limited to innate immunity, because both antigen presentation and T cell mediated immunity were also suppressed. Targeted PGE2 suppression via genetic ablation of microsomal prostaglandin E-synthase 1 (mPGES-1) or by the pharmacological inhibition of PGE2 receptors EP2 and EP4 substantially improved survival against lethal IAV infection whereas PGE2 administration reversed this phenotype. These data demonstrate that the mPGES-1-PGE2 pathway is targeted by IAV to evade host type I IFN-dependent antiviral immunity. We propose that specific inhibition of PGE2 signaling might serve as a treatment for IAV.


Asunto(s)
Dinoprostona/metabolismo , Virus de la Influenza A/fisiología , Interferón Tipo I/metabolismo , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Animales , Presentación de Antígeno/efectos de los fármacos , Apoptosis/efectos de los fármacos , Células Cultivadas , Dinoprostona/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunidad/efectos de los fármacos , Inmunidad/genética , Interferón Tipo I/genética , Oxidorreductasas Intramoleculares/genética , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia Molecular Dirigida , Infecciones por Orthomyxoviridae/inmunología , Prostaglandina-E Sintasas , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Linfocitos T/inmunología , Linfocitos T/virología , Replicación Viral/genética
5.
J Allergy Clin Immunol ; 147(6): 2330-2342, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33453289

RESUMEN

BACKGROUND: Lung nociceptor neurons amplify immune cell activity and mucus metaplasia in response to an inhaled allergen challenge in sensitized mice. OBJECTIVE: We sought to identify the cellular mechanisms by which these sensory neurons are activated subsequent to allergen exposure. METHODS: We used calcium microscopy and electrophysiologic recording to assess whether vagal neurons directly respond to the model allergen ovalbumin (OVA). Next, we generated the first nociceptor-specific FcεR1γ knockdown (TRPV1Cre::FcεR1γfl/fl) mice to assess whether this targeted invalidation would affect the severity of allergic inflammation in response to allergen challenges. RESULTS: Lung-innervating jugular nodose complex ganglion neurons express the high-affinity IgE receptor FcεR1, the levels of which increase in OVA-sensitized mice. FcεR1γ-expressing vagal nociceptor neurons respond directly to OVA complexed with IgE with depolarization, action potential firing, calcium influx, and neuropeptide release. Activation of vagal neurons by IgE-allergen immune complexes, through the release of substance P from their peripheral terminals, directly amplifies TH2 cell influx and polarization in the airways. Allergic airway inflammation is decreased in TRPV1Cre::FcεR1γfl/fl mice and in FcεR1α-/- mice into which bone marrow has been transplanted. Finally, increased in vivo circulating levels of IgE following allergen sensitization enhances the responsiveness of FcεR1 to immune complexes in both mouse jugular nodose complex ganglion neurons and human induced pluripotent stem cell-derived nociceptors. CONCLUSIONS: Allergen sensitization triggers a feedforward inflammatory loop between IgE-producing plasma cells, FcεR1-expressing vagal sensory neurons, and TH2 cells, which helps to both initiate and amplify allergic airway inflammation. These data highlight a novel target for reducing allergy, namely, FcεR1γ expressed by nociceptors.


Asunto(s)
Expresión Génica , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Receptores de IgE/genética , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Alérgenos/inmunología , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Predisposición Genética a la Enfermedad , Hipersensibilidad/genética , Hipersensibilidad/patología , Ratones , Ratones Noqueados , Neuronas/inmunología , Neuronas/metabolismo , Nociceptores/metabolismo , Ovalbúmina/efectos adversos , Ovalbúmina/inmunología , Receptores de IgE/metabolismo , Mucosa Respiratoria/patología , Sustancia P/metabolismo , Nervio Vago
6.
FASEB J ; 32(2): 829-837, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28982732

RESUMEN

Group 2 innate lymphoid cells (ILC2s) represent a rapid source of type 2 cytokines, such as IL-5 and IL-13, and play an important role in orchestrating type 2 immune response. Adenosine is an endogenous purine nucleoside, a catabolite of ATP that binds and activates ≥1 of 4 transmembrane G protein-coupled cell-surface adenosine receptors (ARs)-A1, A2A, A2B, and A3. Here, we studied the role of ARs in the regulation of cytokine production by ILC2s. We found that A2BARs suppress the production of both IL-5 and IL-13 by ILC2s, whereas A2AARs augment IL-5 production and fail to affect IL-13 release. Combined stimulation of all ARs led to the suppression of both IL-5 and IL-13 production, which indicated that A2BARs dominate A2AARs. Both pre- and post-transcriptional processes may be involved in the AR modulation of ILC2 IL-5 and IL-13 production. Thus, we identify adenosine as a novel negative regulator of ILC2 activation.-Csóka, B., Németh, Z. H., Duerr, C. U., Fritz, J. H., Pacher, P., Haskó, G. Adenosine receptors differentially regulate type 2 cytokine production by IL-33-activated bone marrow cells, ILC2s, and macrophages.


Asunto(s)
Células de la Médula Ósea/inmunología , Interleucina-13/inmunología , Interleucina-33/farmacología , Interleucina-5/inmunología , Macrófagos/inmunología , Receptor de Adenosina A2A/inmunología , Receptor de Adenosina A2B/inmunología , Células Th2/inmunología , Animales , Células de la Médula Ósea/citología , Interleucina-13/genética , Interleucina-33/inmunología , Interleucina-5/genética , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Macrófagos/citología , Ratones , Ratones Noqueados , Receptor de Adenosina A2A/genética , Receptor de Adenosina A2B/genética , Células Th2/citología
7.
J Immunol ; 198(1): 71-81, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27881703

RESUMEN

The regulatory properties of B cells have been studied in autoimmune diseases; however, their role in allergic diseases is poorly understood. We demonstrate that Semaphorin 4C (Sema4C), an axonal guidance molecule, plays a crucial role in B cell regulatory function. Mice deficient in Sema4C exhibited increased airway inflammation after allergen exposure, with massive eosinophilic lung infiltrates and increased Th2 cytokines. This phenotype was reproduced by mixed bone marrow chimeric mice with Sema4C deficient only in B cells, indicating that B lymphocytes were the key cells affected by the absence of Sema4C expression in allergic inflammation. We determined that Sema4C-deficient CD19+CD138+ cells exhibited decreased IL-10 and increased IL-4 expression in vivo and in vitro. Adoptive transfer of Sema4c-/- CD19+CD138+ cells induced marked pulmonary inflammation, eosinophilia, and increased bronchoalveolar lavage fluid IL-4 and IL-5, whereas adoptive transfer of wild-type CD19+CD138+IL-10+ cells dramatically decreased allergic airway inflammation in wild-type and Sema4c-/- mice. This study identifies a novel pathway by which Th2-mediated immune responses are regulated. It highlights the importance of plasma cells as regulatory cells in allergic inflammation and suggests that CD138+ B cells contribute to cytokine balance and are important for maintenance of immune homeostasis in allergic airways disease. Furthermore, we demonstrate that Sema4C is critical for optimal regulatory cytokine production in CD138+ B cells.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Células Plasmáticas/inmunología , Hipersensibilidad Respiratoria/inmunología , Semaforinas/inmunología , Traslado Adoptivo , Animales , Western Blotting , Citocinas/biosíntesis , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/inmunología , Sindecano-1/inmunología
8.
J Virol ; 90(20): 9406-19, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27512060

RESUMEN

UNLABELLED: STING has emerged in recent years as a key player in orchestrating innate immune responses to cytosolic DNA and RNA derived from pathogens. However, the regulation of STING still remains poorly defined. In the present study, we investigated the mechanism of the regulation of STING expression in relation to the RIG-I pathway. Our data show that signaling through RIG-I induces STING expression at both the transcriptional and protein levels in various cell types. STING induction by the RIG-I agonist 5'triphosphorylated RNA (5'pppRNA) was recognized to be a delayed event resulting from an autocrine/paracrine mechanism. Indeed, cotreatment with tumor necrosis factor alpha and type I/II interferon was found to have a synergistic effect on the regulation of STING expression and could be potently decreased by impairing NF-κB and/or STAT1/2 signaling. STING induction significantly contributed to sustainment of the immune signaling cascade following 5'pppRNA treatment. Physiologically, this cross talk between the RNA- and DNA-sensing pathways allowed 5'pppRNA to efficiently block infection by herpes simplex virus 1 (HSV-1) both in vitro and in vivo in a STING-dependent fashion. These observations demonstrate that STING induction by RIG-I signaling through the NF-κB and STAT1/2 cascades is essential for RIG-I agonist-mediated HSV-1 restriction. IMPORTANCE: The innate immune system represents the first line of defense against invading pathogens. The dysregulation of this system can result in failure to combat pathogens, inflammation, and autoimmune diseases. Thus, precise regulation at each level of the innate immune system is crucial. Recently, a number of studies have established STING to be a central molecule in the innate immune response to cytosolic DNA and RNA derived from pathogens. Here, we describe the regulation of STING via RIG-I-mediated innate immune sensing. We found that STING is synergistically induced via proinflammatory and antiviral cytokine cascades. In addition, we show that in vivo protection against herpes simplex virus 1 (HSV-1) by a RIG-I agonist required STING. Our study provides new insights into the cross talk between DNA and RNA pathogen-sensing systems via the control of STING.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Herpes Simple/metabolismo , Herpesvirus Humano 1/metabolismo , Proteínas de la Membrana/metabolismo , Regulación hacia Arriba/fisiología , Células A549 , Línea Celular , Línea Celular Tumoral , Citocinas/metabolismo , Humanos , Inmunidad Innata/fisiología , Interferón Tipo I/metabolismo , FN-kappa B/metabolismo , Receptores Inmunológicos , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal/fisiología , Activación Transcripcional/fisiología
9.
J Immunol ; 195(9): 4479-91, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26416282

RESUMEN

Understanding the mechanisms of host macrophage responses to Mycobacterium tuberculosis is essential for uncovering potential avenues of intervention to boost host resistance to infection. Macrophage transcriptome profiling revealed that M. tuberculosis infection strongly induced the expression of several enzymes controlling tryptophan catabolism. These included IDO1 and tryptophan 2,3-dioxygenase, which catalyze the rate-limiting step in the kynurenine pathway, producing ligands for the aryl hydrocarbon receptor (AHR). The AHR and heterodimeric partners AHR nuclear translocator and RELB are robustly expressed, and AHR and RELB levels increased further during infection. Infection enhanced AHR/AHR nuclear translocator and AHR/RELB DNA binding and stimulated the expression of AHR target genes, including that encoding the inflammatory cytokine IL-1ß. AHR target gene expression was further enhanced by exogenous kynurenine, and exogenous tryptophan, kynurenine, or synthetic agonist indirubin reduced mycobacterial viability. Comparative expression profiling revealed that AHR ablation diminished the expression of numerous genes implicated in innate immune responses, including several cytokines. Notably, AHR depletion reduced the expression of IL23A and IL12B transcripts, which encode subunits of IL-23, a macrophage cytokine that stimulates production of IL-22 by innate lymphoid cells. AHR directly induced IL23A transcription in human and mouse macrophages through near-upstream enhancer regions. Taken together, these findings show that AHR signaling is strongly engaged in M. tuberculosis-infected macrophages and has widespread effects on innate immune responses. Moreover, they reveal a cascade of AHR-driven innate immune signaling, because IL-1ß and IL-23 stimulate T cell subsets producing IL-22, another direct target of AHR transactivation.


Asunto(s)
Inmunidad Innata/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Transducción de Señal/inmunología , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Pleiotropía Genética/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Interleucina-23/genética , Interleucina-23/inmunología , Interleucina-23/metabolismo , Interleucinas/genética , Interleucinas/inmunología , Interleucinas/metabolismo , Células L , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Microscopía Fluorescente , Mycobacterium tuberculosis/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/genética , Factor de Transcripción ReIB/genética , Factor de Transcripción ReIB/inmunología , Factor de Transcripción ReIB/metabolismo , Transcriptoma/genética , Transcriptoma/inmunología , Interleucina-22
10.
Nature ; 481(7380): 199-203, 2011 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-22158124

RESUMEN

The largest mucosal surface in the body is in the gastrointestinal tract, a location that is heavily colonized by microbes that are normally harmless. A key mechanism required for maintaining a homeostatic balance between this microbial burden and the lymphocytes that densely populate the gastrointestinal tract is the production and transepithelial transport of poly-reactive IgA (ref. 1). Within the mucosal tissues, B cells respond to cytokines, sometimes in the absence of T-cell help, undergo class switch recombination of their immunoglobulin receptor to IgA, and differentiate to become plasma cells. However, IgA-secreting plasma cells probably have additional attributes that are needed for coping with the tremendous bacterial load in the gastrointestinal tract. Here we report that mouse IgA(+) plasma cells also produce the antimicrobial mediators tumour-necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS), and express many molecules that are commonly associated with monocyte/granulocytic cell types. The development of iNOS-producing IgA(+) plasma cells can be recapitulated in vitro in the presence of gut stroma, and the acquisition of this multifunctional phenotype in vivo and in vitro relies on microbial co-stimulation. Deletion of TNF-α and iNOS in B-lineage cells resulted in a reduction in IgA production, altered diversification of the gut microbiota and poor clearance of a gut-tropic pathogen. These findings reveal a novel adaptation to maintaining homeostasis in the gut, and extend the repertoire of protective responses exhibited by some B-lineage cells.


Asunto(s)
Inmunoglobulina A/inmunología , Intestino Delgado/citología , Intestino Delgado/inmunología , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Animales , Células de la Médula Ósea/citología , Linaje de la Célula , Células Cultivadas , Quimera/inmunología , Citrobacter rodentium/inmunología , Técnicas de Cocultivo , Femenino , Vida Libre de Gérmenes , Granulocitos/citología , Granulocitos/metabolismo , Inmunidad Innata/inmunología , Inmunoglobulina A/biosíntesis , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestino Delgado/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Monocitos/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenotipo , Células Plasmáticas/metabolismo , Bazo/citología , Células del Estroma/citología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
11.
EMBO J ; 31(21): 4153-64, 2012 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-23064150

RESUMEN

Immunity against infection with Listeria monocytogenes is not achieved from innate immune stimulation by contact with killed but requires viable Listeria gaining access to the cytosol of infected cells. It has remained ill-defined how such immune sensing of live Listeria occurs. Here, we report that efficient cytosolic immune sensing requires access of nucleic acids derived from live Listeria to the cytoplasm of infected cells. We found that Listeria released nucleic acids and that such secreted bacterial RNA/DNA was recognized by the cytosolic sensors RIG-I, MDA5 and STING thereby triggering interferon ß production. Secreted Listeria nucleic acids also caused RIG-I-dependent IL-1ß-production and inflammasome activation. The signalling molecule CARD9 contributed to IL-1ß production in response to secreted nucleic acids. In conclusion, cytosolic recognition of secreted bacterial nucleic acids by RIG-I provides a mechanistic explanation for efficient induction of immunity by live bacteria.


Asunto(s)
Citoplasma/metabolismo , ARN Helicasas DEAD-box/fisiología , ADN Bacteriano/inmunología , Inmunidad Celular/inmunología , Inflamación/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , ARN Bacteriano/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Western Blotting , Proteínas Adaptadoras de Señalización CARD , Células Cultivadas , Citoplasma/inmunología , Citoplasma/microbiología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN Bacteriano/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Inflamación/microbiología , Helicasa Inducida por Interferón IFIH1 , Listeria monocytogenes/genética , Listeriosis/genética , Listeriosis/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Bacteriano/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
12.
Cytokine ; 87: 1-8, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27255596

RESUMEN

Group 2 innate lymphoid cells (ILC2) exert critical roles in type 2 immune responses, epithelial repair at mucosal tissues and metabolic homeostasis. ILC2 rapidly provide large amounts of type 2 signature cytokines, thereby driving type 2 immune responses such as the defense against helminths. However, if deregulated, ILC2 facilitate tissue fibrosis and trigger unwanted type 2 immunopathologies such as allergies, asthma and atopic dermatitis. Therefore, ILC2 need to be tightly regulated and we are just beginning to understand which mediators activate or inhibit this rare but important cell population. In this review, we summarize current knowledge about positive and negative regulation of ILC2 and discuss its immunological consequences.


Asunto(s)
Inmunidad Innata , Linfocitos , Células Th2/inmunología , Animales , Asma/inmunología , Citocinas/inmunología , Dermatitis Atópica/inmunología , Humanos , Hipersensibilidad/inmunología , Interleucina-33/inmunología , Ratones , Membrana Mucosa/inmunología
13.
J Immunol ; 192(6): 2787-99, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24534531

RESUMEN

Innate immune responses provoke the accumulation of leukocytes at sites of inflammation. In addition to monocytes and granulocytes, B cells also participate in antimicrobial innate immune responses; however, the mechanisms for accumulation of B cells to sites of inflammation are not well understood. To study B cell accumulation following systemic inflammation, we used a model synthetic ligand that stimulates a specific pattern recognition molecule, nucleotide-binding oligomerization domain-containing protein 1 (Nod1). Upon exposure to Nod1 agonists, both B cells and neutrophils rapidly accumulate within the spleen, and dendritic cells migrate into the periarterial lymphoid sheath. Nod1 stimulation led to a marked increase in several chemokines within the spleen, including CXCL13, CCL2, and CCL20. Whereas the lymphotoxin pathway was critical for the induction of the B cell chemoattractant CXCL13 in response to Nod1 agonists, B cell accumulation within the spleen following Nod1-induced systemic inflammation was independent of the lymphotoxin pathway. In contrast, a CCR6/CCL20 chemokine loop instructed rapid increase of B cells in the spleen in response to systemic administration of Nod1 agonists in a TNF-α-dependent manner. Moreover, CCR6 was required to regulate Nod1-mediated B cell responses. These results reveal a novel mechanism of B cells during inflammation and shed light on how B cells participate in innate immune responses to microbial stimulation.


Asunto(s)
Linfocitos B/inmunología , Quimiocina CCL20/inmunología , Proteína Adaptadora de Señalización NOD1/inmunología , Receptores CCR6/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea/métodos , Línea Celular , Células Cultivadas , Quimiocina CCL20/metabolismo , Ácido Diaminopimélico/análogos & derivados , Ácido Diaminopimélico/farmacología , Femenino , Citometría de Flujo , Recuento de Linfocitos , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD1/metabolismo , Receptores CCR6/genética , Receptores CCR6/metabolismo , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Quimera por Trasplante/sangre , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
14.
Infect Immun ; 83(12): 4541-54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26371125

RESUMEN

The genetic basis of natural susceptibility to progressive Cryptococcus neoformans infection is not well understood. Using C57BL/6 and CBA/J inbred mice, we previously identified three chromosomal regions associated with C. neoformans susceptibility (Cnes1, Cnes2, and Cnes3). To validate and characterize the role of Cnes2 during the host response, we constructed a congenic strain on the C57BL/6 background (B6.CBA-Cnes2). Phenotypic analysis of B6.CBA-Cnes2 mice 35 days after C. neoformans infection showed a significant reduction of fungal burden in the lungs and spleen with higher pulmonary expression of gamma interferon (IFN-γ) and interleukin-12 (IL-12), lower expression of IL-4, IL-5, and IL-13, and an absence of airway epithelial mucus production compared to that in C57BL/6 mice. Multiparameter flow cytometry of infected lungs also showed a significantly higher number of neutrophils, exudate macrophages, CD11b(+) dendritic cells, and CD4(+) cells in B6.CBA-Cnes2 than in C57BL/6 mice. The activation state of recruited macrophages and dendritic cells was also significantly increased in B6.CBA-Cnes2 mice. Taken together, these findings demonstrate that the Cnes2 interval is a potent regulator of host defense, immune responsiveness, and differential Th1/Th2 polarization following C. neoformans infection.


Asunto(s)
Cromosomas de los Mamíferos/inmunología , Criptococosis/genética , Cryptococcus neoformans/inmunología , Sitios Genéticos/inmunología , Predisposición Genética a la Enfermedad , Inmunidad Innata , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/patología , Cromosomas de los Mamíferos/química , Cruzamientos Genéticos , Criptococosis/inmunología , Criptococosis/microbiología , Criptococosis/patología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células Dendríticas/patología , Expresión Génica , Interacciones Huésped-Patógeno , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Interleucina-5/genética , Interleucina-5/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Neutrófilos/inmunología , Neutrófilos/microbiología , Neutrófilos/patología , Fenotipo , Balance Th1 - Th2
15.
PLoS Pathog ; 9(8): e1003575, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23990787

RESUMEN

Aspergillus fumigatus is the most common cause of invasive mold disease in humans. The mechanisms underlying the adherence of this mold to host cells and macromolecules have remained elusive. Using mutants with different adhesive properties and comparative transcriptomics, we discovered that the gene uge3, encoding a fungal epimerase, is required for adherence through mediating the synthesis of galactosaminogalactan. Galactosaminogalactan functions as the dominant adhesin of A. fumigatus and mediates adherence to plastic, fibronectin, and epithelial cells. In addition, galactosaminogalactan suppresses host inflammatory responses in vitro and in vivo, in part through masking cell wall ß-glucans from recognition by dectin-1. Finally, galactosaminogalactan is essential for full virulence in two murine models of invasive aspergillosis. Collectively these data establish a role for galactosaminogalactan as a pivotal bifunctional virulence factor in the pathogenesis of invasive aspergillosis.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Aspergillus fumigatus/patogenicidad , Polisacáridos Fúngicos/inmunología , Polisacáridos/inmunología , Factores de Virulencia/inmunología , beta-Glucanos/inmunología , Animales , Aspergilosis/genética , Aspergilosis/patología , Aspergillus fumigatus/genética , Carbohidrato Epimerasas/genética , Carbohidrato Epimerasas/inmunología , Línea Celular , Modelos Animales de Enfermedad , Polisacáridos Fúngicos/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/inmunología , Humanos , Hifa/genética , Hifa/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Ratones , Polisacáridos/genética , Factores de Virulencia/genética
16.
BMC Microbiol ; 15: 238, 2015 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-26502905

RESUMEN

BACKGROUND: Different species and strains of probiotic bacteria confer distinct immunological responses on immune cells. Lactobacillus rhamnosus GR-1 (GR-1) is a probiotic bacterial strain found in both the intestinal and urogenital tracts, and has immunomodulatory effects on several cell types including macrophages. However, detailed immunological responses and the signaling mechanism involved in the response are largely unknown. RESULTS: We examined the production of GR-1-induced cytokines/chemokines and signaling events in macrophages. Among 84 cytokines and chemokines examined, GR-1 discretely induced granulocyte colony-stimulating factor (G-CSF) mRNA at highest levels (>60-fold) without inducing other cytokines such as IL-1α, IL-1ß, IL-6 and TNF-α (<5-fold). The toll-like receptor (TLR) 2/6-agonist PAM2CSK4, TLR2/1-agonist PAM3CSK4 and TLR4-agonist lipopolysaccharide induced all of these inflammatory cytokines at high levels (>50-fold). The TLR2 ligand lipoteichoic acid activated all mitogen-activated kinases, Akt and NF-κB; whereas, GR-1 selectively activated extracellular regulated kinases and p38, NF-κB and Akt, but not c-Jun N-terminal kinases (JNKs) in a TLR2-dependent manner. Using specific inhibitors, we demonstrated that lack of JNKs activation by GR-1 caused inefficient production of pro-inflammatory cytokines but not G-CSF production. A secreted heat-labile protein-like molecule, 30-100 kDa in size, induced the preferential production of G-CSF. CONCLUSION: This study elucidated unique signaling events triggered by GR-1, resulting in selective production of the immunomodulatory cytokine G-CSF in macrophages.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/biosíntesis , Lacticaseibacillus rhamnosus/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Factores de Virulencia/inmunología , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Factores de Virulencia/metabolismo
17.
J Immunol ; 191(5): 2503-13, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23894196

RESUMEN

Susceptibility to progressive infection with the fungus Cryptococcus neoformans is associated with an allergic pattern of lung inflammation, yet the factors that govern this host response are not clearly understood. Using a clinically relevant mouse model of inhalational infection with virulent C. neoformans H99, we demonstrate a role for IL-33-dependent signaling in host immune defense. Infection of BALB/c mice with 10(4) CFU of C. neoformans H99 caused a time-dependent induction of IL-33 with accumulation of type 2 pulmonary innate lymphoid cells and alternatively activated macrophages in the lungs as well as Th2-polarized CD4(+) T cells in draining lymph nodes. IL-33R subunit T1/ST2-deficient (T1/ST2(-/-)) mice infected with C. neoformans H99 had improved survival with a decreased fungal burden in the lungs, spleen, and brain, compared with wild-type mice. Signaling through T1/ST2 was required for the accumulation and early production of IL-5 and IL-13 by lung type 2 pulmonary innate lymphoid cells. Further analysis of T1/ST2(-/-) mice revealed increased fungicidal exudate macrophages in the lungs and decreased C. neoformans-specific Th2 cells in the mediastinal lymph nodes. T1/ST2 deficiency also diminished goblet cell hyperplasia, mucus hypersecretion, bronchoalveolar lavage eosinophilia, alternative activation of macrophages, and serum IgE. These observations demonstrate that IL-33-dependent signaling contributes to the expansion of innate type 2 immunity and subsequent Th2-biased lung immunopathology that facilitates C. neoformans growth and dissemination.


Asunto(s)
Inmunidad Adaptativa/inmunología , Criptococosis/inmunología , Inmunidad Innata/inmunología , Interleucinas/inmunología , Transducción de Señal/inmunología , Animales , Separación Celular , Criptococosis/metabolismo , Cryptococcus neoformans , Modelos Animales de Enfermedad , Citometría de Flujo , Interleucina-33 , Interleucinas/metabolismo , Enfermedades Pulmonares Fúngicas/inmunología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Immunol ; 191(4): 1657-65, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23836062

RESUMEN

IL-27 is an APC-derived IL-6/IL-12 family composite cytokine with multiple functions such as regulation of Th1, Th17, and regulatory T cell differentiation, B cell proliferation, and Ig class switching. The IL-27 complex is formed by the association of the cytokine p28 with the soluble cytokine receptor EBV-induced gene 3 (EBI3). The IL-27 cytokine and soluble receptor subunits p28 and EBI3 can be secreted independently. The p28 subunit has been shown to have IL-27-independent biological activities. We previously demonstrated that p28 can form an alternative composite cytokine with the EBI3 homolog cytokine-like factor 1 (CLF; CRLF1). p28/CLF modulates NK cell activity and CD4 T cell cytokine production in vitro. In this study we used IL-6-dependent plasmacytoma cell line B9 and CD4 T cells from IL-27Rα-deficient mice to demonstrate that p28/CLF activates IL-27-unresponsive cells, indicating that p28/CLF and IL-27 signal through different receptors. The observation that p28/CLF, unlike IL-27, sustains B9 plasmacytoma cell proliferation prompted us to investigate the effects of p28/CLF on mouse B cells. We observed that p28/CLF induces IgM, IgG2c, and IgG1 production and plasma cell differentiation. p28/CLF therefore has the potential to contribute to B and plasma cell function, differentiation, and proliferation in normal and pathological conditions such as Castelman's disease and multiple myeloma.


Asunto(s)
Linfocitos B/citología , Interleucinas/inmunología , Linfopoyesis/fisiología , Células Plasmáticas/citología , Animales , Linfocitos B/inmunología , División Celular , Línea Celular , Femenino , Inmunoglobulinas/biosíntesis , Interleucinas/genética , Quinasas Janus/fisiología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Células Plasmáticas/inmunología , Procesamiento Proteico-Postraduccional , Receptores de Citocinas/deficiencia , Receptores de Citocinas/genética , Receptores de Interleucina , Proteínas Recombinantes de Fusión/inmunología , Factores de Transcripción STAT/fisiología , Transducción de Señal , Células Th2/inmunología , Transfección
19.
Proc Natl Acad Sci U S A ; 108(36): 14896-901, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21856952

RESUMEN

Although a number of studies have examined the development of T-helper cell type 2 (Th2) immunity in different settings, the mechanisms underlying the initiation of this arm of adaptive immunity are not well understood. We exploited the fact that immunization with antigen plus either nucleotide-binding oligomerization domain-containing proteins 1 (Nod1) or 2 (Nod2) agonists drives Th2 induction to understand how these pattern-recognition receptors mediate the development of systemic Th2 immune responses. Here, we show in bone-marrow chimeric mice that Nod1 and Nod2 expression within the stromal compartment is necessary for priming of effector CD4(+) Th2 responses and specific IgG1 antibodies. In contrast, sensing of these ligands by dendritic cells was not sufficient to induce Th2 immunity, although these cells contribute to the response. Moreover, we determined that CD11c(+) cells were the critical antigen-presenting cells, whereas basophils and B cells did not affect the capacity of Nod ligands to induce CD4(+) Th2 effector function. Finally, we found that full Th2 induction upon Nod1 and Nod2 activation was dependent on both thymic stromal lymphopoietin production by the stromal cells and the up-regulation of the costimulatory molecule, OX40 ligand, on dendritic cells. This study provides in vivo evidence of how systemic Th2 immunity is induced in the context of Nod stimulation. Such understanding will influence the rational design of therapeutics that could reprogram the immune system during an active Th1-mediated disease, such as Crohn's disease.


Asunto(s)
Citocinas/inmunología , Proteína Adaptadora de Señalización NOD1/inmunología , Proteína Adaptadora de Señalización NOD2/inmunología , Células Th2/inmunología , Animales , Linfocitos B/inmunología , Basófilos/inmunología , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/terapia , Citocinas/genética , Células Dendríticas/inmunología , Inmunidad Celular/fisiología , Inmunización , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Noqueados , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/genética , Ligando OX40 , Estructura Terciaria de Proteína , Células TH1/inmunología , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología , Linfopoyetina del Estroma Tímico
20.
Front Immunol ; 15: 1330549, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38433831

RESUMEN

Background: Vaccination against COVID-19 is highly effective in preventing severe disease and hospitalization, but primary COVID mRNA vaccination schedules often differed from those recommended by the manufacturers due to supply chain issues. We investigated the impact of delaying the second dose on antibody responses to COVID mRNA-vaccines in a prospective cohort of health-care workers in Quebec. Methods: We recruited participants from the McGill University Health Centre who provided serum or participant-collected dried blood samples (DBS) at 28-days, 3 months, and 6 months post-second dose and at 28-days after a third dose. IgG antibodies to SARS-CoV2 spike (S), the receptor-binding domain (RBD), nucleocapsid (N) and neutralizing antibodies to the ancestral strain were assessed by enzyme-linked immunosorbent assay (ELISA). We examined associations between long (≤89 days) versus short (<89 days) between-dose intervals and antibody response through multivariable mixed-effects models adjusted for age, sex, prior covid infection status, time since vaccine dose, and assay batch. Findings: The cohort included 328 participants who received up to three vaccine doses (>80% Pfizer-BioNTech). Weighted averages of the serum (n=744) and DBS (n=216) cohort results from the multivariable models showed that IgG anti-S was 31% higher (95% CI: 12% to 53%) and IgG anti-RBD was 37% higher (95% CI: 14% to 65%) in the long vs. short interval participants, across all time points. Interpretation: Our study indicates that extending the covid primary series between-dose interval beyond 89 days (approximately 3 months) provides stronger antibody responses than intervals less than 89 days. Our demonstration of a more robust antibody response with a longer between dose interval is reassuring as logistical and supply challenges are navigated in low-resource settings.


Asunto(s)
Formación de Anticuerpos , COVID-19 , Humanos , Estudios Prospectivos , Vacunas contra la COVID-19 , ARN Viral , COVID-19/prevención & control , SARS-CoV-2 , Anticuerpos Neutralizantes , Inmunoglobulina G , ARN Mensajero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA