Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 57(7): 1549-1566.e8, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38776917

RESUMEN

The activities, ontogeny, and mechanisms of lineage expansion of eosinophils are less well resolved than those of other immune cells, despite the use of biological therapies targeting the eosinophilia-promoting cytokine interleukin (IL)-5 or its receptor, IL-5Rα. We combined single-cell proteomics and transcriptomics and generated transgenic IL-5Rα reporter mice to revisit eosinophilopoiesis. We reconciled human and murine eosinophilopoiesis and provided extensive cell-surface immunophenotyping and transcriptomes at different stages along the continuum of eosinophil maturation. We used these resources to show that IL-5 promoted eosinophil-lineage expansion via transit amplification, while its deletion or neutralization did not compromise eosinophil maturation. Informed from our resources, we also showed that interferon response factor-8, considered an essential promoter of myelopoiesis, was not intrinsically required for eosinophilopoiesis. This work hence provides resources, methods, and insights for understanding eosinophil ontogeny, the effects of current precision therapeutics, and the regulation of eosinophil development and numbers in health and disease.


Asunto(s)
Linaje de la Célula , Eosinófilos , Interleucina-5 , Ratones Transgénicos , Proteómica , Análisis de la Célula Individual , Transcriptoma , Eosinófilos/inmunología , Eosinófilos/metabolismo , Animales , Interleucina-5/metabolismo , Interleucina-5/genética , Humanos , Ratones , Proteómica/métodos , Análisis de la Célula Individual/métodos , Diferenciación Celular/inmunología , Ratones Endogámicos C57BL , Perfilación de la Expresión Génica/métodos , Subunidad alfa del Receptor de Interleucina-5/metabolismo , Subunidad alfa del Receptor de Interleucina-5/genética , Mielopoyesis/genética , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Ratones Noqueados
2.
EMBO J ; 41(18): e109353, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35920020

RESUMEN

Macrophage polarization is a process whereby macrophages acquire distinct effector states (M1 or M2) to carry out multiple and sometimes opposite functions. We show here that translational reprogramming occurs during macrophage polarization and that this relies on the Elongator complex subunit Elp3, an enzyme that modifies the wobble uridine base U34 in cytosolic tRNAs. Elp3 expression is downregulated by classical M1-activating signals in myeloid cells, where it limits the production of pro-inflammatory cytokines via FoxO1 phosphorylation, and attenuates experimental colitis in mice. In contrast, alternative M2-activating signals upregulate Elp3 expression through a PI3K- and STAT6-dependent signaling pathway. The metabolic reprogramming linked to M2 macrophage polarization relies on Elp3 and the translation of multiple candidates, including the mitochondrial ribosome large subunit proteins Mrpl3, Mrpl13, and Mrpl47. By promoting translation of its activator Ric8b in a codon-dependent manner, Elp3 also regulates mTORC2 activation. Elp3 expression in myeloid cells further promotes Wnt-driven tumor initiation in the intestine by maintaining a pool of tumor-associated macrophages exhibiting M2 features. Collectively, our data establish a functional link between tRNA modifications, mTORC2 activation, and macrophage polarization.


Asunto(s)
Histona Acetiltransferasas , Activación de Macrófagos , Transducción de Señal , Animales , Codón/metabolismo , Histona Acetiltransferasas/genética , Activación de Macrófagos/genética , Macrófagos/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones
3.
Immunity ; 46(3): 457-473, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28329706

RESUMEN

Living in a microbe-rich environment reduces the risk of developing asthma. Exposure of humans or mice to unmethylated CpG DNA (CpG) from bacteria reproduces these protective effects, suggesting a major contribution of CpG to microbe-induced asthma resistance. However, how CpG confers protection remains elusive. We found that exposure to CpG expanded regulatory lung interstitial macrophages (IMs) from monocytes infiltrating the lung or mobilized from the spleen. Trafficking of IM precursors to the lung was independent of CCR2, a chemokine receptor required for monocyte mobilization from the bone marrow. Using a mouse model of allergic airway inflammation, we found that adoptive transfer of IMs isolated from CpG-treated mice recapitulated the protective effects of CpG when administered before allergen sensitization or challenge. IM-mediated protection was dependent on IL-10, given that Il10-/- CpG-induced IMs lacked regulatory effects. Thus, the expansion of regulatory lung IMs upon exposure to CpG might underlie the reduced risk of asthma development associated with a microbe-rich environment.


Asunto(s)
Quimiotaxis de Leucocito/inmunología , ADN Bacteriano/inmunología , Hipersensibilidad/inmunología , Macrófagos Alveolares/inmunología , Hipersensibilidad Respiratoria/inmunología , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodesoxirribonucleótidos/inmunología , Bazo/inmunología
4.
Blood ; 141(14): 1708-1717, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-36599086

RESUMEN

The downstream signaling of the interleukin-7 (IL-7) receptor (IL-7R) plays important physiological and pathological roles, including the differentiation of lymphoid cells and proliferation of acute lymphoblastic leukemia cells. Gain-of-function mutations in the IL-7Rα chain, the specific component of the receptor for IL-7, result in constitutive, IL-7-independent signaling and trigger acute lymphoblastic leukemia. Here, we show that the loss of the phosphoinositide 5-phosphatase INPP5K is associated with increased levels of the INPP5K substrate phosphatidylinositol 4,5-bisphosphate (PtdIns[4,5]P2) and causes an altered dynamic structure of the IL-7 receptor. We discovered that the IL-7Rα chain contains a very conserved positively charged polybasic amino acid sequence in its cytoplasmic juxtamembrane region; this region establish stronger ionic interactions with negatively charged PtdIns(4,5)P2 in the absence of INPP5K, freezing the IL-7Rα chain structure. This dynamic structural alteration causes defects in IL-7R signaling, culminating in decreased expressions of EBF1 and PAX5 transcription factors, in microdomain formation, cytoskeletal reorganization, and bone marrow B-cell differentiation. Similar alterations after the reduced INPP5K expression also affected mutated, constitutively activated IL-7Rα chains that trigger leukemia development, leading to reduced cell proliferation. Altogether, our results indicate that the lipid 5-phosphatase INPP5K hydrolyzes PtdIns(4,5)P2, allowing the requisite conformational changes of the IL-7Rα chain for optimal signaling.


Asunto(s)
Interleucina-7 , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Interleucina-7/genética , Interleucina-7/metabolismo , Fosfatidilinositol 4,5-Difosfato , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/metabolismo , Transducción de Señal/genética
5.
J Immunol ; 206(5): 1077-1087, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33483347

RESUMEN

The activation of T cells is accompanied by intensive posttranscriptional remodeling of their proteome. We observed that protein expression of enzymes that modify wobble uridine in specific tRNAs, namely elongator subunit 3 (Elp3) and cytosolic thiouridylase (Ctu)2, increased in the course of T cell activation. To investigate the role of these tRNA epitranscriptomic modifiers in T cell biology, we generated mice deficient for Elp3 in T cells. We show that deletion of Elp3 has discrete effects on T cells. In vitro, Elp3-deficient naive CD4+ T cells polarize normally but are delayed in entering the first cell cycle following activation. In vivo, different models of immunization revealed that Elp3-deficient T cells display reduced expansion, resulting in functional impairment of T follicular helper (TFH) responses, but not of other CD4+ effector T cell responses. Transcriptomic analyses identified a progressive overactivation of the stress-responsive transcription factor Atf4 in Elp3-deficient T cells. Overexpression of Atf4 in wild-type T cells phenocopies the effect of Elp3 loss on T cell cycle entry and TFH cell responses. Reciprocally, partial silencing of Atf4 or deletion of its downstream effector transcription factor Chop rescues TFH responses of Elp3-deficient T cells. Together, our results reveal that specific epitranscriptomic tRNA modifications contribute to T cell cycle entry and promote optimal TFH responses.


Asunto(s)
Factor de Transcripción Activador 4/genética , Histona Acetiltransferasas/genética , ARN de Transferencia/genética , Células T Auxiliares Foliculares/inmunología , Uridina/genética , Factor de Transcripción Activador 4/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Ciclo Celular/genética , Ciclo Celular/inmunología , Femenino , Histona Acetiltransferasas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Procesamiento Postranscripcional del ARN/genética , Procesamiento Postranscripcional del ARN/inmunología , ARN de Transferencia/inmunología , Transcriptoma/genética , Transcriptoma/inmunología , Uridina/inmunología
6.
Cell ; 133(6): 1019-31, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18555778

RESUMEN

Oncogene-induced cellular senescence (OIS) is emerging as a potent cancer-protective response to oncogenic events, serving to eliminate early neoplastic cells from the proliferative pool. Using combined genetic and bioinformatic analysis, we find that OIS is linked specifically to the activation of an inflammatory transcriptome. Induced genes included the pleiotropic cytokine interleukin-6 (IL-6), which upon secretion by senescent cells acted mitogenically in a paracrine fashion. Unexpectedly, IL-6 was also required for the execution of OIS, but in a cell-autonomous mode. Its depletion caused the inflammatory network to collapse and abolished senescence entry and maintenance. Furthermore, we demonstrate that the transcription factor C/EBPbeta cooperates with IL-6 to amplify the activation of the inflammatory network, including IL-8. In human colon adenomas, IL-8 specifically colocalized with arrested, p16(INK4A)-positive epithelium. We propose a model in which the context-dependent cytostatic and promitogenic functions of specific interleukins contribute to connect senescence with an inflammatory phenotype and cancer.


Asunto(s)
Senescencia Celular , Inflamación , Interleucina-6/metabolismo , Adenoma/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proliferación Celular , Neoplasias del Colon/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Perfilación de la Expresión Génica , Heterocromatina , Humanos , Interleucina-8/metabolismo , Interferencia de ARN , Regulación hacia Arriba
7.
Eur Respir J ; 59(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34475229

RESUMEN

Neutralising antibodies against the cytokine interleukin (IL)5 have become widely used for the control of severe eosinophilic asthma. Remarkably, patients receiving neutralising anti-IL5 biological therapies retain a very stable population of residual blood eosinophils. Whether these residual eosinophils are endowed with particular biological activity has not yet been studied, but is of importance in predicting potential long-term effects of IL5 neutralisation in patients. To tackle the effect of IL5 depletion on residual eosinophils, we used a comparative RNA-sequencing approach and compared the gene expression programme of eosinophils arising in IL5-depleted or IL5-replete human or murine hosts, at steady-state in vivo and following in vitro stimulation with the eosinophil-activating alarmin IL33. We compared blood eosinophils from patients with severe allergic eosinophilic asthma treated with anti-IL5 mepolizumab therapy to those of healthy controls and matched asthma patients receiving anti-IgE omalizumab therapy. We made similar comparisons on bone marrow eosinophils from mice genetically deficient or not for IL5. We report that restriction of IL5 availability did not elicit any detectable transcriptional response in steady-state residual eosinophils in mepolizumab-treated patients or IL5-deficient mice, and influenced only a handful of genes in their response to IL33. Together, these results support the notion that treatment with IL5 neutralising antibodies spares a pool of circulating residual eosinophils largely resembling those of healthy individuals.


Asunto(s)
Antiasmáticos , Asma , Eosinofilia Pulmonar , Animales , Antiasmáticos/farmacología , Antiasmáticos/uso terapéutico , Anticuerpos Monoclonales Humanizados , Asma/metabolismo , Eosinófilos , Humanos , Interleucina-5 , Ratones , Eosinofilia Pulmonar/inducido químicamente
8.
Int J Mol Sci ; 22(18)2021 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-34576313

RESUMEN

Asthma is now recognized as a heterogeneous disease, encompassing different phenotypes driven by distinct pathophysiological mechanisms called endotypes. Common phenotypes of asthma, referred to as eosinophilic asthma, are characterized by the presence of eosinophilia. Eosinophils are usually considered invariant, terminally differentiated effector cells and have become a primary therapeutic target in severe eosinophilic asthma (SEA) and other eosinophil-associated diseases (EADs). Biological treatments that target eosinophils reveal an unexpectedly complex role of eosinophils in asthma, including in SEA, suggesting that "not all eosinophils are equal". In this review, we address our current understanding of the role of eosinophils in asthma with regard to asthma phenotypes and endotypes. We further address the possibility that different SEA phenotypes may involve differences in eosinophil biology. We discuss how these differences could arise through eosinophil "endotyping", viz. adaptations of eosinophil function imprinted during their development, or through tissue-induced plasticity, viz. local adaptations of eosinophil function through interaction with their lung tissue niches. In doing so, we also discuss opportunities, technical challenges, and open questions that, if addressed, might provide considerable benefits in guiding the choice of the most efficient precision therapies of SEA and, by extension, other EADs.


Asunto(s)
Asma/metabolismo , Eosinófilos/metabolismo , Animales , Humanos , Inmunoterapia/métodos
9.
Proc Natl Acad Sci U S A ; 114(43): E9056-E9065, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29073102

RESUMEN

It has been shown that γδ T cells protect against the formation of squamous cell carcinoma (SCC) in several models. However, the role of γδ T cells in human papillomavirus (HPV)-associated uterine cervical SCC, the third-leading cause of death by cancer in women, is unknown. Here, we investigated the impact of γδ T cells in a transgenic mouse model of carcinogenesis induced by HPV16 oncoproteins. Surprisingly, γδ T cells promoted the development of HPV16 oncoprotein-induced lesions. HPV16 oncoproteins induced a decrease in epidermal Skint1 expression and the associated antitumor Vγ5+ γδ T cells, which were replaced by γδ T-cell subsets (mainly Vγ6+ γδlowCCR2+CCR6-) actively producing IL-17A. Consistent with a proangiogenic role, γδ T cells promoted the formation of blood vessels in the dermis underlying the HPV-induced lesions. In human cervical biopsies, IL-17A+ γδ T cells could only be observed at the cancer stage (SCC), where HPV oncoproteins are highly expressed, supporting the clinical relevance of our observations in mice. Overall, our results suggest that HPV16 oncoproteins induce a reorganization of the local epithelial-associated γδ T-cell subpopulations, thereby promoting angiogenesis and cancer development.


Asunto(s)
Linfocitos Intraepiteliales/patología , Linfocitos Intraepiteliales/virología , Neoplasias de Células Escamosas/virología , Infecciones por Papillomavirus/patología , Neoplasias del Cuello Uterino/virología , Animales , Cuello del Útero , Epidermis/patología , Epidermis/virología , Femenino , Humanos , Inmunoglobulinas/metabolismo , Interleucina-17/metabolismo , Ratones Transgénicos , Neoplasias de Células Escamosas/patología , Neovascularización Patológica , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Receptores CCR2/metabolismo , Receptores CCR6/metabolismo , Proteínas Represoras/metabolismo , Neoplasias del Cuello Uterino/patología
10.
Cell Immunol ; 330: 91-96, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29458975

RESUMEN

Lung macrophages have mostly been studied considering only their most accessible and well-defined representative, the alveolar macrophage (AM). In contrast, the identity and putative immune functions of their tissue counterpart, the interstitial macrophage (IM), have long remained much more elusive. Yet, recent evidence supports the notion that IMs perform important immune functions in the lung, notably in terms of innate immunoregulation. Here, we review current knowledge on the phenotype, ontogeny and function of IMs and propose strategies for the unambiguous identification and study of this important and dynamic lung innate immune cell population.


Asunto(s)
Interleucina-10/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Macrófagos/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Humanos , Interleucina-10/metabolismo , Pulmón/citología , Pulmón/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Modelos Inmunológicos , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Oligodesoxirribonucleótidos/inmunología , Oligodesoxirribonucleótidos/farmacología
11.
Eur J Immunol ; 46(11): 2614-2628, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27546168

RESUMEN

Very few transcription factors have been identified that are required by antigen-presenting cells (APCs) to induce T helper type 2 (Th2) responses. Because lung CD11b+ conventional dendritic cells (CD11b+ cDCs) are responsible for priming Th2 responses in house-dust mite (HDM)-induced airway allergy, we used them as a model to identify transcriptional events regulating the pro-Th2 activity of cDCs. Transcriptomic profiling of lung CD11b+ cDCs exposed to HDM in vivo revealed first that HDM triggers an antiviral defence-like response, and second that the majority of HDM-induced transcriptional changes depend on the transcription factor Interferon Response Factor-3 (Irf3). Validating the functional relevance of these observations, Irf3-deficient CD11b+ cDCs displayed reduced pro-allergic activity. Indeed, Irf3-deficient CD11b+ cDCs induced less Th2, more regulatory T cell, and similar Th1 differentiation in naïve CD4+ T cells compared to their wild-type counterparts. The altered APC activity of Irf3 CD11b+ cDCs was associated with reduced expression of CD86 and was phenocopied by blocking CD86 activity in wild-type CD11b+ cDCs. Altogether, these results establish Irf3, known mostly for its role in antiviral responses, as a transcription factor involved in the induction of Th2 responses through the promotion of pro-Th2 costimulation in CD11b+ DCs.


Asunto(s)
Alérgenos/inmunología , Antígenos Dermatofagoides/inmunología , Células Dendríticas/inmunología , Factor 3 Regulador del Interferón/metabolismo , Pulmón/inmunología , Células Th2/inmunología , Factores de Transcripción/inmunología , Administración Intranasal , Animales , Antígenos Dermatofagoides/administración & dosificación , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Diferenciación Celular , Factor 3 Regulador del Interferón/deficiencia , Factor 3 Regulador del Interferón/genética , Ratones , Ratones Noqueados , Análisis por Micromatrices , Fenotipo
13.
Proc Natl Acad Sci U S A ; 110(13): 5139-44, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23483055

RESUMEN

Metastasis confronts clinicians with two major challenges: estimating the patient's risk of metastasis and identifying therapeutic targets. Because they are key signal integrators connecting cellular processes to clinical outcome, we aimed to identify transcriptional nodes regulating cancer cell metastasis. Using rodent xenograft models that we previously developed, we identified the transcription factor Fos-related antigen-1 (Fra-1) as a key coordinator of metastasis. Because Fra-1 often is overexpressed in human metastatic breast cancers and has been shown to control their invasive potential in vitro, we aimed to assess the implication and prognostic significance of the Fra-1-dependent genetic program in breast cancer metastasis and to identify potential Fra-1-dependent therapeutic targets. In several in vivo assays in mice, we demonstrate that stable RNAi depletion of Fra-1 from human breast cancer cells strongly suppresses their ability to metastasize. These results support a clinically important role for Fra-1 and the genetic program it controls. We show that a Fra-1-dependent gene-expression signature accurately predicts recurrence of breast cancer. Furthermore, a synthetic lethal drug screen revealed that antagonists of the adenosine receptor A2B (ADORA2B) are preferentially toxic to breast tumor cells expressing Fra-1. Both RNAi silencing and pharmacologic blockade of ADORA2B inhibited filopodia formation and invasive activity of breast cancer cells and correspondingly reduced tumor outgrowth in the lungs. These data show that Fra-1 activity is causally involved in and is a prognostic indicator of breast cancer metastasis. They suggest that Fra-1 activity predicts responsiveness to inhibition of pharmacologically tractable targets, such as ADORA2B, which may be used for clinical interference of metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor de Adenosina A2B/metabolismo , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-fos/genética , Seudópodos/genética , Seudópodos/metabolismo , Seudópodos/patología , Ratas , Receptor de Adenosina A2B/genética , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
PLoS Pathog ; 9(11): e1003773, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278020

RESUMEN

Interferons (IFNs) are a group of cytokines with a well-established antiviral function. They can be induced by viral infection, are secreted and bind to specific receptors on the same or neighbouring cells to activate the expression of hundreds of IFN stimulated genes (ISGs) with antiviral function. Type I IFN has been known for more than half a century. However, more recently, type III IFN (IFNλ, IL-28/29) was shown to play a similar role and to be particularly important at epithelial surfaces. Here we show that airway epithelia, the primary target of influenza A virus, produce both IFN I and III upon infection, and that induction of both depends on the RIG-I/MAVS pathway. While IRF3 is generally regarded as the transcription factor required for initiation of IFN transcription and the so-called "priming loop", we find that IRF3 deficiency has little impact on IFN expression. In contrast, lack of IRF7 reduced IFN production significantly, and only IRF3(-/-)IRF7(-/-) double deficiency completely abolished it. The transcriptional response to influenza infection was largely dependent on IFNs, as it was reduced to a few upregulated genes in epithelia lacking receptors for both type I and III IFN (IFNAR1(-/-)IL-28Rα(-/-)). Wild-type epithelia and epithelia deficient in either the type I IFN receptor or the type III IFN receptor exhibit similar transcriptional profiles in response to virus, indicating that none of the induced genes depends selectively on only one IFN system. In chimeric mice, the lack of both IFN I and III signalling in the stromal compartment alone significantly increased the susceptibility to influenza infection. In conclusion, virus infection of airway epithelia induces, via a RIG-I/MAVS/IRF7 dependent pathway, both type I and III IFNs which drive two completely overlapping and redundant amplification loops to upregulate ISGs and protect from influenza infection.


Asunto(s)
Células Epiteliales/metabolismo , Virus de la Influenza A/metabolismo , Interferón Tipo I/metabolismo , Interleucinas/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Mucosa Respiratoria/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interleucinas/genética , Interleucinas/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Receptores de Superficie Celular , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología
15.
Vet Res ; 46: 3, 2015 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-25595212

RESUMEN

Respiratory diseases, including inflammatory airway disease (IAD), viral and bacterial infections, are common problems in exercising horses. The airway epithelium constitutes a major physical barrier against airborne infections and plays an essential role in the lung innate immune response mainly through toll-like receptor (TLR) activation. The aim of this study was to develop a model for the culture of equine bronchial epithelial cells (EBEC) in vitro and to explore EBEC innate immune responses in trained horses. Bronchial epithelial biopsies were taken from 6 adult horses during lower airway endoscopy. EBEC were grown in vitro by an explant method. The innate immune response of EBEC was evaluated in vitro by treatment with TLR ligands. TLR3 is the most strongly expressed TLR at the mRNA level in EBEC and stimulation of EBEC with Poly(I:C), an analog of viral dsRNA, triggers a strong secretion of IFN-ß, TNF-α, IL-6 and CXCL8. We further evaluated the EBEC innate immune response in horses that underwent a 4-month-training program. While training had no effect on TLR mRNA expression in EBEC as well as in bronchial biopsies, it increased the production of IFN-ß after stimulation with a TLR3 ligand and decreased the secretion of TNF-α and IL-6 after stimulation with a TLR2 and TLR3 ligand. These findings may be implicated in the increased risk for viral and bacterial infections observed in sport horses. Altogether, we report a successful model for the culture of EBEC that can be applied to the investigation of pathophysiologic conditions in longitudinal studies.


Asunto(s)
Caballos/fisiología , Inmunidad Innata , Condicionamiento Físico Animal , Mucosa Respiratoria/inmunología , Receptores Toll-Like/genética , Animales , Bronquios/citología , Bronquios/inmunología , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Citocinas/metabolismo , Femenino , Caballos/genética , Caballos/inmunología , Masculino , Poli I-C/farmacología , Mucosa Respiratoria/citología , Receptores Toll-Like/metabolismo
16.
Am J Respir Cell Mol Biol ; 51(1): 135-42, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24502337

RESUMEN

In humans, strenuous exercise causes increased susceptibility to respiratory infections associated with down-regulated expression of Toll-like receptors (TLRs) and costimulatory and antigen-presenting molecules. Lower airway diseases are also a common problem in sport and racing horses. Because innate immunity plays an essential role in lung defense mechanisms, we assessed the effect of acute exercise and training on innate immune responses in two different compartments. Blood monocytes and pulmonary alveolar macrophages (PAMs) were collected from horses in untrained, moderately trained, intensively trained, and deconditioned states before and after a strenuous exercise test. The cells were analyzed for TLR messenger ribonucleic acid (mRNA) expression by real-time PCR in vitro, and cytokine production after in vitro stimulation with TLR ligands was measured by ELISA. Our results showed that training, but not acute exercise, modified the innate immune responses in both compartments. The mRNA expression of TLR3 was down-regulated by training in both cell types, whereas the expression of TLR4 was up-regulated in monocytes. Monocytes treated with LPS and a synthetic diacylated lipoprotein showed increased cytokine secretion in trained and deconditioned subjects, indicating the activation of cells at the systemic level. The production of TNF-α and IFN-ß in nonstimulated and stimulated PAMs was decreased in trained and deconditioned horses and might therefore explain the increased susceptibility to respiratory infections. Our study reports a dissociation between the systemic and the lung response to training that is probably implicated in the systemic inflammation and in the pulmonary susceptibility to infection.


Asunto(s)
Inmunidad Innata/inmunología , Inflamación/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Condicionamiento Físico Animal , Receptores Toll-Like/metabolismo , Animales , Western Blotting , Femenino , Caballos , Hidrocortisona/metabolismo , Inflamación/metabolismo , Inflamación/patología , Interferón beta/genética , Interferón beta/metabolismo , Estudios Longitudinales , Pulmón/metabolismo , Macrófagos Alveolares/metabolismo , Monocitos/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
J Immunol ; 189(11): 5393-401, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23109724

RESUMEN

Lyme disease is caused by spirochetes of the Borrelia burgdorferi sensu lato complex. They are transmitted mainly by Ixodes ricinus ticks. After a few hours of infestation, neutrophils massively infiltrate the bite site. They can kill Borrelia via phagocytosis, oxidative burst, and hydrolytic enzymes. However, factors in tick saliva promote propagation of the bacteria in the host even in the presence of a large number of neutrophils. The neutrophil extracellular trap (NET) consists in the extrusion of the neutrophil's own DNA, forming traps that can retain and kill bacteria. The production of reactive oxygen species is apparently associated with the onset of NETs (NETosis). In this article, we describe NET formation at the tick bite site in vivo in mice. We show that Borrelia burgdorferi sensu stricto spirochetes become trapped and killed by NETs in humans and that the bacteria do not seem to release significant nucleases to evade this process. Saliva from I. ricinus did not affect NET formation by human neutrophils or its stability. However, it greatly decreased neutrophil reactive oxygen species production, suggesting that a strong decrease of hydrogen peroxide does not affect NET formation. Finally, round bodies trapped in NETs were observed, some of them staining as live bacteria. This observation could help contribute to a better understanding of the early steps of Borrelia invasion and erythema migrans formation after tick bite.


Asunto(s)
Vectores Arácnidos/inmunología , Mordeduras y Picaduras , Grupo Borrelia Burgdorferi/fisiología , Glositis Migratoria Benigna/inmunología , Ixodes/inmunología , Enfermedad de Lyme/inmunología , Neutrófilos/inmunología , Saliva/inmunología , Animales , Vectores Arácnidos/microbiología , ADN/inmunología , Femenino , Glositis Migratoria Benigna/complicaciones , Glositis Migratoria Benigna/microbiología , Glositis Migratoria Benigna/patología , Humanos , Ixodes/microbiología , Enfermedad de Lyme/complicaciones , Enfermedad de Lyme/microbiología , Enfermedad de Lyme/patología , Masculino , Ratones , Infiltración Neutrófila , Neutrófilos/metabolismo , Conejos , Especies Reactivas de Oxígeno/inmunología , Saliva/química
18.
J Immunol ; 187(9): 4517-29, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21948987

RESUMEN

Sirtuins are a unique class of NAD(+)-dependent deacetylases that regulate diverse biological functions such as aging, metabolism, and stress resistance. Recently, it has been shown that sirtuins may have anti-inflammatory activities by inhibiting proinflammatory transcription factors such as NF-κB. In contrast, we report in this study that pharmacological inhibition of sirtuins dampens adaptive Th2 responses and subsequent allergic inflammation by interfering with lung dendritic cell (DC) function in a mouse model of airway allergy. Using genetic engineering, we demonstrate that sirtuin 1 represses the activity of the nuclear receptor peroxisome proliferator-activated receptor-γ in DCs, thereby favoring their maturation toward a pro-Th2 phenotype. This study reveals a previously unappreciated function of sirtuin 1 in the regulation of DC function and Th2 responses, thus shedding new light on our current knowledge on the regulation of inflammatory processes by sirtuins.


Asunto(s)
Asma/inmunología , Células Dendríticas/inmunología , PPAR gamma/antagonistas & inhibidores , Sirtuina 1/fisiología , Células Th2/inmunología , Animales , Asma/enzimología , Asma/patología , Hiperreactividad Bronquial/genética , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Inmunofenotipificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , PPAR gamma/metabolismo , Sirtuina 1/antagonistas & inhibidores , Células Th2/enzimología , Células Th2/patología
19.
Eur J Immunol ; 41(2): 380-91, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21268008

RESUMEN

Asthma is a complex disease linked to various pathophysiological events including the activity of proteinases. The multifunctional A disintegrin and metalloproteinases (ADAMs) displaying the ability to cleave membrane-bound mediators or cytokines appear to be key mediators in various inflammatory processes. In the present study, we investigated ADAM-8 expression and production in a mouse model of allergen-induced airway inflammation. In allergen-exposed animals, increased expression of ADAM-8 was found in the lung parenchyma and in DC purified from the lungs. The potential role of ADAM-8 in the development of allergen-induced airway inflammation was further investigated by the use of an anti-ADAM-8 antibody and ADAM-8 knockout animals. We observed a decrease in allergen-induced acute inflammation both in BALF and the peribronchial area in anti-ADAM-8 antibody-treated mice and in ADAM-8-deficient mice (ADAM-8(-/-) ) after allergen exposure. ADAM-8 depletion led to a significant decrease of the CD11c(+) lung DC. We also report lower levels of CCL11 and CCL22 production in antibody-treated mice and ADAM-8- deficient mice that might be explained by decreased eosinophilic inflammation and lower numbers of DC, respectively. In conclusion, ADAM-8 appears to favour allergen-induced acute airway inflammation by promoting DC recruitment and CCL11 and CCL22 production.


Asunto(s)
Proteínas ADAM/metabolismo , Antígenos CD/metabolismo , Asma/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Antígenos CD/genética , Antígenos CD/inmunología , Asma/inmunología , Asma/patología , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Movimiento Celular/genética , Movimiento Celular/inmunología , Quimiocina CCL11/metabolismo , Quimiocina CCL22/metabolismo , Citocinas/metabolismo , Eosinófilos/metabolismo , Eosinófilos/patología , Expresión Génica/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/inmunología , Vacunación
20.
BMC Vet Res ; 8: 64, 2012 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-22621400

RESUMEN

BACKGROUND: Recurrent airway obstruction (RAO, also known as equine heaves) is an inflammatory condition caused by exposure of susceptible horses to organic dusts in hay. The immunological processes responsible for the development and the persistence of airway inflammation are still largely unknown. Hypoxia-inducible factor (Hif) is mainly known as a major regulator of energy homeostasis and cellular adaptation to hypoxia. More recently however, Hif also emerged as an essential regulator of innate immune responses. Here, we aimed at investigating the potential involvement of Hif1-α in myeloid cells in horse with recurrent airway obstruction. RESULTS: In vitro, we observed that Hif is expressed in equine myeloid cells after hay dust stimulation and regulates genes such as tumor necrosis factor alpha (TNF-α), interleukin-8 (IL-8) and vascular endothelial growth factor A (VEGF-A). We further showed in vivo that airway challenge with hay dust upregulated Hif1-α mRNA expression in myeloid cells from the bronchoalveolar lavage fluid (BALF) of healthy and RAO-affected horses, with a more pronounced effect in cells from RAO-affected horses. Finally, Hif1-α mRNA expression in BALF cells from challenged horses correlated positively with lung dysfunction. CONCLUSION: Taken together, our results suggest an important role for Hif1-α in myeloid cells during hay dust-induced inflammation in horses with RAO. We therefore propose that future research aiming at functional inactivation of Hif1 in lung myeloid cells could open new therapeutic perspectives for RAO.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Enfermedades de los Caballos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Enfermedades Pulmonares Obstructivas/veterinaria , Animales , Antitusígenos/farmacología , Células Cultivadas , Polvo , Caballos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inflamación/metabolismo , Inflamación/veterinaria , Interleucina-8/genética , Interleucina-8/metabolismo , Pulmón/citología , Pulmón/metabolismo , Enfermedades Pulmonares Obstructivas/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Noscapina/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA