Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1238221, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37809058

RESUMEN

Introduction: Previous work in humans has demonstrated that both innate and adaptive immune signaling pathways contribute to the pathogenesis of idiopathic inflammatory myopathy (IIM), a systemic autoimmune disease targeting muscle as well as extra-muscular organs. To better define interactive signaling networks in IIM, we characterized the cellular phenotype and transcriptomic profiles of muscle-infiltrating cells in our established murine model of histidyl-tRNA synthetase (HRS)-induced myositis. Methods: Myositis was induced in wild type (WT) and various congenic/mutant strains of C57BL/6 mice through intramuscular immunization with recombinant HRS. Histopathological, immunohistochemical, flow cytometric, and transcriptomic assessments were used to characterize the functional relationship between muscle-infiltrating cell populations in these strains lacking different components of innate and/or adaptive immune signaling. Results: RAG1 KO mice developed markedly reduced muscle inflammation relative to WT mice, demonstrating a key requirement for T cells in driving HRS-induced myositis. While the reduction of mononuclear cell infiltrates in CD4-Cre.MyD88fl/fl conditional knockout mice and OT-II TCR transgenic mice highlighted roles for both innate and TCR-mediated/adaptive immune signaling in T cells, diminished inflammation in Lyz2-Cre.MyD88fl/fl conditional knockout mice underscored the importance of macrophage/myeloid cell populations in supporting T cell infiltration. Single cell RNA sequencing-based clustering of muscle-infiltrating subpopulations and associated pathway analyses showed that perturbations of T cell signaling/function alter the distribution and phenotype of macrophages, fibroblasts, and other non-lymphoid cell populations contributing to HRS-induced myositis. Discussion: Overall, HRS-induced myositis reflects the complex interplay between multiple cell types that collectively drive a TH1-predominant, pro-inflammatory tissue phenotype requiring antigen-mediated activation of both MyD88- and TCR-dependent T cell signaling pathways.


Asunto(s)
Histidina-ARNt Ligasa , Miositis , Humanos , Ratones , Animales , Linfocitos T , Ratones Endogámicos C57BL , Inmunidad Adaptativa , Macrófagos , Inflamación , Ratones Noqueados , Receptores de Antígenos de Linfocitos T
2.
J Allergy Clin Immunol ; 152(1): 94-106.e12, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36893862

RESUMEN

BACKGROUND: Type 1 (T1) inflammation (marked by IFN-γ expression) is now consistently identified in subsets of asthma cohorts, but how it contributes to disease remains unclear. OBJECTIVE: We sought to understand the role of CCL5 in asthmatic T1 inflammation and how it interacts with both T1 and type 2 (T2) inflammation. METHODS: CCL5, CXCL9, and CXCL10 messenger RNA expression from sputum bulk RNA sequencing, as well as clinical and inflammatory data were obtained from the Severe Asthma Research Program III (SARP III). CCL5 and IFNG expression from bronchoalveolar lavage cell bulk RNA sequencing was obtained from the Immune Mechanisms in Severe Asthma (IMSA) cohort and expression related to previously identified immune cell profiles. The role of CCL5 in tissue-resident memory T-cell (TRM) reactivation was evaluated in a T1high murine severe asthma model. RESULTS: Sputum CCL5 expression strongly correlated with T1 chemokines (P < .001 for CXCL9 and CXCL10), consistent with a role in T1 inflammation. CCL5high participants had greater fractional exhaled nitric oxide (P = .009), blood eosinophils (P < .001), and sputum eosinophils (P = .001) in addition to sputum neutrophils (P = .001). Increased CCL5 bronchoalveolar lavage expression was unique to a previously described T1high/T2variable/lymphocytic patient group in the IMSA cohort, with IFNG trending with worsening lung obstruction only in this group (P = .083). In a murine model, high expression of the CCL5 receptor CCR5 was observed in TRMs and was consistent with a T1 signature. A role for CCL5 in TRM activation was supported by the ability of the CCR5 inhibitor maraviroc to blunt reactivation. CONCLUSION: CCL5 appears to contribute to TRM-related T1 neutrophilic inflammation in asthma while paradoxically also correlating with T2 inflammation and with sputum eosinophilia.


Asunto(s)
Asma , Quimiocina CCL5 , Animales , Humanos , Ratones , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocinas/metabolismo , Eosinófilos , Inflamación/metabolismo , Neutrófilos , Esputo
3.
Mucosal Immunol ; 15(1): 154-164, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34580428

RESUMEN

Immune tolerance to allergens in early-life decreases the risk for asthma in later life. Here we show establishment of stable airway tolerance to the allergen, house dust mite (HDM), by exposing newborn mice repeatedly to a low dose of the allergen. Lung dendritic cells (DCs) from tolerized mice induced a low Th2 response in vitro mirroring impact of tolerance in vivo. In line with our previous finding of increased mitochondrial H2O2 production from lung DCs of mice tolerized to ovalbumin, depletion of mitochondrial H2O2 in MCAT mice abrogated HDM-induced airway tolerance (Tol) with elevated Th2 effector response, airway eosinophilia, and increased airway hyperreactivity. WT-Tol mice displayed a decrease in total, cDC1 and cDC2 subsets in the lung as compared to that in naive mice. In contrast, the lungs of MCAT-Tol mice showed 3-fold higher numbers of cDCs including those of the subsets as compared to that in WT mice. Our study demonstrates an important role of mitochondrial H2O2 in constraining lung DC numbers towards establishment of early-life airway tolerance to allergens.


Asunto(s)
Asma/inmunología , Células Dendríticas/inmunología , Hipersensibilidad/inmunología , Pulmón/patología , Mitocondrias/metabolismo , Células Th2/inmunología , Alérgenos/inmunología , Animales , Animales Recién Nacidos , Antígenos Dermatofagoides/inmunología , Modelos Animales de Enfermedad , Humanos , Peróxido de Hidrógeno/metabolismo , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pyroglyphidae
4.
J Allergy Clin Immunol ; 149(1): 113-124.e7, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34146578

RESUMEN

BACKGROUND: Many patients with severe asthma (SA) fail to respond to type 2 inflammation-targeted therapies. We previously identified a cohort of subjects with SA expressing type 1 inflammation manifesting with IFN-γ expression and variable type 2 responses. OBJECTIVE: We investigated the role of the chemotactic receptors C-X-C chemokine receptor 3 (CXCR3) and C-C chemokine receptor 5 (CCR5) in establishing type 1 inflammation in SA. METHODS: Bronchoalveolar lavage microarray data from the Severe Asthma Research Program I/II were analyzed for pathway expression and paired with clinical parameters. Wild-type, Cxcr3-/-, and Ccr5-/- mice were exposed to a type 1-high SA model with analysis of whole lung gene expression and histology. Wild-type and Cxcr3-/- mice were treated with a US Food and Drug Administration-approved CCR5 inhibitor (maraviroc) with assessment of airway resistance, inflammatory cell recruitment by flow cytometry, whole lung gene expression, and histology. RESULTS: A cohort of subjects with increased IFN-γ expression showed higher asthma severity. IFN-γ expression was correlated with CXCR3 and CCR5 expression, but in Cxcr3-/- and Ccr5-/- mice type 1 inflammation was preserved in a murine SA model, most likely owing to compensation by the other pathway. Incorporation of maraviroc into the experimental model blunted airway hyperreactivity despite only mild effects on lung inflammation. CONCLUSIONS: IFNG expression in asthmatic airways was strongly correlated with expression of both the chemokine receptors CXCR3 and CCR5. Although these pathways provide redundancy for establishing type 1 lung inflammation, inhibition of the CCL5/CCR5 pathway with maraviroc provided unique benefits in reducing airway hyperreactivity. Targeting this pathway may be a novel approach for improving lung function in individuals with type 1-high asthma.


Asunto(s)
Asma/inmunología , Receptores CCR5/inmunología , Receptores CXCR3/inmunología , Adulto , Resistencia de las Vías Respiratorias , Animales , Asma/tratamiento farmacológico , Asma/fisiopatología , Bronquios/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Antagonistas de los Receptores CCR5/uso terapéutico , Femenino , Humanos , Inflamación/inmunología , Inflamación/fisiopatología , Interferón gamma/inmunología , Masculino , Maraviroc/uso terapéutico , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Receptores CCR5/genética , Receptores CXCR3/genética , Mucosa Respiratoria/inmunología , Índice de Severidad de la Enfermedad , Adulto Joven
5.
Cell Rep ; 35(2): 108974, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852838

RESUMEN

Clinical definitions of asthma fail to capture the heterogeneity of immune dysfunction in severe, treatment-refractory disease. Applying mass cytometry and machine learning to bronchoalveolar lavage (BAL) cells, we find that corticosteroid-resistant asthma patients cluster largely into two groups: one enriched in interleukin (IL)-4+ innate immune cells and another dominated by interferon (IFN)-γ+ T cells, including tissue-resident memory cells. In contrast, BAL cells of a healthier population are enriched in IL-10+ macrophages. To better understand cellular mediators of severe asthma, we developed the Immune Cell Linkage through Exploratory Matrices (ICLite) algorithm to perform deconvolution of bulk RNA sequencing of mixed-cell populations. Signatures of mitosis and IL-7 signaling in CD206-FcεRI+CD127+IL-4+ innate cells in one patient group, contrasting with adaptive immune response in T cells in the other, are preserved across technologies. Transcriptional signatures uncovered by ICLite identify T-cell-high and T-cell-poor severe asthma patients in an independent cohort, suggesting broad applicability of our findings.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Aprendizaje Automático , Macrófagos/inmunología , Inmunidad Adaptativa , Corticoesteroides/uso terapéutico , Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Asma/genética , Asma/patología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Estudios de Casos y Controles , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Memoria Inmunológica , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-7/genética , Interleucina-7/inmunología , Macrófagos/patología , Proteómica/métodos , Receptores de IgE/genética , Receptores de IgE/inmunología , Índice de Severidad de la Enfermedad , Transducción de Señal
6.
Curr Opin Immunol ; 72: 34-42, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33773471

RESUMEN

Asthma as a clinical entity manifests with a broad spectrum of disease severity. Unlike milder asthma, severe disease is poorly controlled by inhaled corticosteroids, the current standard of care. Transcriptomic data, along with patient characteristics and response to biologics show that though Type 2 (T2) immune response remains an integral feature of asthma, additional molecular and immunologic factors may play important roles in pathogenesis. Mechanisms of T2 development, cellular sources of T2 cytokines and their relationship to additional immune pathways concurrently activated may distinguish several different subphenotypes, and perhaps endotypes of asthma, with differential response to non-specific and targeted anti-inflammatory therapies. Recent data have also associated non-T2 cytokines derived from T cells, particularly IFN-γ, and epithelial mediators with severe asthma. These topics and their relationships to acute asthma exacerbations are discussed in this review.


Asunto(s)
Asma/diagnóstico , Asma/inmunología , Susceptibilidad a Enfermedades/inmunología , Inmunidad , Alérgenos/inmunología , Animales , Asma/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Eosinófilos/inmunología , Eosinófilos/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunización , Neutrófilos/inmunología , Neutrófilos/metabolismo , Especificidad de Órganos/inmunología , Índice de Severidad de la Enfermedad , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
7.
JCI Insight ; 2(17)2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28878128

RESUMEN

Respiratory syncytial virus (RSV) infects almost all infants by 2 years of age, and severe bronchiolitis resulting from RSV infection is the primary cause of hospitalization in the first year of life. Among infants hospitalized due to RSV-induced bronchiolitis, those with a specific mutation in the chemokine receptor CX3CR1, which severely compromises binding of its ligand CX3CL1, were at a higher risk for more severe viral bronchiolitis than those without the mutation. Here, we show that RSV infection of newborn mice deficient in CX3CR1 leads to significantly greater neutrophilic inflammation in the lungs, accompanied by an increase in mucus production compared with that induced in WT mice. Analysis of innate and adaptive immune responses revealed an early increase in the number of IL-17+ γδ T cells in CX3CR1-deficient mice that outnumbered IFN-γ+ γδ T cells as well as IFN-γ+ NK cells, IFN-γ being host protective in the context of RSV infection. This bias toward IL-17+ γδ T cells persisted at a later time. The lungs of CX3CR1-deficient mice expressed higher levels of IL-1ß mRNA and protein, and blockade of IL-1ß signaling using IL-1 receptor antagonist significantly reduced the number of IL-17+ γδ T cells in the lungs of infected mice. Blockade of IL-17RC abolished RSV-induced lung pathology in infected CX3CR1-deficient mice. We propose that, in infants harboring mutant CX3CR1, targeting the IL-17R may minimize disease severity and hospitalization in early life.


Asunto(s)
Animales Recién Nacidos , Receptor 1 de Quimiocinas CX3C/genética , Inmunidad Innata , Pulmón/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Animales , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ratones , ARN Mensajero/metabolismo , Transducción de Señal
8.
JCI Insight ; 2(13)2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28679952

RESUMEN

We previously showed that Th1/type 1 inflammation marked by increased IFN-γ levels in the airways can be appreciated in 50% of patients with severe asthma, despite high dose corticosteroid (CS) treatment. We hypothesized that a downstream target of IFN-γ, CXCL10, which recruits Th1 cells via the cognate receptor CXCR3, is an important contributor to Th1high asthma and CS unresponsiveness. We show high levels of CXCL10 mRNA closely associated with IFNG levels in the BAL cells of 50% of severe asthmatics and also in the airways of mice subjected to a severe asthma model, both in the context of high-dose CS treatment. The inability of CS to dampen IFNG or CXCL10 expression was not because of impaired nuclear translocation of the glucocorticoid receptor (GR) or its transactivational functions. Rather, in the presence of CS and IFN-γ, STAT1 and GR were recruited on critical regulatory elements in the endogenous CXCL10 promoter in monocytes, albeit without any abatement of CXCL10 gene expression. High CXCL10 gene expression was also associated with a mast cell signature in both humans and mice, CXCR3 being also expressed by mast cells. These findings suggest that the IFN-γ-CXCL10 axis plays a central role in persistent type 1 inflammation that may be facilitated by CS therapy through GR-STAT1 cooperation converging on the CXCL10 promoter.

9.
JCI Insight ; 2(10)2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28515358

RESUMEN

Severe asthma (SA) is a significant problem both clinically and economically, given its poor response to corticosteroids (CS). We recently reported a complex type 1-dominated (IFN-γ-dominated) immune response in more than 50% of severe asthmatics despite high-dose CS treatment. Also, IFN-γ was found to be critical for increased airway hyperreactivity (AHR) in our model of SA. The transcription factor IRF5 expressed in M1 macrophages can induce a Th1/Th17 response in cocultured human T cells. Here we show markedly higher expression of IRF5 in bronchoalveolar lavage (BAL) cells of severe asthmatics as compared with that in cells from milder asthmatics or healthy controls. Using our SA mouse model, we demonstrate that lack of IRF5 in lymph node migratory DCs severely limits their ability to stimulate the generation of IFN-γ- and IL-17-producing CD4+ T cells and IRF5-/- mice subjected to the SA model displayed significantly lower IFN-γ and IL-17 responses, albeit showing a reciprocal increase in Th2 response. However, the absence of IRF5 rendered the mice responsive to CS with suppression of the heightened Th2 response. These data support the notion that IRF5 inhibition in combination with CS may be a viable approach to manage disease in a subset of severe asthmatics.

10.
Nat Commun ; 8: 13944, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28074841

RESUMEN

Bacterial pneumonia is a significant healthcare burden worldwide. Failure to resolve inflammation after infection precipitates lung injury and an increase in morbidity and mortality. Gram-negative bacteria are common in pneumonia and increased levels of the mito-damage-associated molecular pattern (DAMP) cardiolipin can be detected in the lungs. Here we show that mice infected with Klebsiella pneumoniae develop lung injury with accumulation of cardiolipin. Cardiolipin inhibits resolution of inflammation by suppressing production of anti-inflammatory IL-10 by lung CD11b+Ly6GintLy6CloF4/80+ cells. Cardiolipin induces PPARγ SUMOylation, which causes recruitment of a repressive NCOR/HDAC3 complex to the IL-10 promoter, but not the TNF promoter, thereby tipping the balance towards inflammation rather than resolution. Inhibition of HDAC activity by sodium butyrate enhances recruitment of acetylated histone 3 to the IL-10 promoter and increases the concentration of IL-10 in the lungs. These findings identify a mechanism of persistent inflammation during pneumonia and indicate the potential of HDAC inhibition as a therapy.


Asunto(s)
Cardiolipinas/fisiología , Inflamación/metabolismo , Interleucina-10/biosíntesis , Infecciones por Klebsiella/fisiopatología , Klebsiella pneumoniae/aislamiento & purificación , Neumonía Bacteriana/metabolismo , Animales , Cardiolipinas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Interleucina-10/genética , Interleucina-10/metabolismo , Infecciones por Klebsiella/microbiología , Lipopolisacáridos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Supresoras de Origen Mieloide/inmunología , Oxidación-Reducción , PPAR gamma/agonistas , PPAR gamma/metabolismo , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Regiones Promotoras Genéticas , Células RAW 264.7 , Sumoilación , Factor de Necrosis Tumoral alfa/genética
11.
J Clin Invest ; 126(7): 2394-403, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27367183

RESUMEN

The term asthma encompasses a disease spectrum with mild to very severe disease phenotypes whose traditional common characteristic is reversible airflow limitation. Unlike milder disease, severe asthma is poorly controlled by the current standard of care. Ongoing studies using advanced molecular and immunological tools along with improved clinical classification show that severe asthma does not identify a specific patient phenotype, but rather includes patients with constant medical needs, whose pathobiologic and clinical characteristics vary widely. Accordingly, in recent clinical trials, therapies guided by specific patient characteristics have had better outcomes than previous therapies directed to any subject with a diagnosis of severe asthma. However, there are still significant gaps in our understanding of the full scope of this disease that hinder the development of effective treatments for all severe asthmatics. In this Review, we discuss our current state of knowledge regarding severe asthma, highlighting different molecular and immunological pathways that can be targeted for future therapeutic development.


Asunto(s)
Asma/inmunología , Interferón gamma/inmunología , Animales , Antiasmáticos/uso terapéutico , Asma/sangre , Asma/terapia , Exposición a Riesgos Ambientales , Humanos , Sistema Inmunológico , Inflamación , Fenotipo , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
12.
Cell Rep ; 15(8): 1700-14, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27184852

RESUMEN

Inhalation of environmental antigens such as allergens does not always induce inflammation in the respiratory tract. While antigen-presenting cells (APCs), including dendritic cells and macrophages, take up inhaled antigens, the cell-intrinsic molecular mechanisms that prevent an inflammatory response during this process, such as activation of the transcription factor NF-κB, are not well understood. Here, we show that the nuclear receptor PPARγ plays a critical role in blocking NF-κB activation in response to inhaled antigens to preserve immune tolerance. Tolerance induction promoted mitochondrial respiration, generation of H2O2, and suppression of NF-κB activation in WT, but not PPARγ-deficient, APCs. Forced restoration of H2O2 in PPARγ-deficient cells suppressed IκBα degradation and NF-κB activation. Conversely, scavenging reactive oxygen species from mitochondria promoted IκBα degradation with loss of regulatory and promotion of inflammatory T cell responses in vivo. Thus, communication between PPARγ and the mitochondria maintains immune quiescence in the airways.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Peróxido de Hidrógeno/metabolismo , Pulmón/citología , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Animales , Antígeno CD11c/metabolismo , Proliferación Celular , Citocinas/genética , Células Dendríticas/metabolismo , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Tolerancia Inmunológica , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , PPAR gamma/deficiencia , PPAR gamma/metabolismo , Regiones Promotoras Genéticas/genética , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/citología
13.
J Clin Invest ; 125(8): 3037-50, 2015 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-26121748

RESUMEN

Severe asthma (SA) is a challenge to control, as patients are not responsive to high doses of systemic corticosteroids (CS). In contrast, mild-moderate asthma (MMA) is responsive to low doses of inhaled CS, indicating that Th2 cells, which are dominant in MMA, do not solely orchestrate SA development. Here, we analyzed broncholalveolar lavage cells isolated from MMA and SA patients and determined that IFN-γ (Th1) immune responses are exacerbated in the airways of individuals with SA, with reduced Th2 and IL-17 responses. We developed a protocol that recapitulates the complex immune response of human SA, including the poor response to CS, in a murine model. Compared with WT animals, Ifng-/- mice subjected to this SA model failed to mount airway hyperresponsiveness (AHR) without appreciable effect on airway inflammation. Conversely, AHR was not reduced in Il17ra-/- mice, although airway inflammation was lower. Computer-assisted pathway analysis tools linked IFN-γ to secretory leukocyte protease inhibitor (SLPI), which is expressed by airway epithelial cells, and IFN-γ inversely correlated with SLPI expression in SA patients and the mouse model. In mice subjected to our SA model, forced SLPI expression decreased AHR in the absence of CS, and it was further reduced when SLPI was combined with CS. Our study identifies a distinct immune response in SA characterized by a dysregulated IFN-γ/SLPI axis that affects lung function.


Asunto(s)
Asma/inmunología , Regulación de la Expresión Génica/inmunología , Interferón gamma/inmunología , Inhibidor Secretorio de Peptidasas Leucocitarias/inmunología , Células Th2/inmunología , Adolescente , Adulto , Animales , Asma/genética , Asma/patología , Lavado Broncoalveolar , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Humanos , Interferón gamma/genética , Interleucina-17/genética , Interleucina-17/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Persona de Mediana Edad , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Inhibidor Secretorio de Peptidasas Leucocitarias/genética , Índice de Severidad de la Enfermedad , Células TH1/inmunología , Células TH1/patología , Células Th2/patología
14.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L130-40, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25326577

RESUMEN

Although asthma has long been considered a heterogeneous disease, attempts to define subgroups of asthma have been limited. In recent years, both clinical and statistical approaches have been utilized to better merge clinical characteristics, biology, and genetics. These combined characteristics have been used to define phenotypes of asthma, the observable characteristics of a patient determined by the interaction of genes and environment. Identification of consistent clinical phenotypes has now been reported across studies. Now the addition of various 'omics and identification of specific molecular pathways have moved the concept of clinical phenotypes toward the concept of molecular phenotypes. The importance of these molecular phenotypes is being confirmed through the integration of molecularly targeted biological therapies. Thus the global term asthma is poised to become obsolete, being replaced by terms that more specifically identify the pathology associated with the disease.


Asunto(s)
Asma/clasificación , Asma/genética , Citocinas/biosíntesis , Eosinófilos/inmunología , Inflamación/inmunología , Animales , Asma/inmunología , Modelos Animales de Enfermedad , Humanos , Pulmón/inmunología , Pulmón/patología , Ratones , Fenotipo
15.
J Immunol ; 192(12): 5471-5475, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24829419

RESUMEN

We reported previously that c-kit ligation by membrane-bound stem cell factor (mSCF) boosts IL-6 production in dendritic cells (DCs) and a Th17-immune response. However, Th17 establishment also requires heterodimeric IL-23, but the mechanisms that regulate IL-23 gene expression in DCs are not fully understood. We show that IL-23p19 gene expression in lung DCs is dependent on mSCF, which is regulated by the metalloproteinase MMP-9. Th1-inducing conditions enhanced MMP-9 activity, causing cleavage of mSCF, whereas the opposite was true for Th17-promoting conditions. In MMP-9(-/-) mice, a Th1-inducing condition could maintain mSCF and enhance IL-23p19 in DCs, promoting IL-17-producing CD4(+) T cells in the lung. Conversely, mSCF cleavage from bone marrow DCs in vitro by rMMP-9 led to reduced IL-23p19 expression under Th17-inducing conditions, with dampening of intracellular AKT phosphorylation. Collectively, these results show that the c-kit/mSCF/MMP-9 axis regulates IL-23 gene expression in DCs to control IL-17 production in the lung.


Asunto(s)
Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-17/inmunología , Subunidad p19 de la Interleucina-23/inmunología , Pulmón/inmunología , Metaloproteinasa 9 de la Matriz/inmunología , Factor de Células Madre/inmunología , Animales , Membrana Celular/genética , Membrana Celular/inmunología , Membrana Celular/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/genética , Interleucina-17/genética , Interleucina-17/metabolismo , Subunidad p19 de la Interleucina-23/biosíntesis , Subunidad p19 de la Interleucina-23/genética , Pulmón/citología , Pulmón/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Fosforilación/genética , Fosforilación/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
16.
Curr Opin Allergy Clin Immunol ; 14(1): 7-12, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24300419

RESUMEN

PURPOSE OF REVIEW: Binding of the receptor tyrosine kinase, c-kit, to its ligand, stem cell factor (SCF), mediates numerous biological functions. Important roles for c-kit in hematopoiesis, melanogenesis, erythropoiesis, spermatogenesis, and carcinogenesis are well documented. Similarly, activation of mast cells and eosinophils by c-kit ligation has long been known to result in degranulation with concomitant release of pro-inflammatory mediators including cytokines. This review will highlight a recently discovered function of c-kit in regulating the adaptive immune responses with relevance to allergic diseases. RECENT FINDINGS: Recent studies in a number of laboratories including our own highlight the previously unappreciated functions for c-kit in immunological processes. Increased expression of c-kit and its ligand, SCF, on dendritic cells by Th2/Th17-inducing stimuli leads to c-kit activation and immune skewing toward these subsets and away from Th1 responses. Treatment of dendritic cells with inhibitors of c-kit activation such as imatinib mesylate (Gleevec) induces breach of T-cell tolerance, skewing of responses toward Th1, and activation of natural killer cells. SUMMARY: Taken together, these observations suggest that the c-kit/SCF axis may be a useful target for redirecting deleterious immune responses in various disease settings, including allergic diseases that are often associated with Th2 and Th17 responses.


Asunto(s)
Células Dendríticas/inmunología , Hipersensibilidad/metabolismo , Células Asesinas Naturales/inmunología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/metabolismo , Células Th17/inmunología , Células Th2/inmunología , Inmunidad Adaptativa , Animales , Benzamidas/farmacología , Citocinas/inmunología , Humanos , Mesilato de Imatinib , Activación de Linfocitos/efectos de los fármacos , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/inmunología , Pirimidinas/farmacología , Sistema Respiratorio/inmunología , Transducción de Señal , Balance Th1 - Th2/efectos de los fármacos
17.
J Immunol ; 191(1): 25-9, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23733880

RESUMEN

Dendritic cell (DC)-T cell interactions that underlie inducible/adaptive regulatory T cell generation and airway tolerance are not well understood. In this study, we show that mice lacking CD11c(hi) lung DCs, but containing plasmacytoid DCs (pDCs), fail tolerization with inhaled Ag and cannot support Foxp3 induction in vivo in naive CD4(+) T cells. CD103(+) DCs from tolerized mice efficiently induced Foxp3 in cocultured naive CD4(+) T cells but pDCs and lung macrophages failed to do so. CD103(+) DCs, but not pDCs or lung macrophages, upregulated the expression of retinaldehyde dehydrogenase 2 (aldh1a2), which is key for the production of retinoic acid, a cofactor for TGF-ß for Foxp3 induction. Batf3(-/-) mice, selectively lacking CD103(+) DCs, failed tolerization by inhaled Ag. Collectively, our data show that pulmonary tolerance is dependent on CD103(+) DCs, correlating with their ability to upregulate aldh1a2, which can promote Foxp3 expression in T cells.


Asunto(s)
Aldehído Deshidrogenasa/biosíntesis , Antígenos CD/biosíntesis , Antígeno CD11c/fisiología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/biosíntesis , Cadenas alfa de Integrinas/biosíntesis , Pulmón/inmunología , Regulación hacia Arriba/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/enzimología , Células Dendríticas/metabolismo , Tolerancia Inmunológica/inmunología , Pulmón/enzimología , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
18.
Haematologica ; 98(3): 464-72, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22983573

RESUMEN

The intensity of hemolytic anemia has been proposed as an independent risk factor for the development of certain clinical complications of sickle cell disease, such as pulmonary hypertension, hypoxemia and cutaneous leg ulceration. A composite variable derived from several individual markers of hemolysis could facilitate studies of the underlying mechanisms of hemolysis. In this study, we assessed the association of hemolysis with outcomes in sickle cell anemia. A hemolytic component was calculated by principal component analysis from reticulocyte count, serum lactate dehydrogenase, aspartate aminotransferase and total bilirubin concentrations in 415 hemoglobin SS patients. Association of this component with direct markers of hemolysis and clinical outcomes was assessed. As primary validation, both plasma red blood cell microparticles and cell-free hemoglobin concentration were higher in the highest hemolytic component quartile compared to the lowest quartile (P≤0.0001 for both analyses). The hemolytic component was lower with hydroxyurea therapy, higher hemoglobin F, and alpha-thalassemia (P≤0.0005); it was higher with higher systemic pulse pressure, lower oxygen saturation, and greater values for tricuspid regurgitation velocity, left ventricular diastolic dimension and left ventricular mass (all P<0.0001). Two-year follow-up analysis showed that a high hemolytic component was associated with an increased risk of death (hazard ratio, HR 3.44; 95% confidence interval, CI: 1.2-9.5; P=0.02). The hemolytic component reflects direct markers of intravascular hemolysis in patients with sickle cell disease and allows for adjusted analysis of associations between hemolytic severity and clinical outcomes. These results confirm associations between hemolytic rate and pulse pressure, oxygen saturation, increases in Doppler-estimated pulmonary systolic pressures and mortality (Clinicaltrials.gov identifier: NCT00492531).


Asunto(s)
Anemia de Células Falciformes/epidemiología , Hemólisis , Biomarcadores/sangre , Micropartículas Derivadas de Células , Comorbilidad , Índices de Eritrocitos , Europa (Continente)/epidemiología , Humanos , Mortalidad , Factores de Riesgo , Estados Unidos/epidemiología
19.
Nat Med ; 18(10): 1525-30, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22961107

RESUMEN

Immune tolerance is instituted early in life, during which time regulatory T (T(reg)) cells have an important role. Recurrent infections with respiratory syncytial virus (RSV) in early life increase the risk for asthma in adult life. Repeated infection of infant mice tolerized to ovalbumin (OVA) through their mother's milk with RSV induced allergic airway disease in response to OVA sensitization and challenge, including airway inflammation, hyper-reactivity and higher OVA-specific IgE, as compared to uninfected tolerized control mice. Virus infection induced GATA-3 expression and T helper type 2 (T(H)2) cytokine production in forkhead box P3 (FOXP3)(+) T(reg) cells and compromised the suppressive function of pulmonary T(reg) cells in a manner that was dependent on interleukin-4 receptor α (IL-4Rα) expression in the host. Thus, by promoting a T(H)2-type inflammatory response in the lung, RSV induced a T(H)2-like effector phenotype in T(reg) cells and attenuated tolerance to an unrelated antigen (allergen). Our findings highlight a mechanism by which viral infection targets a host-protective mechanism in early life and increases susceptibility to allergic disease.


Asunto(s)
Asma/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Linfocitos T Reguladores/inmunología , Animales , Asma/virología , Factores de Transcripción Forkhead/biosíntesis , Factor de Transcripción GATA3/biosíntesis , Tolerancia Inmunológica , Inmunoglobulina E/sangre , Subunidad alfa del Receptor de Interleucina-4/inmunología , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/metabolismo
20.
Proc Natl Acad Sci U S A ; 108(13): 5360-5, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21402950

RESUMEN

Aspergillus fumigatus is commonly associated with allergic bronchopulmonary aspergillosis in patients with severe asthma in which chronic airway neutrophilia predicts a poor outcome. We were able to recapitulate fungus-induced neutrophilic airway inflammation in a mouse model in our efforts to understand the underlying mechanisms. However, neutrophilia occurred in a mouse strain-selective fashion, providing us with an opportunity to perform a comparative study to elucidate the mechanisms involved. Here we show that TNF-α, largely produced by Ly6c(+)CD11b(+) dendritic cells (DCs), plays a central role in promoting IL-17A from CD4(+) T cells and collaborating with it to induce airway neutrophilia. Compared with C57BL/6 mice, BALB/c mice displayed significantly more TNF-α-producing DCs and macrophages in the lung. Lung TNF-α levels were drastically reduced in CD11c-DTR BALB/c mice depleted of CD11c+ cells, and TNF-α-producing Ly6c(+)CD11b(+) cells were abolished in Dectin-1(-/-) and MyD88(-/-) BALB/c mice. TNF-α deficiency itself blunted accumulation of inflammatory Ly6c(+)CD11b(+) DCs. Also, lack of TNF-α decreased IL-17A but promoted IL-5 levels, switching inflammation from a neutrophil to eosinophil bias resembling that in C57BL/6 mice. The TNF-α(low) DCs in C57BL/6 mice contained more NF-κB p50 homodimers, which are strong repressors of TNF-α transcription. Functionally, collaboration between TNF-α and IL-17A triggered significantly higher levels of the neutrophil chemoattractants keratinocyte cytokine and macrophage inflammatory protein 2 in BALB/c mice. Our study identifies TNF-α as a molecular switch that orchestrates a sequence of events in DCs and CD4 T cells that promote neutrophilic airway inflammation.


Asunto(s)
Células Dendríticas/inmunología , Eosinofilia/inmunología , Interleucina-17/inmunología , Interleucina-5/inmunología , Pulmón/inmunología , Neutrófilos/inmunología , Aspergilosis Pulmonar/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Antígenos CD/inmunología , Linfocitos T CD4-Positivos/inmunología , Humanos , Pulmón/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptor Toll-Like 2/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...