Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 326(3): L313-L329, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38290163

RESUMEN

Respiratory viral infections are one of the major causes of illness and death worldwide. Symptoms associated with respiratory infections can range from mild to severe, and there is limited understanding of why there is large variation in severity. Environmental exposures are a potential causative factor. The aryl hydrocarbon receptor (AHR) is an environment-sensing molecule expressed in all immune cells. Although there is considerable evidence that AHR signaling influences immune responses to other immune challenges, including respiratory pathogens, less is known about the impact of AHR signaling on immune responses during coronavirus (CoV) infection. In this study, we report that AHR activation significantly altered immune cells in the lungs and bone marrow of mice infected with a mouse CoV. AHR activation transiently reduced the frequency of multiple cells in the mononuclear phagocyte system, including monocytes, interstitial macrophages, and dendritic cells in the lung. In the bone marrow, AHR activation altered myelopoiesis, as evidenced by a reduction in granulocyte-monocyte progenitor cells and an increased frequency of myeloid-biased progenitor cells. Moreover, AHR activation significantly affected multiple stages of the megakaryocyte lineage. Overall, these findings indicate that AHR activation modulates multiple aspects of the immune response to a CoV infection. Given the significant burden of respiratory viruses on human health, understanding how environmental exposures shape immune responses to infection advances our knowledge of factors that contribute to variability in disease severity and provides insight into novel approaches to prevent or treat disease.NEW & NOTEWORTHY Our study reveals a multifaceted role for aryl hydrocarbon receptor (AHR) signaling in the immune response to coronavirus (CoV) infection. Sustained AHR activation during in vivo mouse CoV infection altered the frequency of mature immune cells in the lung and modulated emergency hematopoiesis, specifically myelopoiesis and megakaryopoiesis, in bone marrow. This provides new insight into immunoregulation by the AHR and extends our understanding of how environmental exposures can impact host responses to respiratory viral infections.


Asunto(s)
Infecciones por Coronavirus , Receptores de Hidrocarburo de Aril , Infecciones del Sistema Respiratorio , Animales , Humanos , Ratones , Médula Ósea/metabolismo , Infecciones por Coronavirus/metabolismo , Pulmón/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo
2.
Toxicol Appl Pharmacol ; 489: 117010, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38901696

RESUMEN

Humoral responses to respiratory viruses, such as influenza viruses, develop over time and are central to protection from repeated infection with the same or similar viruses. Epidemiological and experimental studies have linked exposures to environmental contaminants that bind the aryl hydrocarbon receptor (AHR) with modulated antibody responses to pathogenic microorganisms and common vaccinations. Other studies have prompted investigation into the potential therapeutic applications of compounds that activate AHR. Herein, using two different AHR ligands [2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and 2-(1H-Indol-3-ylcarbonyl)-4-thiazolecarboxylic acid methyl ester (ITE), to modulate the duration of AHR activity, we show that the humoral response to viral infection is dependent upon the duration and timing of AHR signaling, and that different cellular elements of the response have different sensitivities. When AHR activation was initiated prior to infection with influenza A virus, there was suppression of all measured elements of the humoral response (i.e., the frequency of T follicular helper cells, germinal center B cells, plasma cells, and circulating virus-specific antibody). However, when the timing of AHR activation was adjusted to either early (days -1 to +5 relative to infection) or later (days +5 onwards), then AHR activation affected different aspects of the overall humoral response. These findings highlight the importance of considering the timing of AHR activation in relation to triggering an immune response, particularly when targeting the AHR to manipulate disease processes.


Asunto(s)
Inmunidad Humoral , Dibenzodioxinas Policloradas , Receptores de Hidrocarburo de Aril , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Inmunidad Humoral/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Femenino , Factores de Tiempo , Ratones , Ratones Endogámicos C57BL , Indoles/farmacología , Transducción de Señal/efectos de los fármacos , Anticuerpos Antivirales , Infecciones por Orthomyxoviridae/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/metabolismo , Ligandos , Tiazoles
3.
Mov Disord ; 39(3): 606-613, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38389433

RESUMEN

BACKGROUND: Environmental exposure to trichloroethylene (TCE), a carcinogenic dry-cleaning chemical, may be linked to Parkinson's disease (PD). OBJECTIVE: The objective of this study was to determine whether PD and cancer were elevated among attorneys who worked near a contaminated site. METHODS: We surveyed and evaluated attorneys with possible exposure and assessed a comparison group. RESULTS: Seventy-nine of 82 attorneys (96.3%; mean [SD] age: 69.5 [11.4] years; 89.9% men) completed at least one phase of the study. For comparison, 75 lawyers (64.9 [10.2] years; 65.3% men) underwent clinical evaluations. Four (5.1%) of them who worked near the polluted site reported PD, more than expected based on age and sex (1.7%; P = 0.01) but not significantly higher than the comparison group (n = 1 [1.3%]; P = 0.37). Fifteen (19.0%), compared to four in the comparison group (5.3%; P = 0.049), had a TCE-related cancer. CONCLUSIONS: In a retrospective study, diagnoses of PD and TCE-related cancers appeared to be elevated among attorneys who worked next to a contaminated dry-cleaning site. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Neoplasias , Enfermedad de Parkinson , Tricloroetileno , Masculino , Humanos , Anciano , Femenino , Enfermedad de Parkinson/epidemiología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/diagnóstico , Estudios Retrospectivos , Tricloroetileno/análisis
4.
J Immunol ; 208(10): 2319-2330, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35444027

RESUMEN

T follicular helper (Tfh) cells support Ab responses and are a critical component of adaptive immune responses to respiratory viral infections. Tfh cells are regulated by a network of signaling pathways that are controlled, in part, by transcription factors. The aryl hydrocarbon receptor (AHR) is an environment-sensing transcription factor that modulates many aspects of adaptive immunity by binding a range of small molecules. However, the contribution of AHR signaling to Tfh cell differentiation and function is not known. In this article, we report that AHR activation by three different agonists reduced the frequency of Tfh cells during primary infection of C57BL/6 mice with influenza A virus (IAV). Further, using the high-affinity and AHR-specific agonist 2,3,7,8-tetrachlorodibenzo-p-dioxin, we show that AHR activation reduced Tfh cell differentiation and T cell-dependent B cell responses. Using conditional AHR knockout mice, we demonstrated that alterations of Tfh cells and T cell-dependent B cell responses after AHR activation required the AHR in T cells. AHR activation reduced the number of T follicular regulatory (Tfr) cells; however, the ratio of Tfr to Tfh cells was amplified. These alterations to Tfh and Tfr cells during IAV infection corresponded with differences in expression of BCL6 and FOXP3 in CD4+ T cells and required the AHR to have a functional DNA-binding domain. Overall, these findings support that the AHR modulates Tfh cells during viral infection, which has broad-reaching consequences for understanding how environmental factors contribute to variation in immune defenses against infectious pathogens, such as influenza and severe acute respiratory syndrome coronavirus.


Asunto(s)
Virus de la Influenza A , Infecciones por Orthomyxoviridae , Células T Auxiliares Foliculares , Animales , Diferenciación Celular , Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Receptores de Hidrocarburo de Aril/inmunología , Células T Auxiliares Foliculares/inmunología
5.
Am J Physiol Lung Cell Mol Physiol ; 323(5): L578-L592, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36068185

RESUMEN

Bronchiolitis obliterans (BO) is a debilitating disease of the small airways that can develop following exposure to toxic chemicals as well as respiratory tract infections. BO development is strongly associated with diacetyl (DA) inhalation exposures at occupationally relevant concentrations or severe influenza A viral (IAV) infections. However, it remains unclear whether lower dose exposures or more mild IAV infections can result in similar pathology. In the current work, we combined these two common environmental exposures, DA and IAV, to test whether shorter DA exposures followed by sublethal IAV infection would result in similar airways disease. Adult mice exposed to DA vapors 1 h/day for 5 consecutive days followed by infection with the airway-tropic IAV H3N2 (HKx31) resulted in increased mortality, increased bronchoalveolar lavage (BAL) neutrophil percentage, mixed obstruction and restriction by lung function, and subsequent airway remodeling. Exposure to DA or IAV alone failed to result in significant pathology, whereas mice exposed to DA + IAV showed increased α-smooth muscle actin (αSMA) and epithelial cells coexpressing the basal cell marker keratin 5 (KRT5) with the club cell marker SCGB1A1. To test whether DA exposure impairs epithelial repair after IAV infection, mice were infected first with IAV and then exposed to DA during airway epithelial repair. Mice exposed to IAV + DA developed similar airway remodeling with increased subepithelial αSMA and epithelial cells coexpressing KRT5 and SCGB1A1. Our findings reveal an underappreciated concept that common environmental insults while seemingly harmless by themselves can have catastrophic implications on lung function and long-term respiratory health when combined.


Asunto(s)
Bronquiolitis Obliterante , Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Ratones , Animales , Humanos , Diacetil/toxicidad , Remodelación de las Vías Aéreas (Respiratorias) , Subtipo H3N2 del Virus de la Influenza A , Bronquiolitis Obliterante/patología , Mucosa Respiratoria/patología , Células Epiteliales/patología , Pulmón/patología , Gripe Humana/patología
6.
Toxicol Appl Pharmacol ; 450: 116160, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35817128

RESUMEN

Epidemiological studies associate biomass smoke with an increased risk for respiratory infections in children and adults in the developing world, with 500,000 premature deaths each year attributed to biomass smoke-related acute respiratory infections including infections caused by respiratory viruses. Animal dung is a biomass fuel of particular concern because it generates more toxic compounds per amount burned than wood, and is a fuel of last resort for the poorest households. Currently, there is little biological evidence on the effects of dung biomass smoke exposure on immune responses to respiratory viral infections. Here, we investigated the impact of dung biomass exposure on respiratory infection using a mouse model of dung biomass smoke and cultured primary human small airway epithelial cells (SAECs). Mice infected with influenza A virus (IAV) after dung biomass smoke exposure had increased mortality, lung inflammation and virus mRNA levels, and suppressed expression of innate anti-viral mediators compared to air exposed mice. Importantly, there was still significant tissue inflammation 14 days after infection in dung biomass smoke-exposed mice even after inflammation had resolved in air-exposed mice. Dung biomass smoke exposure also suppressed the production of anti-viral cytokines and interferons in cultured SAECs treated with poly(I:C) or IAV. This study shows that dung biomass smoke exposure impairs the immune response to respiratory viruses and contributes to biomass smoke-related susceptibility to respiratory viral infections, likely due to a failure to resolve the inflammatory effects of biomass smoke exposure.


Asunto(s)
Gripe Humana , Neumonía , Infecciones del Sistema Respiratorio , Animales , Biomasa , Niño , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo
8.
Int J Mol Sci ; 19(11)2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445691

RESUMEN

In a time where "translational" science has become a mantra in the biomedical field, it is reassuring when years of research into a biological phenomenon suddenly points towards novel prevention or therapeutic approaches to disease, thereby demonstrating once again that basic science and translational science are intimately linked. The studies on the aryl hydrocarbon receptor (AHR) discussed here provide a perfect example of how years of basic toxicological research on a molecule, whose normal physiological function remained a mystery for so long, has now yielded a treasure trove of actionable information on the development of targeted therapeutics. Examples are autoimmunity, metabolic imbalance, inflammatory skin and gastro-intestinal diseases, cancer, development and perhaps ageing. Indeed, the AHR field no longer asks, "What does this receptor do in the absence of xenobiotics?" It now asks, "What doesn't this receptor do?".


Asunto(s)
Receptores de Hidrocarburo de Aril/metabolismo , Animales , Senescencia Celular , Dieta , Evolución Molecular , Tracto Gastrointestinal/patología , Humanos , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Obesidad/patología , Paris , Receptores de Hidrocarburo de Aril/genética , Células Madre/metabolismo
9.
Am J Respir Cell Mol Biol ; 56(4): 453-464, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27967234

RESUMEN

An aberrant oxygen environment at birth increases the severity of respiratory viral infections later in life through poorly understood mechanisms. Here, we show that alveolar epithelial cell (AEC) 2 cells (AEC2s), progenitors for AEC1 cells, are depleted in adult mice exposed to neonatal hypoxia or hyperoxia. Airway cells expressing surfactant protein (SP)-C and ATP binding cassette subfamily A member 3, alveolar pod cells expressing keratin (KRT) 5, and pulmonary fibrosis were observed when these mice were infected with a sublethal dose of HKx31, H3N2 influenza A virus. This was not seen in infected siblings birthed into room air. Genetic lineage tracing studies in mice exposed to neonatal hypoxia or hyperoxia revealed pre-existing secretoglobin 1a1+ cells produced airway cells expressing SP-C and ATP binding cassette subfamily A member 3. Pre-existing Kr5+ progenitor cells produced squamous alveolar cells expressing receptor for advanced glycation endproducts, aquaporin 5, and T1α in alveoli devoid of AEC2s. They were not the source of KRT5+ alveolar pod cells. These oxygen-dependent changes in epithelial cell regeneration and fibrosis could be recapitulated by conditionally depleting AEC2s in mice using diphtheria A toxin and then infecting with influenza A virus. Likewise, airway cells expressing SP-C and alveolar cells expressing KRT5 were observed in human idiopathic pulmonary fibrosis. These findings suggest that alternative progenitor lineages are mobilized to regenerate the alveolar epithelium when AEC2s are severely injured or depleted by previous insults, such as an adverse oxygen environment at birth. Because these lineages regenerate AECs in spatially distinct compartments of a lung undergoing fibrosis, they may not be sufficient to prevent disease.


Asunto(s)
Envejecimiento/metabolismo , Células Epiteliales Alveolares/citología , Linaje de la Célula , Células Madre/citología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Animales , Animales Recién Nacidos , Linaje de la Célula/efectos de los fármacos , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Queratina-5/metabolismo , Ratones , Modelos Biológicos , Oxígeno/farmacología , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Uteroglobina/metabolismo
10.
Stem Cells ; 34(5): 1396-406, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26891117

RESUMEN

Alveolar epithelial type II cells (AEC2) maintain pulmonary homeostasis by producing surfactant, expressing innate immune molecules, and functioning as adult progenitor cells for themselves and alveolar epithelial type I cells (AEC1). How the proper number of alveolar epithelial cells is determined in the adult lung is not well understood. Here, BrdU labeling, genetic lineage tracing, and targeted expression of the anti-oxidant extracellular superoxide dismutase in AEC2s are used to show how the oxygen environment at birth influences postnatal expansion of AEC2s and AEC1s in mice. Birth into low (12%) or high (≥60%) oxygen stimulated expansion of AEC2s through self-renewal and differentiation of the airway Scgb1a1 + lineage. This non-linear or hormesis response to oxygen was specific for the alveolar epithelium because low oxygen stimulated and high oxygen inhibited angiogenesis as defined by changes in V-cadherin and PECAM (CD31). Although genetic lineage tracing studies confirmed adult AEC2s are stem cells for AEC1s, we found no evidence that postnatal growth of AEC1s were derived from self-renewing Sftpc + or the Scbg1a1 + lineage of AEC2s. Taken together, our results show how a non-linear response to oxygen at birth promotes expansion of AEC2s through two distinct lineages. Since neither lineage contributes to the postnatal expansion of AEC1s, the ability of AEC2s to function as stem cells for AEC1s appears to be restricted to the adult lung. Stem Cells 2016;34:1396-1406.


Asunto(s)
Envejecimiento/fisiología , Células Epiteliales Alveolares/citología , Células Epiteliales/citología , Pulmón/citología , Oxígeno/farmacología , Células Madre/citología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Modelos Biológicos
11.
J Immunol ; 194(9): 4446-57, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25810390

RESUMEN

Successfully fighting infection requires a properly tuned immune system. Recent epidemiological studies link exposure to pollutants that bind the aryl hydrocarbon receptor (AHR) during development with poorer immune responses later in life. Yet, how developmental triggering of AHR durably alters immune cell function remains unknown. Using a mouse model, we show that developmental activation of AHR leads to long-lasting reduction in the response of CD8(+) T cells during influenza virus infection, cells critical for resolving primary infection. Combining genome-wide approaches, we demonstrate that developmental activation alters DNA methylation and gene expression patterns in isolated CD8(+) T cells prior to and during infection. Altered transcriptional profiles in CD8(+) T cells from developmentally exposed mice reflect changes in pathways involved in proliferation and immunoregulation, with an overall pattern that bears hallmarks of T cell exhaustion. Developmental exposure also changed DNA methylation across the genome, but differences were most pronounced following infection, where we observed inverse correlation between promoter methylation and gene expression. This points to altered regulation of DNA methylation as one mechanism by which AHR causes durable changes in T cell function. Discovering that distinct gene sets and pathways were differentially changed in developmentally exposed mice prior to and after infection further reveals that the process of CD8(+) T cell activation is rendered fundamentally different by early life AHR signaling. These findings reveal a novel role for AHR in the developing immune system: regulating DNA methylation and gene expression as T cells respond to infection later in life.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Metilación de ADN , Receptores de Hidrocarburo de Aril/genética , Virosis/genética , Virosis/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/metabolismo , Virosis/metabolismo
12.
Environ Sci Technol ; 50(13): 7152-62, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27244128

RESUMEN

To determine demographic, reproductive, and maternal dietary factors that predict perfluoroalkyl substance (PFAS) concentrations in breast milk, we measured perfluorooctane sulfonic (PFOS) and perfluorooctanoic acid (PFOA) concentrations, using liquid chromatography-mass spectrometry, in 184 colostrum samples collected from women participating in a cohort study in Eastern Slovakia between 2002 and 2004. During their hospital delivery stay, mothers completed a food frequency questionnaire, and demographic and reproductive data were also collected. PFOS and PFOA predictors were identified by optimizing multiple linear regression models using Akaike's information criterion (AIC). The geometric mean concentration in colostrum was 35.3 pg/mL for PFOS and 32.8 pg/mL for PFOA. In multivariable models, parous women had 40% lower PFOS (95% CI: -56 to -17%) and 40% lower PFOA (95% CI: -54 to -23%) concentrations compared with nulliparous women. Moreover, fresh/frozen fish consumption, longer birth intervals, and Slovak ethnicity were associated with higher PFOS and PFOA concentrations in colostrum. These results will help guide the design of future epidemiologic studies examining milk PFAS concentrations in relation to health end points in children.


Asunto(s)
Ácidos Alcanesulfónicos , Calostro/química , Animales , Caprilatos , Cromatografía Liquida , Estudios de Cohortes , Demografía , Fluorocarburos , Humanos
13.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L76-85, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25381024

RESUMEN

Respiratory distress in preterm or low birth weight infants is often treated with supplemental oxygen. However, this therapy can disrupt normal lung development and architecture and alter responses to respiratory insults. Similarly, exposure of newborn mice to 100% oxygen during saccular lung development leads to permanent alveolar simplification, and upon challenge with influenza A virus, mice exhibit reduced host resistance. Natural killer (NK) cells are key players in antiviral immunity, and emerging evidence suggest they also help to maintain homeostasis in peripheral tissues, including the lung, by promoting epithelial cell regeneration via IL-22. We tested the hypothesis that adult mice exposed to hyperoxia as neonates have modified NK cell responses to infection. We report here that mice exposed to neonatal hyperoxia had fewer IL-22(+) NK cells in their lungs after influenza virus challenge and a parallel increase in IFN-γ(+) NK cells. Using reciprocal bone marrow chimeric mice, we show that exposure of either hematopoietic or nonhematopoietic cells was sufficient to increase the severity of infection and to diminish the frequency of IL-22(+) NK cells in the infected lung. Overall, our findings suggest that neonatal hyperoxia leads to long-term changes in the reparative vs. cytotoxic nature of NK cells and that this is due in part to intrinsic changes in hematopoietic cells. These differences may contribute to how oxygen alters the host response to respiratory viral infections.


Asunto(s)
Hiperoxia/inmunología , Inmunidad Celular , Virus de la Influenza A/inmunología , Células Asesinas Naturales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Alveolos Pulmonares/inmunología , Animales , Animales Recién Nacidos , Hiperoxia/patología , Interferón gamma/inmunología , Interleucinas/inmunología , Células Asesinas Naturales/patología , Ratones , Infecciones por Orthomyxoviridae/patología , Alveolos Pulmonares/patología , Interleucina-22
14.
Am J Physiol Lung Cell Mol Physiol ; 309(3): L305-13, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26071552

RESUMEN

Respiratory infections are a threat to health and economies worldwide, yet the basis for striking variation in the severity of infection is not completely understood. Environmental exposures during development are associated with increased severity and incidence of respiratory infection later in life. Many of these exposures include ligands of the aryl hydrocarbon receptor (AHR), a transcription factor expressed by immune and nonimmune cells. In adult animals, AHR activation alters CD4(+) T cells and changes immunopathology. Developmental AHR activation impacts CD4(+) T-cell responses in lymphoid tissues, but whether skewed responses are also present in the infected lung is unknown. To determine whether pulmonary CD4(+) T-cell responses are modified by developmental AHR activation, mice were exposed to the prototypical AHR ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin during development and infected with influenza virus as adults. Lungs of exposed offspring had greater bronchopulmonary inflammation compared with controls, and activated, virus-specific CD4(+) T cells contributed to the infiltrating leukocytes. These effects were CD4(+) T cell subset specific, with increases in T helper type 1 and regulatory T cells, but no change in the frequency of T helper type 17 cells in the infected lung. This is in direct contrast to prior reports of suppressed conventional CD4(+) T-cell responses in the lymph node. Using adoptive transfers and manipulating the pathogen properties, we determined that developmental exposure influenced factors intrinsic and extrinsic to CD4(+) T cells and may involve developmentally induced changes in signals from infected lung epithelial cells. Thus developmental exposures lead to context-dependent changes in pulmonary CD4(+) T-cell subsets, which may contribute to differential responses to respiratory infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Infecciones del Sistema Respiratorio/inmunología , Animales , Femenino , Virus de la Influenza A/inmunología , Activación de Linfocitos , Masculino , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/virología
15.
J Immunol ; 190(2): 659-68, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23233726

RESUMEN

The underlying reasons for variable clinical outcomes from respiratory viral infections remain uncertain. Several studies suggest that environmental factors contribute to this variation, but limited knowledge of cellular and molecular targets of these agents hampers our ability to quantify or modify their contribution to disease and improve public health. The aryl hydrocarbon receptor (AhR) is an environment-sensing transcription factor that binds many anthropogenic and natural chemicals. The immunomodulatory properties of AhR ligands are best characterized with extensive studies of changes in CD4(+) T cell responses. Yet, AhR modulates other aspects of immune function. We previously showed that during influenza virus infection, AhR activation modulates neutrophil accumulation in the lung, and this contributes to increased mortality in mice. Enhanced levels of inducible NO synthase (iNOS) in infected lungs are observed during the same time frame as AhR-mediated increased pulmonary neutrophilia. In this study, we evaluated whether these two consequences of AhR activation are causally linked. Reciprocal inhibition of AhR-mediated elevations in iNOS and pulmonary neutrophilia reveal that although they are contemporaneous, they are not causally related. We show using Cre/loxP technology that elevated iNOS levels and neutrophil number in the infected lung result from separate, AhR-dependent signaling in endothelial and respiratory epithelial cells, respectively. Studies using mutant mice further reveal that AhR-mediated alterations in these innate responses to infection require a functional nuclear localization signal and DNA binding domain. Thus, gene targets of AhR in non-hematopoietic cells are important new considerations for understanding AhR-mediated changes in innate anti-viral immunity.


Asunto(s)
Neutrófilos/inmunología , Neutrófilos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Orthomyxoviridae/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , ADN/metabolismo , Células Endoteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Transgénicos , Infiltración Neutrófila/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Infecciones por Orthomyxoviridae/genética , Unión Proteica , Receptores de Hidrocarburo de Aril/genética , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología
16.
Pediatr Allergy Immunol ; 25(2): 180-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24520985

RESUMEN

BACKGROUND: Infants born prematurely are often treated with supplemental oxygen, which can increase their risk for airway hyper-responsiveness (AHR), asthma, reduced lung function, and altered responses to respiratory viral infections later in childhood. Likewise, exposure of newborn mice to hyperoxia alters baseline pulmonary mechanics and the host response to influenza A virus infection in adult mice. Here, we use this mouse model to test the hypothesis that neonatal hyperoxia also promotes AHR and exacerbated allergen-induced symptoms in adult mice. METHODS: Baseline lung mechanics and AHR measured by methacholine provocation were assessed in adult male and female mice exposed to room air or 100% oxygen (hyperoxia) between post-natal days 0-4. AHR and lung inflammation were evaluated after adult female mice were sensitized with ovalbumin (OVA) plus alum and challenged with aerosolized OVA. RESULTS: Baseline lung compliance increased and resistance decreased in adult female, but not male, mice exposed to neonatal hyperoxia compared with siblings exposed to room air. Neonatal hyperoxia significantly enhanced methacholine-induced AHR in female mice, but did not affect allergen-induced AHR to methacholine or lung inflammation. CONCLUSION: Increased incidence of AHR and asthma is reported in children born prematurely and exposed to supplemental oxygen. Our findings in adult female mice exposed to hyperoxia as neonates suggest that this AHR reported in children born prematurely may reflect non-atopic wheezing due to intrinsic structural changes in airway development.


Asunto(s)
Hiperreactividad Bronquial/fisiopatología , Broncoconstricción , Hiperoxia/fisiopatología , Pulmón/fisiopatología , Neumonía/fisiopatología , Factores de Edad , Resistencia de las Vías Respiratorias , Animales , Animales Recién Nacidos , Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/diagnóstico , Pruebas de Provocación Bronquial , Broncoconstrictores/farmacología , Modelos Animales de Enfermedad , Femenino , Hiperoxia/complicaciones , Hiperoxia/diagnóstico , Rendimiento Pulmonar , Masculino , Cloruro de Metacolina , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Ovalbúmina , Neumonía/inducido químicamente , Neumonía/diagnóstico , Factores de Riesgo , Factores Sexuales
17.
J Immunotoxicol ; 21(1): 2340495, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38946256

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) are anthropogenic organofluorine compounds that persist indefinitely in the environment and bioaccumulate throughout all trophic levels. Biomonitoring efforts have detected multiple PFAS in the serum of most people. Immune suppression has been among the most consistent effects of exposure to PFAS. PFAS often co-occur as mixtures in the environment, however, few studies have examined immunosuppression of PFAS mixtures or determined whether PFAS exposure affects immune function in the context of infection. In this study, mixtures containing two or four different PFAS and a mouse model of infection with influenza A virus (IAV) were used to assess immunotoxicity of PFAS mixtures. PFAS were administered via the drinking water as either a binary mixture of perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) or quaternary mixture of PFOS, PFOA, perfluorohexane sulfonate (PFHxS), and perfluorononanoic acid (PFNA). The results indicated that the binary mixture affected the T-cell response, while the quaternary mixture affected the B-cell response to infection. These findings indicate that the immunomodulatory effects of PFAS mixtures are not simply additive, and that the sensitivity of immune responses to PFAS varies by cell type and mixture. The study also demonstrates the importance of studying adverse health effects of PFAS mixtures.


Asunto(s)
Ácidos Alcanesulfónicos , Caprilatos , Fluorocarburos , Virus de la Influenza A , Infecciones por Orthomyxoviridae , Fluorocarburos/efectos adversos , Fluorocarburos/toxicidad , Animales , Ratones , Virus de la Influenza A/inmunología , Ácidos Alcanesulfónicos/toxicidad , Ácidos Alcanesulfónicos/efectos adversos , Infecciones por Orthomyxoviridae/inmunología , Caprilatos/toxicidad , Caprilatos/efectos adversos , Humanos , Femenino , Ratones Endogámicos C57BL , Gripe Humana/inmunología , Modelos Animales de Enfermedad , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos
18.
Am J Respir Cell Mol Biol ; 48(2): 258-66, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23258231

RESUMEN

Supplemental oxygen used to treat infants born prematurely constitutes a major risk factor for long-term deficits in lung function and host defense against respiratory infections. Likewise, neonatal oxygen exposure results in alveolar simplification in adult mice, and enhances leukocyte recruitment and fibrosis when adult mice are infected with a sublethal dose of influenza A virus. Because pulmonary fibrosis was not observed in infected adult mice exposed to room air as neonates, previous neonatal oxygen exposure may have reprogrammed how the adult lung responds to epithelial injury. By administering bleomycin to adult mice exposed to room air or hyperoxia as neonates, we tested the hypothesis that neonatal hyperoxia enhances fibrosis when the epithelium is injured by direct fibrotic stimulus. Increased sensitivity to bleomycin-induced lung fibrosis was observed in adult mice exposed to neonatal hyperoxia, and was associated with increased numbers of leukocytes and an accumulation of active transforming growth factor (TGF)-ß1 in the lung. Fate mapping of the respiratory epithelium revealed that the epithelial-mesenchymal transition was not a significant source of fibroblasts in room air-exposed or oxygen-exposed mice treated with bleomycin. Instead, the treatment of mice with anti-Gr-1 antibody that depletes neutrophils and myeloid-derived suppressor cells reduced the early activation of TGF-ß1 and attenuated hyperoxia-enhanced fibrosis. Because bleomycin and influenza A virus both cause epithelial injury, understanding how neonatal hyperoxia reprograms the epithelial response to these two different injurious agents could lead to new therapeutic opportunities for treating lung diseases attributed to prematurity.


Asunto(s)
Animales Recién Nacidos , Hiperoxia/fisiopatología , Fibrosis Pulmonar/inducido químicamente , Animales , Secuencia de Bases , Cartilla de ADN , Inmunohistoquímica , Ratones , Fibrosis Pulmonar/fisiopatología , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
Am J Physiol Lung Cell Mol Physiol ; 305(4): L282-90, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23748535

RESUMEN

Exposing preterm infants or newborn mice to high concentrations of oxygen disrupts lung development and alters the response to respiratory viral infections later in life. Superoxide dismutase (SOD) has been separately shown to mitigate hyperoxia-mediated changes in lung development and attenuate virus-mediated lung inflammation. However, its potential to protect adult mice exposed to hyperoxia as neonates against viral infection is not known. Here, transgenic mice overexpressing extracellular (EC)-SOD in alveolar type II epithelial cells are used to test whether SOD can alleviate the deviant pulmonary response to influenza virus infection in adult mice exposed to hyperoxia as neonates. Fibrotic lung disease, observed following infection in wild-type (WT) mice exposed to hyperoxia as neonates, was prevented by overexpression of EC-SOD. However, leukocyte recruitment remained excessive, and levels of monocyte chemoattractant protein (MCP)-1 remained modestly elevated following infection in EC-SOD Tg mice exposed to hyperoxia as neonates. Because MCP-1 is often associated with pulmonary inflammation and fibrosis, the host response to infection was concurrently evaluated in adult Mcp-1 WT and Mcp-1 knockout mice exposed to neonatal hyperoxia. In contrast to EC-SOD, excessive leukocyte recruitment, but not lung fibrosis, was dependent upon MCP-1. Our findings demonstrate that neonatal hyperoxia alters the inflammatory and fibrotic responses to influenza A virus infection through different pathways. Therefore, these data suggest that multiple therapeutic strategies may be needed to provide complete protection against diseases attributed to prematurity and early life exposure to oxygen.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Hiperoxia/complicaciones , Hiperoxia/virología , Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/virología , Transducción de Señal , Envejecimiento/inmunología , Envejecimiento/patología , Animales , Animales Recién Nacidos , Quimiocina CCL2/metabolismo , Progresión de la Enfermedad , Espacio Extracelular/enzimología , Femenino , Hiperoxia/enzimología , Hiperoxia/patología , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/patología , Alveolos Pulmonares/enzimología , Alveolos Pulmonares/patología , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/enzimología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , Superóxido Dismutasa/metabolismo , Análisis de Supervivencia
20.
Am J Pathol ; 181(2): 441-51, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22677423

RESUMEN

Oxygen exposure in premature infants is a major risk factor for bronchopulmonary dysplasia and can impair the host response to respiratory viral infections later in life. Similarly, adult mice exposed to hyperoxia as neonates display alveolar simplification associated with a reduced number of alveolar epithelial type II cells and exhibit persistent inflammation, fibrosis, and mortality when infected with influenza A virus. Because type II cells participate in innate immunity and alveolar repair, their loss may contribute to oxygen-mediated sensitivity to viral infection. A genomewide screening of type II cells identified eosinophil-associated RNase 1 (Ear1). Ear1 was also detected in airway epithelium and was reduced in lungs of mice exposed to neonatal hyperoxia. Electroporation-mediated gene delivery of Ear1 to the lung before infection successfully reduced viral replication and leukocyte recruitment during infection. It also diminished the enhanced morbidity and mortality attributed to neonatal hyperoxia. These findings demonstrate that novel epithelial expression of Ear1 functions to limit influenza A virus infection, and its loss contributes to oxygen-associated epithelial injury and fibrosis after infection. People born prematurely may have defects in epithelial innate immunity that increase their risk for respiratory viral infections.


Asunto(s)
Neurotoxina Derivada del Eosinófilo/metabolismo , Epitelio/metabolismo , Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Oxígeno/farmacología , Ribonucleasas/metabolismo , Envejecimiento/patología , Aire , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Animales Recién Nacidos , Electroporación , Epitelio/efectos de los fármacos , Epitelio/patología , Epitelio/virología , Femenino , Técnicas de Transferencia de Gen , Hiperoxia/complicaciones , Hiperoxia/patología , Hiperoxia/virología , Virus de la Influenza A/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA