Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Cancer Cell ; 39(8): 1018-1022, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34115988

RESUMEN

Owing to clinical success of immune-checkpoint blockade, immunotherapy is becoming a cornerstone of modern oncology, and immuno-oncology is at the forefront of basic cancer research. This commentary outlines future opportunities for immuno-oncology modeling.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Neoplasias Experimentales , Neoplasias/terapia , Envejecimiento/inmunología , Animales , Bases de Datos Factuales , Perros , Humanos , Oncología Médica/métodos , Ratones , Neoplasias/inmunología , Técnicas de Cultivo de Órganos , Investigación Biomédica Traslacional/métodos
3.
Front Immunol ; 11: 1067, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582173

RESUMEN

The past two decades have brought impressive advancements in immune modulation, particularly with the advent of both cancer immunotherapy and biologic therapeutics for inflammatory conditions. However, the dynamic nature of the immune response often complicates the assessment of therapeutic outcomes. Innovative imaging technologies are designed to bridge this gap and allow non-invasive visualization of immune cell presence and/or function in real time. A variety of anatomical and molecular imaging modalities have been applied for this purpose, with each option providing specific advantages and drawbacks. Anatomical methods including magnetic resonance imaging (MRI), computed tomography (CT), and ultrasound provide sharp tissue resolution, which can be further enhanced with contrast agents, including super paramagnetic ions (for MRI) or nanobubbles (for ultrasound). Conjugation of the contrast material to an antibody allows for specific targeting of a cell population or protein of interest. Protein platforms including antibodies, cytokines, and receptor ligands are also popular choices as molecular imaging agents for positron emission tomography (PET), single-photon emission computerized tomography (SPECT), scintigraphy, and optical imaging. These tracers are tagged with either a radioisotope or fluorescent molecule for detection of the target. During the design process for immune-monitoring imaging tracers, it is important to consider any potential downstream physiologic impact. Antibodies may deplete the target cell population, trigger or inhibit receptor signaling, or neutralize the normal function(s) of soluble proteins. Alternatively, the use of cytokines or other ligands as tracers may stimulate their respective signaling pathways, even in low concentrations. As in vivo immune imaging is still in its infancy, this review aims to describe the modalities and immunologic targets that have thus far been explored, with the goal of promoting and guiding the future development and application of novel imaging technologies.


Asunto(s)
Sistema Inmunológico/diagnóstico por imagen , Imagen Molecular/métodos , Imagen Óptica/métodos , Animales , Anticuerpos/inmunología , Rastreo Celular , Citocinas/inmunología , Genes Reporteros , Humanos , Sistema Inmunológico/citología , Ligandos , Imagen por Resonancia Magnética , Tomografía de Emisión de Positrones , Radiofármacos , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X
4.
Pharmacol Ther ; 197: 212-224, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30759375

RESUMEN

Exposure to air pollution and other environmental inhalation hazards, such as occupational exposures to dusts and fumes, aeroallergens, and tobacco smoke, is a significant cause of chronic lung inflammation leading to respiratory disease. It is now recognized that resolution of inflammation is an active process controlled by a novel family of small lipid mediators termed "specialized pro-resolving mediators" or SPMs, derived mainly from dietary omega-3 polyunsaturated fatty acids. Chronic inflammation results from an imbalance between pro-inflammatory and pro-resolution pathways. Research is ongoing to develop SPMs, and the pro-resolution pathway more generally, as a novel therapeutic approach to diseases characterized by chronic inflammation. Here, we will review evidence that the resolution pathway is dysregulated in chronic lung inflammatory diseases, and that SPMs and related molecules have exciting therapeutic potential to reverse or prevent chronic lung inflammation, with a focus on lung inflammation due to inhalation of environmental hazards including urban particulate matter, organic dusts and tobacco smoke.


Asunto(s)
Contaminación del Aire , Fumar Cigarrillos , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos , Enfermedades Pulmonares/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Enfermedades Pulmonares/tratamiento farmacológico
5.
Cancer Res ; 78(19): 5706-5717, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30115693

RESUMEN

IFNγ is an attractive target for imaging active antitumor immunity due to its function in the T-cell signaling axis. Here, we test an IFNγ immuno-PET (immunoPET) probe for its capacity to identify adaptive immunotherapy response after HER2/neu vaccination in both spontaneous salivary and orthotopic neu+ mouse mammary tumors. IFNγ immunoPET detected elevated cytokine levels in situ after vaccination, which inversely correlated with tumor growth rate, an indicator of response to therapy. In a model of induced T-cell anergy where CD8 T cells infiltrate the tumor, but upregulate PD-1, IFNγ tracer uptake was equivalent to isotype control, illustrating a lack of antitumor T-cell activity. The IFNγ immunoPET tracer detected IFNγ protein sequestered on the surface of tumor cells, likely in complex with the IFNγ receptor, which may explain imaging localization of this soluble factor in vivo Collectively, we find that the activation status of cytotoxic T cells is annotated by IFNγ immunoPET, with reduced off-target binding to secondary lymphoid tissues compared with imaging total CD3+ tumor-infiltrating lymphocytes. Targeting of soluble cytokines such as IFNγ by PET imaging may provide valuable noninvasive insight into the function of immune cells in situ Significance: This study presents a novel approach to monitor therapeutic outcomes via IFNγ-targeted positron emission tomography. Cancer Res; 78(19); 5706-17. ©2018 AACR.


Asunto(s)
Inmunoterapia , Interferón gamma/metabolismo , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Tomografía de Emisión de Positrones , Animales , Unión Competitiva , Complejo CD3/metabolismo , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Islas de CpG , Femenino , Heterocigoto , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Animales/diagnóstico por imagen , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/terapia , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Receptor de Muerte Celular Programada 1/metabolismo , Radiofármacos , Saliva/metabolismo , Linfocitos T Citotóxicos/citología
6.
Am J Physiol Lung Cell Mol Physiol ; 314(3): L505-L513, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351447

RESUMEN

Cigarette smokers and people exposed to second-hand smoke are at an increased risk for pulmonary viral infections, and yet the mechanism responsible for this heightened susceptibility is not understood. To understand the effect of cigarette smoke on susceptibility to viral infection, we used an air-liquid interface culture system and exposed primary human small airway epithelial cells (SAEC) to whole cigarette smoke, followed by treatment with the viral mimetic polyinosinic polycytidylic acid (poly I:C) or influenza A virus (IAV). We found that prior smoke exposure strongly inhibited production of proinflammatory (interleukin-6 and interleukin-8) and antiviral [interferon-γ-induced protein 10 (IP-10) and interferons] mediators in SAECs in response to poly I:C and IAV infection. Impaired antiviral responses corresponded to increased infection with IAV. This was associated with a decrease in phosphorylation of the key antiviral transcription factor interferon response factor 3 (IRF3). Here, we found that cigarette smoke exposure inhibited activation of Toll-like receptor 3 (TLR3) by impairing TLR3 cleavage, which was required for downstream phosphorylation of IRF3 and production of IP-10. These results identify a novel mechanism by which cigarette smoke exposure impairs antiviral responses in lung epithelial cells, which may contribute to increased susceptibility to respiratory infections.


Asunto(s)
Antivirales/metabolismo , Células Epiteliales/inmunología , Gripe Humana/complicaciones , Interferón beta/metabolismo , Sistema Respiratorio/inmunología , Fumar/efectos adversos , Receptor Toll-Like 3/metabolismo , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Virus de la Influenza A/aislamiento & purificación , Gripe Humana/metabolismo , Gripe Humana/virología , Poli I-C/administración & dosificación , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Sistema Respiratorio/virología , Transducción de Señal
7.
Toxicol In Vitro ; 43: 76-86, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28572013

RESUMEN

Worldwide, over 4 million premature deaths each year are attributed to the burning of biomass fuels for cooking and heating. Epidemiological studies associate household air pollution with lung diseases, including chronic obstructive pulmonary disease, lung cancer, and respiratory infections. Animal dung, a biomass fuel used by economically vulnerable populations, generates more toxic compounds per mass burned than other biomass fuels. The type of animal dung used varies widely depending on local agro-geography. There are currently neither standardized experimental systems for dung biomass smoke research nor studies assessing the health impacts of different types of dung smoke. Here, we used a novel reproducible exposure system to assess outcomes related to inflammation and respiratory infections in human airway cells exposed to six different types of dung biomass smoke. We report that dung biomass smoke, regardless of species, is pro-inflammatory and activates the aryl hydrocarbon receptor and JNK transcription factors; however, dung smoke also suppresses interferon responses after a challenge with a viral mimetic. These effects are consistent with epidemiological data, and suggest a mechanism by which the combustion of animal dung can directly cause lung diseases, promote increased susceptibility to infection, and contribute to the global health problem of household air pollution.


Asunto(s)
Estiércol , Humo/efectos adversos , Animales , Biomasa , Línea Celular , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , MAP Quinasa Quinasa 4/metabolismo , Poli I-C/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción ReIA/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 311(6): L1222-L1233, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27836898

RESUMEN

Animal dung is a biomass fuel burned by vulnerable populations who cannot afford cleaner sources of energy, such as wood and gas, for cooking and heating their homes. Exposure to biomass smoke is the leading environmental risk for mortality, with over 4,000,000 deaths each year worldwide attributed to indoor air pollution from biomass smoke. Biomass smoke inhalation is epidemiologically associated with pulmonary diseases, including chronic obstructive pulmonary disease (COPD), lung cancer, and respiratory infections, especially in low and middle-income countries. Yet, few studies have examined the mechanisms of dung biomass smoke-induced inflammatory responses in human lung cells. Here, we tested the hypothesis that dung biomass smoke causes inflammatory responses in human lung cells through signaling pathways involved in acute and chronic lung inflammation. Primary human small airway epithelial cells (SAECs) were exposed to dung smoke at the air-liquid interface using a newly developed, automated, and reproducible dung biomass smoke generation system. The examination of inflammatory signaling showed that dung biomass smoke increased the production of several proinflammatory cytokines and enzymes in SAECs through activation of the activator protein (AP)-1 and arylhydrocarbon receptor (AhR) but not nuclear factor-κB (NF-κB) pathways. We propose that the inflammatory responses of lung cells exposed to dung biomass smoke contribute to the development of respiratory diseases.


Asunto(s)
Biomasa , Células Epiteliales/metabolismo , Células Epiteliales/patología , Inflamación/metabolismo , Inflamación/patología , Pulmón/patología , Transducción de Señal , Humo/efectos adversos , Animales , Compuestos Azo/farmacología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Caballos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Material Particulado/análisis , Pirazoles/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo
9.
Immunology ; 147(1): 41-54, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26555456

RESUMEN

The aryl hydrocarbon receptor (AhR) is a transcription factor that has been extensively studied as a regulator of toxicant metabolism. However, recent evidence indicates that the AhR also plays an important role in immunity. We hypothesized that the AhR is a novel, immune regulator of T helper type 2 (Th2) -mediated allergic airway disease. Here, we report that AhR-deficient mice develop increased allergic responses to the model allergen ovalbumin (OVA), which are driven in part by increased dendritic cell (DC) functional activation. AhR knockout (AhR(-/-) ) mice sensitized and challenged with OVA develop an increased inflammatory response in the lung compared with wild-type controls, with greater numbers of inflammatory eosinophils and neutrophils, greater T-cell proliferation, greater production of Th2 cytokines, and higher levels of OVA-specific IgE and IgG1. Lung DCs from AhR(-/-) mice stimulated antigen-specific proliferation and Th2 cytokine production by naive T cells in vitro. Additionally, AhR(-/-) DCs produced higher levels of tumour necrosis factor-α and interleukin-6, which promote Th2 differentiation, and expressed higher cell surface levels of stimulatory MHC Class II and CD86 molecules. Overall, loss of the AhR was associated with enhanced T-cell activation by pulmonary DCs and heightened pro-inflammatory allergic responses. This suggests that endogenous AhR ligands are involved in the normal regulation of Th2-mediated immunity in the lung via a DC-dependent mechanism. Therefore, the AhR may represent an important target for therapeutic intervention in allergic airways inflammation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Dendríticas/metabolismo , Pulmón/metabolismo , Neumonía/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Hipersensibilidad Respiratoria/metabolismo , Células Th2/metabolismo , Animales , Presentación de Antígeno , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Carbazoles/farmacología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Inmunidad Celular , Inmunidad Mucosa , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Ligandos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina , Neumonía/inducido químicamente , Neumonía/inmunología , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/deficiencia , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/inmunología , Hipersensibilidad Respiratoria/inducido químicamente , Hipersensibilidad Respiratoria/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Factores de Tiempo
10.
Am J Respir Cell Mol Biol ; 50(4): 737-47, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24175906

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is characterized by progressive fibrotic destruction of normal lung architecture. Due to a lack of effective treatment options, new treatment approaches are needed. We previously identified transglutaminase (TG)2, a multifunctional protein expressed by human lung fibroblasts (HLFs), as a positive driver of fibrosis. TG2 catalyzes crosslinking of extracellular matrix proteins, enhances cell binding to fibronectin and integrin, and promotes fibronectin expression. We investigated whether the small electrophilic molecules 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) and 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2) inhibit the expression and profibrotic functions of TG2. CDDO and 15d-PGJ2 reduced expression of TG2 mRNA and protein in primary HLFs from control donors and donors with IPF. CDDO and 15d-PGJ2 also decreased the in vitro profibrotic effector functions of HLFs including collagen gel contraction and cell migration. The decrease in TG2 expression did not occur through activation of the peroxisome proliferator activated receptor γ or generation of reactive oxidative species. CDDO and 15d-PGJ2 inhibited the extracellular signal-regulated kinase pathway, resulting in the suppression of TG2 expression. This is the first study to show that small electrophilic compounds inhibit the expression and profibrotic effector functions of TG2, a key promoter of fibrosis. These studies identify new and important antifibrotic activities of these two small molecules, which could lead to new treatments for fibrotic lung disease.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fibrosis Pulmonar Idiopática/enzimología , Pulmón/efectos de los fármacos , Ácido Oleanólico/análogos & derivados , Prostaglandina D2/análogos & derivados , Transglutaminasas/antagonistas & inhibidores , Estudios de Casos y Controles , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Inhibidores Enzimáticos/química , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/patología , Proteínas de Unión al GTP , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/enzimología , Pulmón/patología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Terapia Molecular Dirigida , Ácido Oleanólico/química , Ácido Oleanólico/farmacología , Fosforilación , Prostaglandina D2/química , Prostaglandina D2/farmacología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Inhibidores de Proteínas Quinasas/farmacología , Transglutaminasas/metabolismo
11.
J Org Chem ; 77(14): 6043-50, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22730896

RESUMEN

Two monosubstituted and one tetrasubstituted N-confused porphyrins (1-3) were prepared in ca. 3-5% yields using a [2 + 2] synthesis. The monosubstituted porphyrins have carbomethoxy (1) or nitro (2) substituents on one of the meso-phenyl groups, while the meso-phenyl groups of the third NCP (3) are substituted with nitro, bromo, and methyl groups in an AB(2)C pattern. The specific regiochemistry of the aryl rings around the macrocycle in each porphyrin was definitively determined using a combination of 1D ((1)H and (13)C) and 2D (gHMBC, gHSQC and ROESY) NMR spectroscopy. The absorption spectra of 1-3 in CH(2)Cl(2) are similar to those of N-confused tetraphenylporphyrin (NCTPP) but have Soret and Q bands that are shifted to lower energies with smaller extinction coefficients in comparison to those for NCTPP.


Asunto(s)
Porfirinas/síntesis química , Ciclización , Espectroscopía de Resonancia Magnética , Estructura Molecular , Porfirinas/química , Espectrofotometría Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...