Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Eur Respir J ; 60(6)2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35777774

RESUMEN

Respiratory diseases account for over 5 million deaths yearly and are a huge burden to healthcare systems worldwide. Murine models have been of paramount importance to decode human lung biology in vivo, but their genetic, anatomical, physiological and immunological differences with humans significantly hamper successful translation of research into clinical practice. Thus, to clearly understand human lung physiology, development, homeostasis and mechanistic dysregulation that may lead to disease, it is essential to develop models that accurately recreate the extraordinary complexity of the human pulmonary architecture and biology. Recent advances in micro-engineering technology and tissue engineering have allowed the development of more sophisticated models intending to bridge the gap between the native lung and its replicates in vitro Alongside advanced culture techniques, remarkable technological growth in downstream analyses has significantly increased the predictive power of human biology-based in vitro models by allowing capture and quantification of complex signals. Refined integrated multi-omics readouts could lead to an acceleration of the translational pipeline from in vitro experimental settings to drug development and clinical testing in the future. This review highlights the range and complexity of state-of-the-art lung models for different areas of the respiratory system, from nasal to large airways, small airways and alveoli, with consideration of various aspects of disease states and their potential applications, including pre-clinical drug testing. We explore how development of optimised physiologically relevant in vitro human lung models could accelerate the identification of novel therapeutics with increased potential to translate successfully from the bench to the patient's bedside.


Asunto(s)
Pulmón , Enfermedades Respiratorias , Humanos , Animales , Ratones , Pulmón/fisiología , Ingeniería de Tejidos/métodos
2.
Immunology ; 146(3): 470-85, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26288256

RESUMEN

Human mast cells (huMCs) are involved in both innate and adaptive immune responses where they release mediators including amines, reactive oxygen species (ROS), eicosanoids and cytokines. We have reported that interferon-γ (IFN-γ) enhances FcγR-dependent ROS production. The aim of this study was to extend these observations by investigating the effect of IFN-γ on the biological responses of huMCs to Staphylococcus aureus. We found that exposure of huMCs to S. aureus generated intracellular and extracellular ROS, which were enhanced in the presence of IFN-γ. IFN-γ also promoted bacteria killing, ß-hexosaminidase release and eicosanoid production. Interferon-γ similarly increased expression of mRNAs encoding CCL1 to CCL4, granulocyte-macrophage colony-stimulating factor (GM-CSF), tumour necrosis factor-α and CXCL8 in S. aureus-stimulated huMCs. The ability of IFN-γ to increase CXCL8 and GM-CSF protein levels was confirmed by ELISA. Fibronectin or a ß1 integrin blocking antibody completely abrogated IFN-γ-dependent S. aureus binding and reduced S. aureus-dependent CXCL8 secretion. These data demonstrate that IFN-γ primes huMCs for enhanced anti-bacterial and pro-inflammatory responses to S. aureus, partially mediated by ß1 integrin.


Asunto(s)
Interferón gamma/inmunología , Mastocitos/inmunología , Mastocitos/microbiología , Staphylococcus aureus/inmunología , Inmunidad Adaptativa , Animales , Células Cultivadas , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Citocinas/genética , Citocinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Integrina beta1/metabolismo , Interferón gamma/farmacología , Interleucina-8/genética , Interleucina-8/metabolismo , Mastocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Staphylococcus aureus/patogenicidad , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
J Allergy Clin Immunol ; 132(3): 665-675.e8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23632299

RESUMEN

BACKGROUND: Because TNF-α is increased in severe asthma, we hypothesized that TNF-α contributes to barrier dysfunction and cell activation in bronchial epithelial cells. We further hypothesized that src-family kinase inhibition would improve barrier function in healthy cells in the presence of TNF-α and directly in cultures of severe asthmatic cells where the barrier is disrupted. OBJECTIVES: We assessed the effect of TNF-α, with or without src-family kinase inhibitor SU6656, on barrier properties and cytokine release in differentiated human bronchial epithelial cultures. Further, we tested the effect of SU6656 on differentiated primary cultures from severe asthma. METHODS: Barrier properties of differentiated human bronchial epithelial air-liquid interface cultures from healthy subjects and subjects with severe asthma were assessed with transepithelial electrical resistance and fluorescent dextran passage. Proteins were detected by immunostaining or Western blot analysis and cytokines by immunoassay. Mechanisms were investigated with src kinase and other inhibitors. RESULTS: TNF-α lowered transepithelial electrical resistance and increased fluorescent dextran permeability, caused loss of occludin and claudins from tight junctions with redistribution of p120 catenin and E-cadherin from adherens junctions, and also increased endogenous TNF-α, IL-6, IL-1ß, IL-8, thymic stromal lymphoprotein, and pro-matrix metalloprotease 9 release. SU6656 reduced TNF-α-mediated paracellular permeability changes, restored occludin, p120, and E-cadherin and lowered autocrine TNF-α release. Importantly, SU6656 improved the barrier properties of severe asthmatic air-liquid interface cultures. Redistribution of E-cadherin and p120 was observed in bronchial biopsies from severe asthmatic airways. CONCLUSIONS: Inhibiting TNF-α or src kinases may be a therapeutic option to normalize barrier integrity and cytokine release in airway diseases associated with barrier dysfunction.


Asunto(s)
Asma/metabolismo , Bronquios/metabolismo , Células Epiteliales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Familia-src Quinasas/metabolismo , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/metabolismo , Bronquios/citología , Cadherinas/metabolismo , Cateninas/metabolismo , Células Cultivadas , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Humanos , Indoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Sulfonamidas/farmacología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Familia-src Quinasas/antagonistas & inhibidores , Catenina delta
4.
Tissue Barriers ; : 2300580, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38179897

RESUMEN

Lipids and their mediators have important regulatory functions in many cellular processes, including the innate antiviral response. The aim of this study was to compare the lipid membrane composition of in vitro differentiated primary bronchial epithelial cells (PBECs) with ex vivo bronchial brushings and to establish whether any changes in the lipid membrane composition affect antiviral defense of cells from donors without and with severe asthma. Using mass spectrometry, we showed that the lipid membrane of in vitro differentiated PBECs was deprived of polyunsaturated fatty acids (PUFAs) compared to ex vivo bronchial brushings. Supplementation of the culture medium with arachidonic acid (AA) increased the PUFA-content to more closely match the ex vivo membrane profile. Rhinovirus (RV16) infection of AA-supplemented cultures from healthy donors resulted in significantly reduced viral replication while release of inflammatory mediators and prostaglandin E2 (PGE2) was significantly increased. Indomethacin, an inhibitor of prostaglandin-endoperoxide synthases, suppressed RV16-induced PGE2 release and significantly reduced CXCL-8/IL-8 release from AA-supplemented cultures indicating a link between PGE2 and CXCL8/IL-8 release. In contrast, in AA-supplemented cultures from severe asthmatic donors, viral replication was enhanced whereas PTGS2 expression and PGE2 release were unchanged and CXCL8/IL-8 was significantly reduced in response to RV16 infection. While the PTGS2/COX-2 pathway is initially pro-inflammatory, its downstream products can promote symptom resolution. Thus, reduced PGE2 release during an RV-induced severe asthma exacerbation may lead to prolonged symptoms and slower recovery. Our data highlight the importance of reflecting the in vivo lipid profile in in vitro cell cultures for mechanistic studies.

5.
Eur Respir J ; 42(1): 87-97, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23143548

RESUMEN

The airway epithelium forms a physical, chemical and immunological barrier against inhaled environmental substances. In asthma, these barrier properties are thought to be abnormal. In this study, we analysed the effect of grass pollen on the physical and immunological barrier properties of differentiated human primary bronchial epithelial cells. Following exposure to Timothy grass (Phleum pratense) pollen extract, the integrity of the physical barrier was not impaired as monitored by measuring the transepithelial resistance and immunofluorescence staining of tight junction proteins. In contrast, pollen exposure affected the immunological barrier properties by modulating vectorial mediator release. CXC chemokine ligand (CXCL)8/interleukin (IL)-8 showed the greatest increase in response to pollen exposure with preferential release to the apical compartment. Inhibition of the extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase pathways selectively blocked apical CXCL8/IL-8 release via a post-transcriptional mechanism. Apical release of CC chemokine ligand (CCL)20/macrophage inflammatory protein-3α, CCL22/monocyte-derived chemokine and tumour necrosis factor-α was significantly increased only in severe asthma cultures, while CCL11/eotaxin-1 and CXCL10/interferon-γ-induced protein-10 were reduced in nonasthmatic cultures. The bronchial epithelial barrier modulates polarised release of mediators in response to pollen without direct effects on its physical barrier properties. The differential response of cells from normal and asthmatic donors suggests the potential for the bronchial epithelium to promote immune dysfunction in asthma.


Asunto(s)
Asma/inmunología , Bronquios/patología , Células Epiteliales/patología , Extractos Vegetales/química , Polen/química , Alérgenos/química , Asma/metabolismo , Broncoscopía , Células Cultivadas , Quimiocinas/inmunología , Humanos , Inflamación , Interleucina-8/inmunología , Ligandos , Poaceae
6.
Brain ; 135(Pt 4): 1102-14, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22436237

RESUMEN

Sporadic inclusion body myositis is a severely disabling myopathy. The design of effective treatment strategies is hampered by insufficient understanding of the complex disease pathology. Particularly, the nature of interrelationships between inflammatory and degenerative pathomechanisms in sporadic inclusion body myositis has remained elusive. In Alzheimer's dementia, accumulation of ß-amyloid has been shown to be associated with upregulation of nitric oxide. Using quantitative polymerase chain reaction, an overexpression of inducible nitric oxide synthase was observed in five out of ten patients with sporadic inclusion body myositis, two of eleven with dermatomyositis, three of eight with polymyositis, two of nine with muscular dystrophy and two of ten non-myopathic controls. Immunohistochemistry confirmed protein expression of inducible nitric oxide synthase and demonstrated intracellular nitration of tyrosine, an indicator for intra-fibre production of nitric oxide, in sporadic inclusion body myositis muscle samples, but much less in dermatomyositis or polymyositis, hardly in dystrophic muscle and not in non-myopathic controls. Using fluorescent double-labelling immunohistochemistry, a significant co-localization was observed in sporadic inclusion body myositis muscle between ß-amyloid, thioflavine-S and nitrotyrosine. In primary cultures of human myotubes and in myoblasts, exposure to interleukin-1ß in combination with interferon-γ induced a robust upregulation of inducible nitric oxide synthase messenger RNA. Using fluorescent detectors of reactive oxygen species and nitric oxide, dichlorofluorescein and diaminofluorescein, respectively, flow cytometry revealed that interleukin-1ß combined with interferon-γ induced intracellular production of nitric oxide, which was associated with necrotic cell death in muscle cells. Intracellular nitration of tyrosine was noted, which partly co-localized with amyloid precursor protein, but not with desmin. Pharmacological inhibition of inducible nitric oxide synthase by 1400W reduced intracellular production of nitric oxide and prevented accumulation of ß-amyloid, nitration of tyrosine as well as cell death inflicted by interleukin-1ß combined with interferon-γ. Collectively, these data suggest that, in skeletal muscle, inducible nitric oxide synthase is a central component of interactions between interleukin-1ß and ß-amyloid, two of the most relevant molecules in sporadic inclusion body myositis. The data further our understanding of the pathology of sporadic inclusion body myositis and may point to novel treatment strategies.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-1beta/farmacología , Células Musculares/metabolismo , Miositis por Cuerpos de Inclusión/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Dactinomicina/análogos & derivados , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Regulación de la Expresión Génica/fisiología , Humanos , Interferón gamma/farmacología , Células Musculares/efectos de los fármacos , Miositis por Cuerpos de Inclusión/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , ARN Mensajero/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
7.
RSC Adv ; 13(21): 14494-14500, 2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37179995

RESUMEN

A droplet generator has been developed that interfaces with a barrier-on-chip platform for temporal analyte compartmentalisation and analysis. Droplets are generated every 20 minutes in 8 separate parallel microchannels, with an average droplet volume of 9.47 ± 0.6 µL, allowing simultaneous analysis of 8 different experiments. The device was tested using an epithelial barrier model by monitoring the diffusion of a fluorescent high molecular weight dextran molecule. The epithelial barrier was perturbed using detergent leading to a peak at 3-4 hours, correlating with simulations. For the untreated (control) a constant, very low level of dextran diffusion was observed. The epithelial cell barrier properties were also continuously measured using electrical impedance spectroscopy to extract an equivalent trans epithelial resistance.

8.
Viruses ; 14(11)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36366528

RESUMEN

Mast cells (MCs) are classically associated with allergic asthma but their role in antiviral immunity is unclear. Human rhinoviruses (HRVs) are a major cause of asthma exacerbations and can infect and replicate within MCs. The primary site of HRV infection is the airway epithelium and MCs localise to this site with increasing asthma severity. The asthma susceptibility gene, IL-33, encodes an epithelial-derived cytokine released following HRV infection but its impact on MC antiviral responses has yet to be determined. In this study we investigated the global response of LAD2 MCs to IL-33 stimulation using RNA sequencing and identified genes involved in antiviral immunity. In spite of this, IL-33 treatment increased permissiveness of MCs to HRV16 infection which, from the RNA-Seq data, we attributed to upregulation of ICAM1. Flow cytometric analysis confirmed an IL-33-dependent increase in ICAM1 surface expression as well as LDLR, the receptors used by major and minor group HRVs for cellular entry. Neutralisation of ICAM1 reduced the IL-33-dependent enhancement in HRV16 replication and release in both LAD2 MCs and cord blood derived MCs. These findings demonstrate that although IL-33 induces an antiviral signature in MCs, it also upregulates the receptors for HRV entry to enhance infection. This highlights the potential for a gene-environment interaction involving IL33 and HRV in MCs to contribute to virus-induced asthma exacerbations.


Asunto(s)
Asma , Infecciones por Picornaviridae , Humanos , Rhinovirus/fisiología , Interleucina-33/farmacología , Mastocitos/metabolismo , Antivirales/farmacología , Tolerancia , Replicación Viral , Células Epiteliales
9.
Lab Chip ; 22(10): 2041-2054, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35485428

RESUMEN

A multichannel microfluidic platform for real-time monitoring of epithelial barrier integrity by electrical impedance has been developed. Growth and polarization of human epithelial cells from the airway or gastrointestinal tract was continuously monitored over 5 days in 8 parallel, individually perfused microfluidic chips. Electrical impedance data were continuously recorded to monitor cell barrier formation using a low-cost bespoke impedance analyser. Data was analysed using an electric circuit model to extract the equivalent transepithelial electrical resistance and epithelial cell layer capacitance. The cell barrier integrity steadily increased overtime, achieving an average resistance of 418 ± 121 Ω cm2 (airway cells) or 207 ± 59 Ω cm2 (gastrointestinal cells) by day 5. The utility of the polarized airway epithelial barrier was demonstrated using a 24 hour challenge with double stranded RNA to mimic viral infection. This caused a rapid decrease in barrier integrity in association with disruption of tight junctions, whereas simultaneous treatment with a corticosteroid reduced this effect. The platform is able to measure barrier integrity in real-time and is scalable, thus has the potential to be used for drug development and testing.


Asunto(s)
Espectroscopía Dieléctrica , Microfluídica , Impedancia Eléctrica , Células Epiteliales , Humanos , Uniones Estrechas
10.
Biotechniques ; 2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34269076

RESUMEN

Single-cell RNA sequencing (scRNA-seq) of the bronchial epithelium enables examination of cellular subtypes and their responses to viral infections. Here, an optimized method for the isolation of virally infected primary bronchial epithelial cells using a commercially available microfluidic device is presented. Using this method single cells can be rapidly isolated with minimal equipment available in most laboratories. Isolation can be carried out inside biological safety cabinets, permitting the use of virally infected cells. Both cell-line and primary cells isolated using the device retained sufficient RNA integrity for the generation of short-read sequencing-compatible cDNA libraries to facilitate scRNA-seq.

11.
Lab Chip ; 10(12): 1611-7, 2010 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-20379587

RESUMEN

A bio-impedance chip has been developed for real-time monitoring of the kinetics of epithelial cell monolayers in vitro. The human bronchial epithelial cell line (16-HBE 14o-) was cultured in Transwells creating a sustainable and interactive model of the airway epithelium. Conducting polymer polypyrrole (PPy) doped with polystyrene sulfonate (PSS) was electrochemically deposited onto the surface of gold-plated electrodes to reduce the influence of the electrical double layer on the impedance measurements. Finite element and equivalent circuit models were used to model and determine the electrical properties of the epithelial cell monolayer from the impedance spectra. Electrically tight, confluent monolayers of 16 HBE 14o- cells were treated with increasing concentrations of either Triton X-100 to solubilize cell membranes or ethylene glycol-bis(2-aminoethyl-ether)-N,N,N'N'-tetraacetic acid (EGTA) to disrupt cell-cell adhesion. Experimental impedance data showed that disruption of epithelial barrier function in response to Triton X-100 and EGTA can be successfully measured by the bio-impedance chip. The results were consistent with the conventional hand-held trans-epithelial electrical resistance measurements. Immunofluorescent staining of the ZO-1 tight junction protein in the untreated and treated 16HBEs was performed to verify the disruption of the tight junctions by EGTA.


Asunto(s)
Células Epiteliales/citología , Procedimientos Analíticos en Microchip , Línea Celular , Impedancia Eléctrica , Electrodos , Humanos , Cinética , Polímeros/química , Poliestirenos/química , Pirroles/química , Factores de Tiempo
12.
J Immunol ; 181(11): 7706-12, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19017959

RESUMEN

Activated mast cells are a major source of the eicosanoids PGD(2) and leukotriene C(4) (LTC(4)), which contribute to allergic responses. These eicosanoids are produced following the ERK1/2-dependent activation of cytosolic phospholipase A(2), thus liberating arachidonic acid, which is subsequently metabolized by the actions of 5-lipoxygenase and cyclooxygenase to form LTC(4) and PGD(2), respectively. These pathways also generate reactive oxygen species (ROS), which have been proposed to contribute to FcepsilonRI-mediated signaling in mast cells. In this study, we demonstrate that, in addition to ERK1/2-dependent pathways, ERK1/2-independent pathways also regulate FcepsilonRI-mediated eicosanoid and ROS production in mast cells. A role for the Tec kinase Btk in the ERK1/2-independent regulatory pathway was revealed by the significantly attenuated FcepsilonRI-dependent PGD(2), LTC(4), and ROS production in bone marrow-derived mast cells of Btk(-/-) mice. The FcepsilonRI-dependent activation of Btk and eicosanoid and ROS generation in bone marrow-derived mast cells and human mast cells were similarly blocked by the PI3K inhibitors, Wortmannin and LY294002, indicating that Btk-regulated eicosanoid and ROS production occurs downstream of PI3K. In contrast to ERK1/2, the PI3K/Btk pathway does not regulate cytosolic phospholipase A(2) phosphorylation but rather appears to regulate the generation of ROS, LTC(4), and PGD(2) by contributing to the necessary Ca(2+) signal for the production of these molecules. These data demonstrate that strategies to decrease mast cell production of ROS and eicosanoids would have to target both ERK1/2- and PI3K/Btk-dependent pathways.


Asunto(s)
Antígenos/farmacología , Leucotrieno C4/inmunología , Mastocitos/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Prostaglandina D2/inmunología , Proteínas Tirosina Quinasas/inmunología , Especies Reactivas de Oxígeno/inmunología , Agammaglobulinemia Tirosina Quinasa , Androstadienos/farmacología , Animales , Antígenos/genética , Antígenos/inmunología , Antígenos/metabolismo , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/inmunología , Araquidonato 5-Lipooxigenasa/metabolismo , Ácido Araquidónico/genética , Ácido Araquidónico/inmunología , Ácido Araquidónico/metabolismo , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/inmunología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Señalización del Calcio/inmunología , Cromonas/farmacología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Activación Enzimática/inmunología , Inhibidores Enzimáticos/farmacología , Humanos , Hipersensibilidad/enzimología , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Leucotrieno C4/biosíntesis , Leucotrieno C4/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Mastocitos/enzimología , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfolipasas A2 Citosólicas , Fosforilación/efectos de los fármacos , Prostaglandina D2/biosíntesis , Prostaglandina D2/genética , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo
13.
J Allergy Clin Immunol ; 124(1): 23-34; quiz 35-6, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19560576

RESUMEN

The bronchial epithelium is pivotally involved in the provision of chemical, physical, and immunologic barriers to the inhaled environment. These barriers serve to maintain normal homeostasis, but when compromised, the immunologic barrier becomes activated to protect the internal milieu of the lung. We discuss what is currently understood about abnormalities in these barrier functions in patients with asthma and consider novel therapeutic opportunities that target this key structure.


Asunto(s)
Asma/fisiopatología , Asma/terapia , Sistema Inmunológico , Preparaciones Farmacéuticas , Mucosa Respiratoria/patología , Virosis , Asma/inmunología , Humanos
14.
Methods Mol Biol ; 2163: 85-89, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32766968

RESUMEN

Mouse bone marrow-derived mast cells (mBMMCs) are an invaluable tool for the study of mast cell function as they represent a primary source of mature mast cells. They can be sourced from wild-type, knockout, and transgenic mice and are used to repopulate mast cell-deficient mice. This method describes the isolation of mast cell hematopoietic progenitors from the bone marrow of mouse femurs and their subsequent culture in an IL-3-rich culture medium. After 4 weeks in culture, mBMMCs are obtained in high number and are of high purity. Assessment of their granularity by toluidine staining and IgE receptor expression by flow cytometry is also described. These cells are a useful tool in the determination of in vitro and in vivo mast cell function in innate and adaptive immunity.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Mastocitos/citología , Células Madre Mesenquimatosas/citología , Cultivo Primario de Células/métodos , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células Cultivadas , Medios de Cultivo/química , Medios de Cultivo/farmacología , Interleucina-3/análisis , Interleucina-3/farmacología , Mastocitos/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Receptores de IgE/genética , Receptores de IgE/metabolismo
16.
J Immunol Methods ; 477: 112703, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31711888

RESUMEN

Dendritic cells (DCs) play a central role in regulating innate and adaptive immune responses. It is well accepted that their regulatory functions change over the life course. In order to study DCs function during early life it is important to characterize the function of neonatal DCs. However, the availability of neonatal DCs is limited due to ethical reasons or relative small samples of cord blood making it difficult to perform large-scale experiments. Our aim was to establish a robust protocol for the generation of neonatal DCs from cord blood derived CD34+ hematopoietic stem cells. For the expansion of DC precursor cells we used a cytokine cocktail containing Flt-3 L, SCF, TPO, IL-3 and IL-6. The presence of IL-3 and IL-6 in the first 2 weeks of expansion culture was essential for the proliferation of DC precursor cells expressing CD14. After 4 weeks in culture, CD14+ precursor cells were selected and functional DCs were generated in the presence of GM-CSF and IL-4. Neonatal DCs were then stimulated with Poly(I:C) and LPS to mimic viral or bacterial infections, respectively. Poly(I:C) induced a higher expression of the maturation markers CD80, CD86 and CD40 compared to LPS. In line with literature data using cord blood DCs, our Poly(I:C) matured neonatal DCs cells showed a higher release of IL-12p70 compared to LPS matured neonatal DCs. Additionally, we demonstrated a higher release of IFN-γ, TNF-α, IL-1ß and IL-6, but lower release of IL-10 in Poly(I:C) matured compared to LPS matured neonatal DCs derived from cord blood CD34+ hematopoietic stem cells. In summary, we established a robust protocol for the generation of large numbers of functional neonatal DCs. In line with previous studies, we showed that neonatal DCs generated form CD34+ cord blood progenitors have a higher inflammatory potential when exposed to viral than bacterial related stimuli.


Asunto(s)
Sangre Fetal/citología , Células Madre Hematopoyéticas/fisiología , Cultivo Primario de Células/métodos , Adyuvantes Inmunológicos/farmacología , Antígenos CD34/metabolismo , Infecciones Bacterianas/inmunología , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Cesárea , Medios de Cultivo/metabolismo , Citocinas/metabolismo , Células Dendríticas , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Recién Nacido , Lipopolisacáridos/inmunología , Poli I-C/inmunología , Virosis/inmunología
17.
Sci Rep ; 9(1): 9789, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31278312

RESUMEN

Engineering tissue structures that mimic those found in vivo remains a challenge for modern biology. We demonstrate a new technique for engineering composite structures of cells comprising layers of heterogeneous cell types. An acoustofluidic bioreactor is used to assemble epithelial cells into a sheet-like structure. On transferring these cell sheets to a confluent layer of fibroblasts, the epithelial cells cover the fibroblast surface by collective migration maintaining distinct epithelial and fibroblast cell layers. The collective behaviour of the epithelium is dependent on the formation of cell-cell junctions during levitation and contrasts with the behaviour of mono-dispersed epithelial cells where cell-matrix interactions dominate and hinder formation of discrete cell layers. The multilayered tissue model is shown to form a polarised epithelial barrier and respond to apical challenge. The method is useful for engineering a wide range of layered tissue types and mechanistic studies on collective cell migration.


Asunto(s)
Ingeniería de Tejidos , Acústica , Animales , Biomarcadores , Reactores Biológicos , Adhesión Celular , Impedancia Eléctrica , Células Epiteliales , Fibroblastos , Humanos
18.
Immun Inflamm Dis ; 5(1): 45-56, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28250924

RESUMEN

INTRODUCTION: The epithelial and endothelial barriers of the airway mucosa are critical for regulation of tissue homeostasis and protection against pathogens or other tissue damaging agents. In response to a viral infection, epithelial cells must signal to the endothelium to initiate immune cell recruitment. This is a highly temporal regulated process; however, the mechanisms of this cross-talk are not fully understood. METHODS: In a close-contact co-culture model of human airway epithelial and endothelial cells, cellular crosstalk was analyzed using transepithelial electrical resistance (TER) measurements, immunofluorescence, electron microscopy, and ELISA. Viral infections were simulated by exposing airway epithelial cells apically to double-stranded RNA (Poly(I:C)). Using a microfluidic culture system, the temporal release of mediators was analyzed in the co-culture model. RESULTS: Within 4 h of challenge, double-stranded RNA induced the release of TNF-α by epithelial cells. This activated endothelial cells by triggering the release of the chemoattractant CX3CL1 (fractalkine) by 8 h post-challenge and expression of adhesion molecules E-selectin and ICAM-1. These responses were significantly reduced by neutralising TNF-α. CONCLUSION: By facilitating kinetic profiling, the microfluidic co-culture system has enabled identification of a key signaling mechanism between the epithelial and endothelial barriers. Better understanding of cell-cell cross-talk and its regulatory mechanisms has the potential to identify new therapeutic strategies to control airway inflammation.


Asunto(s)
Comunicación Celular , Células Epiteliales/fisiología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Bronquios/citología , Línea Celular , Células Cultivadas , Quimiocina CX3CL1/metabolismo , Técnicas de Cocultivo , Selectina E/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Microfluídica , Poli I-C/farmacología , ARN Bicatenario/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
19.
Pathogens ; 5(3)2016 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-27527221

RESUMEN

The bronchial epithelium provides protection against pathogens from the inhaled environment through the formation of a highly-regulated barrier. In order to understand the pulmonary diseases melioidosis and tularemia caused by Burkholderia thailandensis and Fransicella tularensis, respectively, the barrier function of the human bronchial epithelium were analysed. Polarised 16HBE14o- or differentiated primary human bronchial epithelial cells (BECs) were exposed to increasing multiplicities of infection (MOI) of B. thailandensis or F. tularensis Live Vaccine Strain and barrier responses monitored over 24-72 h. Challenge of polarized BECs with either bacterial species caused an MOI- and time-dependent increase in ionic permeability, disruption of tight junctions, and bacterial passage from the apical to the basolateral compartment. B. thailandensis was found to be more invasive than F. tularensis. Both bacterial species induced an MOI-dependent increase in TNF-α release. An increase in ionic permeability and TNF-α release was induced by B. thailandensis in differentiated BECs. Pretreatment of polarised BECs with the corticosteroid fluticasone propionate reduced bacterial-dependent increases in ionic permeability, bacterial passage, and TNF-α release. TNF blocking antibody Enbrel(®) reduced bacterial passage only. BEC barrier properties are disrupted during respiratory bacterial infections and targeting with corticosteroids or anti-TNF compounds may represent a therapeutic option.

20.
Tissue Barriers ; 4(3): e1206378, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27583193

RESUMEN

The bronchial epithelium and underlying fibroblasts form an epithelial mesenchymal trophic unit (EMTU) which controls the airway microenvironment. We hypothesized that cell-cell communication within the EMTU propagates and amplifies the innate immune response to respiratory viral infections. EMTU co-culture models incorporating polarized (16HBE14o-) or differentiated primary human bronchial epithelial cells (HBECs) and fibroblasts were challenged with double-stranded RNA (dsRNA) or rhinovirus. In the polarized EMTU model, dsRNA affected ionic but not macromolecular permeability or cell viability. Compared with epithelial monocultures, dsRNA-stimulated pro-inflammatory mediator release was synergistically enhanced in the basolateral compartment of the EMTU model, with the exception of IL-1α which was unaffected by the presence of fibroblasts. Blockade of IL-1 signaling with IL-1 receptor antagonist (IL-1Ra) completely abrogated dsRNA-induced basolateral release of mediators except CXCL10. Fibroblasts were the main responders to epithelial-derived IL-1 since exogenous IL-1α induced pro-inflammatory mediator release from fibroblast but not epithelial monocultures. Our findings were confirmed in a differentiated EMTU model where rhinovirus infection of primary HBECs and fibroblasts resulted in synergistic induction of basolateral IL-6 that was significantly abrogated by IL-1Ra. This study provides the first direct evidence of integrated IL-1 signaling within the EMTU to propagate inflammatory responses to viral infection.


Asunto(s)
Comunicación Celular , Microambiente Celular , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Interleucina-1alfa/metabolismo , Mucosa Respiratoria/metabolismo , Permeabilidad Capilar , Línea Celular , Células Cultivadas , Quimiocina CXCL10/metabolismo , Células Epiteliales/virología , Fibroblastos/virología , Humanos , Mucosa Respiratoria/citología , Mucosa Respiratoria/virología , Rhinovirus/patogenicidad , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA