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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 21(11): 1327-1335, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32839612

RESUMO

Although animal models have been evaluated for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, none have fully recapitulated the lung disease phenotypes seen in humans who have been hospitalized. Here, we evaluate transgenic mice expressing the human angiotensin I-converting enzyme 2 (ACE2) receptor driven by the cytokeratin-18 (K18) gene promoter (K18-hACE2) as a model of SARS-CoV-2 infection. Intranasal inoculation of SARS-CoV-2 in K18-hACE2 mice results in high levels of viral infection in lungs, with spread to other organs. A decline in pulmonary function occurs 4 days after peak viral titer and correlates with infiltration of monocytes, neutrophils and activated T cells. SARS-CoV-2-infected lung tissues show a massively upregulated innate immune response with signatures of nuclear factor-κB-dependent, type I and II interferon signaling, and leukocyte activation pathways. Thus, the K18-hACE2 model of SARS-CoV-2 infection shares many features of severe COVID-19 infection and can be used to define the basis of lung disease and test immune and antiviral-based countermeasures.


Assuntos
Betacoronavirus/imunologia , Infecções por Coronavirus/patologia , Imunidade Inata/imunologia , Peptidil Dipeptidase A/genética , Pneumonia Viral/patologia , Pneumonia/patologia , Enzima de Conversão de Angiotensina 2 , Animais , COVID-19 , Chlorocebus aethiops , Infecções por Coronavirus/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Interferon Tipo I/imunologia , Interferon gama/imunologia , Queratina-18/genética , Leucócitos/imunologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , NF-kappa B/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Pandemias , Pneumonia/genética , Pneumonia/virologia , Pneumonia Viral/imunologia , Regiões Promotoras Genéticas/genética , SARS-CoV-2 , Linfócitos T/imunologia , Células Vero , Replicação Viral/imunologia
3.
Arch Toxicol ; 97(7): 1847-1858, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37166470

RESUMO

Arsenic trioxide (ATO), an inorganic arsenical, is a toxic environmental contaminant. It is also a widely used chemical with industrial and medicinal uses. Significant public health risk exists from its intentional or accidental exposure. The pulmonary pathology of acute high dose exposure is not well defined. We developed and characterized a murine model of a single inhaled exposure to ATO, which was evaluated 24 h post-exposure. ATO caused hypoxemia as demonstrated by arterial blood-gas measurements. ATO administration caused disruption of alveolar-capillary membrane as shown by increase in total protein and IgM in the bronchoalveolar lavage fluid (BALF) supernatant and an onset of pulmonary edema. BALF of ATO-exposed mice had increased HMGB1, a damage-associated molecular pattern (DAMP) molecule, and differential cell counts revealed increased neutrophils. BALF supernatant also showed an increase in protein levels of eotaxin/CCL-11 and MCP-3/CCL-7 and a reduction in IL-10, IL-19, IFN-γ, and IL-2. In the lung of ATO-exposed mice, increased protein levels of G-CSF, CXCL-5, and CCL-11 were noted. Increased mRNA levels of TNF-a, and CCL2 in ATO-challenged lungs further supported an inflammatory pathogenesis. Neutrophils were increased in the blood of ATO-exposed animals. Pulmonary function was also evaluated using flexiVent. Consistent with an acute lung injury phenotype, respiratory and lung elastance showed significant increase in ATO-exposed mice. PV loops showed a downward shift and a decrease in inspiratory capacity in the ATO mice. Flow-volume curves showed a decrease in FEV0.1 and FEF50. These results demonstrate that inhaled ATO leads to pulmonary damage and characteristic dysfunctions resembling ARDS in humans.


Assuntos
Lesão Pulmonar Aguda , Arsenicais , Humanos , Camundongos , Animais , Modelos Animais de Doenças , Pulmão/patologia , Líquido da Lavagem Broncoalveolar/química
4.
Am J Physiol Lung Cell Mol Physiol ; 323(1): L107-L120, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35670484

RESUMO

Despite decades of research, studies investigating the physiological alterations caused by an acute bout of inflammation induced by exposing the lung to lipopolysaccharide have yielded inconsistent results. This can be attributed to small effects and/or a lack of fitted physiological testing. Herein, a comprehensive investigation of lung mechanics was conducted on 270 male C57BL/6 mice at 24, 48, or 96 h after an intranasal exposure to saline or lipopolysaccharide at either 1 or 3 mg/kg (30 mice per group). Traditional techniques that probe the lung using small-amplitude perturbations (i.e., oscillometry) were used, together with less conventional and new techniques that probe the lung using maneuvers of large amplitudes. The latter include a partial and a full-range pressure-volume maneuvers to measure quasi-static elastance, compliance, total lung volume, vital capacity, and residual volume. The results demonstrate that lung mechanics assessed by oscillometry was only slightly affected by lipopolysaccharide, confirming previous findings. In contradistinction, lipopolysaccharide markedly altered mechanics when the lung was probed with maneuvers of large amplitudes. With the dose of 3 mg/kg at the peak of inflammation (48 h postexposure), lipopolysaccharide increased quasi-static elastance by 26.7% (P < 0.0001) and decreased compliance by 34.5% (P < 0.0001). It also decreased lung volumes, including total lung capacity, vital capacity, and residual volume by 33.3%, 30.5%, and 43.3%, respectively (all P < 0.0001). These newly reported physiological alterations represent sensitive outcomes to efficiently evaluate countermeasures (e.g., drugs) in the context of several lung diseases.


Assuntos
Lipopolissacarídeos , Respiração com Pressão Positiva , Animais , Inflamação , Lipopolissacarídeos/farmacologia , Pulmão/fisiologia , Complacência Pulmonar , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Respiração com Pressão Positiva/métodos , Mecânica Respiratória/fisiologia
5.
Am J Physiol Lung Cell Mol Physiol ; 322(2): L294-L304, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34936511

RESUMO

There are renewed interests in using the parameter K of Salazar-Knowles' equation to assess lung tissue compliance. K either decreases or increases when the lung's parenchyma stiffens or loosens, respectively. However, whether K is affected by other common features of respiratory diseases, such as inflammation and airway smooth muscle (ASM) contraction, is unknown. Herein, male C57BL/6 mice were treated intranasally with either saline or lipopolysaccharide (LPS) at 1 mg/kg to induce pulmonary inflammation. They were then subjected to either a multiple or a single-dose challenge with methacholine to activate ASM to different degrees. A quasi-static pressure-driven partial pressure-volume (P-V) maneuver was performed before and after methacholine. The Salazar-Knowles' equation was then fitted to the deflation limb of the P-V loop to obtain K, as well as the parameter A, an estimate of lung volume (inspiratory capacity). The fitted curve was also used to derive the quasi-static elastance (Est) at 5 cmH2O. The results demonstrate that LPS and both methacholine challenges increased Est. LPS also decreased A, but did not affect K. In contradistinction, methacholine decreased both A and K in the multiple-dose challenge, whereas it decreased K but not A in the single-dose challenge. These results suggest that LPS increases Est by reducing the open lung volume (A) and without affecting tissue compliance (K), whereas methacholine increases Est by decreasing tissue compliance with or without affecting lung volume. We conclude that lung tissue compliance, assessed using the parameter K of Salazar-Knowles' equation, is insensitive to inflammation but sensitive to ASM contraction.


Assuntos
Lipopolissacarídeos , Pulmão , Resistência das Vias Respiratórias , Animais , Inflamação , Lipopolissacarídeos/farmacologia , Complacência Pulmonar , Masculino , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular , Mecânica Respiratória
6.
FASEB J ; 35(3): e21376, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33605487

RESUMO

Emphysema, a component of chronic obstructive pulmonary disease (COPD), is characterized by irreversible alveolar destruction that results in a progressive decline in lung function. This alveolar destruction is caused by cigarette smoke, the most important risk factor for COPD. Only 15%-20% of smokers develop COPD, suggesting that unknown factors contribute to disease pathogenesis. We postulate that the aryl hydrocarbon receptor (AHR), a receptor/transcription factor highly expressed in the lungs, may be a new susceptibility factor whose expression protects against COPD. Here, we report that Ahr-deficient mice chronically exposed to cigarette smoke develop airspace enlargement concomitant with a decline in lung function. Chronic cigarette smoke exposure also increased cleaved caspase-3, lowered SOD2 expression, and altered MMP9 and TIMP-1 levels in Ahr-deficient mice. We also show that people with COPD have reduced expression of pulmonary and systemic AHR, with systemic AHR mRNA levels positively correlating with lung function. Systemic AHR was also lower in never-smokers with COPD. Thus, AHR expression protects against the development of COPD by controlling interrelated mechanisms involved in the pathogenesis of this disease. This study identifies the AHR as a new, central player in the homeostatic maintenance of lung health, providing a foundation for the AHR as a novel therapeutic target and/or predictive biomarker in chronic lung disease.


Assuntos
Doença Pulmonar Obstrutiva Crônica/etiologia , Receptores de Hidrocarboneto Arílico/deficiência , Idoso , Idoso de 80 Anos ou mais , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto/fisiologia , Enfisema/etiologia , Volume Expiratório Forçado , Humanos , Pulmão/fisiopatologia , Masculino , Camundongos , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/fisiologia , Fumar/efeitos adversos
7.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L1038-L1056, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33822645

RESUMO

Airway oscillometry has become the de facto standard for quality assessment of lung physiology in laboratory animals and has demonstrated its usefulness in understanding diseases of small airways. Nowadays, it is seeing extensive use in daily clinical practice and research; however, a question that remains unanswered is how well physiological findings in animals and humans correlate? Methodological and device differences are obvious between animal and human studies. However, all devices deliver an oscillated airflow test signal and output respiratory impedance. In addition, despite analysis differences, there are ways to interpret animal and human oscillometry data to allow suitable comparisons. The potential with oscillometry is its ability to reveal universal features of the respiratory system across species, making translational extrapolation likely to be predictive. This means that oscillometry can thus help determine if an animal model displays the same physiological characteristics as the human disease. Perhaps more importantly, it can also be useful to determine whether an intervention is effective as well as to understand if it affects the desired region of the respiratory system, e.g., the periphery of the lung. Finally, findings in humans can also inform preclinical scientists and give indications as to what type of physiological changes should be observed in animal models to make them relevant as models of human disease. The present article will attempt to demonstrate the potential of oscillometry in respiratory research, an area where the development of novel therapies is plagued with a failure rate higher than in other disease areas.


Assuntos
Asma/tratamento farmacológico , Asma/fisiopatologia , Impedância Elétrica/uso terapêutico , Pulmão/fisiopatologia , Oscilometria , Animais , Humanos , Pulmão/efeitos dos fármacos , Pulmão/fisiologia , Oscilometria/métodos , Testes de Função Respiratória/métodos , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos
8.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L204-L212, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34009049

RESUMO

The quantification of airway compliance (Caw) is essential to the study of airway alterations in disease models. However, the required measurements of airway pressure and volume are difficult to acquire in mice. We hypothesized that the inflation limb of full-range pressure-volume (PV) curves could be used to quantify Caw, as it contains a segment where only the airway tree is distended. The study objective was to assess the feasibility of the approach by analysis of full-range PV curves previously collected in three mouse models: an elastase model of emphysema, a genetic model spontaneously developing emphysema (leukotriene C4 synthase knockout; LTC4S-KO), and a bleomycin model of lung fibrosis. Attempts to validate results included Caw change relative to respiratory system compliance (ΔCaw/ΔC), the minute work of breathing (mWOB), and the elastance at 20.5 Hz (Ers_20.5) from prior respiratory mechanics measurements in the same subjects. Caw was estimated at 3% of total compliance in healthy mice or 2.3 ± 1 µL/cmH2O (n = 17). The technique detected changes in models of respiratory obstructive and restrictive diseases relative to control mice as well as differences in the two emphysema models studied. The changes in Caw were consistent with those seen in ΔCaw/ΔC, mWOB, or Ers_20.5, with some variations according to the model, as well as with results reported in the literature in humans and mice. Direct Caw measurements in subjects as small as mice could prove useful to further characterize other respiratory disease models associated with airway remodeling or to assess treatment effects.


Assuntos
Resistência das Vias Respiratórias , Bleomicina/toxicidade , Enfisema Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia , Transtornos Respiratórios/complicações , Animais , Antibióticos Antineoplásicos/toxicidade , Feminino , Complacência Pulmonar , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Enfisema Pulmonar/etiologia , Fibrose Pulmonar/induzido quimicamente , Mecânica Respiratória
9.
Respir Res ; 18(1): 123, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28629359

RESUMO

BACKGROUND: Pulmonary function measurements are important when studying respiratory disease models. Both resistance and compliance have been used to assess lung function in mice. Yet, it is not always clear how these parameters relate to forced expiration (FE)-related parameters, most commonly used in humans. We aimed to characterize FE measurements in four well-established mouse models of lung diseases. METHOD: Detailed respiratory mechanics and FE measurements were assessed concurrently in Balb/c mice, using the forced oscillation and negative pressure-driven forced expiration techniques, respectively. Measurements were performed at baseline and following increasing methacholine challenges in control Balb/c mice as well as in four disease models: bleomycin-induced fibrosis, elastase-induced emphysema, LPS-induced acute lung injury and house dust mite-induced asthma. RESULTS: Respiratory mechanics parameters (airway resistance, tissue damping and tissue elastance) confirmed disease-specific phenotypes either at baseline or following methacholine challenge. Similarly, lung function defects could be detected in each disease model by at least one FE-related parameter (FEV0.1, FEF0.1, FVC, FEV0.1/FVC ratio and PEF) at baseline or during the methacholine provocation assay. CONCLUSIONS: FE-derived outcomes in four mouse disease models behaved similarly to changes found in human spirometry. Routine combined lung function assessments could increase the translational utility of mouse models.


Assuntos
Resistência das Vias Respiratórias/fisiologia , Testes de Provocação Brônquica/métodos , Modelos Animais de Doenças , Pneumopatias/patologia , Pneumopatias/fisiopatologia , Animais , Enfisema/patologia , Enfisema/fisiopatologia , Volume Expiratório Forçado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fibrose Pulmonar/patologia , Fibrose Pulmonar/fisiopatologia
10.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L837-46, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25637610

RESUMO

Airway hyperresponsiveness often constitutes a primary outcome in respiratory studies in mice. The procedure commonly employs aerosolized challenges, and results are typically reported in terms of bronchoconstrictor concentrations loaded into the nebulizer. Yet, because protocols frequently differ across studies, especially in terms of aerosol generation and delivery, direct study comparisons are difficult. We hypothesized that protocol variations could lead to differences in aerosol delivery efficiency and, consequently, in the dose delivered to the subject, as well as in the response. Thirteen nebulization patterns containing common protocol variations (nebulization time, duty cycle, particle size spectrum, air humidity, and/or ventilation profile) and using increasing concentrations of methacholine and broadband forced oscillations (flexiVent, SCIREQ, Montreal, Qc, Canada) were created, characterized, and studied in anesthetized naïve A/J mice. A delivered dose estimate calculated from nebulizer-, ventilator-, and subject-specific characteristics was introduced and used to account for protocol variations. Results showed that nebulization protocol variations significantly affected the fraction of aerosol reaching the subject site and the delivered dose, as well as methacholine reactivity and sensitivity in mice. From the protocol variants studied, addition of a slow deep ventilation profile during nebulization was identified as a key factor for optimization of the technique. The study also highlighted sensitivity differences within the lung, as well as the possibility that airway responses could be selectively enhanced by adequate control of nebulizer and ventilator settings. Reporting results in terms of delivered doses represents an important standardizing element for assessment of airway hyperresponsiveness in mice.


Assuntos
Cloreto de Metacolina/toxicidade , Hipersensibilidade Respiratória/induzido quimicamente , Administração por Inalação , Aerossóis , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Nebulizadores e Vaporizadores/normas , Padrões de Referência , Projetos de Pesquisa
11.
Exp Lung Res ; 41(2): 84-92, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25398012

RESUMO

The sole commercial system currently employing the forced oscillation technique (FOT) in small laboratory animals (flexiVent; SCIREQ Inc., Canada) was recently redesigned along with its operating software. Yet, many users still work with the legacy version or a mixed configuration. This study aimed to compare result accuracy and precision between three flexiVent system configurations and to quantify the impact of configuration changes on measured parameters. Physiologically relevant resistance or elastance were measured at 2.5 Hz on the following three system configurations using characterized mechanical test loads: (i) legacy flexiVent-flexiVent v5.3.4 (Leg-fV5), (ii) legacy flexiVent-flexiWare v7.2.1 (Leg-fW7), and (iii) flexiVent FX-flexiWare v7.2.1 (FX-fW7). Results demonstrated measurements of high precision that were consistent between system configurations. There was no statistical difference between system configurations in terms of measuring a predicted resistance. Measurements of elastance, on the other hand, were configuration-sensitive with FX-fW7 generating values that were closer to theoretical ones than the other two configurations. The largest impact on measurement outcomes was associated with the most noteworthy configuration change (i.e., software and hardware replacement). This effect was however constrained, with variations in the order of 3-5%, approximately. In conclusion, the latest version of the sole commercial pre-clinical FOT system currently available generated results that were equivalent or better than those acquired with two other system configurations. Given that configuration changes were associated with subtle parameter differences, best practice would recommend consistency within a study and reporting the full details of the system used.


Assuntos
Ventilação de Alta Frequência/métodos , Testes de Função Respiratória/métodos , Resistência das Vias Respiratórias/fisiologia , Animais , Pulmão/fisiologia , Camundongos , Oscilometria/métodos , Mecânica Respiratória/fisiologia , Software
12.
Front Physiol ; 15: 1345488, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38444763

RESUMO

Acute respiratory distress syndrome (ARDS) is characterized by an exacerbated inflammatory response, severe damage to the alveolar-capillary barrier and a secondary infiltration of protein-rich fluid into the airspaces, ultimately leading to respiratory failure. Resolution of ARDS depends on the ability of the alveolar epithelium to reabsorb lung fluid through active transepithelial ion transport, to control the inflammatory response, and to restore a cohesive and functional epithelium through effective repair processes. Interestingly, several lines of evidence have demonstrated the important role of potassium (K+) channels in the regulation of epithelial repair processes. Furthermore, these channels have previously been shown to be involved in sodium/fluid absorption across alveolar epithelial cells, and we have recently demonstrated the contribution of KvLQT1 channels to the resolution of thiourea-induced pulmonary edema in vivo. The aim of our study was to investigate the role of the KCNQ1 pore-forming subunit of KvLQT1 channels in the outcome of ARDS parameters in a model of acute lung injury (ALI). We used a molecular approach with KvLQT1-KO mice challenged with bleomycin, a well-established ALI model that mimics the key features of the exudative phase of ARDS on day 7. Our data showed that KvLQT1 deletion exacerbated the negative outcome of bleomycin on lung function (resistance, elastance and compliance). An alteration in the profile of infiltrating immune cells was also observed in KvLQT1-KO mice while histological analysis showed less interstitial and/or alveolar inflammatory response induced by bleomycin in KvLQT1-KO mice. Finally, a reduced repair rate of KvLQT1-KO alveolar cells after injury was observed. This work highlights the complex contribution of KvLQT1 in the development and resolution of ARDS parameters in a model of ALI.

13.
PLoS One ; 18(2): e0281452, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36757935

RESUMO

The advent of micro-computed tomography (microCT) has provided significant advancement in our ability to generate clinically relevant assessments of lung health and disease in small animal models. As microCT use to generate outcomes analysis in pulmonary preclinical models has increased there have been substantial improvements in image quality and resolution, and data analysis software. However, there are limited published methods for standardized imaging and automated analysis available for investigators. Manual quantitative analysis of microCT images is complicated by the presence of inflammation and parenchymal disease. To improve the efficiency and limit user-associated bias, we have developed an automated pulmonary air and tissue segmentation (PATS) task list to segment lung air volume and lung tissue volume for quantitative analysis. We demonstrate the effective use of the PATS task list using four distinct methods for imaging, 1) in vivo respiration controlled scanning using a flexiVent, 2) longitudinal breath-gated in vivo scanning in resolving and non-resolving pulmonary disease initiated by lipopolysaccharide-, bleomycin-, and silica-exposure, 3) post-mortem imaging, and 4) ex vivo high-resolution scanning. The accuracy of the PATS task list was compared to manual segmentation. The use of these imaging techniques and automated quantification methodology across multiple models of lung injury and fibrosis demonstrates the broad applicability and adaptability of microCT to various lung diseases and small animal models and presents a significant advance in efficiency and standardization of preclinical microCT imaging and analysis for the field of pulmonary research.


Assuntos
Pneumopatias , Camundongos , Animais , Microtomografia por Raio-X/métodos , Pneumopatias/diagnóstico por imagem , Pneumopatias/patologia , Pulmão/diagnóstico por imagem , Pulmão/patologia , Modelos Animais de Doenças , Fibrose
14.
Am J Respir Crit Care Med ; 183(7): 865-75, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20971830

RESUMO

RATIONALE: IL-9 is a pleiotropic cytokine that has multiple effects on structural as well as numerous hematopoietic cells, which are central to the pathogenesis of asthma. OBJECTIVES: The contribution of IL-9 to asthma pathogenesis has thus far been unclear, due to conflicting reports in the literature. These earlier studies focused on the role of IL-9 in acute inflammatory models; here we have investigated the effects of IL-9 blockade during chronic allergic inflammation. METHODS: Mice were exposed to either prolonged ovalbumin or house dust mite allergen challenge to induce chronic inflammation and airway remodeling. MEASUREMENTS AND MAIN RESULTS: We found that IL-9 governs allergen-induced mast cell (MC) numbers in the lung and has pronounced effects on chronic allergic inflammation. Anti-IL-9 antibody-treated mice were protected from airway remodeling with a concomitant reduction in mature MC numbers and activation, in addition to decreased expression of the profibrotic mediators transforming growth factor-ß1, vascular endothelial growth factor, and fibroblast growth factor-2 in the lung. Airway remodeling was associated with impaired lung function in the peripheral airways and this was reversed by IL-9 neutralization. In human asthmatic lung tissue, we identified MCs as the main IL-9 receptor expressing population and found them to be sources of vascular endothelial growth factor and fibroblast growth factor-2. CONCLUSIONS: Our data suggest an important role for an IL-9-MC axis in the pathology associated with chronic asthma and demonstrate that an impact on this axis could lead to a reduction in chronic inflammation and improved lung function in patients with asthma.


Assuntos
Alérgenos/imunologia , Asma/imunologia , Líquido da Lavagem Broncoalveolar/citologia , Interleucina-9/imunologia , Pulmão/imunologia , Pulmão/patologia , Mastócitos/imunologia , Alérgenos/administração & dosagem , Análise de Variância , Animais , Asma/metabolismo , Biomarcadores/metabolismo , Biópsia por Agulha , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/farmacologia , RNA Mensageiro/análise , Distribuição Aleatória , Testes de Função Respiratória , Estatísticas não Paramétricas
15.
Front Physiol ; 13: 1069466, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699692

RESUMO

Alveolar ion and fluid absorption is essential for lung homeostasis in healthy conditions as well as for the resorption of lung edema, a key feature of acute respiratory distress syndrome. Liquid absorption is driven by active transepithelial sodium transport, through apical ENaC Na+ channels and basolateral Na+/K+-ATPase. Our previous work unveiled that KvLQT1 K+ channels also participate in the control of Na+/liquid absorption in alveolar epithelial cells. Our aim was to further investigate the function of KvLQT1 channels and their interplay with other channels/transporters involved in ion/liquid transport in vivo using adult wild-type (WT) and KvLQT1 knock-out (KO) mice under physiological conditions and after thiourea-induced lung edema. A slight but significant increase in water lung content (WLC) was observed in naïve KvLQT1-KO mice, relative to WT littermates, whereas lung function was generally preserved and histological structure unaltered. Following thiourea-induced lung edema, KvLQT1-KO did not worsen WLC or lung function. Similarly, lung edema was not aggravated by the administration of a KvLQT1 inhibitor (chromanol). However, KvLQT1 activation (R-L3) significantly reduced WLC in thiourea-challenged WT mice. The benefits of R-L3 were prevented in KO or chromanol-treated WT mice. Furthermore, R-L3 treatment had no effect on thiourea-induced endothelial barrier alteration but restored or enhanced the levels of epithelial alveolar AQP5, Na+/K+-ATPase, and ENaC expressions. Altogether, the results indicate the benefits of KvLQT1 activation in the resolution of lung edema, probably through the observed up-regulation of epithelial alveolar channels/transporters involved in ion/water transport.

16.
Sci Rep ; 11(1): 7777, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33833346

RESUMO

Due to frequent and often severe lung affections caused by COVID-19, murine models of acute respiratory distress syndrome (ARDS) are increasingly used in experimental lung research. The one induced by a single lipopolysaccharide (LPS) exposure is practical. However, whether it is preferable to administer LPS intranasally or intratracheally remains an open question. Herein, female C57Bl/6 J mice were exposed intranasally or intratracheally to one dose of either saline or 3 mg/kg of LPS. They were studied 24 h later. The groups treated with LPS, either intranasally or intratracheally, exhibited a pronounced neutrophilic inflammation, signs of lung tissue damage and protein extravasation into the alveoli, and mild lung dysfunction. The magnitude of the response was generally not different between groups exposed intranasally versus intratracheally. However, the variability of some the responses was smaller in the LPS-treated groups exposed intranasally versus intratracheally. Notably, the saline-treated mice exposed intratracheally demonstrated a mild neutrophilic inflammation and alterations of the airway epithelium. We conclude that an intranasal exposure is as effective as an intratracheal exposure in a murine model of ARDS induced by LPS. Additionally, the groups exposed intranasally demonstrated less variability in the responses to LPS and less complications associated with the sham procedure.


Assuntos
Inflamação/induzido quimicamente , Lipopolissacarídeos/efeitos adversos , Pulmão/patologia , Síndrome do Desconforto Respiratório/induzido quimicamente , Administração Intranasal , Animais , Modelos Animais de Doenças , Feminino , Inflamação/patologia , Lipopolissacarídeos/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Proteínas/análise , Síndrome do Desconforto Respiratório/patologia
17.
Respir Res ; 11: 82, 2010 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-20565957

RESUMO

BACKGROUND: Pulmonary function has been reported in mice using negative pressure-driven forced expiratory manoeuvres (NPFE) and the forced oscillation technique (FOT). However, both techniques have always been studied using separate cohorts of animals or systems. The objective of this study was to obtain NPFE and FOT measurements at baseline and following bronchoconstriction from a single cohort of mice using a combined system in order to assess both techniques through a refined approach. METHODS: Groups of allergen- or sham-challenged ovalbumin-sensitized mice that were either vehicle (saline) or drug (dexamethasone 1 mg/kg ip)-treated were studied. Surgically prepared animals were connected to an extended flexiVent system (SCIREQ Inc., Montreal, Canada) permitting NPFE and FOT measurements. Lung function was assessed concomitantly by both techniques at baseline and following doubling concentrations of aerosolized methacholine (MCh; 31.25 - 250 mg/ml). The effect of the NPFE manoeuvre on respiratory mechanics was also studied. RESULTS: The expected exaggerated MCh airway response of allergic mice and its inhibition by dexamethasone were detected by both techniques. We observed significant changes in FOT parameters at either the highest (Ers, H) or the two highest (Rrs, RN, G) MCh concentrations. The flow-volume (F-V) curves obtained following NPFE manoeuvres demonstrated similar MCh concentration-dependent changes. A dexamethasone-sensitive decrease in the area under the flow-volume curve at the highest MCh concentration was observed in the allergic mice. Two of the four NPFE parameters calculated from the F-V curves, FEV0.1 and FEF50, also captured the expected changes but only at the highest MCh concentration. Normalization to baseline improved the sensitivity of NPFE parameters at detecting the exaggerated MCh airway response of allergic mice but had minimal impact on FOT responses. Finally, the combination with FOT allowed us to demonstrate that NPFE induced persistent airway closure that was reversible by deep lung inflation. CONCLUSIONS: We conclude that FOT and NPFE can be concurrently assessed in the same cohort of animals to determine airway mechanics and expiratory flow limitation during methacholine responses, and that the combination of the two techniques offers a refined control and an improved reproducibility of the NPFE.


Assuntos
Hiper-Reatividade Brônquica/diagnóstico , Broncoconstrição , Pulmão/fisiopatologia , Testes de Função Respiratória , Corticosteroides/farmacologia , Animais , Hiper-Reatividade Brônquica/imunologia , Hiper-Reatividade Brônquica/fisiopatologia , Hiper-Reatividade Brônquica/prevenção & controle , Testes de Provocação Brônquica , Broncoconstrição/efeitos dos fármacos , Broncoconstritores , Dexametasona/farmacologia , Modelos Animais de Doenças , Feminino , Fluxo Expiratório Forçado , Volume Expiratório Forçado , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Cloreto de Metacolina , Camundongos , Camundongos Endogâmicos BALB C , Oscilometria , Ovalbumina , Mecânica Respiratória , Fatores de Tempo , Capacidade Vital
18.
bioRxiv ; 2020 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-32676600

RESUMO

Severe Acute Respiratory Syndrome Coronavirus -2 (SARS-CoV-2) emerged in late 2019 and has spread worldwide resulting in the Coronavirus Disease 2019 (COVID-19) pandemic. Although animal models have been evaluated for SARS-CoV-2 infection, none have recapitulated the severe lung disease phenotypes seen in hospitalized human cases. Here, we evaluate heterozygous transgenic mice expressing the human ACE2 receptor driven by the epithelial cell cytokeratin-18 gene promoter (K18-hACE2) as a model of SARS-CoV-2 infection. Intranasal inoculation of SARS-CoV-2 in K18-hACE2 mice results in high levels of viral infection in lung tissues with additional spread to other organs. Remarkably, a decline in pulmonary function, as measured by static and dynamic tests of respiratory capacity, occurs 4 days after peak viral titer and correlates with an inflammatory response marked by infiltration into the lung of monocytes, neutrophils, and activated T cells resulting in pneumonia. Cytokine profiling and RNA sequencing analysis of SARS-CoV-2-infected lung tissues show a massively upregulated innate immune response with prominent signatures of NF-kB-dependent, type I and II interferon signaling, and leukocyte activation pathways. Thus, the K18-hACE2 model of SARS-CoV-2 infection recapitulates many features of severe COVID-19 infection in humans and can be used to define the mechanistic basis of lung disease and test immune and antiviral-based countermeasures.

19.
Nat Commun ; 11(1): 5640, 2020 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-33159078

RESUMO

Lymphangioleiomyomatosis (LAM) is a rare fatal cystic lung disease due to bi-allelic inactivating mutations in tuberous sclerosis complex (TSC1/TSC2) genes coding for suppressors of the mechanistic target of rapamycin complex 1 (mTORC1). The origin of LAM cells is still unknown. Here, we profile a LAM lung compared to an age- and sex-matched healthy control lung as a hypothesis-generating approach to identify cell subtypes that are specific to LAM. Our single-cell RNA sequencing (scRNA-seq) analysis reveals novel mesenchymal and transitional alveolar epithelial states unique to LAM lung. This analysis identifies a mesenchymal cell hub coordinating the LAM disease phenotype. Mesenchymal-restricted deletion of Tsc2 in the mouse lung produces a mTORC1-driven pulmonary phenotype, with a progressive disruption of alveolar structure, a decline in pulmonary function, increase of rapamycin-sensitive expression of WNT ligands, and profound female-specific changes in mesenchymal and epithelial lung cell gene expression. Genetic inactivation of WNT signaling reverses age-dependent changes of mTORC1-driven lung phenotype, but WNT activation alone in lung mesenchyme is not sufficient for the development of mouse LAM-like phenotype. The alterations in gene expression are driven by distinctive crosstalk between mesenchymal and epithelial subsets of cells observed in mesenchymal Tsc2-deficient lungs. This study identifies sex- and age-specific gene changes in the mTORC1-activated lung mesenchyme and establishes the importance of the WNT signaling pathway in the mTORC1-driven lung phenotype.


Assuntos
Pulmão/metabolismo , Linfangioleiomiomatose/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Mesoderma/metabolismo , Fatores Etários , Idoso , Animais , Feminino , Humanos , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Linfangioleiomiomatose/tratamento farmacológico , Linfangioleiomiomatose/genética , Linfangioleiomiomatose/fisiopatologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Mesoderma/efeitos dos fármacos , Camundongos , Fatores Sexuais , Sirolimo/administração & dosagem , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo , Via de Sinalização Wnt
20.
Am J Respir Cell Mol Biol ; 39(1): 26-35, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18258919

RESUMO

Interleukin 13 (IL-13) is considered to be a key driver of the development of airway allergic inflammation and remodeling leading to airway hyperresponsiveness (AHR). How precisely IL-13 leads to the development of airway inflammation, AHR, and mucus production is not fully understood. In order to identify key mediators downstream of IL-13, we administered adenovirus IL-13 to specifically induce IL-13-dependent inflammation in the lungs of mice. This approach was shown to induce cardinal features of lung disease, specifically airway inflammation, elevated cytokines, AHR, and mucus secretion. Notably, the model is resistant to corticosteroid treatment and is characterized by marked neutrophilia, two hallmarks of more severe forms of asthma. To identify IL-13-dependent mediators, we performed a limited-scale two-dimensional SDS-PAGE proteomic analysis and identified proteins significantly modulated in this model. Intriguingly, several identified proteins were unique to this model, whereas others correlated with those modulated in a mouse ovalbumin-induced pulmonary inflammation model. We corroborated this approach by illustrating that proteomic analysis can identify known pathways/mediators downstream of IL-13. Thus, we have characterized a murine adenovirus IL-13 lung model that recapitulates specific disease traits observed in human asthma, and have exploited this model to identify effectors downstream of IL-13. Collectively, these findings will enable a broader appreciation of IL-13 and its impact on disease pathways in the lung.


Assuntos
Infecções por Adenoviridae/fisiopatologia , Adenoviridae , Obstrução das Vias Respiratórias/induzido quimicamente , Interleucina-13/efeitos adversos , Adenoviridae/genética , Animais , Técnicas de Cultura de Células , Divisão Celular , Modelos Animais de Doenças , Interleucina-13/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Muco/metabolismo , Ovalbumina/efeitos adversos , Testes de Função Respiratória , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA