Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 6(1)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33232302

RESUMO

Existing animal models of cystic fibrosis (CF) have provided key insights into CF pathogenesis but have been limited by short lifespans, absence of key phenotypes, and/or high maintenance costs. Here, we report the CRISPR/Cas9-mediated generation of CF rabbits, a model with a relatively long lifespan and affordable maintenance and care costs. CF rabbits supplemented solely with oral osmotic laxative had a median survival of approximately 40 days and died of gastrointestinal disease, but therapeutic regimens directed toward restoring gastrointestinal transit extended median survival to approximately 80 days. Surrogate markers of exocrine pancreas disorders were found in CF rabbits with declining health. CFTR expression patterns in WT rabbit airways mimicked humans, with widespread distribution in nasal respiratory and olfactory epithelia, as well as proximal and distal lower airways. CF rabbits exhibited human CF-like abnormalities in the bioelectric properties of the nasal and tracheal epithelia. No spontaneous respiratory disease was detected in young CF rabbits. However, abnormal phenotypes were observed in surviving 1-year-old CF rabbits as compared with WT littermates, and these were especially evident in the nasal respiratory and olfactory epithelium. The CF rabbit model may serve as a useful tool for understanding gut and lung CF pathogenesis and for the practical development of CF therapeutics.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Animais , Sistemas CRISPR-Cas , Fibrose Cística/patologia , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Feminino , Trato Gastrointestinal/patologia , Trato Gastrointestinal/fisiopatologia , Técnicas de Inativação de Genes , Humanos , Masculino , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Coelhos , Sistema Respiratório/patologia , Sistema Respiratório/fisiopatologia , Distribuição Tecidual , Transcriptoma
2.
Am J Respir Cell Mol Biol ; 59(3): 383-396, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29579396

RESUMO

Understanding how expression of airway secretory mucins MUC5B and MUC5AC is regulated in health and disease is important to elucidating the pathogenesis of mucoobstructive respiratory diseases. The transcription factor SPDEF (sterile α-motif pointed domain epithelial specific transcription factor) is a key regulator of MUC5AC, but its role in regulating MUC5B in health and in mucoobstructive lung diseases is unknown. Characterization of Spdef-deficient mice upper and lower airways demonstrated region-specific, Spdef-dependent regulation of basal Muc5b expression. Neonatal Spdef-deficient mice exhibited reductions in BAL Muc5ac and Muc5b. Adult Spdef-deficient mice partially phenocopied Muc5b-deficient mice as they exhibited reduced Muc5b in nasopharyngeal and airway epithelia but not in olfactory Bowman glands, 75% incidence of nasopharyngeal hair/mucus plugs, and mild bacterial otitis media, without defective mucociliary clearance in the nasopharynx. In contrast, tracheal mucociliary clearance was reduced in Spdef-deficient mice in the absence of lung disease. To evaluate the role of Spdef in the development and persistence of Muc5b-predominant mucoobstructive lung disease, Spdef-deficient mice were crossed with Scnn1b-transgenic (Scnn1b-Tg) mice, which exhibit airway surface dehydration-induced airway mucus obstruction and inflammation. Spdef-deficient Scnn1b-Tg mice exhibited reduced Muc5ac, but not Muc5b, expression and BAL content. Airway mucus obstruction was not decreased in Spdef-deficient Scnn1b-Tg mice, consistent with Muc5b-dominant Scnn1b disease, but increased airway neutrophilia was observed compared with Spdef-sufficient Scnn1b-Tg mice. Collectively, these results indicate that Spdef regulates baseline Muc5b expression in respiratory epithelia but does not contribute to Muc5b regulation in a mouse model of Muc5b-predominant mucus obstruction caused by airway dehydration.


Assuntos
Pneumopatias/metabolismo , Mucina-5B/metabolismo , Depuração Mucociliar/fisiologia , Proteínas Proto-Oncogênicas c-ets/genética , Animais , Canais Epiteliais de Sódio/genética , Pneumopatias/genética , Camundongos Transgênicos , Mucina-5B/genética
3.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L318-L331, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29074490

RESUMO

The epithelial Na+ channel (ENaC) regulates airway surface hydration. In mouse airways, ENaC is composed of three subunits, α, ß, and γ, which are differentially expressed (α > ß > γ). Airway-targeted overexpression of the ß subunit results in Na+ hyperabsorption, causing airway surface dehydration, hyperconcentrated mucus with delayed clearance, lung inflammation, and perinatal mortality. Notably, mice overexpressing the α- or γ-subunit do not exhibit airway Na+ hyperabsorption or lung pathology. To test whether overexpression of multiple ENaC subunits produced Na+ transport and disease severity exceeding that of ßENaC-Tg mice, we generated double (αß, αγ, ßγ) and triple (αßγ) transgenic mice and characterized their lung phenotypes. Double αγENaC-Tg mice were indistinguishable from WT littermates. In contrast, double ßγENaC-Tg mice exhibited airway Na+ absorption greater than that of ßENaC-Tg mice, which was paralleled by worse survival, decreased mucociliary clearance, and more severe lung pathology. Double αßENaC-Tg mice exhibited Na+ transport rates comparable to those of ßENaC-Tg littermates. However, αßENaC-Tg mice had poorer survival and developed severe parenchymal consolidation. In situ hybridization (RNAscope) analysis revealed both alveolar and airway αENaC-Tg overexpression. Triple αßγENaC-Tg mice were born in Mendelian proportions but died within the first day of life, and the small sample size prevented analyses of cause(s) of death. Cumulatively, these results indicate that overexpression of ßENaC is rate limiting for generation of pathological airway surface dehydration. Notably, airway co-overexpression of ß- and γENaC had additive effects on Na+ transport and disease severity, suggesting dose dependency of these two variables.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Pneumopatias/patologia , Pneumonia/patologia , Mucosa Respiratória/patologia , Animais , Canais Epiteliais de Sódio/genética , Pneumopatias/etiologia , Pneumopatias/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fenótipo , Pneumonia/etiologia , Pneumonia/metabolismo , Mucosa Respiratória/metabolismo , Transdução de Sinais
4.
Mucosal Immunol ; 10(2): 395-407, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27435107

RESUMO

Airway diseases, including cigarette smoke-induced chronic bronchitis, cystic fibrosis, and primary ciliary dyskinesia are associated with decreased mucociliary clearance (MCC). However, it is not known whether a simple reduction in MCC or concentration-dependent mucus adhesion to airway surfaces dominates disease pathogenesis or whether decreasing the concentration of secreted mucins may be therapeutic. To address these questions, Scnn1b-Tg mice, which exhibit airway mucus dehydration/adhesion, were compared and crossed with Muc5b- and Muc5ac-deficient mice. Absence of Muc5b caused a 90% reduction in MCC, whereas Scnn1b-Tg mice exhibited an ∼50% reduction. However, the degree of MCC reduction did not correlate with bronchitic airway pathology, which was observed only in Scnn1b-Tg mice. Ablation of Muc5b significantly reduced the extent of mucus plugging in Scnn1b-Tg mice. However, complete absence of Muc5b in Scnn1b-Tg mice was associated with increased airway inflammation, suggesting that Muc5b is required to maintain immune homeostasis. Loss of Muc5ac had few phenotypic consequences in Scnn1b-Tg mice. These data suggest that: (i) mucus hyperconcentration dominates over MCC reduction alone to produce bronchitic airway pathology; (ii) Muc5b is the dominant contributor to the Scnn1b-Tg phenotype; and (iii) therapies that limit mucin secretion may reduce plugging, but complete Muc5b removal from airway surfaces may be detrimental.


Assuntos
Brônquios/fisiologia , Bronquite Crônica/imunologia , Fibrose Cística/imunologia , Síndrome de Kartagener/imunologia , Mucina-5AC/metabolismo , Mucina-5B/metabolismo , Depuração Mucociliar , Obstrução das Vias Respiratórias/genética , Animais , Brônquios/patologia , Canais Epiteliais de Sódio/genética , Homeostase , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mucina-5AC/genética , Mucina-5B/genética , Depuração Mucociliar/genética , Fumar/efeitos adversos
5.
Am J Respir Cell Mol Biol ; 54(2): 210-21, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26121027

RESUMO

Resident immune cells (e.g., macrophages [MΦs]) and airway mucus clearance both contribute to a healthy lung environment. To investigate interactions between pulmonary MΦ function and defective mucus clearance, a genetic model of lysozyme M (LysM) promoter-mediated MΦ depletion was generated, characterized, and crossed with the sodium channel ß subunit transgenic (Scnn1b-Tg) mouse model of defective mucus clearance. Diphtheria toxin A-mediated depletion of LysM(+) pulmonary MΦs in wild-type mice with normal mucus clearance resulted in lethal pneumonia in 24% of neonates. The pneumonias were dominated by Pasteurella pneumotropica and accompanied by emaciation, neutrophilic inflammation, and elevated Th1 cytokines. The incidence of emaciation and pneumonia reached 51% when LysM(+) MΦ depletion was superimposed on the airway mucus clearance defect of Scnn1b-Tg mice. In LysM(+) MΦ-depleted Scnn1b-Tg mice, pneumonias were associated with a broader spectrum of bacterial species and a significant reduction in airway mucus plugging. Bacterial burden (CFUs) was comparable between Scnn1b-Tg and nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice. However, the nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice exhibited increased airway inflammation, the presence of neutrophilic infiltration, and increased levels of inflammatory cytokines in bronchoalveolar lavage fluid compared with Scnn1b-Tg mice. Collectively, these data identify key MΦ-mucus clearance interactions with respect to both infectious and inflammatory components of muco-obstructive lung disease.


Assuntos
Pulmão/imunologia , Macrófagos/imunologia , Depuração Mucociliar , Infecções por Pasteurella/imunologia , Pasteurella pneumotropica/imunologia , Pneumonia Bacteriana/imunologia , Animais , Animais Recém-Nascidos , Citocinas/imunologia , Citocinas/metabolismo , Toxina Diftérica/genética , Toxina Diftérica/metabolismo , Modelos Animais de Doenças , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Predisposição Genética para Doença , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Pulmão/metabolismo , Pulmão/microbiologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Muramidase/genética , Infecções por Pasteurella/genética , Infecções por Pasteurella/metabolismo , Infecções por Pasteurella/microbiologia , Pasteurella pneumotropica/patogenicidade , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fenótipo , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/microbiologia , Regiões Promotoras Genéticas
6.
Am J Physiol Lung Cell Mol Physiol ; 304(7): L469-80, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23377346

RESUMO

Airway surface hydration depends on the balance between transepithelial Na(+) absorption and Cl(-) secretion. In adult mice, absence of functional cystic fibrosis transmembrane conductance regulator (Cftr) fails to recapitulate human cystic fibrosis (CF) lung disease. In contrast, overexpression of the epithelial Na(+) channel ß subunit in transgenic mice (ßENaC-Tg) produces unregulated Na(+) hyperabsorption and results in CF-like airway surface dehydration, mucus obstruction, inflammation, and increased neonatal mortality. To investigate whether the combination of airway Na(+) hyperabsorption and absent Cftr-mediated Cl(-) secretion resulted in more severe lung pathology, we generated double-mutant ΔF508 CF/ßENaC-Tg mice. Survival of ΔF508 CF/ßENaC-Tg mice was reduced compared with ßENaC-Tg or ΔF508 CF mice. Absence of functional Cftr did not affect endogenous or transgenic ENaC currents but produced reduced basal components of Cl(-) secretion and tracheal cartilaginous defects in both ΔF508 CF and ΔF508 CF/ßENaC-Tg mice. Neonatal ΔF508 CF/ßENaC-Tg mice exhibited higher neutrophilic pulmonary inflammation and club cell (Clara cell) necrosis compared with ßENaC-Tg littermates. Neonatal ΔF508 CF/ßENaC-Tg mice also exhibited spontaneous bacterial infections, but the bacterial burden was similar to that of ßENaC-Tg littermates. Adult ΔF508 CF/ßENaC-Tg mice exhibited pathological changes associated with eosinophilic crystalline pneumonia, a phenotype not observed in age-matched ßENaC-Tg mice. Collectively, these data suggest that the combined abnormalities in Na(+) absorption and Cl(-) secretion produce more severe lung disease than either defect alone. Airway cartilage abnormalities, airway cell necrosis, and exaggerated neutrophil infiltration likely interact with defective mucus clearance caused by ßENaC overexpression and absent CFTR-mediated Cl(-) secretion to produce the increased neonatal mortality observed in ΔF508 CF/ßENaC-Tg mice.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Canais Epiteliais de Sódio/metabolismo , Pulmão/metabolismo , Eosinofilia Pulmonar/metabolismo , Sódio/metabolismo , Absorção/genética , Animais , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Canais Epiteliais de Sódio/genética , Humanos , Transporte de Íons/genética , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Necrose , Infiltração de Neutrófilos/genética , Neutrófilos/metabolismo , Neutrófilos/patologia , Pneumonia/genética , Pneumonia/metabolismo , Pneumonia/patologia , Eosinofilia Pulmonar/genética , Eosinofilia Pulmonar/patologia
7.
Physiol Genomics ; 44(8): 470-84, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22395316

RESUMO

Mucus clearance is an important airway innate defense mechanism. Airway-targeted overexpression of the epithelial Na(+) channel ß-subunit [encoded by sodium channel nonvoltage gated 1, beta subunit (Scnn1b)] in mice [Scnn1b-transgenic (Tg) mice] increases transepithelial Na(+) absorption and dehydrates the airway surface, which produces key features of human obstructive lung diseases, including mucus obstruction, inflammation, and air-space enlargement. Because the first Scnn1b-Tg mice were generated on a mixed background, the impact of genetic background on disease phenotype in Scnn1b-Tg mice is unknown. To explore this issue, congenic Scnn1b-Tg mice strains were generated on C57BL/6N, C3H/HeN, BALB/cJ, and FVB/NJ backgrounds. All strains exhibited a two- to threefold increase in tracheal epithelial Na(+) absorption, and all developed airway mucus obstruction, inflammation, and air-space enlargement. However, there were striking differences in neonatal survival, ranging from 5 to 80% (FVB/NJ

Assuntos
Obstrução das Vias Respiratórias/genética , Pneumopatias Obstrutivas/genética , Muco/metabolismo , Obstrução das Vias Respiratórias/metabolismo , Animais , Modelos Animais de Doenças , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos
8.
J Immunol ; 182(7): 4357-67, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19299736

RESUMO

Overexpression of the epithelial Na(+) channel beta subunit (Scnn1b gene, betaENaC protein) in transgenic (Tg) mouse airways dehydrates mucosal surfaces, producing mucus obstruction, inflammation, and neonatal mortality. Airway inflammation includes macrophage activation, neutrophil and eosinophil recruitment, and elevated KC, TNF-alpha, and chitinase levels. These changes recapitulate aspects of complex human obstructive airway diseases, but their molecular mechanisms are poorly understood. We used genetic and pharmacologic approaches to identify pathways relevant to the development of Scnn1b-Tg mouse lung pathology. Genetic deletion of TNF-alpha or its receptor, TNFR1, had no measurable effect on the phenotype. Deletion of IL-4Ralpha abolished transient mucous secretory cell (MuSC) abundance and eosinophilia normally observed in neonatal wild-type mice. Similarly, IL-4Ralpha deficiency decreased MuSC and eosinophils in neonatal Scnn1b-Tg mice, which correlated with improved neonatal survival. However, chronic lung pathology in adult Scnn1b-Tg mice was not affected by IL-4Ralpha status. Prednisolone treatment ablated eosinophilia and MuSC in adult Scnn1b-Tg mice, but did not decrease mucus plugging or neutrophilia. These studies demonstrate that: 1) normal neonatal mouse airway development entails an IL-4Ralpha-dependent, transient abundance of MuSC and eosinophils; 2) absence of IL-4Ralpha improved neonatal survival of Scnn1b-Tg mice, likely reflecting decreased formation of asphyxiating mucus plugs; and 3) in Scnn1b-Tg mice, neutrophilia, mucus obstruction, and airspace enlargement are IL-4Ralpha- and TNF-alpha-independent, and only MuSC and eosinophilia are sensitive to glucocorticoids. Thus, manipulation of multiple pathways will likely be required to treat the complex pathogenesis caused by airway surface dehydration.


Assuntos
Imunidade nas Mucosas/fisiologia , Receptores de Superfície Celular/metabolismo , Mucosa Respiratória/fisiopatologia , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Quimiocinas/análise , Desidratação/complicações , Modelos Animais de Doenças , Eosinofilia/tratamento farmacológico , Eosinofilia/etiologia , Canais Epiteliais de Sódio/genética , Glucocorticoides/uso terapêutico , Imunidade nas Mucosas/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neutrófilos/imunologia , Prednisolona/uso terapêutico , Receptores de Superfície Celular/genética , Mucosa Respiratória/crescimento & desenvolvimento , Mucosa Respiratória/metabolismo , Sistema Respiratório/crescimento & desenvolvimento , Sistema Respiratório/imunologia , Sistema Respiratório/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...