11 research outputs found

    The ENaC-overexpressing mouse as a model of cystic fibrosis lung disease

    Get PDF
    AbstractChronic lung disease remains the major cause of morbidity and mortality of cystic fibrosis (CF) patients. Cftr mutant mice developed severe intestinal obstruction, but did not exhibit the characteristic CF ion transport defects (i.e. deficient cAMP-dependent Clāˆ’ secretion and increased Na+ absorption) in the lower airways, and failed to develop CF-like lung disease. These observations led to the generation of transgenic mice with airway-specific overexpression of the epithelial Na+ channel (ENaC) as an alternative approach to mimic CF ion transport pathophysiology in the lung. Studies of the phenotype of Ī²ENaC-transgenic mice demonstrated that increased airway Na+ absorption causes airway surface liquid (ASL) depletion, reduced mucus transport and a spontaneous CF-like lung disease with airway mucus obstruction and chronic airway inflammation. Here, we summarize approaches that can be applied for studies of the complex in vivo pathogenesis and preclinical evaluation of novel therapeutic strategies in this model of CF lung disease

    Insulinā€like growth factorā€1 stimulates regulatory T cells and suppresses autoimmune disease

    No full text
    The recent precipitous rise in autoimmune diseases is placing an increasing clinical and economic burden on health systems worldwide. Current therapies are only moderately efficacious, often coupled with adverse side effects. Here, we show that recombinant human insulinā€like growth factorā€1 (rhIGFā€1) stimulates proliferation of both human and mouse regulatory T (Treg) cells in vitro and when delivered systemically via continuous minipump, it halts autoimmune disease progression in mouse models of type 1 diabetes (STZ and NOD) and multiple sclerosis (EAE) in vivo. rhIGFā€1 administration increased Treg cells in affected tissues, maintaining their suppressive properties. Genetically, ablation of the IGFā€1 receptor specifically on Treg cell populations abrogated the beneficial effects of rhIGFā€1 administration on the progression of multiple sclerotic symptoms in the EAE model, establishing a direct effect of IGFā€1 on Treg cell proliferation. These results establish systemically delivered rhIGFā€1 as a specific, effective stimulator of Treg cell action, underscoring the clinical feasibility of manipulating natural tolerance mechanisms to suppress autoimmune disease

    CFTR Regulates Early Pathogenesis of Chronic Obstructive Lung Disease in Ī²ENaC-Overexpressing Mice

    Get PDF
    <div><h3>Background</h3><p>Factors determining the onset and severity of chronic obstructive pulmonary disease remain poorly understood. Previous studies demonstrated that airway surface dehydration in Ī²ENaC-overexpressing (Ī²ENaC-Tg) mice on a mixed genetic background caused either neonatal mortality or chronic obstructive lung disease suggesting that the onset of lung disease was modulated by the genetic background.</p> <h3>Methods</h3><p>To test this hypothesis, we backcrossed Ī²ENaC-Tg mice onto two inbred strains (C57BL/6 and BALB/c) and studied effects of the genetic background on neonatal mortality, airway ion transport and airway morphology. Further, we crossed Ī²ENaC-Tg mice with CFTR-deficient mice to validate the role of CFTR in early lung disease.</p> <h3>Results</h3><p>We demonstrate that the C57BL/6 background conferred increased CFTR-mediated Cl<sup>āˆ’</sup> secretion, which was associated with decreased mucus plugging and mortality in neonatal Ī²ENaC-Tg C57BL/6 compared to Ī²ENaC-Tg BALB/c mice. Conversely, genetic deletion of CFTR increased early mucus obstruction and mortality in Ī²ENaC-Tg mice.</p> <h3>Conclusions</h3><p>We conclude that a decrease or absence of CFTR function in airway epithelia aggravates the severity of early airway mucus obstruction and related mortality in Ī²ENaC-Tg mice. These results suggest that genetic or environmental factors that reduce CFTR activity may contribute to the onset and severity of chronic obstructive pulmonary disease and that CFTR may serve as a novel therapeutic target.</p> </div

    Genetic background modulates airway ion transport in wild-type and Ī²ENaC-Tg mice.

    No full text
    <p>A) Representative original recordings of the effects of amiloride and cAMP-dependent activation (IBMX/forskolin) on transepithelial voltage (V<sub>te</sub>) and transepithelial resistance (R<sub>te</sub>) across freshly excised tracheal tissues from neonatal (3-day-old) wild-type (WT) and Ī²ENaC-Tg mice on the C57BL/6 and BALB/c background. R<sub>te</sub> was determined from V<sub>te</sub> deflections obtained by pulsed current injection. (Bā€“F) Summary of basal equivalent short-circuit current (I<sub>sc</sub>) (B), amiloride-sensitive I<sub>sc</sub> (C), amiloride-insensitive I<sub>sc</sub> (D), cAMP-induced I<sub>sc</sub> (E) and UTP-induced I<sub>sc</sub> (F) in freshly excised tracheal tissues from neonatal WT and Ī²ENaC-Tg mice on the C57BL/6 and BALB/c background. Data are presented as mean Ā± SEM (<i>n</i>ā€Š=ā€Š8ā€“20 mice per group). *<i>P</i><0.05 and **<i>P</i><0.001 compared with WT mice on same strain background, <sup>ā€ </sup><i>P</i><0.05 and <sup>ā€ ā€ </sup><i>P</i><0.01 compared with mice of same genotype on C57BL/6 background.</p

    Survival of Ī²ENaC-Tg mice is modified by genetic background.

    No full text
    <p>Survival curves of Ī²ENaC-Tg mice and wild-type (WT) littermates on mixed (C3H/He x C57BL/6), C57BL/6 and BALB/c backgrounds (nā€Š=ā€Š36ā€“59 mice per group). *<i>P</i><0.05 and <sup>ā€ </sup><i>P</i><0.01 (Kaplan-Meier survival analysis).</p

    Genetic background modulates CFTR-mediated Cl<sup>āˆ’</sup> secretion in airways of wild-type and Ī²ENaC-Tg mice.

    No full text
    <p>Aā€“C) Effects of genetic background on transepithelial Cl- secretion were determined by adding bumetanide or CFTR<sub>inh</sub>-172 to amiloride-pretreated tracheal tissues from neonatal wild-type (WT) and Ī²ENaC-Tg mice on the C57BL/6 and BALB/c background. A,B) Summary of bumetanide-sensitive I<sub>sc</sub> (A) and CFTR<sub>inh</sub>-172-sensitive I<sub>sc</sub> (B) in the presence of amiloride (<i>n</i>ā€Š=ā€Š12ā€“23 mice per group). C) Summary of CFTR<sub>inh</sub>-172-sensitive I<sub>sc</sub> in the presence of amiloride and cAMP-dependent activation (IBMX/forskolin) (<i>n</i>ā€Š=ā€Š5ā€“11 mice per group). Data are presented as mean Ā± SEM. <sup>ā€ </sup><i>P</i><0.05 and <sup>ā€ ā€ </sup><i>P</i><0.01 compared with mice of same genotype on C57BL/6 background.</p

    Genetic background modifies early airway mucus obstruction and epithelial necrosis in neonatal Ī²ENaC-Tg mice.

    No full text
    <p>A) Longitudinal sections of tracheae from neonatal (3-day-old) wild-type (WT) and Ī²ENaC-Tg mice on the C57BL/6 and BALB/c background. Sections were stained with Alcian blueā€“periodic acid Schiff (AB-PAS) to determine the presence of mucus and goblet cells. Scale bars ā€Š=ā€Š200 Āµm. Bā€“D) Summary of mucus content, as determined from measuring volume density of AB-PAS-positive material in the tracheal lumen (B), goblet cell numbers (C) and Transcript levels of Muc5b (D) (<i>n</i>ā€Š=ā€Š4ā€“13 mice per group). E) Airway histology from neonatal (3-day-old) WT and Ī²ENaC-Tg mice on the C57BL/6 and BALB/c background. Sections were stained with hematoxylin and eosin (H&E) and evaluated for degenerative airway epithelial cells (arrows). Scale bars ā€Š=ā€Š40 Āµm (upper panels) and 20 Āµm (lower panels). F) Summary of airway epithelial necrosis as determined from the number of degenerative epithelial cells per mm of the basement membrane (<i>n</i>ā€Š=ā€Š9ā€“11 mice per group). *<i>P</i><0.001 compared with WT mice on same strain background, <sup>ā€ </sup><i>P</i><0.05 compared with mice of same genotype on C57BL/6 background.</p

    Airway Surface Liquid Volume Regulation Determines Different Airway Phenotypes in Liddle Compared with Ī²ENaC-overexpressing Mice*

    Get PDF
    Studies in cystic fibrosis patients and mice overexpressing the epithelial Na+ channel Ī²-subunit (Ī²ENaC-Tg) suggest that raised airway Na+ transport and airway surface liquid (ASL) depletion are central to the pathogenesis of cystic fibrosis lung disease. However, patients or mice with Liddle gain-of-function Ī²ENaC mutations exhibit hypertension but no lung disease. To investigate this apparent paradox, we compared the airway phenotype (nasal versus tracheal) of Liddle with CFTR-null, Ī²ENaC-Tg, and double mutant mice. In mouse nasal epithelium, the region that functionally mimics human airways, high levels of CFTR expression inhibited Liddle epithelial Nat channel (ENaC) hyperfunction. Conversely, in mouse trachea, low levels of CFTR failed to suppress Liddle ENaC hyperfunction. Indeed, Na+ transport measured in Ussing chambers (ā€œfloodedā€ conditions) was raised in both Liddle and Ī²ENaC-Tg mice. Because enhanced Na+ transport did not correlate with lung disease in these mutant mice, measurements in tracheal cultures under physiologic ā€œthin filmā€ conditions and in vivo were performed. Regulation of ASL volume and ENaC-mediated Na+ absorption were intact in Liddle but defective in Ī²ENaC-Tg mice. We conclude that the capacity to regulate Na+ transport and ASL volume, not absolute Na+ transport rates in Ussing chambers, is the key physiologic function protecting airways from dehydration-induced lung disease
    corecore