Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Front Pharmacol ; 12: 573065, 2021.
Article En | MEDLINE | ID: mdl-34054509

In cystic fibrosis (CF), sustained infection and exuberant inflammation results in debilitating and often fatal lung disease. Advancement in CF therapeutics has provided successful treatment regimens for a variety of clinical consequences in CF; however effective means to treat the pulmonary infection and inflammation continues to be problematic. Even with the successful development of small molecule cystic fibrosis transmembrane conductance regulator (CFTR) correctors and potentiators, there is only a modest effect on established infection and inflammation in CF patients. In the pursuit of therapeutics to treat inflammation, the conundrum to address is how to overcome the inflammatory response without jeopardizing the required immunity to manage pathogens and prevent infection. The key therapeutic would have the capacity to dull the inflammatory response, while sustaining the ability to manage infections. Advances in cell-based therapy have opened up the avenue for dynamic and versatile immune interventions that may support this requirement. Cell based therapy has the capacity to augment the patient's own ability to manage their inflammatory status while at the same time sustaining anti-pathogen immunity. The studies highlighted in this manuscript outline the potential use of cell-based therapy for CF. The data demonstrate that 1) total bone marrow aspirates containing Cftr sufficient hematopoietic and mesenchymal stem cells (hMSCs) provide Cftr deficient mice >50% improvement in survival and improved management of infection and inflammation; 2) myeloid cells can provide sufficient Cftr to provide pre-clinical anti-inflammatory and antimicrobial benefit; 3) hMSCs provide significant improvement in survival and management of infection and inflammation in CF; 4) the combined interaction between macrophages and hMSCs can potentially enhance anti-inflammatory and antimicrobial support through manipulating PPARγ. These data support the development of optimized cell-based therapeutics to enhance CF patient's own immune repertoire and capacity to maintain the balance between inflammation and pathogen management.

2.
J Inflamm (Lond) ; 7: 52, 2010 Oct 30.
Article En | MEDLINE | ID: mdl-21034489

BACKGROUND: 17α-Ethynyl-5-androsten-3ß, 7ß, 17ß-triol (HE3286) is a synthetic derivative of an endogenous steroid androstenetriol (ß-AET), a metabolite of the abundant adrenal steroid deyhdroepiandrosterone (DHEA), with broad anti-inflammatory activities. We tested the ability of this novel synthetic steroid with improved pharmacological properties to limit non-productive lung inflammation in rodents and attempted to gauge its immunological impact. METHODS AND RESULTS: In mice, oral treatment with HE3286 (40 mg/kg) significantly (p < 0.05) decreased neutrophil counts and exudate volumes (~50%) in carrageenan-induced pleurisy, and myeloperoxidase in lipopolysaccharide-induced lung injury. HE3286 (40 mg/kg) was not found to be profoundly immune suppressive in any of the classical animal models of immune function, including those used to evaluate antigen specific immune responses in vivo (ovalbumin immunization). When mice treated for two weeks with HE3286 were challenged with K. pneumoniae, nearly identical survival kinetics were observed in vehicle-treated, HE3286-treated and untreated groups. CONCLUSIONS: HE3286 represents a novel, first-in-class anti-inflammatory agent that may translate certain benefits of ß-AET observed in rodents into treatments for chronic inflammatory pulmonary disease.

3.
J Magn Reson Imaging ; 28(2): 527-32, 2008 Aug.
Article En | MEDLINE | ID: mdl-18666218

PURPOSE: To evaluate magnetic resonance imaging (MRI) in assessing lung inflammation longitudinally in genetic mouse models of cystic fibrosis (CF). MRI is used to view soft tissues noninvasively, but the lung is challenging to image. MATERIALS AND METHODS: Cftr(+/+) (wildtype) and Cftr(-/-) (CF) mice were inoculated with agarose beads laden with Pseudomonas aeruginosa. Longitudinal MR lung images were acquired with cardiac gating. The effects of echo time and respiration gating were evaluated to improve the detection of lung inflammation. RESULTS: Cardiac gating and signal averaging sufficiently suppressed motion artifacts without requiring respiration gating. MRI detected moderate to severe inflammation in infected mice, which was confirmed by histology results. CONCLUSION: In vivo longitudinal MRI methods can assess lung inflammation in P. aeruginosa-infected mice, which obviates serial sacrifice. MRI was able to detect inflammation in the absence of other physiological symptoms.


Cystic Fibrosis/complications , Magnetic Resonance Imaging/methods , Pseudomonas Infections/pathology , Animals , Artifacts , Disease Models, Animal , Inflammation , Lung Diseases/microbiology , Mice , Mice, Inbred C57BL , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa
4.
Am J Physiol Lung Cell Mol Physiol ; 295(2): L303-13, 2008 Aug.
Article En | MEDLINE | ID: mdl-18556801

The pathophysiology of cystic fibrosis (CF) inflammatory lung disease is not well understood. CF airway epithelial cells respond to inflammatory stimuli with increased production of proinflammatory cytokines as a result of increased NF-kappaB activation. Peroxisome proliferator-activated receptor-gamma (PPARgamma) inhibits NF-kappaB activity and is reported to be reduced in CF. If PPARgamma participates in regulatory dysfunction in the CF lung, perhaps PPARgamma ligands might be useful therapeutically. Cell models of CF airway epithelium were used to evaluate PPARgamma expression and binding to NF-kappaB at basal and under conditions of inflammatory stimulation by Pseudomonas aeruginosa or TNFalpha/IL-1beta. An animal model of CF was used to evaluate the potential of PPARgamma agonists as therapeutic agents in vivo. In vitro, PPARgamma agonists reduced IL-8 and MMP-9 release from airway epithelial cells in response to PAO1 or TNFalpha/IL-1beta stimulation. Less NF-kappaB bound to PPARgamma in CF than normal cells, in two different assays; PPARgamma agonists abrogated this reduction. PPARgamma bound less to its target DNA sequence in CF cells. To test the importance of the reported PPARgamma inactivation by phosphorylation, we observed that inhibitors of ERK, but not JNK, were synergistic with PPARgamma agonists in reducing IL-8 secretion. In vivo, administration of PPARgamma agonists reduced airway inflammation in response to acute infection with P. aeruginosa in CF, but not wild-type, mice. In summary, PPARgamma inhibits the inflammatory response in CF, at least in part by interaction with NF-kappaB in airway epithelial cells. PPARgamma agonists may be therapeutic in CF.


Cystic Fibrosis/metabolism , Lung/metabolism , PPAR gamma/metabolism , Respiratory Mucosa/metabolism , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/pharmacology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation Mediators , Lung/pathology , MAP Kinase Kinase 4/antagonists & inhibitors , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pseudomonas Infections/genetics , Pseudomonas Infections/metabolism , Pseudomonas Infections/pathology , Pseudomonas aeruginosa , Respiratory Mucosa/pathology
5.
Nat Med ; 14(4): 382-91, 2008 Apr.
Article En | MEDLINE | ID: mdl-18376404

Microbial lung infections are the major cause of morbidity and mortality in the hereditary metabolic disorder cystic fibrosis, yet the molecular mechanisms leading from the mutation of cystic fibrosis transmembrane conductance regulator (CFTR) to lung infection are still unclear. Here, we show that ceramide age-dependently accumulates in the respiratory tract of uninfected Cftr-deficient mice owing to an alkalinization of intracellular vesicles in Cftr-deficient cells. This change in pH results in an imbalance between acid sphingomyelinase (Asm) cleavage of sphingomyelin to ceramide and acid ceramidase consumption of ceramide, resulting in the higher levels of ceramide. The accumulation of ceramide causes Cftr-deficient mice to suffer from constitutive age-dependent pulmonary inflammation, death of respiratory epithelial cells, deposits of DNA in bronchi and high susceptibility to severe Pseudomonas aeruginosa infections. Partial genetic deficiency of Asm in Cftr(-/-)Smpd1(+/-) mice or pharmacological treatment of Cftr-deficient mice with the Asm blocker amitriptyline normalizes pulmonary ceramide and prevents all pathological findings, including susceptibility to infection. These data suggest inhibition of Asm as a new treatment strategy for cystic fibrosis.


Ceramides/metabolism , Cystic Fibrosis/metabolism , Lung/metabolism , Animals , Bacterial Adhesion , Cell Death , Cystic Fibrosis/complications , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , DNA/metabolism , Humans , Hydrogen-Ion Concentration , Inflammation Mediators/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred CFTR , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/etiology , Pseudomonas Infections/metabolism , Pseudomonas Infections/pathology , Pseudomonas Infections/prevention & control , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics
6.
Am J Respir Crit Care Med ; 173(3): 288-96, 2006 Feb 01.
Article En | MEDLINE | ID: mdl-16272448

RATIONALE: Cystic fibrosis is caused by defects in the cystic fibrosis transmembrane conductance regulator gene, which codes for a chloride channel, but the role of this chloride channel in inflammation induced by lung infection with Pseudomonas aeruginosa remains to be defined. OBJECTIVES: We tested the hypothesis that loss of this chloride channel alone is sufficient to cause excessive inflammation in response to inflammatory stimuli. METHODS: We investigated the response of cystic fibrosis and wild-type mice to mucoid P. aeruginosa administered by insufflation. MEASUREMENTS: The host responses measured included survival, weight change, lung morphometry, bacterial clearance, and inflammatory mediators, and cell counts were assessed in bronchoalveolar lavage fluid. MAIN RESULTS: Depending on the dose administered and frequency of dosing, cystic fibrosis mice experienced significantly higher mortality rates, greater weight loss, higher lung pathology scores, and higher inflammatory mediator and neutrophil levels compared with wild-type mice, even after the bacteria had been cleared. Surprisingly, bacteria were cleared just as rapidly in cystic fibrosis mice as in wild-type mice, and sepsis was not observed. Chronic lung infections could not be established with mucoid P. aeruginosa in either cystic fibrosis or wild-type mice. CONCLUSIONS: Absence of this chloride channel alone appears sufficient for exaggerated inflammation and excess mortality compared with wild-type controls in the face of mucoid P. aeruginosa lung infection. To establish chronic infection, additional factors such as bacterial trapping or poor clearance may be required.


Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/pathology , Pseudomonas Infections/metabolism , Pseudomonas Infections/pathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cytokines/metabolism , Disease Models, Animal , Mice , Mice, Congenic , Mice, Inbred CFTR , Pneumonia, Bacterial/microbiology
7.
Am J Respir Cell Mol Biol ; 34(3): 355-63, 2006 Mar.
Article En | MEDLINE | ID: mdl-16284361

Deletion of phenylalanine 508 (deltaF508) accounts for nearly 70% of all mutations that occur in the cystic fibrosis transmembrane conductance regulator (CFTR). The deltaF508 mutation is a class II processing mutation that results in very little or no mature CFTR protein reaching the apical membrane and thus no cAMP-mediated Cl- conductance. Therapeutic strategies have been developed to enhance processing of the defective deltaF508 CFTR molecule so that a functional cAMP-regulated Cl- channel targets to the apical membrane. Sarcoplasmic/endoplasmic reticulum calcium (SERCA) inhibitors, curcumin and thapsigargin, have been reported to effectively correct the CF ion transport defects observed in the deltaF508 CF mice. We investigated the effect of these compounds in human airway epithelial cells to determine if they could induce deltaF508 CFTR maturation, and Cl- secretion. We also used Baby Hamster Kidney cells, heterologously expressing deltaF508 CFTR, to determine if SERCA inhibitors could interfere with the interaction between calnexin and CFTR and thereby correct the deltaF508 CFTR misfolding defect. Finally, at the whole animal level, we tested the ability of curcumin and thapsigargin to (1) induce Cl- secretion and reduce hyperabsorption of Na+ in the nasal epithelia of the deltaF508 mouse in vivo, and (2) induce Cl- secretion in intestine (jejunum and distal colon) and the gallbladder of the deltaF508 CF mouse. We conclude that curcumin and thapsigargin failed to induce maturation of deltaF508 CFTR, or induce Cl- secretion, as measured by biochemical and electrophysiologic techniques in a variety of model systems ranging from cultured cells to in vivo studies.


Calcium-Transporting ATPases/antagonists & inhibitors , Curcumin/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Cystic Fibrosis/metabolism , Respiratory Mucosa/drug effects , Thapsigargin/pharmacology , Animals , Bronchi/cytology , Calnexin/metabolism , Cells, Cultured , Chlorides/metabolism , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gallbladder/drug effects , Gallbladder/physiology , Humans , Intestines/drug effects , Intestines/physiology , Ion Transport , Mice , Mice, Mutant Strains , Mutation , Protein Folding , Respiratory Mucosa/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium/metabolism
8.
Infect Immun ; 73(1): 661-5, 2005 Jan.
Article En | MEDLINE | ID: mdl-15618213

Tissue inhibitor of metalloproteinase 1 (TIMP-1)-deficient mice are resistant to Pseudomonas aeruginosa corneal infections. Corneas healed completely in TIMP-1-deficient mice, and infections were cleared faster in TIMP-1-deficient mice than in wild-type littermates. Genetic suppression studies using matrix metalloproteinase (MMP)-deficient mice showed that MMP-9, MMP-3, and MMP-7 but not MMP-2 or MMP-12 are needed for resistance. Increased resistance was also seen during pulmonary infections. These results identify a novel pathway regulating infection resistance.


Pseudomonas Infections/immunology , Tissue Inhibitor of Metalloproteinase-1/physiology , Animals , Lung Diseases/immunology , Matrix Metalloproteinase 3/physiology , Matrix Metalloproteinase 9/physiology , Mice , Mice, Inbred C57BL
9.
Am J Physiol Lung Cell Mol Physiol ; 287(5): L944-52, 2004 Nov.
Article En | MEDLINE | ID: mdl-15246977

Patients with cystic fibrosis have a lesion in the cystic fibrosis transmembrane conductance regulator gene (CFTR), which is associated with abnormal regulation of other ion channels, abnormal glycosylation of secreted and cell surface molecules, and vulnerability to bacterial infection and inflammation in the lung usually leading to the death of these patients. The exact mechanism(s) by which mutation in CFTR leads to lung infection and inflammation is not clear. Mice bearing different mutations in the murine homolog to CFTR (Cftr) (R117H, S489X, Y122X, and DeltaF508, all backcrossed to the C57BL/6J background) were compared with respect to growth and in their ability to respond to lung infection elicited with Pseudomonas aeruginosa-laden agarose beads. Body weights of mice bearing mutations in Cftr were significantly smaller than wild-type mice at most ages. The inflammatory responses to P. aeruginosa-laden agarose beads were comparable in mice of all four Cftr mutant genotypes with respect to absolute and relative cell counts in bronchoalveolar lavage fluid, and cytokine levels (TNF-alpha, IL-1beta, IL-6, macrophage inflammatory protein-2, and keratinocyte chemoattractant) and eicosanoid levels (PGE2 and LTB4) in epithelial lining fluid: the few small differences observed occurred only between cystic fibrosis mice bearing the S489X mutation and those bearing the knockout mutation Y122X. Thus we cannot implicate either misprocessing of CFTR or failure of CFTR to reach the plasma membrane in the genesis of the excess inflammatory response of CF mice. Therefore, it appears that any functional defect in CFTR produces comparable inflammatory responses to lung infections with P. aeruginosa.


Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/microbiology , Cystic Fibrosis/physiopathology , Pseudomonas Infections/physiopathology , Pseudomonas aeruginosa , Animals , Chemokine CXCL1 , Chemokine CXCL2 , Chemokines/metabolism , Chemokines, CXC , Chronic Disease , Cystic Fibrosis/immunology , Cytokines/metabolism , Dinoprostone/metabolism , Female , Genotype , Interleukin-1/metabolism , Interleukin-6/metabolism , Leukocyte Count , Leukotriene B4/metabolism , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CFTR , Neutrophils/cytology , Pseudomonas Infections/genetics , Pseudomonas Infections/immunology , Tumor Necrosis Factor-alpha/metabolism , Weight Loss
10.
Infect Immun ; 72(4): 2045-51, 2004 Apr.
Article En | MEDLINE | ID: mdl-15039325

The lung maintains an elevated level of glutathione (GSH) in epithelial lining fluid (ELF) compared to serum. The mechanism(s) by which the lung maintains high levels of ELF GSH and factors that modulate them are largely unexplored. We hypothesized that lung cystic fibrosis transmembrane conductance regulator protein (CFTR) modulates GSH efflux in response to extracellular stress, which occurs with lung infections. Mice were challenged intratracheally with Pseudomonas aeruginosa, and on the third day of infection bronchoalveolar lavage fluid was obtained and analyzed for cytokines and antioxidants. Lung tissue antioxidants and enzyme activities were also assessed. P. aeruginosa lung infection increased levels of inflammatory cytokines and neutrophils in the ELF. This corresponded with a marked threefold increase in GSH and a twofold increase in urate levels in the ELF of P. aeruginosa-infected wild-type mice. A twofold increase in urate levels was also observed among lung tissue antioxidants of P. aeruginosa-infected wild-type mice. There were no changes in markers of lung oxidative stress associated with the P. aeruginosa lung infection. In contrast with wild-type mice, the CFTR knockout mice lacked a significant increase in ELF GSH when challenged with P. aeruginosa, and this correlated with a decrease in the ratio of reduced to oxidized GSH in the ELF, a marker of oxidative stress. These data would suggest that the lung adapts to infectious agents with elevated ELF GSH and urate. Individuals with lung diseases associated with altered antioxidant transport, such as cystic fibrosis, might lack the ability to adapt to the infection and present with a more severe inflammatory response.


Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Glutathione/metabolism , Lung Diseases/metabolism , Lung/metabolism , Pseudomonas Infections/metabolism , Animals , Antioxidants/metabolism , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/immunology , Inflammation , Lung/immunology , Lung/microbiology , Lung Diseases/immunology , Lung Diseases/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CFTR , Mice, Knockout , Oxidative Stress , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity
11.
Infect Immun ; 72(3): 1479-86, 2004 Mar.
Article En | MEDLINE | ID: mdl-14977953

In cystic fibrosis, a recessive genetic disease caused by defects in the cystic fibrosis conductance regulator (CFTR), the main cause of death is lung infection and inflammation. Nutritional deficits have been proposed to contribute to the excessive host inflammatory response in both humans and Cftr-knockout mice. Cftr-knockout mice and gut-corrected Cftr-knockout mice expressing human CFTR primarily in the gut were challenged with Pseudomonas aeruginosa-laden agarose beads; they responded similarly with respect to bronchoalveolar lavage cell counts and levels of the acute-phase cytokines tumor necrosis factor alpha, interleukin-1beta (IL-1beta), and IL-6. Wild-type mice fed the liquid diet used to prevent intestinal obstruction in Cftr-knockout mice had inflammatory responses to P. aeruginosa-laden agarose beads similar to those of wild-type mice fed an enriched solid diet, so dietary effects are unlikely to account for differences between wild-type mice and mice with cystic fibrosis. Finally, since cystic fibrosis patients and Cftr-knockout mice have an imbalance in fatty acids (significantly lower-than-normal levels of docosahexaenoic acid), the effects of specific supplementation with docosahexaenoic acid of wild-type and Cftr-knockout mice on their inflammatory responses to P. aeruginosa-laden agarose beads were tested. There were no significant differences (P = 0.35) in cumulative survival rates between Cftr-knockout mice and wild-type mice provided with either the liquid diet Peptamen or Peptamen containing docosahexaenoic acid. In conclusion, diet and docosahexaenoic acid imbalances alone are unlikely to explain the differences in the host response to lung infections with mucoid P. aeruginosa between mice with cystic fibrosis and their wild-type counterparts.


Cystic Fibrosis/complications , Diet , Pneumonia, Bacterial/etiology , Pseudomonas Infections/etiology , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Digestive System/metabolism , Docosahexaenoic Acids/administration & dosage , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 286(4): L717-26, 2004 Apr.
Article En | MEDLINE | ID: mdl-14514520

In cystic fibrosis (CF) there is an excessive inflammatory response to lung infections with Pseudomonas aeruginosa, which causes significant morbidity and mortality. Mice deficient in the cystic fibrosis conductance transmembrane regulator homolog (Cftr) have exaggerated production of proinflammatory cytokines in epithelial lining fluid and increased mortality in response to chronic bronchopulmonary infection with mucoid P. aeruginosa, compared with infected wild-type littermates. Whether delivery of CFTR to CF airways by an adenoviral vector (Ad2/CFTR-16) decreases cytokine production and mortality in response to chronic bronchopulmonary infection with mucoid P. aeruginosa was tested. CF mice [stock Cftrtm1Unc-TgN(FABPCFTR)#Jaw] were anesthetized with isoflurane and inoculated intranasally with either Ad2/CFTR-16, diluent (sucrose), or empty vector (Ad2/EV). Two weeks later, mice were anesthetized with 2.5% Avertin and inoculated transtracheally with P. aeruginosa-laden agarose beads (PA M57-15). The cumulative 10-day survival of mice pretreated with Ad2/CFTR-16 was significantly higher compared with mice pretreated with sucrose but not significantly higher than mice pretreated with Ad2/EV. After adjusting for differences in experiment, we found weight loss at 3 days for mice treated with Ad2/CFTR-16 to be significantly less than for the sucrose- or Ad2/EV-treated groups. However, cytokine responses were similar in all groups 3 days after infection. In conclusion, the observed survival advantage of adenoviral delivery of CFTR to the CF lung may be due either to CFTR expression or possibly to proinflammatory effects of the adenoviral vector, or both.


Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/therapy , Genetic Therapy , Pneumonia, Bacterial/therapy , Pseudomonas Infections/therapy , Pseudomonas aeruginosa , Adenoviridae/genetics , Administration, Intranasal , Age Factors , Animals , Chronic Disease , Cystic Fibrosis/mortality , Cystic Fibrosis/pathology , Genetic Vectors , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CFTR , Mice, Knockout , Pneumonia, Bacterial/mortality , Pneumonia, Bacterial/pathology , Pseudomonas Infections/mortality , Pseudomonas Infections/pathology , Survival Rate
13.
Lab Anim ; 36(3): 291-312, 2002 Jul.
Article En | MEDLINE | ID: mdl-12144741

The animal model of chronic bronchopulmonary infection using agarose beads laden with Pseudomonas aeruginosa is frequently utilized in cystic fibrosis research, though it is challenging to perform it in mice. This paper reports the most successful methods for the creation of this model. Transtracheal insertion of a 22 G 1" over-the-needle intravenous catheter to preferentially inoculate the right mainstem bronchus using tribromoethanol anaesthesia administered i.p. was better for a successful surgical outcome compared, respectively, to the use of a 27 G (1/2)" needle, bilateral inoculation or an anaesthetic cocktail of xylazine, acepromazine and ketamine administered i.p. Bilateral infection was associated with higher mortality, greater weight loss and higher levels of bronchoalveolar cytokine concentration, compared to mice infected primarily in the right lung. Mucoid clinical strain PA M57-15 was preferred since mucoid clinical strain PA 2192 led to comparatively more severe lesions and higher mortality. Using the same operator for a given task reduced the variability inherent in this model, illustrated using outcome measures such as gross lung pathology. The response of mice inoculated with P. aeruginosa-laden agarose beads was characterized by bronchopulmonary inflammation, high production of cytokines, and significant weight loss; whereas the response to infection with free-living bacteria was characterized by pneumonia, lower production of cytokines and weight loss. The use of free P. aeruginosa pre-mixed with sterile agarose beads may be considered as an alternative to the use of P. aeruginosa-laden agarose beads, since the histopathological features were similar, though further characterization is needed to evaluate its utility as an adequate model of cystic fibrosis.


Disease Models, Animal , Pneumonia, Bacterial/pathology , Pseudomonas Infections/pathology , Animals , Body Weight , Bronchoalveolar Lavage , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Cytokines/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neutrophils/pathology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/classification , Pseudomonas aeruginosa/pathogenicity
...