12 research outputs found

    Expression of VPAC1 in a murine model of allergic asthma

    Get PDF
    Vasoactive intestinal polypeptide (VIP) is a putative neurotransmitter of the inhibitory non-adrenergic non-cholinergic nervous system and influences the mammalian airway function in various ways. Hence known for bronchodilatory, immunomodulatory and mucus secretion modulating effects by interacting with the VIP receptors VPAC1 and VPAC2, it is discussed to be a promising target for pharmaceutical intervention in common diseases such as COPD and bronchial asthma. Here we examined the expression and transcriptional regulation of VPAC1 in the lungs of allergic mice using an ovalbumin (OVA) -induced model of allergic asthma. Mice were sensitized to OVA and challenged with an OVA aerosol. In parallel a control group was sham sensitized with saline. VPAC1 expression was examined using RT-PCR and real time-PCR studies were performed to quantify gene transcription. VPAC1 mRNA expression was detected in all samples of OVA-sensitized and challenged animals and control tissues. Further realtime analysis did not show significant differences at the transcriptional level. Although the present studies did not indicate a major transcriptional regulation of VPAC1 in states of allergic airway inflammation, immunomodulatory effects of VPAC1 might still be present due to regulations at the translational level

    LPS-induced lung inflammation in marmoset monkeys - an acute model for anti-inflammatory drug testing.

    Get PDF
    Increasing incidence and substantial morbidity and mortality of respiratory diseases requires the development of new human-specific anti-inflammatory and disease-modifying therapeutics. Therefore, new predictive animal models that closely reflect human lung pathology are needed. In the current study, a tiered acute lipopolysaccharide (LPS)-induced inflammation model was established in marmoset monkeys (Callithrix jacchus) to reflect crucial features of inflammatory lung diseases. Firstly, in an ex vivo approach marmoset and, for the purposes of comparison, human precision-cut lung slices (PCLS) were stimulated with LPS in the presence or absence of the phosphodiesterase-4 (PDE4) inhibitor roflumilast. Pro-inflammatory cytokines including tumor necrosis factor-alpha (TNF-α) and macrophage inflammatory protein-1 beta (MIP-1β) were measured. The corticosteroid dexamethasone was used as treatment control. Secondly, in an in vivo approach marmosets were pre-treated with roflumilast or dexamethasone and unilaterally challenged with LPS. Ipsilateral bronchoalveolar lavage (BAL) was conducted 18 hours after LPS challenge. BAL fluid was processed and analyzed for neutrophils, TNF-α, and MIP-1β. TNF-α release in marmoset PCLS correlated significantly with human PCLS. Roflumilast treatment significantly reduced TNF-α secretion ex vivo in both species, with comparable half maximal inhibitory concentration (IC(50)). LPS instillation into marmoset lungs caused a profound inflammation as shown by neutrophilic influx and increased TNF-α and MIP-1β levels in BAL fluid. This inflammatory response was significantly suppressed by roflumilast and dexamethasone. The close similarity of marmoset and human lungs regarding LPS-induced inflammation and the significant anti-inflammatory effect of approved pharmaceuticals assess the suitability of marmoset monkeys to serve as a promising model for studying anti-inflammatory drugs

    Dose-response curves of roflumilast in marmoset and human PCLS.

    No full text
    <p>Marmoset and human PCLS were incubated with 100 ng/mL LPS and increasing concentrations of roflumilast (rof, 0.6–310 nM) for 24 hours. The half maximal inhibitory concentrations were detected at 1.3 nM for marmoset and 1.1 nM for human (dashed lines). Data are presented as mean ± SEM, marmoset: n  = 5; human: n  = 4. TNF-α concentration in supernatants was determined by ELISA. Marmoset  =  grey symbols, human  =  black symbols.</p

    Changes in absolute cell numbers in bronchoalveolar lavage (BAL) fluid after LPS challenge.

    No full text
    <p>Sham-treated, dexamethasone (dxm)-treated, and roflumilast (rof)-treated marmosets were intrabronchially challenged with 500 ng LPS. Eighteen hours later, ipsilateral BAL was performed. Total cells (A), neutrophils (B), macrophages (C), and lymphocytes (D) were differentiated and quantified using light microscopy after Pappenheim staining. Data are presented as scatter dot plot with median, *p<0.05, ***p<0.001, one-tailed Mann-Whitney test against sham.</p

    LPS-induced changes in bronchoalveolar lavage (BAL) fluid.

    No full text
    <p>Representative cytospots of BAL at x200 original magnification after Pappenheim staining. (A) Macrophages are the predominant cell type in unchallenged lung lobes. (B) LPS challenge induced strong neutrophilic influx in sham-treated animals. This effect could be significantly attenuated by (C) roflumilast and (D) dexamethasone pre-treatment. Scale bar  = 50 µm.</p

    Demographic data of the study population.

    No full text
    <p>Animals were randomized in each of the two independent study cycles as indicated (a: first study cycle, b: second study cycle). Before each cycle a baseline BAL was performed 3 weeks before LPS challenge and served as control. Altogether, 3 animals had to be excluded.</p><p>Data are given as mean ± S.E.M., dxm  =  dexamethasone, rof  =  roflumilast.</p

    Analysis of bronchoalveolar lavage (BAL) fluid.

    No full text
    <p>TNF-α content in BAL fluid of marmoset monkeys was significantly reduced in dexamethasone-treated (dxm, p = 0.04) and roflumilast-treated (rof, p = 0.049) animals in contrast to sham-treated individuals (A). MIP-1β concentrations in BAL fluid were reduced by trend in both treatment groups (sham vs. dxm: p = 0.27; sham vs. rof: p = 0.17) (B). Data are presented as scatter dot plot with median, *p<0.05, ***p<0.0.01, one-tailed Mann-Whitney test against sham.</p

    LPS-dependent increase in cytokines and chemokines <i>ex vivo</i>.

    No full text
    <p>Ascending TNF-α (A) and intracellular MIP-1β (B) production after 24-hour incubation with increasing concentrations of LPS in marmoset PCLS. LPS-induced increase in TNF-α (C) and intracellular MIP-1β (D) in marmoset PCLS was significantly suppressed by dexamethasone (dxm). Marmoset PCLS and marmoset WBA (E) on the one hand and marmoset PCLS and human PCLS (F) on the other hand showed significant correlations for TNF-α secretion (Spearman’s rank correlation coefficient (r<sub>S</sub>)  = 1.0 with p  = 0.0004, and r<sub>s</sub>  = 0.9 with p  = 0.01, respectively). Symbols: ○: 2.5 ng/mL LPS, □: 5 ng/mL LPS, ▾: 10 ng/mL LPS, ▵: 100 ng/mL LPS, ◊: 250 ng/mL LPS, x: 500 ng/mL LPS. Data are presented as mean ± SEM, *p<0.05, **p<0.01, Mann-Whitney test (TNF-α: n  = 6, MIP-1β: n  = 4). Correlations were evaluated using a linear regression analysis model combined with Spearman’s rank correlation coefficient. NHP  =  non-human primate, int  =  intracellular.</p
    corecore