23 research outputs found

    Comparison of leukocyte cannabinoid receptor mRNA between volunteers with and without motion sickness during the parabolic flight experiment.

    No full text
    <p>RNA was prepared from whole blood and subjected to quantitative RT-PCR (triplicate determinations). Data were calculated relative to GAPDH and RPL13A-mRNA using an efficiency-corrected algorithm. *indicates a significant decline in CB1-receptor mRNA (W = −26.0, T+ = 1.0, T- = −27.0, p = 0.03, Wilcoxon Signed Rank Test) in volunteers with motion sickness when compared to 24 h pre-flight values. CB1-receptor mRNA in individuals without motion sickness remained unchanged over time (W = −8.0, T+ = 10.0, T- = −18.0, p = 0.58) as was CB2-receptor expression before and after the experiment. Horizontal black bars in the boxplots indicate median values; upper and lower lines of the boxes show the 5<sup>th</sup> and 95<sup>th</sup> percentile.</p

    Whole blood endocannabinoid concentrations during a parabolic flight experiment in 21 male volunteers.

    No full text
    <p><i>Panel A</i> compares blood concentrations of the endocannabinoid anandamide between volunteers who developed motion sickness accompanied by severe N&V (n = 7, red lines) and those who did not (n = 14, blue lines). After the 10<sup>th</sup> parabolic maneuver (T1), anandamide blood concentrations dropped in individuals with motion sickness but increased in volunteers without (Mann-Whitney U Statistic  = 14.5, T = 42.5, *p = 0.02). After the 20<sup>th</sup> maneuver (T2), this difference remained significant (Mann-Whitney U Statistic  = 14.0, T = 42.0, *p = 0.01). <i>Panel B</i> shows the same comparison with regard to the endocannabinoid 2-AG. 2-AG values in volunteers with motion sickness remained almost constant and non-reactive but increased in individuals without. *indicates significantly higher 2-AG blood concentrations in individuals without motion sickness after termination of parabolic maneuvers (Mann-Whitney U Statistic = 0.0, T = 85.0, p = 0.04).</p

    Hypoxia Down-Regulates Neutrophils and Protects Lung Tissue from Inflammatory Damage

    No full text
    <div><p>(A) Exposure of IT LPS-injected mice to hypoxic (10%) oxygen levels for 48 h atmosphere leads to a significantly decreased accumulation of PMNs (left graph), production of LPS-triggered oxygen reactive metabolites in lungs (center graph), and improved lung gas exchange (right graph) compared to a control group of endotoxin-treated mice that were kept at ambient (21%) oxygen. To standardize conditions, the arterial blood samples were taken 15 min after return of the previously hypoxia-exposed animals to normal atmosphere.</p> <p>(B) Treatment by a shorter period of hypoxia attenuates PMN sequestration (left graph) and lung vascular permeability (right graph). Hypoxic treatment of mice even for only 24 h was sufficient to delay PMN sequestration and to diminish the increase in lung vascular permeability.</p> <p>(C) Histologic evidence for the hypoxic inhibition of pulmonary PMN sequestration. Quantitative analysis of lung slices by a pathologist blinded to the experimental design revealed inhibition of PMN sequestration in IT LPS-injected mice following 4-h exposure to hypoxia. Hypoxia not only attenuated PMN accumulation, but the lung tissue damage was also significantly decreased as assessed by the LIS (<i>n</i> = 9, mean ± standard deviation). The representative H&E-stained slices in the right two photomicrographs show less intravascular granulocyte sequestration, less thickening of the alveolocapillary membrane, and almost no granulocytes in the alveolar spaces as compared to IT endotoxin-injected animals breathing 21% O<sub>2</sub>. These observations demonstrate that hypoxia also inhibited the transmigration of granulocytes from capillaries into the alveolar spaces.</p></div

    IT Administration of A<sub>2A</sub>R Agonist Protects from Increased Death Rate upon Oxygenation of Mice with Acute Lung Injury

    No full text
    <p>Compensation for the oxygenation-associated loss of the hypoxia → adenosine → A<sub>2A</sub>R signaling pathway by IT injection of CGS21680 significantly decreased the oxygen-exacerbated death rate in mice with acute lung injury induced by IT injection of SEB and LPS. For further explanation, see legend for <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.0030174#pbio-0030174-g001" target="_blank">Figure 1</a>.</p

    Intratracheal Injection of A<sub>2A</sub>R Selective Agonist Mimics Protective Effects of Hypoxia

    No full text
    <div><p>(A) IT injection of the A<sub>2A</sub>R agonist CGS21680 into endotoxin-inflamed lungs provides protection similar to that observed in hypoxia-treated mice. Number of PMNs recovered after 48 h by BAL from endotoxin-injected animals that were kept at normal 21% oxygen atmosphere was significantly diminished by IT injections of CGS21680 compared to placebo-treated mice. Lung PMNs (left graph) from A<sub>2A</sub>R agonist-treated animals also produced lower levels of reactive oxygen metabolites (H<sub>2</sub>O<sub>2</sub>; right graph).</p> <p>(B) Significantly decreased lung vascular permeability (protein in BAL; left graph) and improved lung gas exchange (p<sub>a</sub>O<sub>2</sub>; right graph) in endotoxin-injected mice after treatment with the A<sub>2A</sub>R agonist CGS21680.</p> <p>(C) Histologic evidence for the lung tissue-protecting effects of A<sub>2A</sub>R agonist during endotoxin- and oxygenation-induced lung damage. Quantitative analysis of lung histopathology by a pathologist blinded to the experimental design revealed inhibition of PMN sequestration in IT LPS-injected mice after treatment with the A<sub>2A</sub>R-selective agonist CGS21680 for 48 h. The lung tissue damage was also significantly decreased as assessed by the LIS (<i>n</i> = 9, mean ± standard deviation). Representative H&E-stained slices in the right two photomicrographs show less intracapillary PMN sequestration and almost no intraalveolar accumulation of PMNs in CGS21680-treated mice. These CGS21680-induced changes are similar to those observed for the effects of hypoxia on endotoxin- injected animals (compare with <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.0030174#pbio-0030174-g006" target="_blank">Figure 6</a>C).</p></div

    Stress and nausea during the experiment.

    No full text
    <p>Comparison of KAB stress symptom and nausea scores between participants with and without motion sickness during the parabolic flight experiment.</p><p>*p<0.01 and</p>#<p>p<0.005 when compared to participants without motion sickness. Data are mean±SD.</p>a<p>German Kurzfragebogen zur aktuellen Beanspruchung (short questionnaire of current stress; KAB) <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0010752#pone.0010752-Mueller1" target="_blank">[21]</a>.</p>b<p>Nausea was quantified on a verbal rating scale ranging from 1 =  no nausea to 6 =  maximal nausea.</p

    Role of PMNs in the Pathogenesis of LPS-Induced Lung Injury

    No full text
    <div><p>(A) Depletion of granulocytes attenuates the endotoxin induced rise in alveolocapillary permeability. Pretreatment of mice with anti-Gr-1 was followed by a significant decrease in the number of granulocytes (left graph) and a significant reduction of the total amount of protein (right graph) recovered by BAL 48 h after IT LPS injection.</p> <p>(B) The more granulocytes immigrated into the alveolar spaces, the higher the alveolocapillary permeability rose. Bivariate analysis according to Pearson revealed a statistically significant correlation (<i>p</i> < 0,001) between the number of PMNs and the amount of protein in the BAL fluid 48 h after IT LPS injection, suggesting that inflammatory lung injury after IT injection of LPS is mostly mediated by granulocytes.</p></div

    Evidence for the Up-Regulation of Immunosuppressive A<sub>2A</sub>R Expression on In Vivo-Activated Granulocytes Isolated from Inflamed Lungs

    No full text
    <div><p>(A) The selective A<sub>2A</sub>R agonist CGS21680 inhibited the fMLP-stimulated hydrogen peroxide production by granulocytes in blood of healthy mice to only a small degree, reflecting low levels of expression of A<sub>2A</sub>R on naïve blood granulocytes. In contrast, granulocytes recovered by BAL from inflamed lungs 48 h after IT LPS injection were much more inhibited by CGS21680, demonstrating functional up-regulation of A<sub>2A</sub>R on in vivo-activated cells.</p> <p>(B) CGS21680 induces cAMP accumulation in in vivo-activated granulocytes isolated from lungs 48 h after IT LPS injection. No effects of the A<sub>2A</sub>R agonist were observed in naïve granulocytes obtained from bone marrow of healthy mice (left graph) or in in vivo-activated granulocytes recovered from inflamed lungs of <i>A<sub>2A</sub>R</i> gene-deficient mice (right graph). The CGS21680-stimulated cAMP production observed in lung granulocytes obtained from wild-type mice could also be antagonized by the selective A<sub>2A</sub>R antagonist ZM241385. Naïve bone marrow granulocytes were used for cAMP measurements, since it was impossible to isolate naïve cells from blood of healthy mice in sufficient numbers.</p> <p>(C) Higher levels of A<sub>2A</sub>R-specific mRNA in in vivo-activated granulocytes. In parallel with the much stronger A<sub>2A</sub>R agonist-induced inhibition of hydrogen peroxide production and accumulation of cAMP in in vivo-activated granulocytes, the relative levels of A<sub>2A</sub>R-specific mRNA were much higher in in vivo-activated granulocytes obtained from inflamed lungs 48 h after IT LPS injection, as compared with naïve granulocytes isolated from the bone marrow of healthy mice (left graph). Up-regulation of A<sub>2A</sub>R mRNA in in vivo-activated granulocytes was confirmed in another set of experimental animals breathing 21% oxygen, but was increased to a much lesser extent in animals subjected to 100% oxygen (right graph). Levels of A<sub>1</sub>R mRNA did not change much in inflammatory lung granulocytes from animals breathing normal atmosphere, but were clearly increased in mice exposed to 100% O<sub>2</sub>. In the two sets of experiments (left and right graphs), granulocytes were pooled from five and six mice per treatment, respectively. Taken together, the results demonstrate that granulocytes recovered from alveolar spaces of inflamed lungs did, indeed, up-regulate their A<sub>2A</sub>R expression during these in vivo lung injury assays, thereby confirming and extending previous findings in other inflammation models [<a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.0030174#pbio-0030174-b11" target="_blank">11</a>, <a href="http://www.plosbiology.org/article/info:doi/10.1371/journal.pbio.0030174#pbio-0030174-b53" target="_blank">53</a>]</p></div

    Increased Death Rate upon Oxygenation of Mice with Acute Lung Injury

    No full text
    <p>Mice were IT injected with SEB and LPS to model polymicrobial infection and were exposed to 21% or 100% oxygen for 48–60 h. Determination of time-dependent survival curves was prohibited by considerations of unrelieved severe respiratory distress in NIH-approved animal care protocol, which required termination of experiments immediately after differences between groups became apparent. Major differences between groups occurred within 48–60 hours after IT injection of toxins, when the majority of oxygenated animals with inflamed lungs had died, while the nonoxygenated, obviously sick control mice with inflamed lungs were still alive.</p

    Exacerbation of Inflammatory Lung Injury after Exposure of Mice to Different Concentrations of Oxygen

    No full text
    <div><p>(A) Enhanced lung vascular permeability (left graph) and impairment of lung gas exchange (right graph) in mice breathing 100% O<sub>2</sub> upon induction of acute lung injury. Following IT injection of mice with SEB and LPS, animals breathed 21% or 100% oxygen. After 48 h, lung vascular permeability and lung gas exchange were determined by the amount of protein recovered by BAL or by measuring pO<sub>2</sub> values in arterial blood drawn, respectively, 15 min after return of mice to normal atmosphere. During this equilibration period, three out of seven mice previously exposed to 100% oxygen died, so that no arterial blood gas analyses could be performed, but BAL protein concentrations were determined immediately thereafter.</p> <p>(B) Increased lung vascular permeability (left graph) and impairment of lung gas exchange (right graph) in mice with acute lung injury even upon exposure to lower levels of oxygen (60%), which are considered clinically safe in humans. Experimental conditions were the same as in (A), except oxygen concentration was 60% instead of 100%.</p></div
    corecore