Protective effect of chorioamnionitis on the development of bronchopulmonary dysplasia triggered by postnatal systemic inflammation in neonatal rats

Chang Won Choi, Juyoung Lee, Joo Youn Oh, Seung Hyun Lee, Hyun Ju Lee, Beyong Il Kim, Chang Won Choi, Juyoung Lee, Joo Youn Oh, Seung Hyun Lee, Hyun Ju Lee, Beyong Il Kim

Abstract

Background: Prenatal or postnatal systemic inflammation can contribute to the development of bronchopulmonary dysplasia (BPD). We investigated whether prenatal intra-amniotic (i.a.) inflammation or early postnatal systemic inflammation can induce BPD in a rat model.

Methods: One microgram of lipopolysaccharide (LPS) or vehicle was injected into the amniotic sacs 2 d before delivery (E20). After birth, 0.25 mg/kg of LPS or vehicle was injected into the peritoneum of pups on postnatal day (P)1, P3, and P5. On P7 and P14, peripheral blood (PB), bronchoalveolar lavage fluid (BALF), and lung tissue were obtained and analyzed.

Results: Postnatal i.p. injections of LPS significantly increased neutrophil counts in PB and BALF on P7 and P14. Similarly, proinflammatory cytokine and angiogenic factor transcript levels were increased in the lung by i.p. LPS on P7. Alveolar and pulmonary vascular development was markedly disrupted by i.p. LPS on P14. However, pretreatment with i.a. LPS significantly negated the detrimental effects of postnatal i.p. LPS on PB and BALF neutrophil counts and on lung proinflammatory cytokine expression and histopathological changes.

Conclusion: Exposure to early postnatal systemic LPS induces BPD, an arrest in alveolarization, in neonatal rats. Preceding exposure to i.a. LPS protects the lungs against BPD triggered by postnatal systemic inflammation.

Figures

Figure 1
Figure 1
Schematic outline of the experimental protocol. Intra-amniotic LPS (1.0 μg/sac) or vehicle was administered on E20. Postnatal i.p. LPS (0.25 mg/kg/d) or vehicle was administered on P1, P3, and P5. BALF and PB collection and lung harvesting were performed on P7 and P14. BALF, bronchoalveolar lavage fluid; IA, intra-amniotic; IP, intraperitoneal; PB, peripheral blood; V, vehicle.
Figure 2
Figure 2
Survival rates in the experimental groups. Fetal death mostly occurred in the intra-amniotic (i.a.) LPS-treated groups. After birth, most of the deaths occurred in the i.p. LPS-treated groups. Nearly half of the rats in the i.p. LPS-treated groups died during the first several days after birth. In all groups, no further mortality occurred beyond P7. Gray solid line, V+V; gray dashed line, LPS+V; black dashed line, V+LPS; black solid line, LPS+LPS. Group abbreviation: V+V, i.a. vehicle followed by i.p. vehicle-treated group; LPS+V, i.a. LPS followed by i.p. vehicle-treated group; V+LPS, i.a. vehicle followed by i.p. LPS-treated group; LPS+LPS, i.a. LPS followed by i.p. LPS-treated group. The arrowhead indicates i.a. LPS (1.0 μg/sac), and the arrows indicate postnatal i.p. LPS (0.25 mg/kg/d) administrations.
Figure 3
Figure 3
Bronchoalveolar lavage fluid and peripheral blood white blood cell and neutrophil counts. (a, b, d, e) Gray bars denote P7 and black bars P14. (a) The total BALF WBC counts were higher in the i.p. LPS-treated groups than in the i.p. vehicle-treated groups on P7 and P14. (b) The V+LPS group had higher BALF neutrophil counts than in other groups on P7 and in the V+V group on P14. (c) Representative light microscopic images of the cytospin BALF samples on P7. A marked increase in the number of neutrophils in the BALF cytospin sample from the V+LPS group is observed compared with the other groups. The arrows indicate neutrophils. Diff-Quik (Sysmex, Kobe, Japan) stained. Original magnification ×400. The bar indicates 50 μm. (d, e) The total PB WBC and neutrophil counts were highest in the V+LPS group on P7. On P14, the total WBC and neutrophil counts in PB were reduced compared with P7, and the V+V group had the highest number of total WBC and neutrophil counts in PB. Group abbreviation: V+V, i.a. vehicle followed by i.p. vehicle-treated group; LPS+V, i.a. LPS followed by i.p. vehicle-treated group; V+LPS, i.a. vehicle followed by i.p. LPS-treated group; LPS+LPS, i.a. LPS followed by i.p. LPS-treated group. The data are the mean ± SEM (N = 5–6 in each group). *P < 0.05 vs. V+V, ** P < 0.05 vs. LPS+V, †P < 0.05 vs. LPS+LPS, ‡P < 0.05 vs. V+LPS. BALF, bronchoalveolar lavage fluid; PB, peripheral blood; V, vehicle; WBC, white blood cell.
Figure 4
Figure 4
Alveolar development and morphometric data. (a) Representative light microscopic images of the rat lungs on P7 and P14. Apparently thinned septal walls were observed in the V+LPS group compared with other groups on P7, and markedly large and simple airspaces were observed in the V+LPS group compared with other groups on P14. Hematoxylin and eosin stained; original magnification ×100. Bars, 200 μm. (b–d) Gray bars denote P7 and black bars P14. (b) The mean cord length (Lm), indicating the average alveolar size, was greater in the i.p. LPS-treated groups than in the i.p. vehicle-treated groups on P7. The Lm was decreased on P14 as the lungs developed in all groups, but V+LPS group had the highest Lm among the groups. (c) The alveolar surface area (SA) was smaller in the V+LPS group than in the i.p. vehicle-treated groups on P7. The SA increased on P14 as the lungs developed in all groups, but the i.p. LPS-treated groups had a smaller SA than the i.p. vehicle-treated groups. (d) The alveolar wall thickness (WT) was thinner in the i.p. LPS-treated groups than in the V+V group on P7. The WT was thinned on P14 as the lungs developed in all groups except the V+LPS group, and the V+LPS group had the thickest WT on P14. Group abbreviation: V+V, i.a. vehicle followed by i.p. vehicle-treated group; LPS+V, i.a. LPS followed by i.p. vehicle-treated group; V+LPS, i.a. vehicle followed by i.p. LPS-treated group; LPS+LPS, i.a. LPS followed by i.p. LPS-treated group. The data are the mean ± SEM (N = 6–7 in each group). *P <0.05 vs. V+V, **P < 0.05 vs. LPS+V, †P < 0.05 vs. LPS+LPS. V, vehicle.
Figure 5
Figure 5
Pulmonary vascular development. (a) Representative light microscopic images of von Willebrand factor (vWF) immunohistochemistry of rat lung sections on P7 and P14. Apparently fewer blood vessels and decreased vWF staining were observed in the V+LPS group compared with the other groups on P7 and P14. The arrows indicate vessels. vWF staining was visualized with a DAB reaction (brown color). Light hematoxylin and eosin staining was used as a counterstain. Original magnification, ×400. Bars = 50 μm. (b, c) Gray bars denote P7 and black bars P14. (b) The number of blood vessels per HPF was significantly lower in the i.p. LPS-treated groups than in the i.p. vehicle-treated groups on P7 and P14. On P14, the V+LPS group had a significantly lower number of vessels per HPF than the LPS+LPS group. (c) The vascular density, which was expressed as the ratio of the vWF-positive area to the total area of the lung parenchyma, was significantly lower in the i.p. LPS-treated groups than in the i.p. vehicle-treated groups on P7 and P14. The V+LPS group had a significantly lower vascular density than the LPS+LPS group on P7 and P14. Group abbreviation: V+V, i.a. vehicle followed by i.p. vehicle-treated group; LPS+V, i.a. LPS followed by i.p. vehicle-treated group; V+LPS, i.a. vehicle followed by i.p. LPS-treated group; LPS+LPS, i.a. LPS followed by i.p. LPS-treated group. The data are the mean ± SEM (N = 6–7 in each group). *P < 0.05 vs. V+V, **P < 0.05 vs. LPS+V; †P < 0.05 vs. LPS+LPS. HPF, high-powered field; V, vehicle. DAB, diaminobenzidine.
Figure 6
Figure 6
Inflammatory cytokine and angiogenic factor mRNA expression in lung tissue. The transcript levels of proinflammatory cytokines (TNF-α, IL-1β, IL-6, IL-12A) and angiogenic factors (VEGF, HIF-1α) on (a) P7 (b) and P14. The transcript levels of TNF-α, IL-1β, IL-6, IL-12A, VEGF, and HIF-1α in the lung tissue were significantly higher in the V+LPS group than in the V+V group on P7. The transcript levels of TNF-α, IL-12A, VEGF remained significantly higher in the V+LPS group than in the V+V group on P14. The mRNA expression of each group is presented as the relative value to that of the V+V group. Black bars, V+V; dark gray bars, LPS+V; gray bars, V+LPS; light gray bars, LPS+LPS. Group abbreviation: V+V, i.a. vehicle followed by i.p. vehicle-treated group; LPS+V, i.a. LPS followed by i.p. vehicle-treated group; V+LPS, i.a. vehicle followed by i.p. LPS-treated group; LPS+LPS, i.a. LPS followed by i.p. LPS-treated group. The data are the mean ± SEM (N = 6–7 in each group). *P < 0.05 vs. V+V. HIF, hypoxia-inducible factor.

References

    1. Smith VC, Zupancic JA, McCormick MC, et al. Trends in severe bronchopulmonary dysplasia rates between 1994 and 2002. J Pediatr 2005;146:469–73.
    1. Jobe AH. The new BPD: an arrest of lung development. Pediatr Res 1999;46:641–3.
    1. Rojas MA, Gonzalez A, Bancalari E, Claure N, Poole C, Silva-Neto G. Changing trends in the epidemiology and pathogenesis of neonatal chronic lung disease. J Pediatr 1995;126:605–10.
    1. Van Marter LJ. Epidemiology of bronchopulmonary dysplasia. In: Abman SH, ed. Bronchopulmonary Dysplasia. New York: Informa Healthcare, 2010:223–66.
    1. Charafeddine L, D'Angio CT, Phelps DL. Atypical chronic lung disease patterns in neonates. Pediatrics 1999;103:759–65.
    1. Husain AN, Siddiqui NH, Stocker JT. Pathology of arrested acinar development in postsurfactant bronchopulmonary dysplasia. Hum Pathol 1998;29:710–7.
    1. Speer CP. Inflammation and bronchopulmonary dysplasia: a continuing story. Semin Fetal Neonatal Med 2006;11:354–62.
    1. Wright CJ, Kirpalani H. Targeting inflammation to prevent bronchopulmonary dysplasia: can new insights be translated into therapies? Pediatrics 2011;128:111–26.
    1. Askie LM, Henderson-Smart DJ, Irwig L, Simpson JM. Oxygen-saturation targets and outcomes in extremely preterm infants. N Engl J Med 2003;349:959–67.
    1. Mokres LM, Parai K, Hilgendorff A, et al. Prolonged mechanical ventilation with air induces apoptosis and causes failure of alveolar septation and angiogenesis in lungs of newborn mice. Am J Physiol Lung Cell Mol Physiol 2010;298:L23–35.
    1. Berger J, Bhandari V. Animal models of bronchopulmonary dysplasia. The term mouse models. Am J Physiol Lung Cell Mol Physiol 2014;307:L936–47.
    1. O'Reilly M, Thébaud B. Animal models of bronchopulmonary dysplasia. The term rat models. Am J Physiol Lung Cell Mol Physiol 2014;307:L948–58.
    1. Bhandari V. Postnatal inflammation in the pathogenesis of bronchopulmonary dysplasia. Birth Defects Res A Clin Mol Teratol 2014;100:189–201.
    1. Choi CW, Kim BI, Hong JS, Kim EK, Kim HS, Choi JH. Bronchopulmonary dysplasia in a rat model induced by intra-amniotic inflammation and postnatal hyperoxia: morphometric aspects. Pediatr Res 2009;65:323–7.
    1. Wilson TC, Bachurski CJ, Ikegami M, Jobe AH, Kallapur SG. Pulmonary and systemic induction of SAA3 after ventilation and endotoxin in preterm lambs. Pediatr Res 2005;58:1204–9.
    1. Kroon AA, Wang J, Huang Z, Cao L, Kuliszewski M, Post M. Inflammatory response to oxygen and endotoxin in newborn rat lung ventilated with low tidal volume. Pediatr Res 2010;68:63–9.
    1. Trummer-Menzi E, Gremlich S, Schittny JC, et al. Evolution of gene expression changes in newborn rats after mechanical ventilation with reversible intubation. Pediatr Pulmonol 2012;47:1204–14.
    1. Schittny JC, Burri PH. Development and growth of the lung. In: Fishman AP, Elias JA, Fishman JA, Grippi MA, Senior RM, Pack AI, eds. Fishman's Pulmonary Diseases and Disorders. 4th edn. New York: McGraw-Hill, 2008:91–114.
    1. Romero A, Villamayor F, Grau MT, Sacristán A, Ortiz JA. Relationship between fetal weight and litter size in rats: application to reproductive toxicology studies. Reprod Toxicol 1992;6:453–6.
    1. Kallapur SG, Jobe AH, Ball MK, et al. Pulmonary and systemic endotoxin tolerance in preterm fetal sheep exposed to chorioamnionitis. J Immunol 2007;179:8491–9.
    1. Jobe AH, Newnham JP, Willet KE, et al. Endotoxin-induced lung maturation in preterm lambs is not mediated by cortisol. Am J Respir Crit Care Med 2000;162:1656–61.
    1. Kramer BW, Ikegami M, Moss TJ, Nitsos I, Newnham JP, Jobe AH. Endotoxin-induced chorioamnionitis modulates innate immunity of monocytes in preterm sheep. Am J Respir Crit Care Med 2005;171:73–7.
    1. Kramer BW, Kallapur SG, Moss TJ, Nitsos I, Newnham JP, Jobe AH. Intra-amniotic LPS modulation of TLR signaling in lung and blood monocytes of fetal sheep. Innate Immun 2009;15:101–7.
    1. Willet KE, Jobe AH, Ikegami M, Newnham J, Brennan S, Sly PD. Antenatal endotoxin and glucocorticoid effects on lung morphometry in preterm lambs. Pediatr Res 2000;48:782–8.
    1. Kallapur SG, Nitsos I, Moss TJ, et al. Chronic endotoxin exposure does not cause sustained structural abnormalities in the fetal sheep lungs. Am J Physiol Lung Cell Mol Physiol 2005;288:L966–74.
    1. Benjamin JT, Carver BJ, Plosa EJ, et al. NF-kappaB activation limits airway branching through inhibition of Sp1-mediated fibroblast growth factor-10 expression. J Immunol 2010;185:4896–903.
    1. Pollet I, Opina CJ, Zimmerman C, Leong KG, Wong F, Karsan A. Bacterial lipopolysaccharide directly induces angiogenesis through TRAF6-mediated activation of NF-kappaB and c-Jun N-terminal kinase. Blood 2003;102:1740–2.
    1. Stoll BJ, Hansen N, Fanaroff AA, et al. Changes in pathogens causing early-onset sepsis in very-low-birth-weight infants. N Engl J Med 2002;347:240–7.
    1. Stoll BJ, Hansen N, Fanaroff AA, et al. Late-onset sepsis in very low birth weight neonates: the experience of the NICHD Neonatal Research Network. Pediatrics 2002;110:285–91.
    1. Lahra MM, Beeby PJ, Jeffery HE. Intrauterine inflammation, neonatal sepsis, and chronic lung disease: a 13-year hospital cohort study. Pediatrics 2009;123:1314–9.
    1. Gisslen T, Hillman NH, Musk GC, et al. Repeated exposure to intra-amniotic LPS partially protects against adverse effects of intravenous LPS in preterm lambs. Innate Immun 2014;20:214–24.
    1. Lee HJ, Lee YJ, Jo HS, et al. Double exposure to intra-amniotic lipopolysaccharide and maternal betamethasone induces sustained increase of neutrophils in the lungs and disrupts alveolarization in newborn rats. J Perinat Med 2013;41:711–8.
    1. Lee HJ, Lee YJ, Choi CW, et al. Rosiglitazone, a peroxisome proliferator-activated receptor-γ agonist, restores alveolar and pulmonary vascular development in a rat model of bronchopulmonary dysplasia. Yonsei Med J 2014;55:99–106.

Source: PubMed

3
Abonnieren