Hyperoxia causes angiopoietin 2-mediated acute lung injury and necrotic cell death

Vineet Bhandari, Rayman Choo-Wing, Chun G Lee, Zhou Zhu, Jonathan H Nedrelow, Geoffrey L Chupp, Xucher Zhang, Michael A Matthay, Lorraine B Ware, Robert J Homer, Patty J Lee, Anke Geick, Antonin R de Fougerolles, Jack A Elias, Vineet Bhandari, Rayman Choo-Wing, Chun G Lee, Zhou Zhu, Jonathan H Nedrelow, Geoffrey L Chupp, Xucher Zhang, Michael A Matthay, Lorraine B Ware, Robert J Homer, Patty J Lee, Anke Geick, Antonin R de Fougerolles, Jack A Elias

Abstract

The angiogenic growth factor angiopoietin 2 (Ang2) destabilizes blood vessels, enhances vascular leak and induces vascular regression and endothelial cell apoptosis. We considered that Ang2 might be important in hyperoxic acute lung injury (ALI). Here we have characterized the responses in lungs induced by hyperoxia in wild-type and Ang2-/- mice or those given either recombinant Ang2 or short interfering RNA (siRNA) targeted to Ang2. During hyperoxia Ang2 expression is induced in lung epithelial cells, while hyperoxia-induced oxidant injury, cell death, inflammation, permeability alterations and mortality are ameliorated in Ang2-/- and siRNA-treated mice. Hyperoxia induces and activates the extrinsic and mitochondrial cell death pathways and activates initiator and effector caspases through Ang2-dependent pathways in vivo. Ang2 increases inflammation and cell death during hyperoxia in vivo and stimulates epithelial necrosis in hyperoxia in vitro. Ang2 in plasma and alveolar edema fluid is increased in adults with ALI and pulmonary edema. Tracheal Ang2 is also increased in neonates that develop bronchopulmonary dysplasia. Ang2 is thus a mediator of epithelial necrosis with an important role in hyperoxic ALI and pulmonary edema.

Figures

Figure 1
Figure 1
Effect of hyperoxia on Ang-1, Ang-2 and Tie 2. Mice were exposed to room air (RA) or 100% O2 for up to 72 h. (a–c) Pulmonary Ang and Tie2 mRNA (a), pulmonary and extrapulmonary organ Ang mRNA (b) and BAL fluid Ang2 protein (c) were assessed by RT-PCR or western blotting. (d) In situ hybridization was used to localize Ang2 mRNA in the airways and parenchyma of mice exposed to room air or 100% O2 for 72 h. Arrows highlight airway epithelial cells (top) and type II cells (bottom). (e,f) Immunohistochemical analysis was used to localize Ang2 protein in mice exposed to room air or 100% O2 for 72 h. Single labeling experiments highlight prominent staining in airway and alveolar epithelial cells (e). Double labeling experiments highlight staining with anti-Ang2 (red) and anti–SP-C (green) antibodies (f). Arrow indicates a cell labeled with both antibodies. Original magnification, ×40 (d–f).
Figure 2
Figure 2
Effect of hyperoxia on survival, inflammation, permeability and tissue injury. Wild-type (Ang2+/+), Ang2+/– and Ang2–/– mice were exposed to 100% O2, and survival (a), BAL total cell recovery (b), differential cell recovery (c), BAL protein (d) and lung tissue injury (assessed by light microscopy, hematoxylin and eosin stain; e) were assessed. In b–e, mice were exposed to room air or 100% O2 for 72 h. Data represent assessments in a minimum of n = 8 mice. Original magnification, ×40 (e). *P < 0.01, #P ≤ 0.02, **P = 0.03, ##P < 0.04.
Figure 3
Figure 3
Role of Ang2 in hyperoxia-induced oxidant and DNA injury. (a,b) Wild-type and Ang2–/– mice were exposed to room air (–) or 100% O2 (+) for 72 h, and subjected to both staining for 8-OHdG (a) and TUNEL evaluation (b). (c) Percentage of TUNEL-positive cells. Original magnification, ×10 (a, b, top); ×40 (a, b, bottom). *P < 0.0001, **P < 0.03.
Figure 4
Figure 4
Effects of rAng2 and Ang2 siRNA on hyperoxia-induced responses. (a–c) Wild-type mice were exposed to 100% O2 and were randomized to receive rAng2 or vehicle control. After 72 h in hyperoxia, vascular congestion (a), BAL cellularity (b) and TUNEL-positive cell death (c) were evaluated. (d–g) Mice were randomized to receive an Ang2 or control (scrambled) siRNA, and Ang2 mRNA (d,e), BAL cellularity (f) and TUNEL-positive cell death (g) were evaluated. Two doses of Ang2 siRNA, low (LD) and high (HD), were used (Methods). Data represent assessments in a minimum of n = 5 mice. Original magnification, ×40 (a). *P < 0.0001, **P ≤ 0.01, #P ≤ 0.02, ##P < 0.03.
Figure 5
Figure 5
Role of Ang2 in hyperoxia-induced alterations in apoptosis and angiogenic regulators and the effect of rAng2 on MLE-12 cell survival. (a–d) Wild-type, Ang2+/– and Ang2–/– mice were exposed to room air or 100% O2 for 72 h, and the indicated mRNAs (a) and caspase-3, caspase-8 and caspase-9 bioactivity (b–d) were assessed. (e,f) MLE-12 cells were cultured for up to 48 h in 5% CO2 and air, or 95% O2 in the presence and absence of the indicated concentration of rAng2. (e) Apoptosis and necrosis were evaluated by Annexin V and propidium iodide (PI) staining and expressed as a percentage of the total cell number. (f) Percentage of cells undergoing pure necrosis (PI-positive, Annexin V–negative). *P < 0.01, #P ≤ 0.02, **P ≤ 0.03, ##P < 0.05.
Figure 6
Figure 6
Ang2 in biological fluids from human adults and neonates. (a) Concentration of Ang2 in the plasma and undiluted AEF of individuals with ALI and healthy controls. Con, healthy adults, Hyd, adults with hydrostatic edema; ALI, adults with acute lung injury. n = 3–4 per group; *P < 0.0001, #P = 0.005, **P = 0.02, ##P = 0.05. (b) Concentration of Ang2 in the tracheal aspirate of premature babies affected with RDS with and without an adverse outcome (bronchopulmonary dysplasia and/or death). n = 5–9 per group; *P < 0.01.

References

    1. Barazzone C, Horowitz S, Donati YR, Rodriguez I, Piguet PF. Oxygen toxicity in mouse lung: pathways to cell death. Am. J. Respir. Cell Mol. Biol. 1998;19:573–581.
    1. Barazzone C, White CW. Mechanisms of cell injury and death in hyperoxia: role of cytokines and Bcl-2 family proteins. Am. J. Respir. Cell Mol. Biol. 2000;22:517–519.
    1. Crapo JD. Morphologic changes in pulmonary oxygen toxicity. Annu. Rev. Physiol. 1986;48:721–731.
    1. O'Reilly MA, et al. Bcl-2 family gene expression during severe hyperoxia induced lung injury. Lab. Invest. 2000;80:1845–1854.
    1. Ward NS, et al. Interleukin-6-induced protection in hyperoxic acute lung injury. Am. J. Respir. Cell Mol. Biol. 2000;22:535–542.
    1. Waxman AB, et al. Targeted lung expression of interleukin-11 enhances murine tolerance of 100% oxygen and diminishes hyperoxia-induced DNA fragmentation. J. Clin. Invest. 1998;101:1970–1982.
    1. Mantell LL, Horowitz S, Davis JM, Kazzaz JA. Hyperoxia-induced cell death in the lung—the correlation of apoptosis, necrosis, and inflammation. Ann. NY Acad. Sci. 1999;887:171–180.
    1. Carmeliet P. Angiogenesis in health and disease. Nat. Med. 2003;9:653–660.
    1. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat. Med. 2003;9:669–676.
    1. Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13:9–22.
    1. McDonald DM. Angiogenesis and remodeling of airway vasculature in chronic inflammation. Am. J. Respir. Crit. Care Med. 2001;164:S39–S45.
    1. Thurston G, et al. Angiopoietin-1 protects the adult vasculature against plasma leakage. Nat. Med. 2000;6:460–463.
    1. Thurston G, et al. Leakage-resistant blood vessels in mice transgenically overexpressing angiopoietin-1. Science. 1999;286:2511–2514.
    1. Yancopoulos GD, et al. Vascular-specific growth factors and blood vessel formation. Nature. 2000;407:242–248.
    1. Jain RK. Molecular regulation of vessel maturation. Nat. Med. 2003;9:685–693.
    1. Sandhu R, et al. Reciprocal regulation of angiopoietin-1 and angiopoietin-2 following myocardial infarction in the rat. Cardiovasc. Res. 2004;64:115–124.
    1. Tait CR, Jones PF. Angiopoietins in tumours: the angiogenic switch. J. Pathol. 2004;204:1–10.
    1. Bloch W, et al. The angiogenesis inhibitor endostatin impairs blood vessel maturation during wound healing. FASEB J. 2000;14:2373–2376.
    1. He CH, et al. Bcl-2-related protein A1 is an endogenous and cytokine-stimulated mediator of cytoprotection in hyperoxic acute lung injury. J. Clin. Invest. 2005;115:1039–1048.
    1. Gale NW, et al. Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by angiopoietin-1. Dev. Cell. 2002;3:411–423.
    1. Horowitz S. Pathways to cell death in hyperoxia. Chest. 1999;116:64S–67S.
    1. O'Reilly MA. DNA damage and cell cycle checkpoints in hyperoxic lung injury: braking to facilitate repair. Am. J. Physiol. Lung Cell. Mol. Physiol. 2001;281:L291–L305.
    1. Raffray M, Cohen GM. Apoptosis and necrosis in toxicology: a continuum or distinct modes of cell death? Pharmacol. Ther. 1997;75:153–177.
    1. Richard C, et al. Androgens modulate the balance between VEGF and angiopoietin expression in prostate epithelial and smooth muscle cells. Prostate. 2002;50:83–91.
    1. Oshima Y, et al. Angiopoietin-2 enhances retinal vessel sensitivity to vascular endothelial growth factor. J. Cell. Physiol. 2004;199:412–417.
    1. Tsigkos S, Koutsilieris M, Papapetropoulos A. Angiopoietins in angiogenesis and beyond. Expert Opin. Investig. Drugs. 2003;12:933–941.
    1. Karmpaliotis D, et al. Angiogenic growth factors in the pathophysiology of a murine model of acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 2002;283:L585–L595.
    1. Joza N, Kroemer G, Penninger JM. Genetic analysis of the mammalian cell death machinery. Trends Genet. 2002;18:142–149.
    1. Lin EY, Orlofsky A, Berger MS, Prystowsky MB. Characterization of A1, a novel hemopoietic-specific early-response gene with sequence similarity to bcl-2. J. Immunol. 1993;151:1979–1988.
    1. Werner AB, de Vries E, Tait SW, Bontjer I, Borst J. Bcl-2 family member Bfl-1/A1 sequesters truncated bid to inhibit is collaboration with pro-apoptotic Bak or Bax. J. Biol. Chem. 2002;277:22781–22788.
    1. Wang X, et al. Necrotic cell death in response to oxidant stress involves the activation of the apoptogenic caspase-8/bid pathway. J. Biol. Chem. 2003;278:29184–29191.
    1. O'Reilly MA, Staversky RJ, Watkins RH, Maniscalco WM. Accumulation of p21Cip1/WAF1 during hyperoxic lung injury in mice. Am. J. Respir. Cell Mol. Biol. 1998;19:777–785.
    1. Mitsutake N, et al. Tie-2 and angiopoietin-1 expression in human thyroid tumors. Thyroid. 2002;12:95–99.
    1. Nakayama T, et al. Expression of Tie-1 and 2 receptors, and angiopoietin-1, 2 and 4 in gastric carcinoma; immunohistochemical analyses and correlation with clinicopathological factors. Histopathology. 2004;44:232–239.
    1. Wurmbach JH, Hammerer P, Sevinc S, Huland H, Ergun S. The expression of angiopoietins and their receptor Tie-2 in human prostate carcinoma. Anticancer Res. 2000;20:5217–5220.
    1. Kim I, et al. The angiopoietin-tie2 system in coronary artery endothelium prevents oxidized low-density lipoprotein-induced apoptosis. Cardiovasc. Res. 2001;49:872–881.
    1. Lobov IB, Brooks PC, Lang RA. Angiopoietin-2 displays VEGF-dependent modulation of capillary structure and endothelial cell survival in vivo. Proc. Natl. Acad. Sci. USA. 2002;99:11205–11210.
    1. Beck H, Acker T, Wiessner C, Allegrini PR, Plate KH. Expression of angiopoietin-1, angiopoietin-2, and tie receptors after middle cerebral artery occlusion in the rat. Am. J. Pathol. 2000;157:1473–1483.
    1. Zagzag D, et al. Vascular apoptosis and involution in gliomas precede neovascularization: a novel concept for glioma growth and angiogenesis. Lab. Invest. 2000;80:837–849.
    1. Ware LB, Matthay MA. Alveolar fluid clearance is impaired in the majority of patients with acute lung injury and the acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2001;163:1376–1383.
    1. Anandarajah AP, Ritchlin CT. Pathogenesis of psoriatic arthritis. Curr. Opin. Rheumatol. 2004;16:338–343.
    1. Fearon U, et al. Angiopoietins, growth factors, and vascular morphology in early arthritis. J. Rheumatol. 2003;30:260–268.
    1. Kimura H, Mochida S, Inao M, Matsui A, Fujiwara K. Angiopoietin/tie receptors system may play a role during reconstruction and capillarization of the hepatic sinusoids after partial hepatectomy and liver necrosis in rats. Hepatol. Res. 2004;29:51–59.
    1. Brat DJ, Van Meir EG. Vaso-occlusive and prothrombotic mechanisms associated with tumor hypoxia, necrosis, and accelerated growth in glioblastoma. Lab. Invest. 2004;84:397–405.
    1. Zhu S, Ware LB, Geiser T, Matthay MA, Matalon S. Increased levels of nitrate and surfactant protein a nitration in the pulmonary edema fluid of patients with acute lung injury. Am. J. Respir. Crit. Care Med. 2001;163:166–172.
    1. Pugin J, Verghese G, Widmer MC, Matthay MA. The alveolar space is the site of intense inflammatory and profibrotic reactions in the early phase of acute respiratory distress syndrome. Crit. Care Med. 1999;27:304–312.
    1. Koga K, et al. Expression of angiopoietin-2 in human glioma cells and its role for angiogenesis. Cancer Res. 2001;61:6248–6254.
    1. Lip PL, et al. Plasma vascular endothelial growth factor, angiopoietin-2, and soluble angiopoietin receptor tie-2 in diabetic retinopathy: effects of laser photocoagulation and angiotensin receptor blockade. Br. J. Ophthalmol. 2004;88:1543–1546.
    1. Ohashi H, et al. Alterations in expression of angiopoietins and the Tie-2 receptor in the retina of streptozotocin induced diabetic rats. Mol. Vis. 2004;10:608–617.
    1. Bhandari A, Bhandari V. Pathogenesis, pathology and pathophysiology of pulmonary sequelae of bronchopulmonary dysplasia in premature infants. Front. Biosci. 2003;8:e370–e380.

Source: PubMed

3
Tilaa