Arginase 2 deficiency prevents oxidative stress and limits hyperoxia-induced retinal vascular degeneration

Jutamas Suwanpradid, Modesto Rojas, M Ali Behzadian, R William Caldwell, Ruth B Caldwell, Jutamas Suwanpradid, Modesto Rojas, M Ali Behzadian, R William Caldwell, Ruth B Caldwell

Abstract

Background: Hyperoxia exposure of premature infants causes obliteration of the immature retinal microvessels, leading to a condition of proliferative vitreoretinal neovascularization termed retinopathy of prematurity (ROP). Previous work has demonstrated that the hyperoxia-induced vascular injury is mediated by dysfunction of endothelial nitric oxide synthase resulting in peroxynitrite formation. This study was undertaken to determine the involvement of the ureahydrolase enzyme arginase in this pathology.

Methods and findings: Studies were performed using hyperoxia-treated bovine retinal endothelial cells (BRE) and mice with oxygen-induced retinopathy (OIR) as experimental models of ROP. Treatment with the specific arginase inhibitor 2(S)-amino-6-boronohexanoic acid (ABH) prevented hyperoxia-induced apoptosis of BRE cells and reduced vaso-obliteration in the OIR model. Furthermore, deletion of the arginase 2 gene protected against hyperoxia-induced vaso-obliteration, enhanced physiological vascular repair, and reduced retinal neovascularization in the OIR model. Additional deletion of one copy of arginase 1 did not improve the vascular pathology. Analyses of peroxynitrite by quantitation of its biomarker nitrotyrosine, superoxide by dihydroethidium imaging and NO formation by diaminofluoroscein imaging showed that the protective actions of arginase 2 deletion were associated with blockade of superoxide and peroxynitrite formation and normalization of NOS activity.

Conclusions: Our data demonstrate the involvement of arginase activity and arginase 2 expression in hyperoxia-induced vascular injury. Arginase 2 deletion prevents hyperoxia-induced retinal vascular injury by preventing NOS uncoupling resulting in decreased reactive oxygen species formation and increased nitric oxide bioavailability.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Inhibiting arginase inhibits hyperoxia-induced cell…
Figure 1. Inhibiting arginase inhibits hyperoxia-induced cell death and preserves nitrite accumulation in vitro and vaso-obliteration in vivo.
For in vitro studies, BRECs were treated with hyperoxia (40% O2, 5% CO2) or normoxia (21% O2, 5% CO2) with and without ABH (100 µM) for 48 hours. Groups of cells were stained with propidium iodide and the percentage of apoptotic nuclei (hypodiploid, M1) and normal nuclei (diploid, M2) were quantified by flow cytometry (A, B). n = 4–5, * P≤0.05 vs normoxia and hyperoxia treated with ABH, # P≤0.05 vs hyperoxic and normoxic treatment. NO release was detected using chemiluminescence to measure nitrite levels in conditioned media from the treated cells (C. n = 6–9. * P≤0.05 vs normoxia with or without ABH, # P≤0.05 vs hyperoxia without ABH. For in vivo analyses, wild type mice were treated with daily i.p. injections of vehicle (saline) or ABH (15 mg/kg) from P7 to P9 or P12. Retinal vessels were visualized by lectin labeling (D) and the area of capillary dropout (yellow) was quantified in fluorescence micrographs using ImageJ (E). n = 7–9, *P≤0.05 vs saline.
Figure 2. Arginase 2 deletion limits hyperoxia-induced…
Figure 2. Arginase 2 deletion limits hyperoxia-induced retinal vaso-obliteration.
Wild type (WT), arginase 2-deficient mice (A1+/+A2−/−) and arginase-deficient mice lacking one copy of arginase 1 (A1+/−A2−/−) were placed in 70% oxygen on P7 and prepared for analysis on P8, P9 or P12. Retinal vessels were visualized by lectin labeling (A) and the area of capillary dropout (yellow) was quantified in fluorescence micrographs using ImageJ (B, C). n = 5–9, *P≤0.05 vs WT. Images of hematoxylin and eosin stained cryostat sections from adult mice show comparable retinal morphology in WT, A2−/− and A1+/−A2−/− retinas (D, GCL: ganglion cell layer, IPL: inner plexiform layer, INL: inner nuclear layer, OPL: outer plexiform layer, ONL: outer nuclear layer, RPE: retinal pigment epithelium, scale bar  = 50 µm).
Figure 3. Arginase 2 deletion limits pathological…
Figure 3. Arginase 2 deletion limits pathological vitreo-retina neovascularization while enhancing intra-retinal neovascularization.
Wild type (WT), arginase 2-deficient mice (A1+/+A2−/−) and arginase 2-deficient mice lacking one copy of arginase 1 (A1+/−A2−/−) were maintained in 70% oxygen from P7 to P12, returned to normoxia for 5 days and prepared for analysis on P17. Retinal vessels were visualized by lectin labeling (A) and areas of vitreoretinal neovascular tufts (arrows) and capillary dropout (yellow) were quantified in fluorescence micrographs using ImageJ (B,C). n = 11–13, *P≤0.05 vs WT and A1+/−A2−/− in B and WT in C.
Figure 4. Hyperoxia treatment and Arginase 2…
Figure 4. Hyperoxia treatment and Arginase 2 expression in OIR retina.
Wild type mice (WT) and mice wild type for arginase 1 and deficient in arginase 2 (A2−/−) were placed in 70% oxygen (OIR) or room air (RA) on P7 and prepared for analysis on after 24 (A) or 48 hr (B). Immunofluorescence imaging (A) of arginase 2 and calbindin in retinal cryosections shows that arginase 2 (red) is highly expressed in horizontal cells (green). Scale bar  = 50 µm, n = 3. Western blot analysis (B) showing arginase 1 and 2 levels following normoxia and hyperoxia treatment, n = 5.
Figure 5. Arginase 2 deletion prevents hyperoxia-induced…
Figure 5. Arginase 2 deletion prevents hyperoxia-induced increases in retinal nitrotyrosine (A) and superoxide (C) levels and preserves NO formation (E).
Wild type (WT) and mice wild type for arginase 1 and deficient in arginase 2 (A2−/−) were placed in 70% oxygen (OIR) or room air (RA) on P7 and prepared for analysis on P8. Nitrotyrosine levels in retina samples were detected by slot blot (A) and quantified using ImageJ software (B). n = 6, *P≤0.05 vs WT-RA, # P≤0.05 vs WT-OIR. DHE imaging of superoxide formation was performed using flash frozen retinal sections (C). Results were quantified using Metamorph Imaging System (D). n = 3–5, scale bar  = 50 µm, *P≤0.05 WT-OIR vs all groups; # = P≤0.05 vs WT-OIR. † P

References

    1. Premature Babies. U.S. National Library of Medicine (2013)
    1. Fulton AB, Hansen RM, Moskowitz A, Akula JD (2009) The neurovascular retina in retinopathy of prematurity. Prog Retin Eye Res 28: 452–482.
    1. Ashton N, Ward B, Serpell G (1954) Effect of oxygen on developing retinal vessels with particular reference to the problem of retrolental fibroplasia. Br J Ophthalmol 38: 397–432.
    1. Smith LE (2008) Through the eyes of a child: understanding retinopathy through ROP the Friedenwald lecture. Invest Ophthalmol Vis Sci 49: 5177–5182.
    1. Smith LE, Wesolowski E, McLellan A, Kostyk SK, D'Amato R, et al. (1994) Oxygen-induced retinopathy in the mouse. Invest Ophthalmol Vis Sci 35: 101–111.
    1. Wu G, Jaeger LA, Bazer FW, Rhoads JM (2004) Arginine deficiency in preterm infants: biochemical mechanisms and nutritional implications. J Nutr Biochem 15: 442–451.
    1. Neu J, Afzal A, Pan H, Gallego E, Li N, et al. (2006) The dipeptide Arg-Gln inhibits retinal neovascularization in the mouse model of oxygen-induced retinopathy. Invest Ophthalmol Vis Sci 47: 3151–3155.
    1. Zhang W, Baban B, Rojas M, Tofigh S, Virmani SK, et al. (2009) Arginase activity mediates retinal inflammation in endotoxin-induced uveitis. Am JPathol 175: 891–902.
    1. Jenkinson CP, Grody WW, Cederbaum SD (1996) Comparative properties of arginases. Comp Biochem Physiol B Biochem Mol Biol 114: 107–132.
    1. Kielczewski JL, Jarajapu YP, McFarland EL, Cai J, Afzal A, et al. (2009) Insulin-like growth factor binding protein-3 mediates vascular repair by enhancing nitric oxide generation. Circ Res 105: 897–905.
    1. Hardy P, Dumont I, Bhattacharya M, Hou X, Lachapelle P, et al. (2000) Oxidants, nitric oxide and prostanoids in the developing ocular vasculature: a basis for ischemic retinopathy. Cardiovasc Res 47: 489–509.
    1. Sessa WC (2009) Molecular control of blood flow and angiogenesis: role of nitric oxide. J Thromb Haemost 7 Suppl 1 35–37.
    1. Brooks SE, Gu X, Samuel S, Marcus DM, Bartoli M, et al. (2001) Reduced severity of oxygen-induced retinopathy in eNOS-deficient mice. Invest Ophthalmol Vis Sci 42: 222–228.
    1. Gu X, El-Remessy AB, Brooks SE, Al-Shabrawey M, Tsai NT, et al. (2003) Hyperoxia induces retinal vascular endothelial cell apoptosis through formation of peroxynitrite. Am J Physiol Cell Physiol 285: C546–554.
    1. Romero MJ, Platt DH, Tawfik HE, Labazi M, El-Remessy AB, et al. (2008) Diabetes-induced coronary vascular dysfunction involves increased arginase activity. Circ Res 102: 95–102.
    1. Romero MJ, Iddings JA, Platt DH, Ali MI, Cederbaum SD, et al. (2012) Diabetes-induced vascular dysfunction involves arginase I. Am J Physiol Heart Circ Physiol 302: H159–166.
    1. Narayanan SP, Suwanpradid J, Saul A, Xu Z, Still A, et al. (2011) Arginase 2 deletion reduces neuro-glial injury and improves retinal function in a model of retinopathy of prematurity. PLoS One 6: e22460.
    1. Narayanan SP, Xu Z, Putluri N, Sreekumar A, Lemtalsi T, et al. (2014) Arginase 2 deficiency reduces hyperoxia-mediated retinal neurodegeneration through the regulation of polyamine metabolism. Cell Death Dis 5: e1075.
    1. Morris SM Jr (2009) Recent advances in arginine metabolism: roles and regulation of the arginases. Br J Pharmacol 157: 922–930.
    1. Christianson DW (2005) Arginase: structure, mechanism, and physiological role in male and female sexual arousal. Acc Chem Res 38: 191–201.
    1. Durante W, Liao L, Reyna SV, Peyton KJ, Schafer AI (2001) Transforming growth factor-beta(1) stimulates L-arginine transport and metabolism in vascular smooth muscle cells: role in polyamine and collagen synthesis. Circulation 103: 1121–1127.
    1. Xia Y, Dawson VL, Dawson TM, Snyder SH, Zweier JL (1996) Nitric oxide synthase generates superoxide and nitric oxide in arginine-depleted cells leading to peroxynitrite-mediated cellular injury. Proc Natl Acad Sci U S A 93: 6770–6774.
    1. Beckman JS, Koppenol WH (1996) Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly. Am J Physiol 271: C1424–1437.
    1. Gow AJ, Duran D, Malcolm S, Ischiropoulos H (1996) Effects of peroxynitrite-induced protein modifications on tyrosine phosphorylation and degradation. FEBS Lett 385: 63–66.
    1. Zhao H, Joseph J, Fales HM, Sokoloski EA, Levine RL, et al. (2005) Detection and characterization of the product of hydroethidine and intracellular superoxide by HPLC and limitations of fluorescence. Proc Natl Acad Sci U S A 102: 5727–5732.
    1. Brooks SE, Gu X, Kaufmann PM, Marcus DM, Caldwell RB (1998) Modulation of VEGF production by pH and glucose in retinal Muller cells. Curr Eye Res 17: 875–882.
    1. Krotova K, Patel JM, Block ER, Zharikov S (2010) Hypoxic upregulation of arginase II in human lung endothelial cells. Am J Physiol Cell Physiol 299: C1541–1548.
    1. Shatanawi A, Romero MJ, Iddings JA, Chandra S, Umapathy NS, et al. (2011) Angiotensin II-induced vascular endothelial dysfunction through RhoA/Rho kinase/p38 mitogen-activated protein kinase/arginase pathway. Am J Physiol Cell Physiol 300: C1181–1192.
    1. Chandra S, Romero MJ, Shatanawi A, Alkilany AM, Caldwell RB, et al. (2012) Oxidative species increase arginase activity in endothelial cells through the RhoA/Rho kinase pathway. Br J Pharmacol 165: 506–519.
    1. Pourcet B, Feig JE, Vengrenyuk Y, Hobbs AJ, Kepka-Lenhart D, et al. (2011) LXRalpha regulates macrophage arginase 1 through PU.1 and interferon regulatory factor 8. Circ Res 109: 492–501.
    1. Pandey D, Sikka G, Bergman Y, Kim JH, Ryoo S, et al. (2014) Transcriptional regulation of endothelial arginase 2 by histone deacetylase 2. Arterioscler Thromb Vasc Biol 34: 1556–1566.
    1. Sahin E, Haubenwallner S, Kuttke M, Kollmann I, Halfmann A, et al. (2014) Macrophage PTEN Regulates Expression and Secretion of Arginase I Modulating Innate and Adaptive Immune Responses. J Immunol.
    1. Stevenson L, Matesanz N, Colhoun L, Edgar K, Devine A, et al. (2010) Reduced nitro-oxidative stress and neural cell death suggests a protective role for microglial cells in TNFalpha-/- mice in ischemic retinopathy. Invest Ophthalmol Vis Sci 51: 3291–3299.
    1. Knabe W, Ochs M (1999) Horizontal cells invest retinal capillaries in the tree shrew Tupaia belangeri. Cell Tissue Res 298: 33–43.
    1. Mojumder DK (2008) Capillary-contacting horizontal cells in the rodent retina. J Anat Soc India 57: 34–36.
    1. Poche RA, Reese BE (2009) Retinal horizontal cells: challenging paradigms of neural development and cancer biology. Development 136: 2141–2151.
    1. Narayanan SP, Rojas M, Suwanpradid J, Toque HA, Caldwell RW, et al. (2013) Arginase in Retinopathy. Prog Retin Eye Res.
    1. Ali NK, Jafri A, Sopi RB, Prakash YS, Martin RJ, et al. (2012) Role of arginase in impairing relaxation of lung parenchyma of hyperoxia-exposed neonatal rats. Neonatology 101: 106–115.
    1. Ryoo S, Lemmon CA, Soucy KG, Gupta G, White AR, et al. (2006) Oxidized low-density lipoprotein-dependent endothelial arginase II activation contributes to impaired nitric oxide signaling. Circ Res 99: 951–960.
    1. Ryoo S, Gupta G, Benjo A, Lim HK, Camara A, et al. (2008) Endothelial arginase II: a novel target for the treatment of atherosclerosis. Circ Res 102: 923–932.
    1. Shin WS, Berkowitz DE, Ryoo SW (2012) Increased arginase II activity contributes to endothelial dysfunction through endothelial nitric oxide synthase uncoupling in aged mice. Exp Mol Med 44: 594–602.
    1. Elms SC, Toque HA, Rojas M, Xu Z, Caldwell RW, et al. (2013) The role of arginase I in diabetes-induced retinal vascular dysfunction in mouse and rat models of diabetes. Diabetologia 56: 654–662.
    1. Patel C, Rojas M, Narayanan SP, Zhang W, Xu Z, et al. (2013) Arginase as a mediator of diabetic retinopathy. Front Immunol 4: 173.
    1. Que LG, Kantrow SP, Jenkinson CP, Piantadosi CA, Huang YC (1998) Induction of arginase isoforms in the lung during hyperoxia. Am J Physiol 275: L96–102.
    1. Forstermann U, Sessa WC (2012) Nitric oxide synthases: regulation and function. Eur Heart J 33: 829–837, 837a–837d.
    1. Edgar K, Gardiner TA, van Haperen R, de Crom R, McDonald DM (2012) eNOS overexpression exacerbates vascular closure in the obliterative phase of OIR and increases angiogenic drive in the subsequent proliferative stage. Invest Ophthalmol Vis Sci 53: 6833–6850.
    1. Feng Y, Venema VJ, Venema RC, Tsai N, Behzadian MA, et al. (1999) VEGF-induced permeability increase is mediated by caveolae. Invest Ophthalmol Vis Sci 40: 157–167.
    1. Riccardi C, Nicoletti I (2006) Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat Protoc 1: 1458–1461.
    1. Jagnandan D, Sessa WC, Fulton D (2005) Intracellular location regulates calcium-calmodulin-dependent activation of organelle-restricted eNOS. Am J Physiol Cell Physiol 289: C1024–1033.
    1. Deignan JL, Livesay JC, Yoo PK, Goodman SI, O'Brien WE, et al. (2006) Ornithine deficiency in the arginase double knockout mouse. Mol Genet Metab 89: 87–96.
    1. Shi O, Morris SM Jr, Zoghbi H, Porter CW, O'Brien WE (2001) Generation of a mouse model for arginase II deficiency by targeted disruption of the arginase II gene. Mol Cell Biol 21: 811–813.
    1. Connor KM, Krah NM, Dennison RJ, Aderman CM, Chen J, et al. (2009) Quantification of oxygen-induced retinopathy in the mouse: a model of vessel loss, vessel regrowth and pathological angiogenesis. Nat Protoc 4: 1565–1573.

Source: PubMed

3
Iratkozz fel