Minocycline inhibits PARP‑1 expression and decreases apoptosis in diabetic retinopathy

Ying Wu, Yongdong Chen, Qiang Wu, Lili Jia, Xinhua Du, Ying Wu, Yongdong Chen, Qiang Wu, Lili Jia, Xinhua Du

Abstract

The present study aimed to investigate the mechanism underlying the effects of minocycline on diabetic retinopathy‑associated cellular apoptosis. A total of 40 Sprague Dawley (SD) rats were used as a diabetic retinopathy model following injection with streptozotocin. Among the 34 rats in which the diabetes model was successfully established, 24 rats were divided into two experimental groups: I and II (T1 and T2, respectively), and orally administered with various doses of minocycline. The remaining 10 rats served as the diabetic retinopathy control group. An additional group of 10 healthy SD rats with comparable weight served as normal controls. The rats in T1 and T2 groups were treated daily for eight consecutive weeks with minocycline at a dose of 2.5 mg/kg and 5 mg/kg, respectively. The mRNA expression levels of poly (ADP‑ribose) polymerase‑1 (PARP‑1) were subsequently measured by reverse transcription‑quantitative polymerase chain reaction, and the protein expression levels of poly‑ADP‑ribose were measured by western blot analysis and immunohistochemistry. Retinal morphology was observed following hematoxylin and eosin staining, and retinal cell apoptosis was measured by terminal deoxynucleotidyl transferase dUTP nick end labeling and caspase‑3 activity assays. The amplitudes of the electroretinogram (ERG) b‑wave and oscillary potentials (OPs) were measured using visual electrophysiology, and compared among the four groups. The results of the present study demonstrated that in the diabetic rats, retinal PARP‑1 gene expression was markedly upregulated, the number of apoptotic cells and the activity levels of caspase‑3 were increased, and the amplitude of the ERG b‑wave and the OPs were markedly lower as compared with the normal rats. Following treatment with minocycline, the abnormal expression of PARP‑1 in the retina was inhibited, and cellular apoptosis was decreased. In conclusion, the results of the present study suggest that PARP‑1 is involved in the development of diabetic retinopathy, and minocycline is able to inhibit PARP‑1 expression and decrease cellular apoptosis, suggesting that minocycline may prove to be a promising drug for the treatment of diabetic retinopathy.

Figures

Figure 1
Figure 1
mRNA expression levels of poly (ADP-ribose) polymerase-1 (PARP-1) in the retinal tissue samples of the CON, DM, T1, and T2 groups. The mRNA expression levels of PARP-1 in the retinal tissue samples of the DM rats were significantly higher, as compared with the CON group. Following minocycline treatment, the abnormally elevated mRNA expression levels of PARP-1 in the retinal tissue samples of the DM rats were suppressed (F=12.56, P=0.0021), as compared with the DM group. No statistically significant difference was observed in PARP-1 gene expression between the T1 and T2 treatment groups (P>0.05). *P<0.01 vs. the other groups; **P<0.05 vs. CON. CON, normal control group; DM, diabetic retinopathy group; T1, 2.5 mg/kg minocycline treatment group; T2, 5 mg/kg minocycline treatment group.
Figure 2
Figure 2
Expression levels of PAR proteins in the retinal tissue samples of DM rats were significantly higher, as comapred with the CON group. Following minocycline treatment, the expression levels of the PAR proteins were downregulated in the DM rats, as compared with the DM group. (A and B) Western blot analysis demonstrated a significant increase in the protein expression levels of the PAR proteins of the DM group, as compared with the other three groups, however no significant difference was observed between the CON, T1 and T2 groups (F=39.02). *P<0.01 vs. CON; **P<0.01 vs. DM. (C) PAR expression was observed in the retinal ganglion cell layer, inner nuclear layer, and outer nuclear layer, as determined by immunohistochemistry. The PAR protein staining in the retinal tissue samples of the CON group was weakly positive. (D) Positive PAR expression increased significantly in the DM rats. (E and F) Following minocycline treatment, the expression levels of the PAR proteins were downregulated in the T1 and T2 groups. Scale bars, 50 µm. CON, normal control group; DM, diabetic retinopathy group; T1, 2.5 mg/kg minocycline treatment group; T2, 5 mg/kg minocycline treatment group; PAR, poly (ADP-ribose).
Figure 3
Figure 3
The number of apoptotic cells increased significantly following 10 weeks in the DM rats, and decreased following minocycline treatment. (A) A small number of apoptotic cells were present in the rat retinal tissue samples of the CON group. (B) The number of apoptotic cells increased significantly following 10 weeks in the DM rats. The apoptotic cells were located in the GCL, INL and ONL. (C and D) Following minocycline treatment, significantly less apoptotic cells were present in the retinal tissue samples of the DM rats in the T1 and T2 groups. (E) The apoptotic index in the GCL of the DM rats was significantly higher, as compared with the CON group. Following minocycline treatment, the elevated apoptotic index in the GCL of the retinal tissue samples of the DM rats was suppressed (F=53.57; *P<0.01 vs. CON). No significant difference was observed between the GCL apoptotic index of the T1 and T2 treatment groups (**P>0.05). (F) Apoptotic index of the INL of the DM rats was significantly elevated, as compared with the CON group. Following minocycline treatment, the elevated apoptotic index in the INL of the retinal tissue samples of the DM rats was unchanged (F=28.32; *P<0.01 vs. CON). No significant difference was observed between the INL apoptotic index of the T1 and T2 treatment groups (P>0.05). (Scale bars, 25 µm). CON, control group; DM, diabetic retinopathy group; T1, 2.5 mg/kg minocycline treatment group; T2, 5 mg/kg minocycline treatment group; GCL, ganglion cell layer; ONL, outer nuclear layer; INL, inner nuclear layer.
Figure 4
Figure 4
The activity levels of caspase-3 in the retinal tissue samples of the CON, DM, T1 and T2 groups. The activity levels of caspase-3 were significantly elevated in the retinal tissue samples of the DM rats, as compared with the CON group. Following minocycline treatment, the elevated activity levels of caspase-3 in the retinal tissue samples of the DM rats was suppressed (F=27.24; *P<0.01 vs. CON and **P<0.01 vs. DM). No statistically significant difference in the activity levels of caspase-3 was observed between the T1 and T2 treatment groups (P>0.05) CON, normal control group; DM, diabetic retinopathy group; T1, 2.5 mg/kg minocycline treatment group; T2, 5 mg/kg minocycline treatment group.
Figure 5
Figure 5
OPs of the retina in the CON, DM, T1 and T2 groups. (A) OPs of the rats in the CON, DM, T1 and T2 groups. (B) Amplitude analysis of the rat OPs in the CON, DM, T1 and T2 groups. The OP amplitudes in the DM group was significantly lower, as compared with the CON group (F=10.46; P=0.0011). *P<0.01 vs. CON. Following minocycline treatment, the OP amplitude increased, but remained lower as compared with the CON group. No statistically significant difference was observed between the OP amplitudes of the T1 and T2 groups (**P>0.05). CON, normal control group; DM, diabetic retinopathy group; T1, 2.5 mg/kg minocycline treatment group; T2, 5 mg/kg minocycline treatment group; OP, oscillary potential.
Figure 6
Figure 6
B-waves of the rats in the CON, DM, T1 and T2 groups, as demonstrated by ERG. (A) B-waves of the rats in the CON, DM, T1 and T2 groups. (B) Amplitude analysis of the ERG b-waves of the rats in all four groups. The b-wave amplitude in the DM group was significantly lower, as compared with the CON group (F=32.52; *P<0.0001 vs. CON), and following minocycline treatment, the b-wave amplitude increased, but remained lower than that of the CON group. The b-waves in the T2 groups were significantly greater, as compared with the T1 group (#P<0.001 vs. T1). CON, normal control group; DM, diabetic retinopathy group; T1, 2.5 mg/kg minocycline treatment group; T2, 5 mg/kg minocycline treatment group; ERG, electroretinogram.

References

    1. Engerman RL, Kern TS. Retinopathy in animal models of diabetes. Diabetes Metab Rev. 1995;11:109–120. doi: 10.1002/dmr.5610110203.
    1. Mizutani M, Kern TS, Lorenzi M. Accelerated death of retinal microvascular cells in human and experimental diabetic retinopathy. J Clin Invest. 1996;97:2883–2890. doi: 10.1172/JCI118746.
    1. Hammes HP, Federoff HJ, Brownlee M. Nerve growth factor prevents both neuroretinal programmed cell death and capillary pathology in experimental diabetes. Mol Med. 1995;1:527–534.
    1. Barber AJ, Lieth E, Khin SA, Antonetti DA, Buchanan AG, Gardner TW. Neural apoptosis in the retina during experimental and human diabetes. Early onset and effect of insulin. J Clin Invest. 1998;102:783–791. doi: 10.1172/JCI2425.
    1. Szabó C, Ohshima H. DNA damage induced by peroxynitrite: Subsequent biological effects. Nitric Oxide. 1997;1:373–385. doi: 10.1006/niox.1997.0143.
    1. Szabó C, Dawson VL. Role of poly(ADP-ribose) synthetase in inflammation and ischaemia-reperfusion. Trends Pharmacol Sci. 1998;19:287–298. doi: 10.1016/S0165-6147(98)01193-6.
    1. Heller B, Wang ZQ, Wagner EF, Radons J, Bürkle A, Fehsel K, Burkart V, Kolb H. Inactivation of the poly(ADP-ribose) polymerase gene affects oxygen radical and nitric oxide toxicity in islet cells. J Biol Chem. 1995;270:11176–11180. doi: 10.1074/jbc.270.19.11176.
    1. Li GY, Fan B, Su GF. Acute energy reduction induces caspase-dependent apoptosis and activates p53 in retinal ganglion cells (RGC-5) Exp Eye Res. 2009;89:581–589. doi: 10.1016/j.exer.2009.06.004.
    1. Zheng L, Kern TS. Role of nitric oxide, superoxide, peroxynitrite and PARP in diabetic retinopathy. Front Biosci (Landmark Ed) 2009;14:3974–3987. doi: 10.2741/3505.
    1. Virág L, Szabó C. The therapeutic potential of poly(ADP-ribose) polymerase inhibitors. Pharmacol Rev. 2002;54:375–429. doi: 10.1124/pr.54.3.375.
    1. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414:813–820. doi: 10.1038/414813a.
    1. Yrjänheikki J, Tikka T, Keinänen R, Goldsteins G, Chan PH, Koistinaho J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci USA. 1999;96:13496–13500. doi: 10.1073/pnas.96.23.13496.
    1. Ryu JK, Franciosi S, Sattayaprasert P, Kim SU, McLarnon JG. Minocycline inhibits neuronal death and glial activation induced by beta-amyloid peptide in rat hippocampus. Glia. 2004;48:85–90. doi: 10.1002/glia.20051.
    1. Krady JK, Basu A, Allen CM, Xu Y, LaNoue KF, Gardner TW, Levison SW. Minocycline reduces proinflammatory cytokine expression, microglial activation, and caspase-3 activation in a rodent model of diabetic retinopathy. Diabetes. 2005;54:1559–1565. doi: 10.2337/diabetes.54.5.1559.
    1. Vincent JA, Mohr S. Inhibition of caspase-1/interleukin-1beta signaling prevents degeneration of retinal capillaries in diabetes and galactosemia. Diabetes. 2007;56:224–230. doi: 10.2337/db06-0427.
    1. Chang CJ, Cherng CH, Liou WS, Liao CL. Minocycline partially inhibits caspase-3 activation and photoreceptor degeneration after photic injury. Ophthalmic Res. 2005;37:202–213. doi: 10.1159/000086610.
    1. Obrosova IG, Minchenko AG, Frank RN, Seigel GM, Zsengeller Z, Pacher P, Stevens MJ, Szabó C. Poly(ADP-ribose) polymerase inhibitors counteract diabetes- and hypoxia-induced retinal vascular endothelial growth factor overexpression. Int J Mol Med. 2004;14:55–64.
    1. Santiago AR, Cristóvão AJ, Santos PF, Carvalho CM, Ambrósio AF. High glucose induces caspase-independent cell death in retinal neural cells. Neurobiol Dis. 2007;25:464–472. doi: 10.1016/j.nbd.2006.10.023.
    1. Zhu B, Wang W, Gu Q, Xu X. Erythropoietin protects retinal neurons and glial cells in early-stage streptozotocin-induced diabetic rats. Exp Eye Res. 2008;86:375–382. doi: 10.1016/j.exer.2007.11.010.
    1. Hotta N, Koh N, Sakakibara F, Nakamura J, Hamada Y, Naruse K, Sasaki H, Mizuno K, Matsubara A, Kakuta H, et al. Effect of an aldose reductase inhibitor, SNK-860, on deficits in the electroretinogram of diabetic rats. Exp Physiol. 1995;80:981–989. doi: 10.1113/expphysiol.1995.sp003909.
    1. Wurziger K, Lichtenberger T, Hanitzsch R. On-bipolar cells and depolarising third-order neurons as the origin of the ERG-b-wave in the RCS rat. Vision Res. 2001;41:1091–1101. doi: 10.1016/S0042-6989(01)00026-8.
    1. Tzekov R, Arden GB. The electroretinogram in diabetic retinopathy. Surv Ophthalmol. 1999;44:53–60. doi: 10.1016/S0039-6257(99)00063-6.
    1. Ogden TE. The oscillatory waves of the primate electroretinogram. Vision Res. 1973;13:1059–1074. doi: 10.1016/0042-6989(73)90144-2.
    1. Wachtmeister L, Dowling JE. The oscillatory potentials of the mudpuppy retina. Invest Ophthalmol Vis Sci. 1978;17:1176–1188.
    1. Li Q, Zemel E, Miller B, Perlman I. Early retinal damage in experimental diabetes: Electroretinographical and morphological observations. Exp Eye Res. 2002;74:615–625. doi: 10.1006/exer.2002.1170.
    1. Kline RP, Ripps H, Dowling JE. Generation of b-wave currents in the skate retina. Proc Natl Acad Sci USA. 1978;75:5727–5731. doi: 10.1073/pnas.75.11.5727.
    1. Klemp K, Sander B, Brockhoff PB, Vaag A, Lund-Andersen H, Larsen M. The multifocal ERG in diabetic patients without retinopathy during euglycemic clamping. Invest Ophthalmol Vis Sci. 2005;46:2620–2626. doi: 10.1167/iovs.04-1254.
    1. Chen BH, Jiang DY, Tang LS. Advanced glycation end-products induce apoptosis involving the signaling pathways of oxidative stress in bovine retinal pericytes. Life Sci. 2006;79:1040–1048. doi: 10.1016/j.lfs.2006.03.020.
    1. Du Y, Miller CM, Kern TS. Hyperglycemia increases mitochondrial superoxide in retina and retinal cells. Free Radic Biol Med. 2003;35:1491–1499. doi: 10.1016/j.freeradbiomed.2003.08.018.
    1. Drel VR, Xu W, Zhang J, Kador PF, Ali TK, Shin J, Julius U, Slusher B, El-Remessy AB, Obrosova IG. Poly(ADP-ribose)polymerase inhibition counteracts cataract formation and early retinal changes in streptozotocin-diabetic rats. Invest Ophthalmol Vis Sci. 2009;50:1778–1790. doi: 10.1167/iovs.08-2191.
    1. Obrosova IG, Julius UA. Role for poly(ADP-ribose) polymerase activation in diabetic nephropathy, neuropathy and retinopathy. Curr Vasc Pharmacol. 2005;3:267–283. doi: 10.2174/1570161054368634.
    1. Zheng L, Szabó C, Kern TS. Poly(ADP-ribose) polymerase is involved in the development of diabetic retinopathy via regulation of nuclear factor-kappaB. Diabetes. 2004;53:2960–2967. doi: 10.2337/diabetes.53.11.2960.
    1. Li GY, Osborne NN. Oxidative-induced apoptosis to an immortalized ganglion cell line is caspase independent but involves the activation of poly(ADP-ribose)polymerase and apoptosis-inducing factor. Brain Res. 2008;1188:35–43. doi: 10.1016/j.brainres.2007.10.073.
    1. Miki K, Uehara N, Shikata N, Matsumura M, Tsubura A. Poly (ADP-ribose) polymerase inhibitor 3-aminobenzamide rescues N-methyl-N-nitrosourea-induced photoreceptor cell apoptosis in Sprague-Dawley rats through preservation of nuclear factor-kappaB activity. Exp Eye Res. 2007;84:285–292. doi: 10.1016/j.exer.2006.09.023.
    1. Oku H, Goto W, Okuno T, Kobayashi T, Sugiyama T, Ota T, Yoneda S, Hara H, Ikeda T. Effects of poly(ADP-ribose) polymerase inhibitor on NMDA-induced retinal injury. Curr Eye Res. 2004;29:403–411. doi: 10.1080/02713680490517917.
    1. Cukras CA, Petrou P, Chew EY, Meyerle CB, Wong WT. Oral minocycline for the treatment of diabetic macular edema (DME): Results of a phase I/II clinical study. Invest Ophthalmol Vis Sci. 2012;53:3865–3874. doi: 10.1167/iovs.11-9413.
    1. Kraus RL, Pasieczny R, Lariosa-Willingham K, Turner MS, Jiang A, Trauger JW. Antioxidant properties of minocycline: Neuroprotection in an oxidative stress assay and direct radical-scavenging activity. J Neurochem. 2005;94:819–827. doi: 10.1111/j.1471-4159.2005.03219.x.
    1. Choi SH, Lee DY, Chung ES, Hong YB, Kim SU, Jin BK. Inhibition of thrombin-induced microglial activation and NADPH oxidase by minocycline protects dopaminergic neurons in the substantia nigra in vivo. J Neurochem. 2005;95:1755–1765. doi: 10.1111/j.1471-4159.2005.03503.x.
    1. Sinha-Hikim I, Shen R, Nzenwa I, Gelfand R, Mahata SK, Sinha-Hikim AP. Minocycline suppresses oxidative stress and attenuates fetal cardiac myocyte apoptosis triggered by in utero cocaine exposure. Apoptosis. 2011;16:563–573. doi: 10.1007/s10495-011-0590-4.
    1. Cieslik M, Pyszko J, Strosznajder JB. Docosahexaenoic acid and tetracyclines as promising neuroprotective compounds with poly(ADP-ribose) polymerase inhibitory activities for oxidative/genotoxic stress treatment. Neurochem Int. 2013;62:626–636. doi: 10.1016/j.neuint.2013.02.016.

Source: PubMed

3
Abonner