Skip to main content
Top
Published in: BMC Ophthalmology 1/2018

Open Access 01-12-2018 | Research article

P66Shc expression in diabetic rat retina

Authors: Ming-Hui Zhao, Jianyan Hu, Shufeng Li, Qiang Wu, Peirong Lu

Published in: BMC Ophthalmology | Issue 1/2018

Login to get access

Abstract

Background

P66Shc is partially localised within the mitochondrial fraction. It is primarily related to the generation of mitochondrial reactive oxygen species and apoptosis. Based on previous studies, we hypothesize that in the retina, p66Shc may exist and affect the development of diabetic retinopathy. The purpose of this study was to investigate p66Shc expression in retinal in streptozotocin-induced diabetic (SD) rats, which may provide a pathway to study the pathogenesis of diabetic retinopathy.

Methods

Reverse transcription-polymerase chain reaction (RT-PCR) and western blot were used to detect retinal p66Shc mRNA and protein expression in SD rats, respectively. Immunohistochemical staining was applied to detect the location of rat retinal p66Shc expression. TUNEL assay was applied to detect the number of apoptotic cells.

Results

P66Shc expression was found in the retina of normal and diabetic rats, and the level of mRNA and protein expression increased with the progression of diabetes mellitus (DM). P66Shc expression was mainly located in the retinal ganglion cell layer and inner nuclear layer. Compared with the normal group, retinal cell tissue apoptosis rate in the D12w group was significantly increased.

Conclusion

Rat retinal p66Shc expression was mainly in the ganglion cell layer and inner nuclear layer. As the degree of DM progressed, p66Shc expression gradually increased, and the number of apoptotic cells also increased.
Literature
1.
go back to reference Xu X, Zhu Q, Xia X, Zhang S, Gu Q, Luo D. Blood retinal barrier breakdown induced by activation of protein kinase C via vascular endothelial growth factor in streptozotocin induced diabetic rats. Curr Eye Res. 2004;28:251–6.CrossRef Xu X, Zhu Q, Xia X, Zhang S, Gu Q, Luo D. Blood retinal barrier breakdown induced by activation of protein kinase C via vascular endothelial growth factor in streptozotocin induced diabetic rats. Curr Eye Res. 2004;28:251–6.CrossRef
2.
go back to reference Sheetz MJ, King GL. Molecular understanding of hyperglycemia’s adverse effects for diabetic complications. JAMA. 2002;288:2579–88.CrossRef Sheetz MJ, King GL. Molecular understanding of hyperglycemia’s adverse effects for diabetic complications. JAMA. 2002;288:2579–88.CrossRef
3.
go back to reference Naruse K, Nakamura J, Hamada Y, Nakayama M, Chaya S, Komori T, et al. Aldose reductase inhibition prevents glucose induced apoptosis in cultured bovine retinal microvascular pericytes. Exp Eye Res. 2000;71:309–15.CrossRef Naruse K, Nakamura J, Hamada Y, Nakayama M, Chaya S, Komori T, et al. Aldose reductase inhibition prevents glucose induced apoptosis in cultured bovine retinal microvascular pericytes. Exp Eye Res. 2000;71:309–15.CrossRef
4.
go back to reference Yao D, Taguchi T, Matsumura T, Pestell R, Giardino L, Suske G, et al. High glucose increases angiopoietin 2 transcription in microvascular endothelial cells through methylglyoxal modification of mSin3A. J Biol Chem. 2007;282:31038–45.CrossRef Yao D, Taguchi T, Matsumura T, Pestell R, Giardino L, Suske G, et al. High glucose increases angiopoietin 2 transcription in microvascular endothelial cells through methylglyoxal modification of mSin3A. J Biol Chem. 2007;282:31038–45.CrossRef
5.
go back to reference Pfister F, Feng Y, vom Hagen F, Hoffmann S, Molema G, Hillebrands JL, et al. Pericyte migration: a novel mechanism of pericyte loss in experimental diabetic retinopathy. Diabetes. 2008;57:2495–502.CrossRef Pfister F, Feng Y, vom Hagen F, Hoffmann S, Molema G, Hillebrands JL, et al. Pericyte migration: a novel mechanism of pericyte loss in experimental diabetic retinopathy. Diabetes. 2008;57:2495–502.CrossRef
6.
go back to reference Di Mario U, Pugliese G. 15th Golgi lecture: from hyperglycaemia to the dysregulation of vascular remodelling in diabetes. Diabetologia. 2001;44:674–92.CrossRef Di Mario U, Pugliese G. 15th Golgi lecture: from hyperglycaemia to the dysregulation of vascular remodelling in diabetes. Diabetologia. 2001;44:674–92.CrossRef
7.
go back to reference Njie-Mbye YF, Kulkarni-Chitnis M, Opere CA, Barrett A, Ohia SE. Lipid peroxidation: pathophysiological and pharmacological implications in the eye. Front Physiol. 2013;4:366.CrossRef Njie-Mbye YF, Kulkarni-Chitnis M, Opere CA, Barrett A, Ohia SE. Lipid peroxidation: pathophysiological and pharmacological implications in the eye. Front Physiol. 2013;4:366.CrossRef
8.
go back to reference Fan TJ, Han LH, Cong RS, Liang J. Caspase family proteases and apoptosis. Acta Biochim Biophys Sin. 2005;37:719–27.CrossRef Fan TJ, Han LH, Cong RS, Liang J. Caspase family proteases and apoptosis. Acta Biochim Biophys Sin. 2005;37:719–27.CrossRef
9.
go back to reference Yang JH, Kwak HW, Kim TG, Han J, Moon SW, Yu SY. Retinal neurodegeneration in type II diabetic otsuka long-evans Tokushima fatty rats. IOVS. 2013;54:3844–51. Yang JH, Kwak HW, Kim TG, Han J, Moon SW, Yu SY. Retinal neurodegeneration in type II diabetic otsuka long-evans Tokushima fatty rats. IOVS. 2013;54:3844–51.
10.
go back to reference Bringmann A, Wiedemann P. Muller glial cells in retinal disease. Ophthalmologica. 2012;227:1–19.CrossRef Bringmann A, Wiedemann P. Muller glial cells in retinal disease. Ophthalmologica. 2012;227:1–19.CrossRef
11.
go back to reference Barber AJ, Gardner TW, Abcouwer SF. The significance of vascular and neural apoptosis to the pathology of diabetic retinopathy. IOVS. 2011;52:1156–63. Barber AJ, Gardner TW, Abcouwer SF. The significance of vascular and neural apoptosis to the pathology of diabetic retinopathy. IOVS. 2011;52:1156–63.
12.
go back to reference Rungger-Brandle E, Dosso AA, Leuenberger PM. Glial reactivity, an early feature of diabetic retinopathy. IOVS. 2000;41:1971–80. Rungger-Brandle E, Dosso AA, Leuenberger PM. Glial reactivity, an early feature of diabetic retinopathy. IOVS. 2000;41:1971–80.
13.
go back to reference Du X, Matsumura T, Edelstein D, Rossetti L, Zsengellér Z, Szabó C, Brownlee M. Inhibition of GAPDH activity by ploy (ADP-ribose) polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Invest. 2003;112:1049–57.CrossRef Du X, Matsumura T, Edelstein D, Rossetti L, Zsengellér Z, Szabó C, Brownlee M. Inhibition of GAPDH activity by ploy (ADP-ribose) polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Invest. 2003;112:1049–57.CrossRef
14.
go back to reference Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012;24:981–90.CrossRef Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012;24:981–90.CrossRef
15.
go back to reference Trinei M, Berniakovich L, Beltrami E, Migliaccio E, Fassina A, Pelicci P, et al. P66Shc signals to age. Aging. 2009;1:503–10.CrossRef Trinei M, Berniakovich L, Beltrami E, Migliaccio E, Fassina A, Pelicci P, et al. P66Shc signals to age. Aging. 2009;1:503–10.CrossRef
16.
go back to reference Giorgio M, Migliaccio E, Orsini F, Paolucci D, Moroni M, Contursi C, et al. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell. 2005;122:221–33.CrossRef Giorgio M, Migliaccio E, Orsini F, Paolucci D, Moroni M, Contursi C, et al. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell. 2005;122:221–33.CrossRef
17.
go back to reference Pinton P, Rimessi A, Marchi S, Orsini F, Migliaccio E, Giorgio M, et al. Protein kinase C beta and prolyl isomerase 1 regulate mitochondrial effects of the life-span determinant p66Shc. Science. 2007;315:659–63.CrossRef Pinton P, Rimessi A, Marchi S, Orsini F, Migliaccio E, Giorgio M, et al. Protein kinase C beta and prolyl isomerase 1 regulate mitochondrial effects of the life-span determinant p66Shc. Science. 2007;315:659–63.CrossRef
18.
go back to reference Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, Pandolfi PP, et al. The p66Shc adaptor protein controls oxidative stress response and life span in mammals. Nature. 1999;402:309–13.CrossRef Migliaccio E, Giorgio M, Mele S, Pelicci G, Reboldi P, Pandolfi PP, et al. The p66Shc adaptor protein controls oxidative stress response and life span in mammals. Nature. 1999;402:309–13.CrossRef
19.
go back to reference Menini S, Amadio L, Oddi G, Ricci C, Pesce C, Pugliese F, et al. Deletion of p66Shc longevity gene protects against experimental diabetic glomerulopathy by preventing diabetes-induced oxidative stress. Diabetes. 2006;55:1642–50.CrossRef Menini S, Amadio L, Oddi G, Ricci C, Pesce C, Pugliese F, et al. Deletion of p66Shc longevity gene protects against experimental diabetic glomerulopathy by preventing diabetes-induced oxidative stress. Diabetes. 2006;55:1642–50.CrossRef
20.
go back to reference Graiani G, Lagrasta C, Migliaccio E, Spillmann F, Meloni M, Madeddu P, et al. Genetic deletion of the p66Shc adaptor protein protects from angiotensin II-induced myocardial damage. Hypertension. 2005;46:433–40.CrossRef Graiani G, Lagrasta C, Migliaccio E, Spillmann F, Meloni M, Madeddu P, et al. Genetic deletion of the p66Shc adaptor protein protects from angiotensin II-induced myocardial damage. Hypertension. 2005;46:433–40.CrossRef
21.
go back to reference Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414:813–20.CrossRef Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414:813–20.CrossRef
22.
go back to reference Jain SK, Kannan K, Lim G, McVie R, Bocchini JA Jr. Hyperketonemia increases tumor necrosis factor-α secretion in cultured U937 monocytes and type 1 diabetic patients and is apparently mediated by oxidative stress and cAMP deficiency. Diabetes. 2002;51:2287–93.CrossRef Jain SK, Kannan K, Lim G, McVie R, Bocchini JA Jr. Hyperketonemia increases tumor necrosis factor-α secretion in cultured U937 monocytes and type 1 diabetic patients and is apparently mediated by oxidative stress and cAMP deficiency. Diabetes. 2002;51:2287–93.CrossRef
23.
go back to reference Jain SK, Kannan K, Lim G, Matthews-Greer J, McVie R, Bocchini JA Jr. Elevated blood interleukin-6 levels in hyperketonemic type 1 diabetic patients and secretion by acetoacetate-treated cultured U937 monocytes. Diabetes Care. 2003;26:2139–43.CrossRef Jain SK, Kannan K, Lim G, Matthews-Greer J, McVie R, Bocchini JA Jr. Elevated blood interleukin-6 levels in hyperketonemic type 1 diabetic patients and secretion by acetoacetate-treated cultured U937 monocytes. Diabetes Care. 2003;26:2139–43.CrossRef
24.
go back to reference Camici GG, Schiavoni M, Francia P, Bachschmid M, Martin-padura I, Hersberger M, et al. Genetic deletion of p66Shc adaptor protein prevents hyperglycemia-induced endothelial dysfunction and oxidative stress. Proc Natl Acad Sci. 2007;104:5217–22.CrossRef Camici GG, Schiavoni M, Francia P, Bachschmid M, Martin-padura I, Hersberger M, et al. Genetic deletion of p66Shc adaptor protein prevents hyperglycemia-induced endothelial dysfunction and oxidative stress. Proc Natl Acad Sci. 2007;104:5217–22.CrossRef
25.
go back to reference Tomilov AA, Bicocca V, Schoenfeld RA, Giorgio M, Migliaccio E, Ramsey JJ, et al. Decreased superoxide production in macrophages of long-lived p66Shc knock-out mice. J Biol Chem. 2010;285:1153–65.CrossRef Tomilov AA, Bicocca V, Schoenfeld RA, Giorgio M, Migliaccio E, Ramsey JJ, et al. Decreased superoxide production in macrophages of long-lived p66Shc knock-out mice. J Biol Chem. 2010;285:1153–65.CrossRef
26.
go back to reference Cesselli D, Jakoniuk I, Barlucchi L, Beltrami AP, Hintze TH, Nadal-Ginard B, et al. Oxidative stress-mediated cardiac cell death is a major determinant of ventricular dysfunction and failure in dog dilated cardiomyopathy. Circ Res. 2001;89:279–86.CrossRef Cesselli D, Jakoniuk I, Barlucchi L, Beltrami AP, Hintze TH, Nadal-Ginard B, et al. Oxidative stress-mediated cardiac cell death is a major determinant of ventricular dysfunction and failure in dog dilated cardiomyopathy. Circ Res. 2001;89:279–86.CrossRef
28.
go back to reference Zaccagnini G, Martelli F, Fasanaro P, Magenta A, Gaetano C, Di Carlo A, et al. P66Shc a modulates tissue response to hindlimb ischemia. Circulation. 2004;109:2917–23.CrossRef Zaccagnini G, Martelli F, Fasanaro P, Magenta A, Gaetano C, Di Carlo A, et al. P66Shc a modulates tissue response to hindlimb ischemia. Circulation. 2004;109:2917–23.CrossRef
29.
go back to reference Napoli C, Martin-Padura I, de Nigris F, Giorgio M, Mansueto G, Somma P, et al. Deletion of the p66Shc longevity gene reduces systemic and tissue oxidative stress, vascular cell apoptosis, and early atherogenesis in mice fed a high-fat diet. Proc Natl Acad Sci U S A. 2003;100:2112–6.CrossRef Napoli C, Martin-Padura I, de Nigris F, Giorgio M, Mansueto G, Somma P, et al. Deletion of the p66Shc longevity gene reduces systemic and tissue oxidative stress, vascular cell apoptosis, and early atherogenesis in mice fed a high-fat diet. Proc Natl Acad Sci U S A. 2003;100:2112–6.CrossRef
30.
go back to reference Francia P, delli Gatti C, Bachschmid M, Martin-Padura I, Savoia C, Migliaccio E, et al. Deletion of p66Shc gene protects against age-related endothelial dysfunction. Circulation. 2004;110:2889–95.CrossRef Francia P, delli Gatti C, Bachschmid M, Martin-Padura I, Savoia C, Migliaccio E, et al. Deletion of p66Shc gene protects against age-related endothelial dysfunction. Circulation. 2004;110:2889–95.CrossRef
31.
go back to reference Sun L, Xiao L, Nie J, Liu FY, Ling GH, Zhu XJ, et al. P66Shc mediates high-glucose and angiotensin II-induced oxidative stress renal tubular injury via mitochondrial-dependent apoptotic pathway. Am J Physiology-Renal Physiol. 2010;299:F1014–25.CrossRef Sun L, Xiao L, Nie J, Liu FY, Ling GH, Zhu XJ, et al. P66Shc mediates high-glucose and angiotensin II-induced oxidative stress renal tubular injury via mitochondrial-dependent apoptotic pathway. Am J Physiology-Renal Physiol. 2010;299:F1014–25.CrossRef
32.
go back to reference Barber AJ, Antonetti DA, Kern TS, Reiter CE, Soans RS, Krady JK, et al. The Ins2Akita mouse as a model of early retinal complications in diabetes. IOVS. 2005;46:2210–8. Barber AJ, Antonetti DA, Kern TS, Reiter CE, Soans RS, Krady JK, et al. The Ins2Akita mouse as a model of early retinal complications in diabetes. IOVS. 2005;46:2210–8.
33.
go back to reference Rozakis-Adcock M, McGlade J, Mbamalu G, Pelicci G, Daly R, Li W, et al. Association of the Shc and Grb2/Sem5 SH2-containing proteins is implicated in activation of the Ras pathway by tyrosine kinases. Nature. 1992;360:689–92.CrossRef Rozakis-Adcock M, McGlade J, Mbamalu G, Pelicci G, Daly R, Li W, et al. Association of the Shc and Grb2/Sem5 SH2-containing proteins is implicated in activation of the Ras pathway by tyrosine kinases. Nature. 1992;360:689–92.CrossRef
34.
go back to reference Mishra R, Emancipator SN, Kern T, Simonson MS. High glucose evokes an intrinsic proapoptotic pathway in mesangial cells. Kidney Int. 2005;57:82–93.CrossRef Mishra R, Emancipator SN, Kern T, Simonson MS. High glucose evokes an intrinsic proapoptotic pathway in mesangial cells. Kidney Int. 2005;57:82–93.CrossRef
35.
go back to reference Lee FT, Cao Z, Long DM, Panagiotopoulos S, Jerums G, Cooper ME, et al. Interactions between angiotensin II and NF-kappaB-dependent pathways in modulating macrophage infiltration in experimental diabetic nephropathy. J Am Soc Nephrol. 2004;15:2139–51.CrossRef Lee FT, Cao Z, Long DM, Panagiotopoulos S, Jerums G, Cooper ME, et al. Interactions between angiotensin II and NF-kappaB-dependent pathways in modulating macrophage infiltration in experimental diabetic nephropathy. J Am Soc Nephrol. 2004;15:2139–51.CrossRef
36.
go back to reference Innocenti M, Tenca P, Frittoli E, Faretta M, Tocchetti A, Di Fiore PP, et al. Mechanisms through which Sos-1 coordinates the activation of Ras and Rac. J Cell Biol. 2002;156:125–36.CrossRef Innocenti M, Tenca P, Frittoli E, Faretta M, Tocchetti A, Di Fiore PP, et al. Mechanisms through which Sos-1 coordinates the activation of Ras and Rac. J Cell Biol. 2002;156:125–36.CrossRef
37.
go back to reference Pacini S, Pellegrini M, Migliaccio E, Patrussi L, Ulivieri C, Ventura A, et al. P66SHC promotes apoptosis and antagonizes mitogenic signaling in T cells. Mol Cell Biol. 2004;24:1747–57.CrossRef Pacini S, Pellegrini M, Migliaccio E, Patrussi L, Ulivieri C, Ventura A, et al. P66SHC promotes apoptosis and antagonizes mitogenic signaling in T cells. Mol Cell Biol. 2004;24:1747–57.CrossRef
38.
go back to reference Arany I, Faisal A, Nagamine Y, Safirstein RL. P66Shc inhibits pro-survival epidermal growth factor receptor/ERK signaling during severe oxidative stress in mouse renal proximal tubule cells. J Biol Chem. 2008;283:6110–7.CrossRef Arany I, Faisal A, Nagamine Y, Safirstein RL. P66Shc inhibits pro-survival epidermal growth factor receptor/ERK signaling during severe oxidative stress in mouse renal proximal tubule cells. J Biol Chem. 2008;283:6110–7.CrossRef
Metadata
Title
P66Shc expression in diabetic rat retina
Authors
Ming-Hui Zhao
Jianyan Hu
Shufeng Li
Qiang Wu
Peirong Lu
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Ophthalmology / Issue 1/2018
Electronic ISSN: 1471-2415
DOI
https://doi.org/10.1186/s12886-018-0724-3

Other articles of this Issue 1/2018

BMC Ophthalmology 1/2018 Go to the issue