全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

p21WAF1/CIP1 Expression is Differentially Regulated by Metformin and Rapamycin

DOI: 10.1155/2014/327640

Full-Text   Cite this paper   Add to My Lib

Abstract:

The mammalian target of rapamycin (mTOR) pathway plays an important role in the development of diabetic nephropathy and other age-related diseases. One of the features of DN is the elevated expression of p21WAF1/CIP1. However, the importance of the mTOR signalling pathway in p21 regulation is poorly understood. Here we investigated the effect of metformin and rapamycin on mTOR-related phenotypes in cell lines of epithelial origin. This study reports that metformin inhibits high glucose-induced p21 expression. High glucose opposed metformin in regulating cell size, proliferation, and protein synthesis. These effects were associated with reduced AMPK activation, affecting downstream mTOR signalling. However, the inhibition of the mTOR pathway by rapamycin did not have a negative effect on p21 expression, suggesting that metformin regulates p21 upstream of mTOR. These findings provide support for the hypothesis that AMPK activation may regulate p21 expression, which may have implications for diabetic nephropathy and other age-related pathologies. 1. Introduction There has been a dramatic increase in the prevalence of diabetes mellitus in recent years [1]. The chronic effects of diabetes may manifest in macro- and microvascular complications that are the major causes of morbidity and mortality in patients with diabetes. Diabetic nephropathy (DN), one of the microvascular complications, is a leading cause of death from kidney failure [2, 3]. Apart from haemodynamic factors, hyperglycaemia has been shown to be an underlying cause of pathogenesis in DN. The damaging effects of hyperglycaemia have been partly attributed to increased cellular glucose uptake in cells that are not protected from high ambient glucose levels. Early cellular changes in the development of DN include hyperplasia and hypertrophy [4]. Several investigators have associated the expression of Cip/Kip cyclin-dependent kinase (CDK) inhibitors, p21 and p27, with glomerular hypertrophy [5–7]. It has been proposed that p21 and p27 may be involved in hypertrophy independently of their cell cycle regulatory properties (Monkawa 2002). Furthermore, the induction of p21 and p27 is also required for senescent arrest, a molecular signature of hypertrophic changes in the early stages of the development of diabetic kidney disease [8]. The fact that p21 null mice do not develop glomerular hypertrophy supports the importance of p21 in DN [9]. The activation of the mammalian target of rapamycin (mTOR), a serine/threonine kinase, plays a pivotal role in the pathologic forms of hypertrophy in the kidneys

References

[1]  E. Adeghate, P. Schattner, and E. Dunn, “An update on the etiology and epidemiology of diabetes mellitus,” Annals of the New York Academy of Sciences, vol. 1084, no. 1, pp. 1–29, 2006.
[2]  Z. Cao and M. E. Cooper, “Pathogenesis of diabetic nephropathy,” Journal of Diabetes Investigation, vol. 2, no. 4, pp. 243–247, 2011.
[3]  M. Kitada, Z. Zhang, A. Mima, and G. L. King, “Molecular mechanisms of diabetic vascular complications,” Journal of Diabetes Investigation, vol. 1, no. 3, pp. 77–89, 2010.
[4]  A. Arya, S. Aggarwal, and H. N. Yadav, “Pathogenesis of diabetic nephropathy,” International Journal of Pharmacy and Pharmaceutical Sciences, vol. 2, no. 4, pp. 24–29, 2010.
[5]  G. Wolf, “Molecular mechanisms of diabetic mesangial cell hypertrophy: a proliferation of novel factors,” Journal of the American Society of Nephrology, vol. 13, no. 10, pp. 2611–2613, 2002.
[6]  S. V. Griffin and S. J. Shankland, “Not just an inhibitor: a role for p21 beyond the cell cycle—“the truth is rarely pure and never simple”,” Journal of the American Society of Nephrology, vol. 15, no. 3, pp. 825–826, 2004.
[7]  M. M. Poplawski, J. W. Mastaitis, F. Isoda, F. Grosjean, F. Zheng, and C. V. Mobbs, “Reversal of diabetic nephropathy by a ketogenic diet,” PLoS ONE, vol. 6, no. 4, Article ID e18604, 2011.
[8]  J. Satriano, H. Mansoury, A. Deng et al., “Transition of kidney tubule cells to a senescent phenotype in early experimental diabetes,” The American Journal of Physiology: Cell Physiology, vol. 299, no. 2, pp. C374–C380, 2010.
[9]  H. Zheng, S. A. Whitman, W. Wu et al., “Therapeutic potential of Nrf2 activators in streptozotocin-induced diabetic nephropathy,” Diabetes, vol. 60, no. 11, pp. 3055–3066, 2011.
[10]  W. Lieberthal and J. S. Levine, “The role of the mammalian target of rapamycin (mTOR) in renal disease,” Journal of the American Society of Nephrology, vol. 20, no. 12, pp. 2493–2502, 2009.
[11]  M. Haneda, D. Koya, M. Isono, and R. Kikkawa, “Overview of glucose signaling in mesangial cells in diabetic nephropathy,” Journal of the American Society of Nephrology, vol. 14, no. 5, pp. 1374–1382, 2003.
[12]  V. P. Houde, S. Br?lé, W. T. Festuccia et al., “Chronic rapamycin treatment causes glucose intolerance and hyperlipidemia by upregulating hepatic gluconeogenesis and impairing lipid deposition in adipose tissue,” Diabetes, vol. 59, no. 6, pp. 1338–1348, 2010.
[13]  K. Inoki, “Role of TSC-mTOR pathway in diabetic nephropathy,” Diabetes Research and Clinical Practice, vol. 82, no. 1, pp. S59–S62, 2008.
[14]  B. Vodenik, J. Rovira, and J. M. Campistol, “Mammalian target of rapamycin and diabetes: what does the current evidence tell us?” Transplantation Proceedings, vol. 41, no. 6, pp. S31–S38, 2009.
[15]  A. M. Miller, J. R. Brestoff, C. B. Phelps, E. Z. Berk, and T. H. Reynolds IV, “Rapamycin does not improve insulin sensitivity despite elevated mammalian target of rapamycin complex 1 activity in muscles of ob/ob mice,” The American Journal of Physiology: Regulatory Integrative and Comparative Physiology, vol. 295, no. 5, pp. R1431–R1438, 2008.
[16]  H. Mori, K. Inoki, K. Masutani et al., “The mTOR pathway is highly activated in diabetic nephropathy and rapamycin has a strong therapeutic potential,” Biochemical and Biophysical Research Communications, vol. 384, no. 4, pp. 471–475, 2009.
[17]  A. Kalender, A. Selvaraj, S. Y. Kim et al., “Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner,” Cell Metabolism, vol. 11, no. 5, pp. 390–401, 2010.
[18]  X. Wang, X. Pan, and J. Song, “AMP-activated protein kinase is required for induction of apoptosis and epithelial-to-mesenchymal transition,” Cellular Signalling, vol. 22, no. 11, pp. 1790–1797, 2010.
[19]  M. R. Owen, E. Doran, and A. P. Halestrap, “Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain,” Biochemical Journal, vol. 348, no. 3, pp. 607–614, 2000.
[20]  B. A. Kefas, Y. Cai, K. Kerckhofs et al., “Metformin-induced stimulation of AMP-activated protein kinase in β-cells impairs their glucose responsiveness and can lead to apoptosis,” Biochemical Pharmacology, vol. 68, no. 3, pp. 409–416, 2004.
[21]  B. Viollet, B. Guigas, N. Sanz Garcia, J. Leclerc, M. Foretz, and F. Andreelli, “Cellular and molecular mechanisms of metformin: an overview,” Clinical Science, vol. 122, no. 6, pp. 253–270, 2012.
[22]  P. Ruggenenti and G. Remuzzi, “Nephropathy of type 1 and type 2 diabetes: diverse pathophysiology same treatment?” Nephrology Dialysis Transplantation, vol. 15, no. 12, pp. 1900–1902, 2000.
[23]  L. Wu and R. Derynck, “Essential role of TGF-β signaling in glucose-induced cell hypertrophy,” Developmental Cell, vol. 17, no. 1, pp. 35–48, 2009.
[24]  C. L. Buller, C. W. Heilig, and F. C. Brosius III, “GLUT1 enhances mTOR activity independently of TSC2 and AMPK,” The American Journal of Physiology: Renal Physiology, vol. 301, no. 3, pp. F588–F596, 2011.
[25]  J. I. Kreisberg and S. H. Ayo, “The glomerular mesangium in diabetes mellitus,” Kidney International, vol. 43, no. 1, pp. 109–113, 1993.
[26]  M. Zakikhani, M.-J. Blouin, E. Piura, and M. N. Pollak, “Metformin and rapamycin have distinct effects on the AKT pathway and proliferation in breast cancer cells,” Breast Cancer Research and Treatment, vol. 123, no. 1, pp. 271–279, 2010.
[27]  R. J. O. Dowling, M. Zakikhani, I. G. Fantus, M. Pollak, and N. Sonenberg, “Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells,” Cancer Research, vol. 67, no. 22, pp. 10804–10812, 2007.
[28]  L. Tosca, C. Ramé, C. Chabrolle, S. Tesseraud, and J. Dupont, “Metformin decreases IGF1-induced cell proliferation and protein synthesis through AMP-activated protein kinase in cultured bovine granulosa cells,” Reproduction, vol. 139, no. 2, pp. 409–418, 2010.
[29]  Y. Zhuang and W. K. Keith, “Cell cycle arrest in Metformin treated breast cancer cells involves activation of AMPK, downregulation of cyclin D1, and requires p27Kip1 or p21Cip1,” Journal of Molecular Signaling, vol. 3, article 18, 2008.
[30]  S. O. Marx, T. Jayaraman, L. O. Go, and A. R. Marks, “Rapamycin-FKBP inhibits cell cycle regulators of proliferation in vascular smooth muscle cells,” Circulation Research, vol. 76, no. 3, pp. 412–417, 1995.
[31]  J. W. Russell, D. Golovoy, A. M. Vincent et al., “High glucose-induced oxidative stress and mitochondrial dysfunction in nuerons,” FASEB Journal, vol. 16, no. 13, pp. 1738–1748, 2002.
[32]  D. C. Han, M. Isono, B. B. Hoffman, and F. N. Ziyadeh, “High glucose stimulates proliferation and collagen type I synthesis in renal cortical fibroblasts: Mediation by autocrine activation of TGF-β,” Journal of the American Society of Nephrology, vol. 10, no. 9, pp. 1891–1899, 1999.
[33]  M. V. Rocco, Y. Chen, S. Goldfarb, and F. N. Ziyadeh, “Elevated glucose stimulates TGE-β gene expression and bioactivity in proximal tubule,” Kidney International, vol. 41, no. 1, pp. 107–114, 1992.
[34]  G. Wolf, K. Sharma, Y. Chen, M. Ericksen, and F. N. Ziyadeh, “High glucose-induced proliferation in mesangial cells is reversed by autocrine TGF-β,” Kidney International, vol. 42, no. 3, pp. 647–656, 1992.
[35]  F. N. Ziyadeh, K. Sharma, M. Ericksen, and G. Wolf, “Stimulation of collagen gene expression and protein synthesis in murine mesangial cells by high glucose is mediated by autocrine activation of transforming growth factor-β,” Journal of Clinical Investigation, vol. 93, no. 2, pp. 536–542, 1994.
[36]  Y.-J. Choi, H.-S. Lim, J.-S. Choi et al., “Blockade of chronic high glucose-induced endothelial apoptosis by Sasa borealis bamboo extract,” Experimental Biology and Medicine, vol. 233, no. 5, pp. 580–591, 2008.
[37]  A. Duffy, A. Liew, J. O'Sullivan, G. Avalos, A. Samali, and T. O'Brien, “Distinct effects of high-glucose conditions on endothelial cells of macrovascular and microvascular origins,” Endothelium-Journal of Endothelial Cell Research, vol. 13, no. 1, pp. 9–16, 2006.
[38]  S. McGinn, S. Saad, P. Poronnik, and C. A. Pollock, “High glucose-mediated effects on endothelial cell proliferation occur via p38 MAP kinase,” The American Journal of Physiology: Endocrinology and Metabolism, vol. 285, no. 4, pp. E708–E717, 2003.
[39]  J.-D. Liu, Y.-J. Wang, C.-H. Chen et al., “Molecular mechanisms of G0/G1 cell-cycle arrest and apoptosis induced by terfenadine in human cancer cells,” Molecular Carcinogenesis, vol. 37, no. 1, pp. 39–50, 2003.
[40]  C. C. Williams, B. A. Singleton, S. D. Llopis, E. V. Skripnikova, L. Jack Jr., and K. Kennedy, “Metformin induces a senescence-associated gene signature in breast cancer cells,” Journal of Health Care for the Poor and Underserved, vol. 24, no. 1, pp. 93–103, 2013.
[41]  A. Tzatsos and P. N. Tsichlis, “Energy depletion inhibits phosphatidylinositol 3-kinase/Akt signaling and induces apoptosis via AMP-activated protein kinase-dependent phosphorylation of IRS-1 at Ser-794,” Journal of Biological Chemistry, vol. 282, no. 25, pp. 18069–18082, 2007.
[42]  B. Guigas, D. Detaille, C. Chauvin et al., “Metformin inhibits mitochondrial permeability transition and cell death: a pharmacological in vitro study,” Biochemical Journal, vol. 382, no. 3, pp. 877–884, 2004.
[43]  A. L. Edinger, C. M. Linardic, G. G. Chiang, C. B. Thompson, and R. T. Abraham, “Differential effects of rapamycin on mammalian target of rapamycin signaling functions in mammalian cells,” Cancer Research, vol. 63, no. 23, pp. 8451–8460, 2003.
[44]  C. U. Niesler, K. H. Myburgh, and F. Moore, “The changing AMPK expression profile in differentiating mouse skeletal muscle myoblast cells helps confer increasing resistance to apoptosis,” Experimental Physiology, vol. 92, no. 1, pp. 207–217, 2007.
[45]  M. M. Shaw, W. K. Gurr, R. J. McCrimmon, D. F. Schorderet, and R. S. Sherwin, “5′AMP-activated protein kinase α deficiency enhances stress-induced apoptosis in BHK and PC12 cells,” Journal of Cellular and Molecular Medicine, vol. 11, no. 2, pp. 286–298, 2007.
[46]  M. L. Coleman, C. J. Marshall, and M. F. Olson, “Ras promotes p21Waf1/Cip1 protein stability via a cyclin D1-imposed block in proteasome-mediated degradation,” The EMBO Journal, vol. 22, no. 9, pp. 2036–2046, 2003.
[47]  D. Moreno, R. Viana, and P. Sanz, “Two-hybrid analysis identifies PSMD11, a non-ATPase subunit of the proteasome, as a novel interaction partner of AMP-activated protein kinase,” International Journal of Biochemistry and Cell Biology, vol. 41, no. 12, pp. 2431–2439, 2009.
[48]  R. Viana, C. Aguado, I. Esteban et al., “Role of AMP-activated protein kinase in autophagy and proteasome function,” Biochemical and Biophysical Research Communications, vol. 369, no. 3, pp. 964–968, 2008.
[49]  V. Y. Yazbeck, D. Buglio, G. V. Georgakis et al., “Temsirolimus downregulates p21 without altering cyclin D1 expression and induces autophagy and synergizes with vorinostat in mantle cell lymphoma,” Experimental Hematology, vol. 36, no. 4, pp. 443–450, 2008.
[50]  A.-M. Gabeni, C. Saucier, M. Bedini, V. Barbu, and J. Mester, “Rapamycin inhibits cdk4 activation, p21Waf1/Cip1 expression and G1-phase progression in transformed mouse fibroblasts,” International Journal of Cancer, vol. 108, no. 2, pp. 200–206, 2004.
[51]  F. C. Brosius III, C. C. Khoury, C. L. Buller, and S. Chen, “Abnormalities in signaling pathways in diabetic nephropathy,” Expert Review of Endocrinology and Metabolism, vol. 5, no. 1, pp. 51–64, 2010.
[52]  C. L. Buller, R. D. Loberg, M.-H. Fan et al., “A GSK-3/TSC2/mTOR pathway regulates glucose uptake and GLUT1 glucose transporter expression,” The American Journal of Physiology: Cell Physiology, vol. 295, no. 3, pp. C836–C843, 2008.
[53]  K. Inoki, M. Haneda, S. Maeda, D. Koya, and R. Kikkawa, “TGF-β1 stimulates glucose uptake by enhancing GLUT1 expression in mesangial cells,” Kidney International, vol. 55, no. 5, pp. 1704–1712, 1999.
[54]  R. Osterby, “Glomerular structural changes in Ttype 1 (insulin-dependent) diabetes mellitus: causes, consequences, and prevention,” Diabetologia, vol. 35, no. 9, pp. 803–812, 1992.
[55]  G. Wolf, “Cell cycle regulation in diabetic nephropathy,” Kidney International, Supplement, vol. 58, no. 77, pp. S59–S66, 2000.
[56]  T. Monkawa, K. Hiromura, G. Wolf, and S. J. Shankland, “The hypertrophic effect of transforming growth factor-β is reduced in the absence of cyclin-dependent kinase-inhibitors p21 and p27,” Journal of the American Society of Nephrology, vol. 13, no. 5, pp. 1172–1178, 2002.
[57]  G. Wolf, R. Reinking, G. Zahner, R. A. K. Stahl, and S. J. Shankland, “Erk 1,2 phosphorylates p27Kip1: functional evidence for a role in high glucose-induced hypertrophy of mesangial cells,” Diabetologia, vol. 46, no. 8, pp. 1090–1099, 2003.
[58]  J. Megyesi, P. M. Price, E. Tamayo, and R. L. Safirstein, “The lack of a functional p21Waf1/Cip1 gene ameliorates progression to chronic renal failure,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 19, pp. 10830–10835, 1999.
[59]  M. V. Blagosklonny, “Cell cycle arrest is not yet senescence, which is not just cell cycle arrest: terminology for TOR-driven aging,” Aging-Us, vol. 4, no. 3, pp. 159–165, 2012.
[60]  C. Deng, P. Zhang, J. W. Herper, S. J. Elledge, and P. Leder, “Mice lacking p21(CIP1/WAF1) undergo normal development, but are defective in G1 checkpoint control,” Cell, vol. 82, no. 4, pp. 675–684, 1995.
[61]  G. Soria and V. Gottifredi, “PCNA-coupled p21 degradation after DNA damage: the exception that confirms the rule?” DNA Repair, vol. 9, no. 4, pp. 358–364, 2010.
[62]  M. M. Kavurma and L. M. Khachigian, “Sp1 inhibits proliferation and induces apoptosis in vascular smooth muscle cells by repressing p21Waf1/Cip1 transcription and cyclin D1-Cdk4-p21Waf1/Cip1 complex formation,” Journal of Biological Chemistry, vol. 278, no. 35, pp. 32537–32543, 2003.
[63]  A. G. Muntean, L. Pang, M. Poncz, S. F. Dowdy, G. A. Blobel, and J. D. Crispino, “Cyclin D-Cdk4 is regulated by GATA-1 and required for megakaryocyte growth and polyploidization,” Blood, vol. 109, no. 12, pp. 5199–5207, 2007.
[64]  D. Féliers, M. A. Frank, and D. J. Riley, “Activation of cyclin D1-Cdk4 and Cdk4-directed phosphorylation of RB protein in diabetic mesangial hypertrophy,” Diabetes, vol. 51, no. 11, pp. 3290–3299, 2002.
[65]  Y. Jiang, D. W. Cheng, E. Levi, and L. P. Singh, “IGF-1 increases laminin, cyclin D1, and P21Cip1 expression in glomerular mesangial cells: An investigation of the intracellular signaling pathway and cell-cycle progression,” Journal of Cellular Biochemistry, vol. 98, no. 1, pp. 208–220, 2006.
[66]  M. Tamamori-Adachi, H. Ito, K. Nobori et al., “Expression of cyclin D1 and CDK4 causes hypertrophic growth of cardiomyocytes in culture: a possible implication for cardiac hypertrophy,” Biochemical and Biophysical Research Communications, vol. 296, no. 2, pp. 274–280, 2002.
[67]  H. Motoshima, B. J. Goldstein, M. Igata, and E. Araki, “AMPK and cell proliferation—AMPK as a therapeutic target for atherosclerosis and cancer,” Journal of Physiology, vol. 574, no. 1, pp. 63–71, 2006.
[68]  Y.-N. Fu, H. Xiao, X.-W. Ma, S.-Y. Jiang, M. Xu, and Y.-Y. Zhang, “Metformin attenuates pressure overload-induced cardiac hypertrophy via AMPK activation,” Acta Pharmacologica Sinica, vol. 32, no. 7, pp. 879–887, 2011.
[69]  K. R. Laderoute, K. Amin, J. M. Calaoagan et al., “5′-AMP-activated protein kinase (AMPK) is induced by low-oxygen and glucose deprivation conditions found in solid-tumor microenvironments,” Molecular and Cellular Biology, vol. 26, no. 14, pp. 5336–5347, 2006.
[70]  H. Safayhi, H. Haase, U. Kramer et al., “L-type calcium channels in insulin-secreting cells: biochemical characterization and phosphorylation in RINm5F cells,” Molecular Endocrinology, vol. 11, no. 5, pp. 619–629, 1997.
[71]  A. Danchin and H. Buc, “Proton magnetic resonance studies on 5′- AMP site in glycogen phosphorylase b,” FEBS Letters, vol. 22, no. 3, pp. 289–293, 1972.
[72]  S. A. Hawley, A. E. Gadalla, G. S. Olsen, and D. Grahame Hardie, “The antidiabetic drug metformin activates the AMP-activated protein kinase cascade via an adenine nucleotide-independent mechanism,” Diabetes, vol. 51, no. 8, pp. 2420–2425, 2002.
[73]  K. Kishi, T. Yuasa, A. Minami et al., “AMP-activated protein kinase is activated by the stimulations of G(q)-coupled receptors,” Biochemical and Biophysical Research Communications, vol. 276, no. 1, pp. 16–22, 2000.
[74]  S. Theodoropoulou, K. Brodowska, M. Kayama, et al., “Aminoimidazole carboxamide ribonucleotide (AICAR) inhibits the growth of retinoblastoma in vivo by decreasing angiogenesis and inducing apoptosis,” PLoS ONE, vol. 8, no. 1, Article ID e52852, 2013.
[75]  D. L. Williamson, D. C. Butler, and S. E. Alway, “AMPK inhibits myoblast differentiation through a PGC-1α-dependent mechanism,” The American Journal of Physiology: Endocrinology and Metabolism, vol. 297, no. 2, pp. E304–E314, 2009.
[76]  W. H. Gotlieb, J. Saumet, M.-C. Beauchamp et al., “In vitro metformin anti-neoplastic activity in epithelial ovarian cancer,” Gynecologic Oncology, vol. 110, no. 2, pp. 246–250, 2008.
[77]  Y. Takiyama, T. Harumi, J. Watanabe et al., “Tubular injury in a rat model of type 2 diabetes is prevented by metformin: a possible role of HIF-1α expression and oxygen metabolism,” Diabetes, vol. 60, no. 3, pp. 981–992, 2011.
[78]  J. M. M. Evans, L. A. Donnelly, A. M. Emslie-Smith, D. R. Alessi, and A. D. Morris, “Metformin and reduced risk of cancer in diabetic patients,” British Medical Journal, vol. 330, no. 7503, pp. 1304–1305, 2005.
[79]  K. Janjetovic, L. Harhaji-Trajkovic, M. Misirkic-Marjanovic et al., “In vitro and in vivo anti-melanoma action of metformin,” European Journal of Pharmacology, vol. 668, no. 3, pp. 373–382, 2011.
[80]  G. Wolf and S. J. Shankland, “p27Kip1: the “Rosebud” of diabetic nephropathy?” Journal of the American Society of Nephrology, vol. 14, no. 3, pp. 819–822, 2003.
[81]  G. Wolf and F. N. Ziyadeh, “Cellular and molecular mechanisms of proteinuria in diabetic nephropathy,” Nephron Physiology, vol. 106, no. 2, pp. 26–31, 2007.
[82]  X. Zhang, X. Chen, D. Wu et al., “Downregulation of connexin 43 expression by high glucose induces senescence in glomerular mesangial cells,” Journal of the American Society of Nephrology, vol. 17, no. 6, pp. 1532–1542, 2006.
[83]  O. Cazzalini, A. I. Scovassi, M. Savio, L. A. Stivala, and E. Prosperi, “Multiple roles of the cell cycle inhibitor p21CDKN1A in the DNA damage response,” Mutation Research, vol. 704, no. 1–3, pp. 12–20, 2010.
[84]  Y. Wang, Y. Liang, and P. M. Vanhoutte, “SIRT1 and AMPK in regulating mammalian senescence: a critical review and a working model,” FEBS Letters, vol. 585, no. 7, pp. 986–994, 2011.
[85]  A. Morocutti, K. A. Earle, H. P. Rodemann, and G. C. Viberti, “Premature cell ageing and evolution of diabetic nephropathy,” Diabetologia, vol. 40, no. 2, pp. 244–246, 1997.
[86]  A. Morocutti, K. A. Earle, M. Sethi et al., “Premature senescence of skin fibroblasts from insulin-dependent diabetic patients with kidney disease,” Kidney International, vol. 50, no. 1, pp. 250–256, 1996.
[87]  D. Verzola, M. T. Gandolfo, G. Gaetani et al., “Accelerated senescence in the kidneys of patients with type 2 diabetic nephropathy,” The American Journal of Physiology: Renal Physiology, vol. 295, no. 5, pp. F1563–F1573, 2008.
[88]  K. Sawai, M. Mukoyama, K. Mori et al., “Redistribution of connexin43 expression in glomerular podocytes predicts poor renal prognosis in patients with type 2 diabetes and overt nephropathy,” Nephrology Dialysis Transplantation, vol. 21, no. 9, pp. 2472–2477, 2006.
[89]  L. Liu, X. Hu, G.-Y. Cai et al., “High glucose-induced hypertrophy of mesangial cells is reversed by connexin43 overexpression via PTEN/Akt/mTOR signaling,” Nephrology Dialysis Transplantation, vol. 27, no. 1, pp. 90–100, 2012.
[90]  P. Atadja, H. Wong, C. Veillete, and K. Riabowol, “Overexpression of cyclin D1 blocks proliferation of normal diploid fibroblasts,” Experimental Cell Research, vol. 217, no. 2, pp. 205–216, 1995.
[91]  T. Yokoi, K. Fukuo, O. Yasuda et al., “Apoptosis signal-regulating kinase 1 mediates cellular senescence induced by high glucose in endothelial cells,” Diabetes, vol. 55, no. 6, pp. 1660–1665, 2006.
[92]  A. Vogetseder, T. Palan, D. Bacic, B. Kaissling, and M. Le Hir, “Proximal tubular epithelial cells are generated by division of differentiated cells in the healthy kidney,” The American Journal of Physiology: Cell Physiology, vol. 292, no. 2, pp. C807–C813, 2007.
[93]  A. Vogetseder, N. Picard, A. Gaspert, M. Walch, B. Kaissling, and M. Le Hir, “Proliferation capacity of the renal proximal tubule involves the bulk of differentiated epithelial cells,” The American Journal of Physiology: Cell Physiology, vol. 294, no. 1, pp. C22–C28, 2008.
[94]  T. Kumazaki, R. S. Robetorye, S. C. Robetorye, and J. R. Smith, “Fibronectin expression increases during in vitro cellular senescence: correlation with increased cell area,” Experimental Cell Research, vol. 195, no. 1, pp. 13–19, 1991.
[95]  N. Chondrogianni, F. L. L. Stratford, I. P. Trougakos, B. Friguet, A. J. Rivett, and E. S. Gonos, “Central role of the proteasome in senescence and survival of human fibroblasts. Induction of a senescence-like phenotype upon its inhibition and resistance to stress upon its activation,” Journal of Biological Chemistry, vol. 278, no. 30, pp. 28026–28037, 2003.
[96]  D.-S. Jung, S. H. Lee, S.-J. Kwak et al., “Apoptosis occurs differentially according to glomerular size in diabetic kidney disease,” Nephrology Dialysis Transplantation, vol. 27, no. 1, pp. 259–266, 2012.
[97]  D. Kumar, S. Robertson, and K. D. Burns, “Evidence of apoptosis in human diabetic kidney,” Molecular and Cellular Biochemistry, vol. 259, no. 1-2, pp. 67–70, 2004.
[98]  M. D. Sanchez-Ni? O, A. B. Sanz, C. Lorz et al., “BASP1 promotes apoptosis in diabetic nephropathy,” Journal of the American Society of Nephrology, vol. 21, no. 4, pp. 610–621, 2010.
[99]  J. M. Vicencio, L. Galluzzi, N. Tajeddine et al., “Senescence, apoptosis or autophagy? When a damaged cell must decide its path—a mini-review,” Gerontology, vol. 54, no. 2, pp. 92–99, 2008.
[100]  M. E. Pagtalunan, P. L. Miller, S. Jumping-Eagle et al., “Podocyte loss and progressive glomerular injury in type II diabetes,” Journal of Clinical Investigation, vol. 99, no. 2, pp. 342–348, 1997.
[101]  M. W. Steffes, D. Schmidt, R. McCrery et al., “Glomerular cell number in normal subjects and in type 1 diabetic patients,” Kidney International, vol. 59, no. 6, pp. 2104–2113, 2001.
[102]  Z. N. Demidenko, S. G. Zubova, E. I. Bukreeva, V. A. Pospelov, T. V. Pospelova, and M. V. Blagosklonny, “Rapamycin decelerates cellular senescence,” Cell Cycle, vol. 8, no. 12, pp. 1888–1895, 2009.
[103]  S. Cufí, A. Vazquez-Martin, C. Oliveras-Ferraros et al., “Metformin lowers the threshold for stress-induced senescence: a role for the microRNA-200 family and miR-205,” Cell Cycle, vol. 11, no. 6, pp. 1235–1246, 2012.
[104]  M. Law, E. Forrester, A. Chytil et al., “Rapamycin disrupts cyclin/cyclin-dependent kinase/p21/proliferating cell nuclear antigen complexes and cyclin D1 reverses rapamycin action by stabilizing these complexes,” Cancer Research, vol. 66, no. 2, pp. 1070–1080, 2006.
[105]  A. Salminen and K. Kaarniranta, “AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network,” Ageing Research Reviews, vol. 11, no. 2, pp. 230–241, 2012.
[106]  K. R. Hallows, P. F. Mount, N. M. Pastor-Soler, and D. A. Power, “Role of the energy sensor AMP-activated protein kinase in renal physiology and disease,” The American Journal of Physiology: Renal Physiology, vol. 298, no. 5, pp. F1067–F1077, 2010.
[107]  R. S. Balaban and L. J. Mandel, “Metabolic substrate utilization by rabbit proximal tubule - an NADH fluorescence,” The American Journal of Physiology: Renal Fluid and Electrolyte Physiology, vol. 254, no. 3, pp. F407–F416, 1988.
[108]  L. Meury, J. Noel, A. Tejedor, J. Senecal, A. Gougoux, and P. Vinay, “Glucose metabolism in dog inner medullary collecting ducts,” Renal Physiology and Biochemistry, vol. 17, no. 5, pp. 246–266, 1994.
[109]  J. A. Menendez, L. Vellon, C. Oliveras-Ferraros, S. Cufí, and A. Vazquez-Martin, “mTOR-regulated senescence and autophagy during reprogramming of somatic cells to pluripotency: a roadmap from energy metabolism to stem cell renewal and aging,” Cell Cycle, vol. 10, no. 21, pp. 3658–3677, 2011.
[110]  A. Prigione, B. Fauler, R. Lurz, H. Lehrach, and J. Adjaye, “The senescence-related mitochondrial/oxidative stress pathway is repressed in human induced pluripotent stem cells,” Stem Cells, vol. 28, no. 4, pp. 721–733, 2010.
[111]  H. Kondoh, M. E. Lleonart, J. Gil et al., “Glycolytic enzymes can modulate cellular life span,” Cancer Research, vol. 65, no. 1, pp. 177–185, 2005.
[112]  I. B. Sahra, Y. L. Marchand-Brustel, J.-F. Tanti, and F. Bost, “Metformin in cancer therapy: a new perspective for an old antidiabetic drug?” Molecular Cancer Therapeutics, vol. 9, no. 5, pp. 1092–1099, 2010.
[113]  O. Moiseeva, X. Deschênes-Simard, E. St-Germain et al., “Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation,” Aging Cell, vol. 12, no. 3, pp. 489–498, 2013.
[114]  A. A. Alhaider, H. M. Korashy, M. M. Sayed-Ahmed, M. Mobark, H. Kfoury, and M. A. Mansour, “Metformin attenuates streptozotocin-induced diabetic nephropathy in rats through modulation of oxidative stress genes expression,” Chemico-Biological Interactions, vol. 192, no. 3, pp. 233–242, 2011.
[115]  M.-J. Lee, D. Feliers, M. M. Mariappan et al., “A role for AMP-activated protein kinase in diabetes-induced renal hypertrophy,” The American Journal of Physiology: Renal Physiology, vol. 292, no. 2, pp. F617–F627, 2007.
[116]  M. Kaur, G. Deep, A. K. Jain et al., “Bitter melon juice activates cellular energy sensor AMP-activated protein kinase causing apoptotic death of human pancreatic carcinoma cells,” Carcinogenesis, vol. 34, no. 7, pp. 1585–1592, 2013.
[117]  S. Kume, M. C. Thomas, and D. Koya, “Nutrient sensing, autophagy, and diabetic nephropathy,” Diabetes, vol. 61, no. 1, pp. 23–29, 2012.
[118]  R. J. Shaw and L. C. Cantley, “Ancient sensor for ancient drug,” Science, vol. 336, no. 6083, pp. 813–814, 2012.
[119]  R. Scacchi, G. Gambina, G. Moretto, and R. M. Corbo, “P21 gene variation and late-onset Alzheimer's disease in the Italian population,” Dementia and Geriatric Cognitive Disorders, vol. 35, no. 1-2, pp. 51–57, 2013.
[120]  M. E. Seward, E. Swanson, A. Norambuena et al., “Amyloid-beta signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer's disease,” Journal of Cell Science, vol. 126, no. 5, pp. 1278–1286, 2013.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133