Skip to main content

Secreted Klotho and Chronic Kidney Disease

  • Chapter
Endocrine FGFs and Klothos

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 728))

Abstract

Soluble Klotho (sKl) in the circulation can be generated directly by alterative splicing of the Klotho transcript or the extracellular domain of membrane Klotho can be released from membrane-anchored Klotho on the cell surface. Unlike membrane Klotho which functions as a coreceptor for fibroblast growth factor-23 (FGF23), sKl, acts as hormonal factor and plays important roles in anti-aging, anti-oxidation, modulation of ion transport, and Wnt signaling. Emerging evidence reveals that Klotho deficiency is an early biomarker for chronic kidney diseases as well as a pathogenic factor. Klotho deficiency is associated with progression and chronic complications in chronic kidney disease including vascular calcification, cardiac hypertrophy, and secondary hyperparathyroidism. In multiple experimental models, replacement of sKl, or manipulated up-regulation of endogenous Klotho protect the kidney from renal insults, preserve kidney function, and suppress renal fibrosis, in chronic kidney disease. Klotho is a highly promising candidate on the horizon as an early biomarker, and as a novel therapeutic agent for chronic kidney disease.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 129.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 169.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Koh N, Fujimori T, Nishiguchi S et al. Severely reduced production of klotho in human chronic renal failure kidney. Biochem Biophys Res Commun 2001; 280(4):1015–1020.

    PubMed  CAS  Google Scholar 

  2. Hu MC, Shi M, Zhang J et al. Klotho deficiency causes vascular calcification in chronic kidney disease. J Am Soc Nephrol 2011; 22(1):124–136.

    PubMed  CAS  Google Scholar 

  3. Aizawa H, Saito Y, Nakamura T et al. Downregulation of the Klotho gene in the kidney under sustained circulatory stress in rats. Biochem Biophys Res Commun 1998; 249(3):865–871.

    PubMed  CAS  Google Scholar 

  4. Matsumura Y, Aizawa H, Shiraki-Iida T et al. Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein. Biochem Biophys Res Commun 1998; 242(3):626–630.

    PubMed  CAS  Google Scholar 

  5. Shiraki-Iida T, Aizawa H, Matsumura Y et al. Structure of the mouse klotho gene and its two transcripts encoding membrane and secreted protein. FEBS Lett 1998; 424(1–2):6–10.

    PubMed  CAS  Google Scholar 

  6. Kuro-o M, Matsumura Y, Aizawa H et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997; 390(6655):45–51.

    PubMed  CAS  Google Scholar 

  7. Tohyama O, Imura A, Iwano A et al. Klotho is a novel beta-glucuronidase capable of hydrolyzing steroid beta-glucuronides. J Biol Chem 2004; 279(11):9777–9784.

    PubMed  CAS  Google Scholar 

  8. Chen CD, Podvin S, Gillespie E et al. Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci USA 2007; 104(50):19796–19801.

    PubMed  CAS  Google Scholar 

  9. Bloch L, Sineshchekova O, Reichenbach D et al. Klotho is a substrate for alpha-, beta-and gamma-secretase. FEBS Lett 2009; 583(19):3221–3224.

    PubMed  CAS  Google Scholar 

  10. Huang CL. Regulation of ion channels by secreted Klotho: mechanisms and implications. Kidney Int 2010; 77(10):855–860.

    PubMed  Google Scholar 

  11. Liu H, Fergusson MM, Castilho RM et al. Augmented Wnt signaling in a mammalian model of accelerated aging. Science 2007; 317(5839):803–806.

    PubMed  CAS  Google Scholar 

  12. Goetz R, Nakada Y, Hu MC et al. Isolated C-terminal tail of FGF23 alleviates hypophosphatemia by inhibiting FGF23-FGFR-Klotho complex formation. Proc Natl Acad Sci USA 2010; 107(1):407–412.

    PubMed  CAS  Google Scholar 

  13. Carpenter TO, Insogna KL, Zhang JH et al. Circulating Levels of Soluble Klotho and FGF23 in X-Linked Hypophosphatemia: Circadian Variance, Effects of Treatment and Relationship to Parathyroid Status. J Clin Endocrinol Metab 2010; 95(11):E352–E357.

    PubMed  Google Scholar 

  14. Hu MC, Shi M, Zhang J et al. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J 2010; 24(9):3438–3450.

    PubMed  CAS  Google Scholar 

  15. Cha SK, Hu MC, Kurosu H et al. Regulation of renal outer medullary potassium channel and renal K(?) excretion by Klotho. Mol Pharmacol 2009; 76(1):38–46.

    PubMed  CAS  Google Scholar 

  16. Cha SK, Ortega B, Kurosu H et al. Removal of sialic acid involving Klotho causes cell-surface retention of TRPV5 channel via binding to galectin-1. Proc Natl Acad Sci USA 2008; 105(28):9805–9810.

    PubMed  CAS  Google Scholar 

  17. Kato Y, Arakawa E, Kinoshita S et al. Establishment of the anti-Klotho monoclonal antibodies and detection of Klotho protein in kidneys. Biochem Biophys Res Commun 2000; 267(2):597–602.

    PubMed  CAS  Google Scholar 

  18. Haruna Y, Kashihara N, Satoh M et al. Amelioration of progressive renal injury by genetic manipulation of Klotho gene. Proc Natl Acad Sci USA 2007; 104(7):2331–2336.

    PubMed  CAS  Google Scholar 

  19. Saito Y, Nakamura T, Ohyama Y et al. In vivo klotho gene delivery protects against endothelial dysfunction in multiple risk factor syndrome. Biochem Biophys Res Commun 2000; 276(2):767–772.

    PubMed  CAS  Google Scholar 

  20. Kurosu H, Yamamoto M, Clark JD et al. Suppression of aging in mice by the hormone Klotho. Science 2005; 309(5742):1829–1833.

    PubMed  CAS  Google Scholar 

  21. Mitani H, Ishizaka N, Aizawa T et al. In vivo klotho gene transfer ameliorates angiotensin II-induced renal damage. Hypertension 2002; 39(4):838–843.

    PubMed  CAS  Google Scholar 

  22. Mitobe M, Yoshida T, Sugiura H et al. Oxidative stress decreases klotho expression in a mouse kidney cell line. Nephron Exp Nephrol 2005; 101(2):e67–e74.

    PubMed  CAS  Google Scholar 

  23. Sugiura H, Yoshida T, Tsuchiya K et al. Klotho reduces apoptosis in experimental ischaemic acute renal failure. Nephrol Dial Transplant 2005; 20(12):2636–2645.

    PubMed  CAS  Google Scholar 

  24. Chang Q, Hoefs S, van der Kemp AW et al. The beta-glucuronidase klotho hydrolyzes and activates the TRPV5 channel. Science 2005; 310(5747):490–493.

    PubMed  CAS  Google Scholar 

  25. Yoshida T, Fujimori T, Nabeshima Y. Mediation of unusually high concentrations of 1,25-dihydroxy vitamin D in homozygous klotho mutant mice by increased expression of renal 1alpha-hydroxylase gene. Endocrinology 2002; 143(2):683–689.

    PubMed  CAS  Google Scholar 

  26. Tsujikawa H, Kurotaki Y, Fujimori T et al. Klotho, a gene related to a syndrome resembling human premature aging, functions in a negative regulatory circuit of vitamin D endocrine system. Mol Endocrinol 2003; 17(12):2393–2403.

    PubMed  CAS  Google Scholar 

  27. Silver J, Naveh-Many T. FGF23 and the parathyroid glands. Pediatr Nephrol 2010; 25(11):2241–2245.

    PubMed  Google Scholar 

  28. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V et al. The parathyroid is a target organ for FGF23 in rats. J Clin Invest 2007; 117(12):4003–4008.

    PubMed  CAS  Google Scholar 

  29. Invidia L, Salvioli S, Altilia S et al. The frequency of Klotho KL-VS polymorphism in a large Italian population, from young subjects to centenarians, suggests the presence of specific time windows for its effect. Biogerontology 2010; 11(1):67–73.

    PubMed  Google Scholar 

  30. Zhang F, Zhai G, Kato BS et al. Association between KLOTHO gene and hand osteoarthritis in a female Caucasian population. Osteoarthritis Cartilage 2007.

    Google Scholar 

  31. Rhee EJ, Oh KW, Lee WY et al. The differential effects of age on the association of KLOTHO gene polymorphisms with coronary artery disease. Metabolism 2006; 55(10):1344–1351.

    PubMed  CAS  Google Scholar 

  32. Rhee EJ, Oh KW, Yun EJ et al. Relationship between polymorphisms G395A in promoter and C1818T in exon 4 of the KLOTHO gene with glucose metabolism and cardiovascular risk factors in Korean women. J Endocrinol Invest 2006; 29(7):613–618.

    PubMed  CAS  Google Scholar 

  33. Nolan VG, Adewoye A, Baldwin C et al. Sickle cell leg ulcers: associations with haemolysis and SNPs in Klotho, TEK and genes of the TGF-beta/BMP pathway. Br J Haematol 2006; 133(5):570–578.

    PubMed  CAS  Google Scholar 

  34. Arking DE, Becker DM, Yanek LR et al. KLOTHO allele status and the risk of early-onset occult coronary artery disease. Am J Hum Genet 2003; 72(5):1154–1161.

    PubMed  CAS  Google Scholar 

  35. Arking DE, Krebsova A, Macek M Sr et al. Association of human aging with a functional variant of klotho. Proc Natl Acad Sci USA 2002; 99(2):856–861.

    PubMed  CAS  Google Scholar 

  36. Kawano K, Ogata N, Chiano M et al. Klotho gene polymorphisms associated with bone density of aged postmenopausal women. J Bone Miner Res 2002; 17(10):1744–1751.

    PubMed  CAS  Google Scholar 

  37. Yu J, Deng M, Zhao J et al. Decreased expression of klotho gene in uremic atherosclerosis in apolipoprotein E-deficient mice. Biochem Biophys Res Commun 2010; 391(1):261–266.

    PubMed  CAS  Google Scholar 

  38. Friedman DJ, Afkarian M, Tamez H et al. Klotho variants and chronic hemodialysis mortality. J Bone Miner Res 2009; 24(11):1847–1855.

    PubMed  CAS  Google Scholar 

  39. Saito K, Ishizaka N, Mitani H et al. Iron chelation and a free radical scavenger suppress angiotensin II-induced downregulation of klotho, an anti-aging gene, in rat. FEBS Lett 2003; 551(1–3):58–62.

    PubMed  CAS  Google Scholar 

  40. Cheng MF, Chen LJ, Cheng JT. Decrease of Klotho in the kidney of streptozotocin-induced diabetic rats. J Biomed Biotechnol 2010; 2010:513853.

    PubMed  Google Scholar 

  41. Han DH, Piao SG, Song JH et al. Effect of sirolimus on calcineurin inhibitor-induced nephrotoxicity using renal expression of KLOTHO, an antiaging gene. Transplantation 2010; 90(2):135–141.

    PubMed  CAS  Google Scholar 

  42. Yoon HE, Ghee JY, Piao S et al. Angiotensin II blockade upregulates the expression of Klotho the anti-ageing gene, in an experimental model of chronic cyclosporine nephropathy. Nephrol Dial Transplant 2011; 26(3):800–813.

    PubMed  CAS  Google Scholar 

  43. Yamazaki Y, Imura A, Urakawa I et al. Establishment of sandwich ELISA for soluble alpha-Klotho measurement: Age-dependent change of soluble alpha-Klotho levels in healthy subjects. Biochem Biophys Res Commun 2010; 398(3):513–518.

    PubMed  CAS  Google Scholar 

  44. Zuo Z, Lei H, Wang X et al. Aging-related kidney damage is associated with a decrease in klotho expression and an increase in superoxide production. Age (Dordr) 2010; Epub ahead of press.

    Google Scholar 

  45. Ichikawa S, Imel EA, Kreiter ML et al. A homozygous missense mutation in human KLOTHO causes severe tumoral calcinosis. J Clin Invest 2007; 117(9):2684–2691.

    PubMed  CAS  Google Scholar 

  46. Manya H, Akasaka-Manya K, Endo T. Klotho protein deficiency and aging. Geriatr Gerontol Int 2010; 10 Suppl 1:S80–S87.

    PubMed  Google Scholar 

  47. Brownstein CA, Adler F, Nelson-Williams C et al. A translocation causing increased alpha-klotho level results in hypophosphatemic rickets and hyperparathyroidism. Proc Natl Acad Sci USA 2008; 105(9):3455–3460.

    PubMed  CAS  Google Scholar 

  48. Takeshita K, Fujimori T, Kurotaki Y et al. Sinoatrial node dysfunction and early unexpected death of mice with a defect of klotho gene expression. Circulation 2004; 109(14):1776–1782.

    PubMed  Google Scholar 

  49. Imura A, Iwano A, Tohyama O et al. Secreted Klotho protein in sera and CSF: implication for posttranslational cleavage in release of Klotho protein from cell membrane. FEBS Lett 2004; 565(1–3):143–147.

    PubMed  CAS  Google Scholar 

  50. Hu MC, Shi M, Zhang J et al. Klotho deficiency is an early biomarker of renal ischemia-reperfusion injury and its replacement is protective. Kidney Int 2010; 78(12):1240–1251.

    PubMed  CAS  Google Scholar 

  51. Schramme A, Abdel-Bakky MS, Gutwein P et al. Characterization of CXCL16 and ADAM10 in the normal and transplanted kidney. Kidney Int 2008; 74(3):328–338.

    PubMed  CAS  Google Scholar 

  52. Kuro-o M. Klotho as a regulator of oxidative stress and senescence. Biol Chem 2008; 389(3):233–241.

    PubMed  CAS  Google Scholar 

  53. Huang BS, Leenen FH. Dietary Na, age and baroreflex control of heart rate and renal sympathetic nerve activity in rats. Am J Physiol 1992; 262(5 Pt 2):H1441–H1448.

    PubMed  CAS  Google Scholar 

  54. Yamazaki M, Ozono K, Okada T et al. Both FGF23 and extracellular phosphate activate Raf/MEK/ERK pathway via FGF receptors in HEK293 cells. J Cell Biochem 2010; 111(5):1210–1221.

    PubMed  CAS  Google Scholar 

  55. Mazzaferro S, Pasquali M, Pirro G et al. The bone and the kidney. Arch Biochem Biophys 2010; 503(1):95–102.

    PubMed  CAS  Google Scholar 

  56. Keisala T, Minasyan A, Lou YR et al. Premature aging in vitamin D receptor mutant mice. J Steroid Biochem Mol Biol 2009; 115(3-5):91–97.

    PubMed  CAS  Google Scholar 

  57. Chattopadhyay N, Baum M, Bai M et al. Ontogeny of the extracellular calcium-sensing receptor in rat kidney. Am J Physiol 1996; 271(3 Pt 2):F736–F743.

    Google Scholar 

  58. Biber J, Hernando N, Forster I et al. Regulation of phosphate transport in proximal tubules. Pflugers Arch 2009; 458(1):39–52.

    PubMed  CAS  Google Scholar 

  59. Moe OW. PiT-2 coming out of the pits. Am J Physiol Renal Physiol 2009; 296(4):F689–F690.

    PubMed  CAS  Google Scholar 

  60. Panda DK, Al Kawas S, Seldin MF et al. 25-hydroxyvitamin D 1alpha-hydroxylase: structure of the mouse gene, chromosomal assignment, and developmental expression. J Bone Miner Res 2001; 16(1):46–56.

    PubMed  CAS  Google Scholar 

  61. Lai WP, Chau TS, Cheung PY et al. Adaptive responses of 25-hydroxyvitamin D3 1-alpha hydroxylase expression to dietary phosphate restriction in young and adult rats. Biochim Biophys Acta 2003; 1639(1):34–42.

    PubMed  CAS  Google Scholar 

  62. Shimada T, Urakawa I, Isakova T et al. Circulating fibroblast growth factor 23 in patients with end-stage renal disease treated by peritoneal dialysis is intact and biologically active. J Clin Endocrinol Metab 2010; 95(2):578–585.

    PubMed  CAS  Google Scholar 

  63. Fukagawa M, Kazama JJ. With or without the kidney: the role of FGF23 in CKD. Nephrol Dial Transplant 2005; 20(7):1295–1298.

    PubMed  CAS  Google Scholar 

  64. Weber TJ, Liu S, Indridason OS et al. Serum FGF23 levels in normal and disordered phosphorus homeostasis. J Bone Miner Res 2003; 18(7):1227–1234.

    PubMed  CAS  Google Scholar 

  65. Nakatani T, Sarraj B, Ohnishi M et al. In vivo genetic evidence for klotho-dependent, fibroblast growth factor 23 (Fgf23) — mediated regulation of systemic phosphate homeostasis. FASEB J 2009; 23(2):433–441.

    PubMed  CAS  Google Scholar 

  66. Jono S, Shioi A, Ikari Y et al. Vascular calcification in chronic kidney disease. J Bone Miner Metab 2006; 24(2):176–181.

    PubMed  Google Scholar 

  67. London GM. Cardiovascular calcifications in uremic patients: clinical impact on cardiovascular function. J Am Soc Nephrol 2003; 14(9 Suppl 4):S305–S309.

    PubMed  Google Scholar 

  68. Weinstein JR, Anderson S. The aging kidney: physiological changes. Adv Chronic Kidney Dis 2010; 17(4):302–307.

    PubMed  Google Scholar 

  69. Martin JE, Sheaff MT. Renal ageing. J Pathol 2007; 211(2):198–205.

    Google Scholar 

  70. Lerma EV. Anatomic and physiologic changes of the aging kidney. Clin Geriatr Med 2009; 25(3):325–329.

    PubMed  Google Scholar 

  71. Chen G, Bridenbaugh EA, Akintola AD et al. Increased susceptibility of aging kidney to ischemic injury: identification of candidate genes changed during aging, but corrected by caloric restriction. Am J Physiol Renal Physiol 2007; 293(4):F1272–F1281.

    PubMed  CAS  Google Scholar 

  72. Musso CG, Liakopoulos V, Ioannidis I et al. Acute renal failure in the elderly: particular characteristics. Int Urol Nephrol 2006; 38(3–4):787–793.

    PubMed  Google Scholar 

  73. Campbell KH, O’Hare AM. Kidney disease in the elderly: update on recent literature. Curr Opin Nephrol Hypertens 2008; 17(3):298–303.

    PubMed  Google Scholar 

  74. Hu MC, Bankir L, Michelet S et al. Massive reduction of urea transporters in remnant kidney and brain of uremic rats. Kidney Int 2000; 58(3):1202–1210.

    PubMed  CAS  Google Scholar 

  75. Tamura K, Suzuki Y, Matsushita M et al. Prevention of aortic calcification by etidronate in the renal failure rat model. Eur J Pharmacol 2007; 558(1–3):159–166.

    PubMed  CAS  Google Scholar 

  76. Peralta CA, Kurella M, Lo JC et al. The metabolic syndrome and chronic kidney disease. Curr Opin Nephrol Hypertens 2006; 15(4):361–365.

    PubMed  CAS  Google Scholar 

  77. Nagase M, Yoshida S, Shibata S et al. Enhanced aldosterone signaling in the early nephropathy of rats with metabolic syndrome: possible contribution of fat-derived factors. J Am Soc Nephrol 2006; 17(12):3438–3446.

    PubMed  CAS  Google Scholar 

  78. Locatelli F, Pozzoni P, Del Vecchio L. Renal manifestations in the metabolic syndrome. J Am Soc Nephrol 2006; 17(4 Suppl 2):S81–S85.

    PubMed  Google Scholar 

  79. Kawano K, Hirashima T, Mori S et al. Spontaneous long-term hyperglycemic rat with diabetic complications. Otsuka Long-Evans Tokushima Fatty (OLETF) strain. Diabetes 1992; 41(11):1422–1428.

    PubMed  CAS  Google Scholar 

  80. Yamagishi T, Saito Y, Nakamura T et al. Troglitazone improves endothelial function and augments renal klotho mRNA expression in Otsuka Long-Evans Tokushima Fatty (OLETF) rats with multiple atherogenic risk factors. Hypertens Res 2001; 24(6):705–709.

    PubMed  CAS  Google Scholar 

  81. Ohyama Y, Kurabayashi M, Masuda H et al. Molecular cloning of rat klotho cDNA: markedly decreased expression of klotho by acute inflammatory stress. Biochem Biophys Res Commun 1998; 251(3):920–925.

    PubMed  CAS  Google Scholar 

  82. Thurston RD, Larmonier CB, Majewski PM et al. Tumor necrosis factor and interferon-gamma down-regulate Klotho in mice with colitis. Gastroenterology 2010; 138(4):1384–1394.

    PubMed  CAS  Google Scholar 

  83. Webster AC, Woodroffe RC, Taylor RS et al. Tacrolimus versus ciclosporin as primary immunosuppression for kidney transplant recipients: meta-analysis and meta-regression of randomised trial data. BMJ 2005; 331(7520):810.

    PubMed  CAS  Google Scholar 

  84. Jiang H, Sakuma S, Fujii Y et al. Tacrolimus versus cyclosporin A: a comparative study on rat renal allograft survival. Transpl Int 1999; 12(2):92–99.

    PubMed  CAS  Google Scholar 

  85. Gulati S, Prasad N, Sharma RK et al. Tacrolimus: a new therapy for steroid-resistant nephrotic syndrome in children. Nephrol Dial Transplant 2008; 23(3):910–913.

    PubMed  CAS  Google Scholar 

  86. Tang S, Tang AW, Tam MK et al. Use of tacrolimus in steroid-and cyclophosphamide-resistant minimal change nephrotic syndrome. Am J Kidney Dis 2003; 42(5):E13–E15.

    PubMed  Google Scholar 

  87. Ziolkowski J, Paczek L, Senatorski G et al. Renal function after liver transplantation: calcineurin inhibitor nephrotoxicity. Transplant Proc 2003; 35(6):2307–2309.

    PubMed  CAS  Google Scholar 

  88. Naesens M, Kuypers DR, Sarwal M. Calcineurin inhibitor nephrotoxicity. Clin J Am Soc Nephrol 2009; 4(2):481–508.

    PubMed  CAS  Google Scholar 

  89. Formica RN Jr, Lorber KM, Friedman AL et al. Sirolimus-based immunosuppression with reduce dose cyclosporine or tacrolimus after renal transplantation. Transplant Proc 2003; 35(3 Suppl):95S–98S.

    PubMed  CAS  Google Scholar 

  90. Kestenbaum B, Sampson JN, Rudser KD et al. Serum phosphate levels and mortality risk among people with chronic kidney disease. J Am Soc Nephrol 2005; 16(2):520–528.

    PubMed  CAS  Google Scholar 

  91. Cannata-Andia JB, Rodriguez-Garcia M. Hyperphosphataemia as a cardiovascular risk factor — how to manage the problem. Nephrol Dial Transplant 2002; 17 Suppl 11:16–19.

    PubMed  CAS  Google Scholar 

  92. Isakova T, Gutierrez OM, Chang Y et al. Phosphorus binders and survival on hemodialysis. J Am Soc Nephrol 2009; 20(2):388–396.

    Google Scholar 

  93. Kuro-o M. A potential link between phosphate and aging-lessons from Klotho-deficient mice. Mech Ageing Dev 2010; 131(4):270–275.

    PubMed  CAS  Google Scholar 

  94. Morishita K, Shirai A, Kubota M et al. The progression of aging in klotho mutant mice can be modified by dietary phosphorus and zinc. J Nutr 2001; 131(12):3182–3188.

    PubMed  CAS  Google Scholar 

  95. Lips P. Vitamin D deficiency and secondary hyperparathyroidism in the elderly: consequences for bone loss and fractures and therapeutic implications. Endocr Rev 2001; 22(4):477–501.

    PubMed  CAS  Google Scholar 

  96. Gutierrez OM. Fibroblast growth factor 23 and disordered vitamin D metabolism in chronic kidney disease: updating the “trade-off” hypothesis. Clin J Am Soc Nephrol 2010; 5(9):1710–1716.

    PubMed  CAS  Google Scholar 

  97. Liu S, Tang W, Zhou J et al. Fibroblast growth factor 23 is a counter-regulatory phosphaturic hormone for vitamin D. J Am Soc Nephrol 2006; 17(5):1305–1315.

    PubMed  CAS  Google Scholar 

  98. Palm F, Nangaku M, Fasching A et al. Uremia induces abnormal oxygen consumption in tubules and aggravates chronic hypoxia of the kidney via oxidative stress. Am J Physiol Renal Physiol 2010; 299(2):F380–F386.

    PubMed  CAS  Google Scholar 

  99. Polichnowski AJ, Jin C, Yang C et al. Role of renal perfusion pressure versus angiotensin II on renal oxidative stress in angiotensin II-induced hypertensive rats. Hypertension 2010; 55(6):1425–1430.

    Google Scholar 

  100. Yu M, Kim YJ, Kang DH. Indoxyl Sulfate-Induced Endothelial Dysfunction in Patients with Chronic Kidney Disease via an Induction of Oxidative Stress. Clin J Am Soc Nephrol 2011: 6(1):30–39.

    PubMed  CAS  Google Scholar 

  101. Barreto FC, Barreto DV, Liabeuf S et al. Serum indoxyl sulfate is associated with vascular disease and mortality in chronic kidney disease patients. Clin J Am Soc Nephrol 2009; 4(10):1551–1558.

    PubMed  CAS  Google Scholar 

  102. Niwa T, Adijiang A, Higuchi Y et al. Indoxyl Sulfate Reduces Klotho Expression and Promotes Senescence in the Kidney of Hypertensive Rats (Abstract). J Am Soc Nephrol 2010; 21:746A.

    Google Scholar 

  103. Jennings P, Koppelstaetter C, Aydin S et al. Cyclosporine A induces senescence in renal tubular epithelial cells. Am J Physiol Renal Physiol 2007; 293(3):F831–F838.

    PubMed  CAS  Google Scholar 

  104. Kailong L, Du X, Yani H et al. P53-Rb signaling pathway is involved in tubular cell senescence in renal ischemia/reperfusion injury. Biocell 2007; 31(2):213–223.

    PubMed  Google Scholar 

  105. Dmitrieva NI, Burg MB. High NaCl promotes cellular senescence. Cell Cycle 2007; 66(24):3108–3113.

    Google Scholar 

  106. Yang H, Fogo AB. Cell senescence in the aging kidney. J Am Soc Nephrol 2010; 21(9):1436–1439.

    PubMed  Google Scholar 

  107. Nakano-Kurimoto R, Ikeda K, Uraoka M et al. Replicative senescence of vascular smooth muscle cells enhances the calcification through initiating the osteoblastic transition. Am J Physiol Heart Circ Physiol 2009; 297(5):H1673–H1684.

    PubMed  CAS  Google Scholar 

  108. Yang HC, Deleuze S, Zuo Y et al. The PPARgamma agonist pioglitazone ameliorates aging-related progressive renal injury. J Am Soc Nephrol 2009; 20(11):2380–2388.

    Google Scholar 

  109. Shimizu H, Bolati D, Adijiang A et al. Senescence and dysfunction of proximal tubular cells are associated with activated p53 expression by indoxyl sulfate. Am J Physiol Cell Physiol 2010; 299(5):C1110–C1117.

    PubMed  CAS  Google Scholar 

  110. De Oliveira RM. Klotho RNAi induces premature senescence of human cells via a p53/p21 dependent pathway. FEBS Lett 2006; 580(24):5753–5758.

    PubMed  Google Scholar 

  111. Ikushima M, Rakugi H, Ishikawa K et al. Anti-apoptotic and anti-senescence effects of Klotho on vascular endothelial cells. Biochem Biophys Res Commun 2006; 339(3):827–832.

    PubMed  CAS  Google Scholar 

  112. Taniyama Y, Morishita R. Does therapeutic angiogenesis overcome CKD? Hypertens Res 2010; 33(2):114–115.

    PubMed  CAS  Google Scholar 

  113. Mu W, Long DA, Ouyang X et al. Angiostatin overexpression is associated with an improvement in chronic kidney injury by an anti-inflammatory mechanism. Am J Physiol Renal Physiol 2009; 296(1):F145–F152.

    PubMed  CAS  Google Scholar 

  114. Westerweel PE, Hoefer IE, Blankestijn PJ et al. End-stage renal disease causes an imbalance between endothelial and smooth muscle progenitor cells. Am J Physiol Renal Physiol 2007; 292(4):F1132–F1140.

    PubMed  CAS  Google Scholar 

  115. Chade AR, Zhu X, Mushin OP et al. Simvastatin promotes angiogenesis and prevents microvascular remodeling in chronic renal ischemia. FASEB J 2006; 20(10):1706–1708.

    PubMed  CAS  Google Scholar 

  116. Reinders ME, Rabelink TJ, Briscoe DM. Angiogenesis and endothelial cell repair in renal disease and allograft rejection. J Am Soc Nephrol 2006; 17(4):932–942.

    PubMed  CAS  Google Scholar 

  117. Nakamura T, Saito Y, Ohyama Y et al. Production of nitric oxide, but not prostacyclin, is reduced in klotho mice. Jpn J Pharmacol 2002; 89(2):149–156.

    PubMed  CAS  Google Scholar 

  118. Shimada T, Takeshita Y, Murohara T et al. Angiogenesis and vasculogenesis are impaired in the precocious-aging klotho mouse. Circulation 2004; 110(9):1148–1155.

    PubMed  Google Scholar 

  119. Fukino K, Suzuki T, Saito Y et al. Regulation of angiogenesis by the aging suppressor gene klotho. Biochem Biophys Res Commun 2002; 293(1):332–337.

    PubMed  CAS  Google Scholar 

  120. Maekawa Y, Ishikawa K, Yasuda O et al. Klotho suppresses TNF-alpha-induced expression of adhesion molecules in the endothelium and attenuates NF-kappaB activation. Endocrine 2009; 35(3):341–346.

    PubMed  CAS  Google Scholar 

  121. Zeisberg M, Duffield JS. Resolved: EMT produces fibroblasts in the kidney. J Am Soc Nephrol 2010; 21(8):1247–1253.

    PubMed  Google Scholar 

  122. Liu Y. New insights into epithelial-mesenchymal transition in kidney fibrosis. J Am Soc Nephrol 2010; 21(2):212–222.

    PubMed  CAS  Google Scholar 

  123. Ardura JA, Rayego-Mateos S, Ramila D et al. Parathyroid hormone-related protein promotes epithelial-mesenchymal transition. J Am Soc Nephrol 2010; 21(2):237–248.

    PubMed  CAS  Google Scholar 

  124. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003; 112(12):1776–1784.

    PubMed  CAS  Google Scholar 

  125. Zeisberg M, Hanai J, Sugimoto H et al. BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat Med 2003; 9(7):964–968.

    PubMed  CAS  Google Scholar 

  126. Iwano M, Plieth D, Danoff TM et al. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest 2002; 110(3):341–350.

    PubMed  CAS  Google Scholar 

  127. Iwano M. EMT and TGF-beta in renal fibrosis. Front Biosci (Schol Ed) 2010; 2:229–238.

    Google Scholar 

  128. Hills CE, Squires PE. TGF-beta1-induced epithelial-to-mesenchymal transition and therapeutic intervention in diabetic nephropathy. Am J Nephrol 2010; 31(1):68–74.

    PubMed  CAS  Google Scholar 

  129. Sato M, Muragaki Y, Saika S et al. Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest 2003; 112(10):1486–1494.

    PubMed  CAS  Google Scholar 

  130. Sugiura H, Yoshida T, Kohei J et al. TGF-??Was Upregulated in Renal Fibrosis Model of Klotho Defect Mouse and Affected Renal Klotho Expression Level (Abstract). J Am Soc Nephrol 2010; 21:376A.

    Google Scholar 

  131. Doi S, Yorioka N, Kuroo M. Secreted Klotho Protein Counteracts Renal Fibrosis through Inhibiting TGF-b1 Signaling (Abstract). J Am Soc Nephrol 2010; 21:270A.

    Google Scholar 

  132. Yamamoto K, Loskutoff DJ. The kidneys of mice with autoimmune disease acquire a hypofibrinolytic/ procoagulant state that correlates with the development of glomerulonephritis and tissue microthrombosis. Am J Pathol 1997; 151(3):725–734.

    PubMed  CAS  Google Scholar 

  133. Yamamoto K, Loskutoff DJ, Saito H. Renal expression of fibrinolytic genes and tissue factor in a murine model of renal disease as a function of age. Semin Thromb Hemost 1998; 24(3):261–268.

    PubMed  CAS  Google Scholar 

  134. Eddy AA, Fogo AB. Plasminogen activator inhibitor-1 in chronic kidney disease: evidence and mechanisms of action. J Am Soc Nephrol 2006; 17(11):2999–3012.

    PubMed  CAS  Google Scholar 

  135. Takeshita K, Yamamoto K, Ito M et al. Increased expression of plasminogen activator inhibitor-1 with fibrin deposition in a murine model of aging, "Klotho" mouse. Semin Thromb Hemost 2002; 28(6):545–554.

    PubMed  CAS  Google Scholar 

  136. Moe SM, Chen NX. Mechanisms of vascular calcification in chronic kidney disease. J Am Soc Nephrol 2008; 19(2):213–216.

    PubMed  CAS  Google Scholar 

  137. Cannata-Andia JB, Rodriguez-Garcia M, Carrillo-Lopez N et al. Vascular calcifications: pathogenesis, management, and impact on clinical outcomes. J Am Soc Nephrol 2006; 17(12 Suppl 3):S267–S273.

    PubMed  Google Scholar 

  138. Mizobuchi M, Towler D, Slatopolsky E. Vascular calcification: the killer of patients with chronic kidney disease. J Am Soc Nephrol 2009; 20(7):1453–1464.

    PubMed  CAS  Google Scholar 

  139. Li X, Yang HY, Giachelli CM. Role of the sodium-dependent phosphate cotransporter, Pit-1, in vascular smooth muscle cell calcification. Circ Res 2006; 98(7):905–912.

    PubMed  CAS  Google Scholar 

  140. Lim K, Lu T-S, Zehnder D et al. Development of Klotho-FGFR1/3 Dependent Resistance to FGF-23 in Human Aortic Smooth Muscle Cells Exposed to Calcifying Stress. J Am Soc Nephrol 2010; 21:140A.

    Google Scholar 

  141. London GM, Marchais SJ, Guerin AP et al. Contributive factors to cardiovascular hypertrophy in renal failure. Am J Hypertens 1989; 2(11 Pt 2):261S–263S.

    PubMed  CAS  Google Scholar 

  142. Guerin AP, Pannier B, Marchais SJ et al. Cardiovascular disease in the dialysis population: prognostic significance of arterial disorders. Curr Opin Nephrol Hypertens 2006; 15(2):105–110.

    PubMed  Google Scholar 

  143. Cerasola G, Nardi E, Palermo A et al. Epidemiology and pathophysiology of left ventricular abnormalities in chronic kidney disease: a review. J Nephrol 2011; 24(1):1–10.

    PubMed  Google Scholar 

  144. Rinat C, Becker-Cohen R, Nir A et al. A comprehensive study of cardiovascular risk factors, cardiac function and vascular disease in children with chronic renal failure. Nephrol Dial Transplant 2010; 25(3):785–793.

    PubMed  Google Scholar 

  145. Edwards NC, Steeds RP, Stewart PM et al. Effect of spironolactone on left ventricular mass and aortic stiffness in early-stage chronic kidney disease: a randomized controlled trial. J Am Coll Cardiol 2009; 54(6):505–512.

    PubMed  CAS  Google Scholar 

  146. Burrell LM, Johnston CI. Angiotensin II receptor antagonists. Potential in elderly patients with cardiovascular disease. Drugs Aging 1997; 10(6):421–434.

    PubMed  CAS  Google Scholar 

  147. Achinger SG, Ayus JC. The role of vitamin D in left ventricular hypertrophy and cardiac function. Kidney Int 2005; 95 Suppl:S37–S42.

    CAS  Google Scholar 

  148. Mirza MA, Larsson A, Melhus H et al. Serum intact FGF23 associate with left ventricular mass, hypertrophy and geometry in an elderly population. Atherosclerosis 2009; 207(2):546–551.

    PubMed  CAS  Google Scholar 

  149. Galitzer H, Ben-Dov IZ, Silver J et al. Parathyroid cell resistance to fibroblast growth factor 23 in secondary hyperparathyroidism of chronic kidney disease. Kidney Int 2010; 77(3):211–218.

    PubMed  CAS  Google Scholar 

  150. Khan S, Vitamin D deficiency and secondary hyperparathyroidism among patients with chronic kidney disease. Am J Med Sci 2007; 333(4):201–207.

    PubMed  Google Scholar 

  151. Krajisnik T, Olauson H, Mirza MA et al. Parathyroid Klotho and FGF-receptor 1 expression decline with renal function in hyperparathyroid patients with chronic kidney disease and kidney transplant recipients. Kidney Int 2010; 78(10):1024–1032.

    PubMed  CAS  Google Scholar 

  152. Canalejo R, Canalejo A, Martinez-Moreno JM et al. FGF23 fails to inhibit uremic parathyroid glands. J Am Soc Nephrol 2010; 21(7):1125–1135.

    PubMed  CAS  Google Scholar 

  153. Hofman-Bang J, Martuseviciene G, Santini MA et al. Increased parathyroid expression of klotho in uremic rats. Kidney Int 2010; 78(11):1119–1127.

    PubMed  CAS  Google Scholar 

  154. Gattineni J, Baum M. Regulation of phosphate transport by fibroblast growth factor 23 (FGF23): implications for disorders of phosphate metabolism. Pediatr Nephrol 2010; 25(4):591–601.

    PubMed  Google Scholar 

  155. Carter JL, O’Riordan SE, Eaglestone GL et al. Bone mineral metabolism and its relationship to kidney disease in a residential care home population: a cross-sectional study. Nephrol Dial Transplant 2008; 23(11):3554–3565.

    PubMed  CAS  Google Scholar 

  156. Oliveira RB, Cancela AL, Graciolli FG et al. Early control of PTH and FGF23 in normophosphatemic CKD patients: a new target in CKD-MBD therapy? Clin J Am Soc Nephrol 2010; 5(2):286–291.

    PubMed  CAS  Google Scholar 

  157. Fukagawa M, Kazama JJ. FGF23: its role in renal bone disease. Pediatr Nephrol 2006; 21(12):1802–1806.

    PubMed  Google Scholar 

  158. Tomida K, Hamano T, Mikami S et al. Serum 25-hydroxyvitamin D as an independent determinant of 1-84 PTH and bone mineral density in nondiabetic predialysis CKD patients. Bone 2009; 44(4):678–683.

    PubMed  CAS  Google Scholar 

  159. Jassal SK, von Muhlen D, Barrett-Connor E. Measures of renal function, BMD, bone loss, and osteoporotic fracture in older adults: the Rancho Bernardo study. J Bone Miner Res 2007; 22(2):203–210.

    PubMed  CAS  Google Scholar 

  160. Kawaguchi H, Manabe N, Miyaura C et al. Independent impairment of osteoblast and osteoclast differentiation in klotho mouse exhibiting low-turnover osteopenia. J Clin Invest 1999; 104(3):229–237.

    PubMed  CAS  Google Scholar 

  161. Kawaguchi H, Manabe N, Chikuda H et al. Cellular and molecular mechanism of low-turnover osteopenia in the klotho-deficient mouse. Cell Mol Life Sci 2000; 57(5):731–737.

    PubMed  CAS  Google Scholar 

  162. Shiraki-Iida T, Iida A, Nabeshima Y et al. Improvement of multiple pathophysiological phenotypes of klotho (kl/kl) mice by adenovirus-mediated expression of the klotho gene. J Gene Med 2000; 2(4):233–242.

    PubMed  CAS  Google Scholar 

  163. Segawa H, Yamanaka S, Ohno Y et al. Correlation between hyperphosphatemia and type II Na-Pi cotransporter activity in klotho mice. Am J Physiol Renal Physiol 2007; 292(2):F769–F779.

    PubMed  CAS  Google Scholar 

  164. Sorribas V, Markovich D, Hayes G et al. Cloning of a Na/Pi cotransporter from opossum kidney cells. J Biol Chem 1994; 269(9):6615–6621.

    PubMed  CAS  Google Scholar 

  165. Ramos AM, Albalate M, Vazquez S et al. Hyperphosphatemia and hyperparathyroidism in incident chronic kidney disease patients. Kidney Int 2008; 111(Suppl):S88–S93.

    CAS  Google Scholar 

  166. Koizumi T, Murakami K, Nakayama H et al. Role of dietary phosphorus in the progression of renal failure. Biochem Biophys Res Commun 2002; 295(4):917–921.

    PubMed  CAS  Google Scholar 

  167. Tonelli M, Wang W, Hemmelgarn B et al. Phosphate removal with several thrice-weekly dialysis methods in overweight hemodialysis patients. Am J Kidney Dis 2009; 54(6):1108–1115.

    PubMed  CAS  Google Scholar 

  168. Marshall CB, Pippin JW, Krofft RD et al. Puromycin aminonucleoside induces oxidant-dependent DNA damage in podocytes in vitro and in vivo. Kidney Int 2006; 70(11):1962–1973.

    PubMed  CAS  Google Scholar 

  169. Diamond JR. Reactive oxygen species and progressive glomerular disease. J Lab Clin Med 1994; 124(4):468–469.

    PubMed  CAS  Google Scholar 

  170. Shih PH, Yen GC. Differential expressions of antioxidant status in aging rats: the role of transcriptional factor Nrf2 and MAPK signaling pathway. Biogcrontology 2007; 8(2):71–80.

    CAS  Google Scholar 

  171. Kachiwala SJ, Harris SE, Wright AF et al. Genetic influences on oxidative stress and their association with normal cognitive ageing. Neurosci Lett 2005; 386(2):116–120.

    PubMed  CAS  Google Scholar 

  172. Yamamoto M, Clark ID, Pastor JV et al. Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem 2005; 280(45):38029–38034.

    PubMed  CAS  Google Scholar 

  173. Feng R, He W, Ochi H. A new murine oxidative stress model associated with senescence. Mech Ageing Dev 2001; 122(6):547–559.

    PubMed  CAS  Google Scholar 

  174. Melk A, Schmidt BM, Braun H et al. Effects of donor age and cell senescence on kidney allograft survival. Am J Transplant 2009; 9(1):114–123.

    PubMed  CAS  Google Scholar 

  175. Yang F, Huang XR, Chung AC et al. Essential role for Smad3 in angiotensin II-induced tubular epithelial-mesenchymal transition. J Pathol 2010; 221(4):390-401.

    Google Scholar 

  176. Liang XB, Ma LJ, Naito T et al. Angiotensin type I receptor blocker restores podocyte potential to promote glomerular endothelial cell growth. J Am Soc Nephrol 2006; 17(7):1886–1895.

    PubMed  CAS  Google Scholar 

  177. Park SY, Song CY, Kim BC et al. Angiotensin II mediates LDL-inducedsuperoxide generation in mesangial cells. Am J Physiol Renal Physiol 2003; 285(5):F909–F915.

    PubMed  CAS  Google Scholar 

  178. Stevenson KM, Edgley AJ, Bergstrom G et al. Angiotensin II infused intrarenally causes preglomerular vascular changes and hypertension. Hypertension 2000; 36(5):839–844.

    PubMed  CAS  Google Scholar 

  179. Peters H, Border WA, Noble NA. Angiotensin II blockade and low-protein diet produce additive therapeutic effects in experimental glomerulonephritis. Kidney Int 2000; 57(4):1493–1501.

    PubMed  CAS  Google Scholar 

  180. Zhou Q, Lin S, Tang R et al. Role ofFosinopril and Valsartan on Klotho Gene Expression Induced by Angiotensin II in Rat Renal Tubular Epithelial Cells. Kidney Blood Press Res 2010; 33(3):186–192.

    PubMed  CAS  Google Scholar 

  181. Narumiya H, Sasaki S, Kuwahara N et al. HMG-CoA reductase inhibitors up-regulate anti-aging klotho mRNA via RhoA inactivation in IMCD3 cells. Cardiovasc Res 2004; 64(2):331–336.

    PubMed  CAS  Google Scholar 

  182. Rosner MH. Urinary biomarkers for the detection of renal injury. Adv Clin Chem 2009; 49:73–97.

    PubMed  CAS  Google Scholar 

  183. Perco P, Wilflingseder J, Bernthaler A et al. Biomarker candidates for cardiovascular disease and bone metabolism disorders in chronic kidney disease: a systems biology perspective. J Cell Mol Med 2008; 12(4): 1177–1187.

    PubMed  CAS  Google Scholar 

  184. Kovesdy CP, Kalantar-Zadeh K. Review article: Biomarkers of clinical outcomes in advanced chronic kidney disease. Nephrology (Carlton) 2009; 14(4):408–415.

    Google Scholar 

  185. Devarajan P. The use of targeted biomarkers for chronic kidney disease. Adv Chronic Kidney Dis 2010; 17(6):469–479.

    PubMed  Google Scholar 

  186. Guh JY. Proteinuria versusalbuminuriainchronickidneydisease. Nephrology(Carlton) 2010; 15(Suppl2):53–56.

    CAS  Google Scholar 

  187. Bokenkamp A, Domanetzki M, Zinck R et al. Reference values for cystatin C serum concentrations in children. Pediatr Nephrol 1998; 12(2):125–129.

    PubMed  CAS  Google Scholar 

  188. Galteau MM, Guyon M, Gueguen R et al. Determination of serum cystatin C: biological variation and reference values. Clin Chem Lab Med 2001; 39(9):850–857.

    PubMed  CAS  Google Scholar 

  189. Shlipak MG, Wassel Fyr CL, Chertow GM et al. Cystatin C and mortality risk in the elderly: the health, aging, and body composition study. J Am Soc Nephrol 2006; 17(1):254–261.

    PubMed  CAS  Google Scholar 

  190. Djousse L, Kurth T, Gaziano JM. Cystatin C and risk of heart failure in the Physicians' Health Study (PHS). Am Heart J 2008; 155(1):82–86.

    PubMed  CAS  Google Scholar 

  191. Drechsler C, Krane V, Winkler K et al. Changes in adiponcctin and the risk of sudden death, stroke, myocardial infarction, and mortality in hemodialysis patients. Kidney Int 2009; 76(5):567–575.

    PubMed  CAS  Google Scholar 

  192. Saraheimo M, Forsblom C, Thorn L et al. Serum adiponectin and progression of diabetic nephropathy in patients with type I diabetes. Diabetes Care 2008; 31(6): 1165–1169.

    PubMed  CAS  Google Scholar 

  193. Baggio B, Gambaro G, Briani G et al. Urinary excretion of glycosaminoglycans and brush border and lysosomal enzymes as markers of glomerular and tubular involvement in kidney diseases. Contrib Nephrol 1984; 42:107–110.

    PubMed  CAS  Google Scholar 

  194. Ryu S, Chang Y, Kim DI et al. gamma-Glutamyltransferase as a predictor of chronic kidney disease in nonhypcrtcnsivc and nondiabetic Korean men. Clin Chem 2007; 53(1):71–77.

    PubMed  CAS  Google Scholar 

  195. Dhaun N, Lilitkarntakul P, Macintyre IM et al. Urinary endothelin-1 in chronic kidney disease and as a marker of disease activity in lupus nephritis. Am J Physiol Renal Physiol 2009; 296(6):Fl477–Fl483.

    Google Scholar 

  196. Cottone S, Mule G, Guarneri M et al. Endothelin-1 and F2-isoprostanerelate to and predict renal dysfunction in hypertensive patients. Nephrol Dial Transplant 2009; 24(2):497–503.

    PubMed  CAS  Google Scholar 

  197. Chang HY, Tung CW, Lee PH et al. Hyperuricemia as an independent risk factor of chronic kidney disease in middle-aged and elderly population. Am J Med Sci 2010; 339(6):509–515.

    PubMed  Google Scholar 

  198. Feig DI. Uric acid: a novel mediator and marker of risk in chronic kidney disease? Curr Opin Nephrol Hypertens 2009; 18(6):526–530.

    PubMed  CAS  Google Scholar 

  199. Madero M, Samak MJ, Wang X et al. Uric acid and long-term outcomes in CKD. Am J Kidney Dis 2009; 53(5):796–803.

    PubMed  CAS  Google Scholar 

  200. Yilmaz MI, Sonmez A, Saglam M et al. Fgf-23 And Vascular Dysfunction In Patients With Stage 3 And 4 Chronic Kidney Disease. Kidney Int 2010; 78(7):679–685.

    PubMed  CAS  Google Scholar 

  201. Appledorn DM, Seregin S, Amalfitano A. Adenovirus vectors for renal-targeted gene delivery. Contrib Nephrol 2008; 159:47–62.

    PubMed  CAS  Google Scholar 

  202. Imai E. Gene therapy approach inrenal disease in the 21st century. Nephrol Dial Transplant 2001; 16Suppl5:26–34.

    PubMed  CAS  Google Scholar 

  203. Wang Y, Sun Z. Klotho gene delivery prevents the progression of spontaneous hypertension and renal damage. Hypertension 2009; 54(4):810–817.

    PubMed  CAS  Google Scholar 

  204. Brown AJ, Slatopolsky E. Drug insight: vitamin D analogs in the treatment of secondary hyperparathyroidism in patients with chronic kidney disease. Nat Clin Pract Endocrinol Metab 2007; 3(2): 134–144.

    PubMed  CAS  Google Scholar 

  205. Gal-Moscovici A, Sprague SM. Role of vitamin D deficiency in chronic kidney disease. J Bone Miner Res 2007; 22(Suppl2):V91–V94.

    PubMed  CAS  Google Scholar 

  206. Gal-Moscovici A, Sprague SM. Use of vitaminD in chronic kidney disease patients. Kidney Int 2010; 78(2): 146–151.

    PubMed  CAS  Google Scholar 

  207. Andress DL. Vitamin D treatment in chronic kidney disease. Semin Dial 2005; 18(4):315–321.

    PubMed  Google Scholar 

  208. Chen HH, Tarng DC, Lee KF et al. Epoetin alfa and darbepoetin alfa: effects on ventricular hypertrophy in patients with chronic kidney disease. J Nephrol 2008; 21(4):543–549.

    PubMed  CAS  Google Scholar 

  209. Coles GA, Cavill I. Erythropoiesis in the anaemia of chronic renal failure: the response to CAPD. Nephrol Dial Transplant 1986; 1(3): 170–174.

    PubMed  CAS  Google Scholar 

  210. Dharmarajan TS, Widjaja D. Erythropoiesis-stimulating agents in anemia: use and misuse. J Am Med Dir Assoc 2009; 10(9):607–616.

    PubMed  CAS  Google Scholar 

  211. Kurosu H, Ogawa Y, Miyoshi M et al. Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem 2006; 281(10):6120–6123.

    PubMed  CAS  Google Scholar 

  212. Hall RL, Wilke WL, Fettman MJ. Captopril slows the progression of chronic renal disease in partially nephrectomized rats. Toxicol Appl Pharmacol 1985; 80(3):517–526.

    PubMed  CAS  Google Scholar 

  213. Brezis M, Greenfeld Z, Shina A et al. Angiotensin II augments medullary hypoxia and predisposes to acute renal failure. Eur J Clin Invest 1990; 20(2): 199–207.

    PubMed  CAS  Google Scholar 

  214. Hali JE. The renin-angiotensin system: renal actions and blood pressure regulation. Compr Ther 1991; 17(5):8–17.

    Google Scholar 

  215. Nabokov A, Amann K, Gassmann P et al. The renoprotective effect of angiotensin-converting enzyme inhibitors in experimental chronic renal failure is not dependent on enhanced kinin activity. Nephrol Dial Transplant 1998; 13(1): 173–176.

    PubMed  CAS  Google Scholar 

  216. Tojo A, Onozato ML, Kobayashi N et al. Angiotensin II and oxidative stress in Dahl Salt-sensitive rat with heart failure. Hypertension 2002; 40(6):834–839.

    PubMed  CAS  Google Scholar 

  217. Muller DN, Mullally A, Dechend R et al. Endothelin-converting enzyme inhibition ameliorates angiotensin IT-induced cardiac damage. Hypertension 2002; 40(6):840–846.

    PubMed  CAS  Google Scholar 

  218. Agarwal R. Proinflammatory effects of oxidative stress in chronic kidney disease: role of additional angiotensin II blockade. Am J Physiol Renal Physiol 2003; 284(4):F863–F869.

    PubMed  CAS  Google Scholar 

  219. Remuzzi G, Perico N, Macia M et al. The role of renin-angiotensin-aldosterone system in the progression of chronic kidney disease. Kidney Int 2005; 99(Suppl):S57–S65.

    CAS  Google Scholar 

  220. Wenzel RR. Renal protection in hypertensive patients: selection of antihypertensive therapy. Drugs 2005; 65Suppl 2:29–39.

    PubMed  CAS  Google Scholar 

  221. Mizuno M, Sada T, Kato M et al. The effect of angiotensin II receptor blockade on an end-stage renal failure model of type 2 diabetes. J Cardiovasc Pharmacol 2006; 48(4): 135–142.

    Google Scholar 

  222. Lebel M, Rodrigue ME, Agharazii M et al. Antihypertensive and renal protective effects ofrenin-angiotens in system blockade in uremic rats treated with erythropoietin. Am J Hypertens 2006; 19(12): 1286–1292.

    PubMed  CAS  Google Scholar 

  223. Negri AL. The klotho gene: a gene predominantly expressed in the kidney is a fundamental regulator of aging and calcium/phosphorus metabolism. J Nephrol 2005; 18(6):654–658.

    PubMed  CAS  Google Scholar 

  224. Kuwahara N, Sasaki S, Kobara M et al. HMG-CoA reductase inhibition improves anti-aging klotho protein expression and arteriosclerosis in rats with chronic inhibition of nitric oxide synthesis. Int J Cardiol 2008; 123(2):84–90.

    PubMed  Google Scholar 

  225. Wang X, Liu X, Zhan Y et al. Pharmacogenomic, physiological, and biochemical investigations on safety and efficacy biomarkers associated with the peroxisome proliferator-activated receptor-gamma activator rosiglitazone in rodents: a translational medicine investigation. J Pharmacol Exp Ther 2010; 334(3):820–829.

    PubMed  CAS  Google Scholar 

  226. Zhang H, Li Y, Fan Y et al. Klotho is a target gene of PPAR-gamma. Kidney Int 2008; 74(6):732–739.

    PubMed  CAS  Google Scholar 

  227. Zhang R, Zheng F. PPAR-gamma and aging: one link through klotho? Kidney Int 2008; 74(6):702–704.

    PubMed  CAS  Google Scholar 

  228. Lopez-Hernandez FJ, Lopez-Novoa JM. Potential utility of PPARalpha activation in the prevention of ischemic and drug-induced acute renal damage. Kidney Int 2009; 76(10): 1022–1024.

    PubMed  CAS  Google Scholar 

  229. Li S, Nagothu KK, Desai V et al. Transgenic expression of proximal tubule peroxisome proliferator-activated receptor-alpha in mice confers protection during acute kidney injury. Kidney Int 2009; 76(10): 1049–1062.

    PubMed  CAS  Google Scholar 

  230. Rakugi H, Matsukawa N, Ishikawa K et al. Anti-oxidative effect of Klotho on endothelial cells through cAMP activation. Endocrine 2007; 31(1):82–87.

    PubMed  CAS  Google Scholar 

  231. Shah SV, Baliga R, Rajapurkar M et al. Oxidants in chronic kidney disease. J Am Soc Nephrol 2007; 18(1): 16–28.

    PubMed  CAS  Google Scholar 

  232. Sugiura H, Yoshida T, Mitobe M et al. Klotho reduces apoptosis in experimental ischaemic acute kidney injury via HSP-70. Nephrol Dial Transplant 2010; 25(1):60–68.

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Orson W. Moe .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2012 Landes Bioscience and Springer Science+Business Media

About this chapter

Cite this chapter

Hu, M.C., Kuro-o, M., Moe, O.W. (2012). Secreted Klotho and Chronic Kidney Disease. In: Kuro-o, M. (eds) Endocrine FGFs and Klothos. Advances in Experimental Medicine and Biology, vol 728. Springer, New York, NY. https://doi.org/10.1007/978-1-4614-0887-1_9

Download citation

Publish with us

Policies and ethics