Skip to main content

Advertisement

Log in

Serum levels of soluble secreted α-Klotho are decreased in the early stages of chronic kidney disease, making it a probable novel biomarker for early diagnosis

  • Original Article
  • Published:
Clinical and Experimental Nephrology Aims and scope Submit manuscript

Abstract

Background

α-Klotho was first identified as an aging gene and was later shown to be a regulator of phosphate metabolism. Fibroblast growth factor 23 (FGF23) is the key regulator of phosphate metabolism. Serum levels of soluble α-Klotho in chronic kidney disease (CKD) patients have not previously been determined, especially in relation with FGF23 and creatinine levels. This study was designed to investigate whether serum soluble α-Klotho levels are modulated by renal function, age, and FGF23 level in CKD patients. This study is the first report on the utility of measuring soluble α-Klotho levels in human CKD.

Methods

A total of 292 CKD patients were enrolled. Serum samples were collected, and FGF23 and soluble α-Klotho levels were measured using enzyme-linked immunosorbent assay kits. In addition, serum creatinine, hemoglobin, albumin, calcium, and phosphate levels were measured.

Results

Serum soluble α-Klotho levels were associated positively with estimated glomerular filtration rate (eGFR) (P < 0.0001) and inversely with serum creatinine level (P < 0.01). Interestingly, α-Klotho levels were significantly decreased in stage 2 CKD compared with stage 1 (P = 0.0001). Serum FGF23 levels were associated positively with serum creatinine and negatively with eGFR. FGF23 levels were significantly increased in stage 5 compared with stage 1 CKD. Soluble α-Klotho was associated inversely with log-transformed FGF23 level (P < 0.01).

Conclusion

Our data indicate that soluble α-Klotho levels are significantly decreased in stage 2 CKD compared to stage 1, and not only in the advanced stages of the disease. Soluble α-Klotho may thus represent a new biomarker for the diagnosis of CKD, especially in the early stage.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Zhang QL, Rothenbacher D. Prevalence of chronic kidney disease in population-based studies: systematic review. BMC Public Health. 2008;8:117.

    Article  PubMed  Google Scholar 

  2. Manjunath G, Tighiouart H, Ibrahim H, Mac LB, Salem DN, Griffiht JL, et al. Level of kidney function as s risk factor for atherosclerotic cardiovascular outcomes in the community. J Am Coll Cardiol. 2003;41:47–55.

    Article  PubMed  Google Scholar 

  3. Baigent C, Burbury K, Wheeler D. Premature cardiovascular disease in chronic renal failure. Lancet. 2000;356:147–52.

    Article  PubMed  CAS  Google Scholar 

  4. Coresh J, Astor BC, Greene T, Eknoyan G, Levey AS. Prevalence of chronic kidney disease and decreased kidney function in the adult US population. Third National Health and Nutrition Examination Survey. Am J Kidney Dis. 2003;41:1–12.

    Article  PubMed  Google Scholar 

  5. Coresh J, Selvin E, Stevens LA, Manzi J, Kusek JW, Eggers P, et al. Prevalence of chronic kidney disease in the United States. JAMA. 2007;298:2038–47.

    Article  PubMed  CAS  Google Scholar 

  6. Menon V, Shlipak MG, Wang X, Coresh J, Greene T, Stevens L, et al. Cystatin C as a risk factor for outcomes in chronic kidney disease. Ann Intern Med. 2007;147:19–27.

    PubMed  Google Scholar 

  7. Tamara I, Huiliang X, Wei Y, Dawei X, Amanda HA, Julia S, et al. Fibroblast growth factor 23 and risks of mortality and end-stage disease in patients with chronic kidney disease. JAMA. 2011;305:2432–9.

    Article  Google Scholar 

  8. Silvia MT, Roberto Z, Fabiliana GG, Luciene MR, Rui TB, Vanda J, et al. FGF23 as a predictor of renal outcome in diabetic nephropathy. J Am Soc Nephrol. 2011;6:241–7.

    Article  Google Scholar 

  9. Sarah S, Birgit R, Daniel R, Eric S, Danilo F, Gunnar H. FGF-23 and future cardiovascular events in patients with chronic kidney disease before initiation of dialysis treatment. Nephrol Dial Transplant. 2010;25:3983–9.

    Article  Google Scholar 

  10. Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosenblantt KP, et al. Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem. 2006;281:6120–3.

    Article  PubMed  CAS  Google Scholar 

  11. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, et al. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 2006;444:770–4.

    Article  PubMed  CAS  Google Scholar 

  12. Nakatani T, Sarraj B, Ohnishi M, Densmore MJ, Taguchi T, Goetz R, et al. In vivo genetic evidence for klotho-dependent, fibroblast growth factor 23 (Fgf23)-mediated regulation of systemic phosphate homeostasis. FASEB J. 2009;23:433–41.

    Article  PubMed  CAS  Google Scholar 

  13. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.

    Article  PubMed  CAS  Google Scholar 

  14. Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, et al. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J. 2010;24:3438–50.

    Article  PubMed  CAS  Google Scholar 

  15. Kato Y, Arakawa E, Kinoshita S, Shirai A, Furuya A, Yamano K, et al. Establishment of the anti-Klotho monoclonal antibodies and detection of Klotho protein in kidneys. BBRC. 2000;267:597–602.

    PubMed  CAS  Google Scholar 

  16. Chen CD, Podvin S, Gillespie E, Leeman SE, Abraham CR. Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci USA. 2007;104:19796–801.

    Article  PubMed  CAS  Google Scholar 

  17. Bloch L, Sineshchekova O, Reichenbach D, Reiss K, Saftig P, Kuro-o M, et al. Klotho is a substrate for alpha-, beta- and gamma-secretase. FEBS Lett. 2009;583:3221–4.

    Article  PubMed  CAS  Google Scholar 

  18. Imura A, Iwano A, Tohyama O, Tsuji Y, Nozaki K, Hashimoto N, et al. Secreted Klotho protein in sera and CSF: Implication for post-translational cleavage in release of Klotho protein from cell membrane. FEBS Lett. 2004;565:143–7.

    Article  PubMed  CAS  Google Scholar 

  19. Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ, Hoenderop JG. The beta-glucosidase klotho hydrolyzes and activates the TRPV5 channel. Science. 2005;310:490–3.

    Article  PubMed  CAS  Google Scholar 

  20. Cha SK, Ortega B, Kurose H, Rosenblatt KP, Kuro-o M, Huang CL. Removal of sialic acid involving Klotho causes cell-surface retention of TRPV5 channel via binding to galactin-1. Proc Natl Acad Sci USA. 2008;105:9805–10.

    Article  PubMed  CAS  Google Scholar 

  21. Cha SK, Hu MC, Kurosu H, Kuro-o M, Moe O, Huang CL. Regulation of renal outer medullary potassium channel and renal K(+) excretion by klotho. Mol Pharmacol. 2009;76:38–46.

    Article  PubMed  CAS  Google Scholar 

  22. Yamazaki Y, Imura A, Urakawa I, Shimada T, Murakami J, Aono Y, et al. Establishment of sandwich ELISA for soluble alpha-Klotho measurement: age-dependent change of soluble alpha-Klotho levels in healthy subjects. BBRC. 2010;398:513–8.

    PubMed  CAS  Google Scholar 

  23. Matsuo S, Imai E, Horio M, Yasuda Y, Tomita K, Nitta K, et al. Revised equations for estimated GFR from serum creatinine in Japan. Am J Kidney Dis. 2009;53:982–92.

    Article  PubMed  CAS  Google Scholar 

  24. Yamazaki Y, Okazaki R, Shibata M, Hasegawa Y, Satoh K, Tajima T, et al. Increased circulatory level of biologically active full-length FGF-23 in patients with hypophosphatemic rickets/osteomalacia. J Clin Endocrinol Metab. 2002;87:4957–60.

    Article  PubMed  CAS  Google Scholar 

  25. Martin B, Marc V, Piet W, Gerjan N. Cross talk between the renin–angiotensin–aldosterone system and vitamin D–FGF-23–klotho in chronic kidney disease. J Am Soc Nephrol. 2011;22:1603–9.

    Article  Google Scholar 

  26. Aizawa H, Saito Y, Nakamura T, Inoue M, Imanari T, Ohyama Y, et al. Downregulation of the Klotho gene in the kidney under sustained circulatory stress in rats. BBRC. 1998;249:865–71.

    PubMed  CAS  Google Scholar 

  27. Koh N, Fujimori T, Nishiguchi S, Tamori A, Shiomi S, Nakatani T, et al. Severely reduced production of klotho in human chronic renal failure kidney. BBRC. 2001;280:1015–20.

    PubMed  CAS  Google Scholar 

  28. Haruna Y, Kashihara N, Satoh M, Tomita N, Namikoshi T, Sasaki T, et al. Amelioration of progressive renal injury by genetic manipulation of Klotho gene. Proc Natl Acad Sci USA. 2007;104:2331–6.

    Article  PubMed  CAS  Google Scholar 

  29. Hu MC, Shi M, Zhang J, Quinones H, Griffith C, Kuro-o M, et al. Klotho deficiency causes vascular calcification in chronic kidney disease. J Am Soc Nephrol. 2011;22:124–36.

    Article  PubMed  CAS  Google Scholar 

  30. Tomiyama K, Maeda R, Urakawa I, Yamazaki Y, Tanaka T, Ito S, et al. Relevant use of Klotho in FGF19 subfamily signaling system in vivo. Proc Natl Acad Sci USA. 2010;107:1666–71.

    Article  PubMed  CAS  Google Scholar 

  31. Segawa H, Yamanaka S, Ohno Y, Onitsuka A, Shiozawa K, Aranami F, et al. Correlation between hyperphosphatemia and type II Na–Pi cotransporter activity in klotho mice. Am J Physiol Renal Physiol. 2007;292:F769–79.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This work was supported by Daiwa Memorial foundation, Japanese Kidney foundation, and a grant from the Ministry of Education, Science, Culture and Sports of Japan (to Y. S., K. I., K. O., S. F., and Y. T.) and a grant of Kochi Organization for Medical Reformation and Renewal to Y.T. We thank Ms. Reiko Matumoto, Ms. Sekie Saito for technical assistances.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yoshio Terada.

About this article

Cite this article

Shimamura, Y., Hamada, K., Inoue, K. et al. Serum levels of soluble secreted α-Klotho are decreased in the early stages of chronic kidney disease, making it a probable novel biomarker for early diagnosis. Clin Exp Nephrol 16, 722–729 (2012). https://doi.org/10.1007/s10157-012-0621-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10157-012-0621-7

Keywords

Navigation