Korean J Intern Med > Volume 25(1); 2010 > Article
Park, Kim, Kang, and Lee: Application of Cystatin C Reduction Ratio to High-Flux Hemodialysis as an Alternative Indicator of the Clearance of Middle Molecules

Abstract

Background/Aims

Although high-flux (HF) dialyzers with enhanced membrane permeability are widely used in current hemodialysis (HD) practice, urea kinetic modeling is still being applied to indicate the adequacy of both low-flux (LF) and HF HD. In comparison with urea (molecular weight, 60 Da) and β2-microglobulin (β2MG, 12 kDa), cystatin C (CyC, 13 kDa) is a larger molecule that has attractive features as a marker for assessing solute clearance. We postulated that CyC might be an alternative for indicating the clearance of middle molecules (MMs), especially with HF HD.

Methods

Eighty-nine patients were divided into LF and HF groups. Using single pool urea kinetic modeling, the urea reduction ratio (URR) and equilibrated Kt/Vurea (eKt/Vurea) were calculated. The serum CyC concentrations were measured using particle-enhanced immunonephelometry. As indices of the middle molecular clearance, the reduction ratios of β2MG and CyC were calculated.

Results

The β2MG reduction ratio (β2MGRR) and CyC reduction ratio (CyCRR) were higher in the HF group compared to the LF group. However, the URR and eKt/Vurea did not differ between the two groups. The CyCRR was significantly correlated with the eKt/Vurea and β2MGRR (r = 0.47 and 0.69, respectively, both p < 0.0001).

Conclusions

Compared to the LF dialyzer, the HF dialyzer removed CyC and β2MG more efficiently. Unlike the β2MGRR, the CyCRR was correlated with the eKt/Vurea and β2MGRR. This study suggests a role for the CyCRR as an alternative indicator of the removal of MMs.

INTRODUCTION

The uremic syndrome is attributable to the progressive retention of a large number of compounds, which are called uremic retention solutes or uremic toxins, when they interact negatively with physiologic function. They include not only small plasma solutes, but also protein-bound solutes and middle molecules (MMs) [1]. Their retention may lead to dialysis morbidity and mortality, which are poorly resolved with conventional hemodialysis (HD).
Since the beginning of the 1980s, urea kinetic modeling has been applied to quantify the removal of small solutes by conventional HD. In current practice, high-flux (HF) dialyzers may allow more efficient removal of larger molecules, which is expected to reduce the risk of HD-related morbidity and mortality substantially. Conversely, no consensus exists concerning the best dialysis dose index and method for calculating it [2,3].
Of the MMs, cystatin C (CyC, molecular weight [MW], 13 kDa) is freely filtered at the level of the glomerulus and virtually all is reabsorbed and metabolized by proximal tubular cells [4-7]. Due to its molecular characteristics, numerous studies have shown that CyC is the most promising endogenous marker of the glomerular filtration rate [8]. In addition, several studies have suggested that CyC is useful as a marker of HD toxin removal, since the plasma CyC has attractive features as a representative MM [9,10].
For these reasons, we hypothesized that the CyC reduction ratio (CyCRR) might reflect the removal of MMs and could be applied to the dialysis dose index in HD, and we designed this single-center, case-control study to examine our hypotheses.

METHODS

Patients

All of the patients underwent maintenance HD at Hanyang University Hospital three times per week. Previously, all 89 patients had chosen low-flux (LF) HD as the dialysis modality. The dialysis modality was chosen according to patient preference, and the dialyzers used were F6HPS for LF and F60S for HF (Fresenius Medical Care, Bad Homburg, Germany). We assumed that no residual renal function was preserved in any patient. Patients were excluded if they were medically unstable, younger than 18 years of age, or mentally disabled.

Measurements

All blood samples were taken before and after the second HD session of the week, according to the guidelines for HD adequacy [11]. Blood samples were collected in tubes without additional anticoagulant and allowed to stand at room temperature for 30 to 60 minutes. Then, the samples were centrifuged to collect serum, which was stored at -70℃ until assayed. Urea nitrogen was measured using a Hitachi 7600 series automatic biochemical analyzer (Hitachi High-Technologies, Tokyo, Japan). Serum β2-microglobulin (β2MG, MW, 12 kDa) concentrations were measured using a chemiluminescent enzyme immunoassay (DPC IMMUNLITE 2000, Diagnostic Products, Los Angeles, CA, USA). CyC was assayed using particle-enhanced immunonephelometry (BN II nephelometer, Dade Behring, Reuil Malmaison, France). All other measurements were made using routine laboratory methods.
We calculated the reduction ratios of solutes for urea, β2MG, and CyC during the treatment. The single-pool urea Kt/V (spKt/Vurea) values were calculated using the second-generation natural logarithmic formula described by Daugirdas [12], and we transformed the spKt/Vurea to the equilibrated Kt/Vurea (eKt/Vurea) [13].
The clearance of β2MG (Kβ2MG) was determined using the formula described by Cheung et al. [14] and then the Kt/Vβ2MG was calculated by multiplying the Kβ2MG by the treatment time and dividing the result by the post-dialysis volume.

Statistical analysis

Baseline characteristics are presented as the mean ± SD or as the proportions of patients in the groups. The Shapiro-Wilk test was used to test for normality. Continuous variables were compared using the Mann-Whitney U test. The chi-square test with Fisher's exact test was applied to evaluate associations between nominal variables. The Spearman correlation coefficient was used to study correlations between variables. All p values were analyzed using two-sided tests, and p values < 0.05 were considered statistically significant. All analyses were performed with SAS version 9.1 (SAS Institute, Cary, NC, USA).

RESULTS

Eighty-nine patients were included and divided into LF (n = 37) and HF (n = 52) groups. Dialysis was performed for 4.0 ± 0.2 hr/session with blood flow rates of 237 ± 23 mL/min and dialysate flow rates of 500 mL/min using bicarbonate dialysate. The dialysis machine, water treatment circuit, and tank were sterilized regularly.
No differences existed between the two groups with respect to the demographic characteristics and the cause of end-stage renal disease (Table 1). The C-reactive protein was higher in the LF HD group (p = 0.0039), but was not correlated with the indices of solute removal in this study (data not shown). The etiology of renal failure in the patients included diabetes mellitus (n = 29, 32.6%), hypertension (n = 18, 20.2%), chronic glomerulonephritis (n = 12, 13.5%), and shrunken kidneys of unknown etiology (n = 23, 25.8%).
The eKt/Vurea in the LF and HF HD groups was 1.19 ± 0.32 and 1.29 ± 0.23, respectively. No differences were observed in the urea reduction ratio (URR) and eKt/Vurea between the two groups (Table 2). However, β2MG and CyC were removed more efficiently by the HF dialyzer (p < 0.0001).
When all patients were considered, the CyCRR was strongly correlated with the β2MG reduction ratio (β2MGRR) and Kt/Vβ2MG and weakly correlated with the URR and eKt/Vurea (Table 3). The pre-dialysis CyC and β2MG levels, and their reduction ratios were not correlated with the albumin, normalized protein catabolic rate, or C-reactive protein (data not shown).

DISCUSSION

We assessed the performance of the CyCRR as representative of the clearance of MMs. As in previous reports [14,15], no difference existed between the two groups in terms of the URR and eKt/Vurea. In addition, the more efficient removal of β2MG in the HF HD group concurred with a past study [14]. However, we found that the CyCRR had a prominent relationship with the β2MGRR and Kt/Vβ2MG in HD.
Most of the small water-soluble uremic retention compounds and MMs that have been shown to exert biologic action have an intradialytic kinetic behavior that is indisputably different from that of urea [16], and their retention is poorly resolved with conventional HD. Therefore, many past studies have emphasized that the removal of small water-soluble uremic retention compounds and MMs using a HF dialyzer may ensure excellent dialysis quality, influence the clinical outcomes of patients in several areas, and reduce the acute and long-term HD-related complications [1,17-20]. These may be particularly important in patients with no residual renal function, who depend completely on dialysis efficiency. Assessing dialysis adequacy using several molecules simultaneously is suitable to reflect small solute and MM clearance [21].
β2MG had been accepted as representative of MMs and is widely recognized as a key component in the development of dialysis-associated amyloidosis [22]. Several studies demonstrated that the use of a HF dialyzer might improve the clearance of β2MG and decrease pre-dialysis plasma levels [23,24]. They expected that HD with a HF dialyzer could reduce the development of amyloidosis and improve patient quality of life. Given the fluctuating β2MG production rate, nonrenal route of elimination, other factors influencing its bloodstream delivery, and intercompartmental transport, β2MG removal is recognized as not being a good indicator [25,26].
After exploring the interrelationship of low-molecular-weight proteins with the glomerular filtration rate, an increasing serum CyC level was found to be related to decreasing renal function and to be associated with some cerebral amyloid angiopathies, tumor progression, and inflammatory processes [27-30]. In addition, some attractive features as a representative MM made us assess the performance of CyC during HD: because CyC should be distributed strictly in extracellular fluid, various kinetic models are not required to describe its kinetics during HD; its production rate is relatively constant or minimally variable; CyC is a free-circulating, unbounded form, and its elimination from the circulation is almost entirely through glomerular filtration; and CyC may be a prognostic biomarker of the risk of death and cardiovascular disease [31,32].
Starting from the hypothesis that the clearance of CyC is representative of the removal of MMs and its reduction ratio is a useful marker of toxin removal in HD, we performed this study to compare the CyCRR with the most widely used methods for measuring HD adequacy. Since CyC has several merits as a representative MM - it is similar to β2MG in molecular weight and clearance during HD, and its reduction ratio has some relationship with the URR and eKt/Vurea - the CyCRR should be useful as an alternative to the β2MGRR and Kt/Vβ2MG for estimating the dialysis clearance of MMs. However, further studies with larger sample sizes should be performed to draw a firm conclusion as to whether the CyCRR is useful indicator of the clearance of MMs.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

References

1. Vanholder R, De Smet R, Glorieux G, et al. Review on uremic toxins: classification, concentration, and interindividual variability. Kidney Int 2003;63:1934–1943PMID : 12675874.
crossref pmid
2. Prado M, Roa LM, Palma A, Milan JA. Double target comparison of blood-side methods for measuring the hemodialysis dose. Kidney Int 2005;68:2863–2876PMID : 16316364.
crossref pmid
3. Vienken J, Bowry S. Quo vadis dialysis membrane? Artif Organs 2002;26:152–159PMID : 11879244.
crossref pmid
4. Mussap M, Plebani M. Biochemistry and clinical role of human cystatin C. Crit Rev Clin Lab Sci 2004;41:467–550PMID : 15603510.
crossref pmid
5. Rule AD, Bergstralh EJ, Slezak JM, Bergert J, Larson TS. Glomerular filtration rate estimated by cystatin C among different clinical presentations. Kidney Int 2006;69:399–405PMID : 16408133.
crossref pmid
6. Shlipak MG, Praught ML, Sarnak MJ. Update on cystatin C: new insights into the importance of mild kidney dysfunction. Curr Opin Nephrol Hypertens 2006;15:270–275PMID : 16609294.
crossref pmid
7. Knight EL, Verhave JC, Spiegelman D, et al. Factors influencing serum cystatin C levels other than renal function and the impact on renal function measurement. Kidney Int 2004;65:1416–1421PMID : 15086483.
crossref pmid
8. Mojiminiyi OA, Abdella N. Evaluation of cystatin C and beta-2 microglobulin as markers of renal function in patients with type 2 diabetes mellitus. J Diabetes Complications 2003;17:160–168PMID : 12738401.
crossref pmid
9. Campo A, Lanfranco G, Gramaglia L, Goia F, Cottino R, Giusto V. Could plasma cystatin C be useful as a marker of hemodialysis low molecular weight proteins removal? Nephron Clin Pract 2004;98:c79–c82PMID : 15528941.
crossref pmid
10. Thysell H, Grubb A, Lindholm T, Ljunggren L, Martensson L. Cystain C: a new marker of biocompatibility or a good marker for the redistribution of LMW proteins during hemodialysis? ASAIO Trans 1988;34:202–204PMID : 3058174.
pmid
11. Hemodialysis Adequacy 2006 Work Group. Clinical practice guidelines for hemodialysis adequacy, update 2006. Am J Kidney Dis 2006;48(Suppl 1):S2–S90PMID : 16813990.
crossref pmid
12. Daugirdas JT. Second generation logarithmic estimates of single-pool variable volume Kt/V: an analysis of error. J Am Soc Nephrol 1993;4:1205–1213PMID : 8305648.
crossref pmid
13. Diaz-Buxo JA, Loredo JP. Standard Kt/V: comparison of calculation methods. Artif Organs 2006;30:178–185PMID : 16480392.
crossref pmid
14. Cheung AK, Levin NW, Greene T, et al. Effects of high-flux hemodialysis on clinical outcomes: results of the HEMO study. J Am Soc Nephrol 2003;14:3251–3263PMID : 14638924.
crossref pmid
15. Ayli D, Ayli M, Azak A, et al. The effect of high-flux hemodialysis on renal anemia. J Nephrol 2004;17:701–706PMID : 15593038.
pmid
16. Vanholder R, Glorieux G, De Smet R, Lameire N. New insights in uremic toxins. Kidney Int Suppl 2003;(84):S6–S10PMID : 12694297.
crossref
17. Basile C. The effect of convection on the nutritional status of haemodialysis patients. Nephrol Dial Transplant 2003;18(Suppl 7):vii46–vii49PMID : 12953030.
crossref pmid
18. Leypoldt JK, Cheung AK. Removal of high-molecular-weight solutes during high-efficiency and high-flux haemodialysis. Nephrol Dial Transplant 1996;11:329–335PMID : 8671788.
crossref pmid
19. Clark WR, Gao D. Determinants of uraemic toxin removal. Nephrol Dial Transplant 2002;17(Suppl 3):30–34PMID : 11912297.
crossref
20. Locatelli F, Manzoni C, Di Filippo S. The importance of convective transport. Kidney Int Suppl 2002;(80):115–120PMID : 11982825.
crossref pmid
21. Lindsay RM, Henderson LW. Adequacy of dialysis. Kidney Int Suppl 1988;24:S92–S99PMID : 3283413.
pmid
22. Evenepoel P, Bammens B, Verbeke K, Vanrenterghem Y. Superior dialytic clearance of beta(2)-microglobulin and p-cresol by high-flux hemodialysis as compared to peritoneal dialysis. Kidney Int 2006;70:794–799PMID : 16820785.
crossref pmid
23. Ayli M, Ayli D, Azak A, et al. The effect of high-flux hemodialysis on dialysis-associated amyloidosis. Ren Fail 2005;27:31–34PMID : 15717632.
crossref pmid
24. Locatelli F, Andrulli S, Pecchini F, et al. Effect of high-flux dialysis on the anaemia of haemodialysis patients. Nephrol Dial Transplant 2000;15:1399–1409PMID : 10978398.
crossref pmid
25. Leypoldt JK, Cheung AK, Deeter RB. Rebound kinetics of beta2-microglobulin after hemodialysis. Kidney Int 1999;56:1571–1577PMID : 10504510.
crossref pmid
26. Ward RA, Greene T, Hartmann B, Samtleben W. Resistance to intercompartmental mass transfer limits beta2-microglobulin removal by post-dilution hemodiafiltration. Kidney Int 2006;69:1431–1437PMID : 16395268.
crossref pmid
27. Newman DJ. Cystatin C. Ann Clin Biochem 2002;39(Pt 2):89–104PMID : 11928770.
crossref pmid
28. Levy E, Jaskolski M, Grubb A. The role of cystatin C in cerebral amyloid angiopathy and stroke: cell biology and animal models. Brain Pathol 2006;16:60–70PMID : 16612983.
crossref pmid
29. Reed CH. Diagnostic applications of cystatin C. Br J Biomed Sci 2000;57:323–329PMID : 11204865.
pmid
30. Revesz T, Ghiso J, Lashley T, et al. Cerebral amyloid angiopathies: a pathologic, biochemical, and genetic view. J Neuropathol Exp Neurol 2003;62:885–898PMID : 14533778.
crossref pmid
31. Ix JH, Shlipak MG, Chertow GM, Whooley MA. Association of cystatin C with mortality, cardiovascular events, and incident heart failure among persons with coronary heart disease: data from the Heart and Soul Study. Circulation 2007;115:173–179PMID : 17190862.
crossref pmid pmc
32. Shlipak MG, Katz R, Sarnak MJ, et al. Cystatin C and prognosis for cardiovascular and kidney outcomes in elderly persons without chronic kidney disease. Ann Intern Med 2006;145:237–246PMID : 16908914.
crossref pmid
Table 1
Demographic characteristics
kjim-25-77-i001.jpg

Values are presented as mean ± SD for continuous variables and number (%) for nominal variables.

NS, not significant.

Table 2
Clearance according to dialyzer
kjim-25-77-i002.jpg

Values are presented as the mean ± SD.

eKt/Vurea, equilibrated Kt/Vurea; β2MG, β2-microglobulin; CyC, cystatin C; NS, not significant.

Table 3
Correlation coefficient between clearance
kjim-25-77-i003.jpg

eKt/Vurea, equilibrated Kt/Vurea; β2MG, β2-microglobulin; CyC, cystatin C.

ap < 0.05.

bp < 0.01.

TOOLS
METRICS Graph View
  • 14 Crossref
  • 16 Scopus
  • 9,133 View
  • 62 Download
Related articles

Editorial Office
101-2501, Lotte Castle President, 109 Mapo-daero, Mapo-gu, Seoul 04146, Korea
Tel: +82-2-2271-6792    Fax: +82-2-790-0993    E-mail: kaim@kams.or.kr                

Copyright © 2024 by Korean Association of Internal Medicine.

Close layer
prev next