Skip to main content
Erschienen in: BMC Nephrology 1/2014

Open Access 01.12.2014 | Research article

Hyperuricemia and deterioration of renal function in autosomal dominant polycystic kidney disease

verfasst von: Miyeun Han, Hayne Cho Park, Hyunsuk Kim, Hyung Ah Jo, Hyuk Huh, Joon Young Jang, Ah-Young Kang, Seung Hyup Kim, Hae Il Cheong, Duk-Hee Kang, Jaeseok Yang, Kook-Hwan Oh, Young-Hwan Hwang, Curie Ahn

Erschienen in: BMC Nephrology | Ausgabe 1/2014

Abstract

Background

The role of hyperuricemia in disease progression of autosomal dominant polycystic kidney disease (ADPKD) has not been defined well. We investigated the association of serum uric acid (sUA) with renal function and the effect of hypouricemic treatment on the rate of renal function decline.

Methods

This is a single-center, retrospective, observational cohort study. A total of 365 patients with ADPKD who had estimated glomerular filtration rate (eGFR) ≥ 15 mL/min/1.73 m2 and who were followed up for > 1 year were included in our analysis. Hyperuricemia was defined by a sUA level of ≥ 7.0 mg/dL in male and ≥ 6.0 mg/dL in female or when hypouricemic medications were prescribed.

Results

Hyperuricemia was associated with reduced initial eGFR, independent of age, sex, hypertension, albuminuria, and total kidney volume. During a median follow-up period of over 6 years, patients with hyperuricemia showed a faster annual decline in eGFR (−6.3% per year vs. −0.9% per year, p = 0.008). However, after adjusting for age, sex, hypertension and initial eGFR, sUA was no longer associated with either annual eGFR decline or the development of ESRD. Among 53 patients who received hypouricemic treatment, the annual eGFR decline appeared to be attenuated after hypouricemic treatment (pretreatment vs. posttreatment: −5.3 ± 8. 2 vs. 0.2 ± 6.2 mL/min/1.73 m2 per year, p = 0.001 by Wilcoxon signed-rank test).

Conclusions

Although hyperuricemia was associated with reduced eGFR, it was not an independent factor for renal progression in ADPKD. However, the correction of hyperuricemia may attenuate renal function decline in some patients with mild renal insufficiency.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2369-15-63) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MH collected the data and drafted the manuscript. HP participated in the design of the study and drafted the manuscript. HK participated in the acquisition of data. HC, HH participated in the analysis and interpretation of data. JJ, AK carried out immunohistochemical staining. SK carried out kidney volume measurement. HC, DK participated in critical revising the manuscript. JY, KO participated in the design of the study and interpretation of data. YH helped interpretation of data and drafted the manuscript. CA conceived of the study, interpretation of data and revising the manuscript. All authors read and approved the final manuscript.
Abkürzungen
ACR
Albumin to creatinine ratio
ADPKD
Autosomal dominant polycystic kidney disease
CKD
Chronic kidney disease
CKD-EPI
Chronic kidney disease epidemiology collaboration
CT
Computed tomography
ESRD
End-stage renal disease
GFR
Glomerular filtration rate
GWAS
Genome-wide association studies
IDMS
Isotope dilution Mass Spectrometry
KDIGO
Kidney disease improving global outcomes
RAS
Renin-angiotensin aldosterone system
sCr
Serum creatinine
sUA
Serum uric acid
TGF-β
Transforming growth factor beta
TKV
Total kidney volume
TNF-α
Tumor necrosis factor alpha.

Background

Uric acid has been regarded as a marker rather than a risk factor for the development of chronic kidney disease (CKD) because a low glomerular filtration rate (GFR) induces elevation of serum uric acid (sUA) level despite compensatory increases in urinary and gastrointestinal urate excretions. However, recent studies suggested an independent role of uric acid in the development of CKD. For instance, hyperuricemia is shown to be associated with an increased risk of CKD in large cohort studies such as the Atherosclerosis Risk in Communities and the Cardiovascular Health Study [1] and the Vienna Health Screening Project [2]. Moreover, hyperuricemia has been reported to be associated with the development of end-stage renal disease (ESRD) [3, 4]. However, the pathogenic role of hyperuricemia in the progression of CKD is still controversial. Hyperuricemia has been reported as a risk factor for renal progression in IgA nephropathy [5], whereas sUA level was not associated with disease progression or kidney failure in general CKD population [6, 7].
Association between autosomal dominant polycystic kidney disease (ADPKD) and hyperuricemia was first described by Rivera et al. [8]. ADPKD is frequently associated with hyperuricemia and gout [9], although fractional excretion of uric acid was not different from CKD groups of different etiologies [10, 11]. Recent retrospective studies reported the association of high sUA levels with early-onset hypertension, large kidney volume, and increased risk of ESRD [12] or progression of renal dysfunction [13]. Considering that hyperuricemia can be a possible correctable risk factor for ADPKD progression, we postulated that 1) hyperuricemia is associated with concurrent renal function, 2) hyperuricemia contributes independently to deterioration of renal function, 3) and the correction of hyperuricemia attenuates renal function decline in ADPKD.

Subjects and methods

Study subjects

A total of 612 patients were screened at the ADPKD clinic in Seoul National University Hospital. ADPKD was diagnosed according to the unified criteria proposed by Pei et al. [14]. We selected 365 patients aged > 18 years with an estimated GFR (eGFR) of > 15 mL/min/1.73 m2 at the initial evaluation and who were followed up for > 1 year. Patients with certain conditions that can independently influence renal function such as diabetes [15], pregnancy, or malignancy were excluded from the analysis. The data were collected retrospectively from each patient between August 1999 and March 2012. The patients underwent a standardized evaluation including detailed family history, renal function, and computed tomography (CT) scan, which was obtained using a multidetector CT scanner (Somatom Sensation 16, Siemens; LightSpeed Ultra 8, GE; Brilliance CT 64, Philips; Somatom Definition, Siemens). The following clinical data and information were collected every 3–6 months: baseline epidemiologic profiles (age, sex, and body weight), medical history including diabetes, hypertension, and gout, medication history including antihypertensive medications and hypouricemic agents, blood pressure, and laboratory results (serum creatinine (sCr), sUA, hemoglobin, serum albumin levels, and urine dipstick). The CT scan was performed every 2 years and total kidney volume (TKV) was calculated using the modified ellipsoid method [16]. This study was approved by the Institutional Review Board of Seoul National University Hospital (H-1002-028-309). Informed consent was obtained from the subjects in accordance with the Declaration of Helsinki.

Evaluation of renal function and hypertension

The sCr was measured at 3- or 6-month interval using the Jaffe method by Hitachi 7600 and Toshiba-200FR, which was calculated to Isotope Dilution Mass Spectrometry (IDMS)-traceable sCr. The Chronic Kidney Disease Epidemiology collaboration (CKD-EPI) formula was used to calculate eGFR. Delta eGFR (ΔeGFR/year) was calculated using the equation (recent eGFR − eGFR at initial visit)/follow-up duration (years). The CKD stage was classified according to the Kidney Disease Improving Global Outcomes (KDIGO) guidelines [17]. ESRD was defined by an eGFR of <15 mL/min/1.73 m2 or initiation of renal replacement therapy. Albuminuria was defined as a stick albumin level of >1+ by urine dipstick test. Urine albumin was quantified by the immunoturbidimetric assay using Toshiba-120FR. Hypertension was defined by a systolic blood pressure of >140 mmHg, diastolic blood pressure of >90 mmHg, or current use of antihypertensive medication.

Uric acid measurement and hyperuricemia management

The sUA level was determined using the uricase method (Hitachi 7600 and Toshiba-200FR). Hyperuricemia was defined by a sUA level of ≥ 7.0 mg/dL in males and ≥ 6.0 mg/dL in females or when patients have received hypouricemic treatment. Hypouricemic medications were prescribed in patients with gout, a history of uric acid stones, or persistent elevation of sUA level of > 8.0 mg/dL that was not controlled by dietary modification in 2 successive visits.

Statistical analyses

Continuous variables were expressed as mean ± standard deviation. Student t -test was used to compare the continuous variables between the groups. Linear regression analysis was used to find the association between sUA and clinical variables. Cox proportional hazard model was used to compare renal survival between groups. Wilcoxon signed-rank test was used to evaluate the effect of hypouricemic medication on the rate of renal progression before and after the start of medication. P < 0.05 was considered statistically significant. All the statistical analyses were performed using SPSS version 19.0 (SPSS Inc., Chicago, IL).

Results

Baseline clinical characteristics according to the presence of hyperuricemia

The clinical characteristics of 365 patients on initial evaluation are summarized in Table 1. They were divided into normouricemic (Group A, n = 278) and hyperuricemic (Group B, n = 87) groups. In Group B, 12 patients (3.3%) underwent hypouricemic treatment. The prevalence of hyperuricemia increased according to CKD stage as follows: 6.3% (6/95) in stage 1, 15.2% (28/184) in stage 2, 52.4% (33/63) in stage 3, and 87.0% (20/23) in stage 4.
Table 1
Baseline characteristics of participants at initial evaluation
 
Total (n = 365)
Group A*(n = 278)
Group B(n = 87)
p-value
Male (%)
183 (50.1%)
122 (43.9%)
61 (70.1%)
< 0.001
Age (yrs)
43.5 ± 11.9
42.4 ± 11.3
47.1 ± 13.2
0.003
 18-39
159 (43.6%)
128 (46.0%)
31 (35.6%)
 
 40-59
177 (48.5%)
133 (47.8%)
44 (50.6%)
 
 ≥ 60
29 (7.9%)
17 (6.1%)
12 (13.8%)
 
Follow up time (months)
73.5 ± 43.4
76.2 ± 43.5
64.7 ± 42.1
0.031
Hypertension (%)
257 (70.4%)
186 (66.9%)
71 (81.6%)
0.004
Systolic BP (mmHg)
136.4 ± 19.9
136.4 ± 20.3
136.3 ± 18.8
0.967
Diastolic BP (mmHg)
85.6 ± 13.9
86.1 ± 14.2
83.6 ± 12.8
0.190
Urinary stone (%)
96 (26.3%)
71 (25.5%)
25 (28.7%)
0.556
sUA (mg/dL)
5.51 ± 1.71
4.80 ± 1.11
7.76 ± 1.26
< 0.001
sCr (mg/dL)
1.17 ± 0.48
1.01 ± 0.23
1.67 ± 0.69
< 0.001
eGFR (ml/min/1.73 m2)
75.1 ± 24.1
81.6 ± 19.3
54.5 ± 26.3
< 0.001
CKD stage
   
< 0.001
 Stage 1
95 (26.0%)
53 (19.1%)
6 (6.9%)
 
 Stage 2
184 (50.4%)
189 (68.0%)
28 (32.2%)
 
 Stage 3
63 (17.3%)
35 (12.6%)
33 (37.9%)
 
 Stage 4
23 (6.3%)
1 (0.4%)
20 (23.0%)
 
Urine pH
6.03 ± 0.73
6.13 ± 0.73
5.74 ± 0.66
< 0.001
Dipstick albumin
   
0.017
 None to 1+
342 (93.7%)
266 (95.7%)
76 (87.4%)
 
 > 1+
23 (6.3%)
12 (4.3%)
11 (12.6%)
 
TKV (mL)
1,524 ± 1,171
1,416 ± 1,050
1,963 ± 1,500
0.013
Losartan (%)
58 (15.9%)
50 (18.0%)
8 (9.2%)
0.025
Diuretic (%)
36 (9.9%)
27 (9.7%)
9 (10.3%)
0.863
*Group A : sUA < 7.0 mg/dL (Male) or sUA < 6.0 mg/dL (Female), Group B: sUA ≥ 7.0 mg/dL (Male) or sUA ≥ 6.0 mg/dL (Female) or on hypouricemic medication. BP, blood pressure; sUA, serum uric acid; sCr, serum creatinine; eGFR, estimated glomerular filtration rate; CKD, chronic kidney disease; TKV, total kidney volume.
When compared with those in Group A, the patients in Group B were slightly older (47.1 ± 13.2 vs. 42.4 ± 11.3 years, p = 0.003), had higher prevalence of hypertension (81.6% vs. 66.9%, p = 0.004), showed male predominance (70.1% vs. 43.5%, p < 0.001), and had shorter follow-up duration (64.7 ± 42.1 vs. 76.2 ± 43.5 months, p = 0.031). Group B had higher sCr levels (1.67 ± 0.69 vs. 1.01 ± 0.23 mg/dL, p < 0.001) and lower eGFR (54.5 ± 26.3 vs. 81.6 ± 19.3 mL/min/1.73 m2, p < 0.001) compared to Group A.

Hyperuricemia was negatively correlated with initial eGFR

Associations between sUA and sCr levels, eGFR, TKV, and albumin to creatinine ratio (ACR) were analysed (Figure 1). The sUA level was positively correlated with sCr level (R 2 = 0.370, p < 0.001), ACR (R 2 = 0.011, p = 0.111) and TKV (R 2 = 0.045, p < 0.001) but negatively correlated with eGFR (R 2 = 0.202, p < 0.001). Simple linear regression analysis in 353 patients without hypouricemic treatments at the initial evaluation revealed that age, albuminuria, TKV, and sUA level were correlated with reduced initial eGFR. After adjustment for age, male sex, blood pressure, albuminuria, and TKV, sUA levels still remained a significant factor (β = −5.117, p < 0.001; Table 2).
Table 2
Factors associated with eGFR in ADPKD patients *
 
   Univariate
 
   Multivariate
 
 
Beta ± SE
p-value
Beta ± SE
p-value
Age (yr)
−1.208 ± 0.087
< 0.001
−0.986 ± 0.086
< 0.001
Female (vs. male)
−2.714 ± 2.520
0.282
−9.217 ± 2.224
< 0.001
Mean BP (mm Hg)
−0.229 ± 0.089
0.010
0.043 ± 0.073
0.558
sUA (mg/dL)
−6.282 ± 0.666
< 0.001
−5.117 ± 0.666
< 0.001
Albuminuria
−14.687 ± 5.160
0.005
−4.956 ± 3.890
0.204
TKV (mL)
−19.524 ± 4.060
< 0.001
−12.782 ± 3.245
< 0.001
*353 patients without hypouricemic medication. transformed in logarithmic scale. eGFR, estimated glomerular filtration rate; ADPKD, autosomal dominant polycystic kidney disease; SE, standard error; BP, blood pressure; sUA, serum uric acid; TKV, total kidney volume.

Hyperuricemia was not independently associated with annual eGFR decline and ESRD progression

Among 365 patients, 42 patients (11.5%; 20 in Group A and 22 in Group B) progressed to ESRD during the follow-up (mean duration, 73.5 ± 43.4 months). When evaluating the deterioration of eGFR, we examined 296 patients not taking hypouricemic medication (255 patients from Group A and 41 patients from Group B) to exclude the influence of hypouricemic agents (Table 3). The mean sUA level was 4.69 ± 1.08 mg/dL in normouricemic Group A and 7.61 ± 0.95 mg/dL in Group B (p < 0.001), indicating a difference of 2.9 mg/dL. Progression to ESRD occurred in 14 (34.1%) out of 41 patients in Group B and 14 (5.4%) out of 255 patients in Group A (p < 0.001). Group B showed a faster annual decline in eGFR (ΔeGFR/year) than Group A in absolute (−1.87 ± 3.30 vs. −0.29 ± 4.22 mL/min/1.73 m2 per year, p = 0.026) and relative values (−6.23% ± 9.84% vs. −0.72% ± 6.03% per year, p = 0.001).
Table 3
Annual eGFR decline (ΔeGFR) in ADPKD patients according to the presence of hyperuricemia *
 
Group A(n = 255)
Group B(n = 41)
p-value
Mean sUA level (mg/dL)
4.69 ± 1.08
7.61 ± 0.95
< 0.001
F/U time (months)
73.0 ± 42.2
62.0 ± 46.8
0.134
Initial eGFR (mL/min/1.73 m2)
82.0 ± 19.6
52.2 ± 27.4
< 0.001
Final eGFR (mL/min/1.73 m2)
75.6 ± 27.9
48.3 ± 31.7
< 0.001
ΔeGFR (mL/min/1.73 m2/yr)
−0.29 ± 4.22
−1.87 ± 3.30
0.026
ΔeGFR (%/yr)
−0.72 ± 6.03
−6.23 ± 9.84
0.001
ESRD progression, n (%)
14 (5.4%)
14 (34.1%)
< 0.001
*A total of 296 patients without hypouricemic medications were included in the analysis. Group A: sUA < 7.0 mg/dL (Male) or < 6.0 mg/dL (Female), Group B: sUA ≥ 7.0 mg/dL (Male) or ≥ 6.0 mg/dL. sUA, serum uric acid; ADPKD, autosomal dominant polycystic kidney disease; F/U, follow-up; eGFR, estimated glomerular filtration rate; ESRD, end-stage renal disease.
Linear regression analysis was performed to determine the factors influencing ΔeGFR/year. In univariate analysis, age, hypertension, initial eGFR, and sUA level were significantly associated with ΔeGFR/year. However, after adjusting for age, sex, blood pressure, and initial eGFR, neither sUA level nor hyperuricemia was significantly associated with ΔeGFR/year (Table 4). In the same manner, multivariate Cox regression analysis showed that hyperuricemia was not an independent risk factor for ESRD (Table 5).
Table 4
Factors affecting annual eGFR decline (ΔeGFR) in patients with ADPKD
 
   Univariate
 
   Multivariate
 
Beta ± SE
p-value
Beta ± SE
p-value
Age (yr)
−0.067 ± 0.020
0.001
−0.071 ± 0.034
0.036
Female
0.369 ± 0.491
0.453
0.775 ± 0.731
0.290
Mean BP (mm Hg)
−0.065 ± 0.018
0.001
−0.056 ± 0.019
0.003
History of stone
0.260 ± 0.377
0.491
-
-
sUA (mg/dL)
−0.330 ± 0.165
0.047
−0.212 ± 0.275
0.443
Initial eGFR (mL/min/1.73 m2)
0.036 ± 0.010
0.001
−0.001 ± 0.020
0.971
eGFR, estimated glomerular filtration rate; ADPKD, autosomal dominant polycystic kidney disease; BP, blood pressure; sUA, serum uric acid; SE, standard error.
Table 5
Multivariate cox regression for development of end-stage renal disease
 
Hazard ratio
   95% CI
p-value
Age (year)
0.931
0.864, 1.003
0.060
Female
3.664
0.011, 1239.509
0.662
Initial eGFR (mL/min/1.73 m2)
0.720
0.620, 0.836
< 0.001
Mean BP (mm Hg)
1.023
0.978, 1.070
0.331
Hyperuricemia (Group B vs. A)
3.082
0.008, 1259.413
0.714
Group A: sUA <7.0 mg/dL, Group B: sUA ≥7.0 mg/dL. CI, confidence interval; eGFR, estimated glomerular filtration rate; BP, blood pressure.

Correction of hyperuricemia may attenuate renal function decline

Although we were not able to show the independent association of sUA level with renal progression in ADPKD patients who were not taking hypouricemic agents, we did exploratory analysis in a subgroup of our cohort to investigate the effect of hypouricemic treatment on eGFR. Among 57 patients newly starting hypouricemic treatment during follow-up period, 53 patients had follow-up period more than 1 year of pre- and post-treatment. Serum UA and eGFR were measured every 3 months. The slopes of the annual change in eGFR before and after hypouricemic treatment were analyzed in 53 patients receiving either allopurinol (n = 12) or benzbromarone (n = 41) more than a year.
Among them, 13 patients were included in CKD stage 1 or 2, 19 in CKD stage 3a, 11 in CKD stage 3b, and 10 in CKD stage 4 before treatment initiation. Mean sUA was 8.70 ± 0.78 mg/dL and mean eGFR was 47.9 ± 20.3 mL/min/1.73 m2 at the time of hypouricemic treatment initiation. The change in sUA and eGFR before and after 1 year of treatment initiation was analyzed (Table 6). After the hypouricemic treatment, the annual decline of renal function (ΔGFR/year) slowed from −5.3 ± 8.2 to 0.2 ± 6.2 mL/min/1.73 m2 per year (Wilcoxon signed-rank test, p = 0.001). Further analysis showed that correction of hyperuricemia may attenuate renal function decline in early CKD stages (1 ~ 3a) whereas it may not attenuate renal function decline in advanced CKD stages (3b ~ 4). Serial changes in sUA and eGFR are presented in Figure 2 and Additional file 1: Table S1. There was no difference between allopurinol and benzbromarone group (data not shown). However, significant changes in systolic blood pressure were also observed after hypouricemic treatment (Additional file 1: Table S2). Because lowering blood pressure itself and change of anti-hypertensive medication (32.1% during the observation period), especially of renin-angiotensin system blockers may affect eGFR, the effect of blood pressure on eGFR was examined by using generalized estimating equation, showing that no significant effect over 2-year observation period (data not shown). In summary, blood pressure changes did not significantly influence eGFR changes after hypouricemic treatment.
Table 6
Annual eGFR change (ΔeGFR) before and after hypouricemic treatment
CKD stage
PreTx (1 yr)
Initiation of Tx
PostTx (1 yr)
p-value*
Total (n = 53)
 sUA (mg/dL)
7.88 ± 1.09
8.70 ± 0.78
6.22 ± 1.40
 
 eGFR (mL/min/1.73 m2)
53.2 ± 20.3
47.9 ± 20.3
48.1 ± 22.9
 
 ΔeGFR (mL/min/1.73 m2/yr)
−5.3 ± 8.2
 
0.2 ± 6.2
0.001
Stage 1-3a (n = 32)
 sUA (mg/dL)
8.06 ± 1.15
8.66 ± 0.90
5.70 ± 1.41
 
 eGFR (mL/min/1.73 m2)
64.8 ± 16.3
60.3 ± 16.1
62.6 ± 16.4
 
 ΔeGFR (mL/min/1.73 m2/yr)
−4.5 ± 9.4
 
2.3 ± 5.7
0.001
Stage 3b-4 (n = 21)
 sUA (mg/dL)
7.62 ± 0.97
8.77 ± 0.59
7.03 ± 0.94
 
 eGFR (mL/min/1.73 m2)
35.6 ± 10.9
29.1 ± 7.2
26.0 ± 10.0
 
 ΔeGFR (mL/min/1.73 m2/yr)
−6.6 ± 5.7
 
−3.1 ± 5.6
0.465
*Wilcoxon signed-rank test. eGFR, estimated glomerular filtration rate; CKD, chronic kidney disease; Tx, treatment; sUA, serum uric acid.

Discussion

In our retrospective cohort study, we demonstrated that sUA level is associated with renal function in ADPKD. However, we were not able to show that hyperuricemia independently contributes to the renal progression.
In our study, sUA level was associated with concurrent eGFR, independent of age, sex, albuminuria, and TKV. This result is consistent with those from previous studies reporting that hyperuricemia is associated with early-onset hypertension, TKV, and increased risk of ESRD [12]. Hyperuricemia also showed negative correlation with ΔeGFR/year. However, after adjusting age, sex, blood pressure, and initial eGFR, the influence of hyperuricemia was not statistically significant. This result may be partly explained by the small difference in sUA level between patients with and without hyperuricemia because we excluded high-risk patients with hyperuricemia who were already being treated. Moreover, the follow-up duration was relatively short. The median follow-up duration was only 73.5 months, and 41.6% of the patients were followed up for <5 years.
Although we failed to show independent effect of hyperuricemia on renal progression, we examined the effect of hypouricemic medication on the slope of annual eGFR change. Correction of hyperuricemia appeared to attenuate ΔeGFR/year, especially for the patients in early CKD stages (1~3a), suggesting that hyperuricemia may influence renal function deterioration in some patients. Although our result is limited by small number of patients and short observation time, this is consistent with the previous finding that allopurinol therapy preserved sCr level and lowered the risk of renal progression in hyperuricemic patients with mild to moderate CKD [18, 19]. On the contrary, hypouricemic treatment had no effect on preserving eGFR in patients with CKD stage 3b to 4. This lack of effect may be related to the lower efficacy of uricosuric agents (e.g. benzbromarone) in the advance kidney failure. Mean sUA level decreased from 8.9 ± 0.9 mg/dL to 5.7 ± 1.4 mg/dL in stage 1-3a group, whereas from 8.8 ± 0.6 mg/dL to 7.0 ± 0.9 mg/dL in stage 3b-4 group. The effect of hypouricemic agents for renal function preservation in advanced CKD stage should be evaluated with more potent hypouricemic agents such as febuxostat in long-term prospective studies.
Several mechanisms were proposed to explain renal dysfunction by hyperuricemia. First, association between increased sUA level and cardiovascular disease has been reported [20]. In our study, no difference in cardiovascular event was observed between the normouricemic and hyperuricemic groups (data not shown). However, the possibility of hyperuricemia causing renal function decline through cardiovascular events cannot be excluded because of the small sample size and short-term follow-up [21]. Second, hyperuricemia may induce direct renal injury through the activation of the renin-angiotensin aldosterone system (RAS). Renal cyst enlargement in ADPKD is known to be associated with stimulation of the circulating and intrarenal RAS [22]. Helal et al. [12] speculated that endothelial dysfunction, which is a well-known characteristic of ADPKD [23], and activation of RAS induced by hyperuricemia would contribute to the progression of ESRD in ADPKD. Endothelial dysfunction and early-onset hypertension are important prognostic factors for the deterioration of renal function in ADPKD [24]. In addition, soluble uric acid might activate inflammatory pathways such as tumor necrosis factor alpha (TNF-α) and chemokines [25] and C-reactive protein [26], possibly leading to interstitial fibrosis. In CKD patients, association between hyperuricemia and increased urinary transforming growth factor beta (TGF-β) was also reported [27].
The pathogenic role of hyperuricemia in renal progression needs to be further investigated in regard to urate handling in ADPKD. In ADPKD, altered tubular membrane transport process might affect renal urate handling and homeostasis. Compared to general population, a higher prevalence of uric acid stone was noted in ADPKD. However, previous studies showed inconsistent data about urate handling in ADPKD [10, 11]. We also performed immunohistochemical staining of four major urate transporters (URAT1, GLUT9, NPT4, and OAT3) in 3 ADPKD kidneys and 1 normal control kidney, which revealed strong expression of all 4 urate transporters along the compressed proximal tubules nearby renal cysts [28] (Additional file 2: Figure S1). It may suggest the possibility of altered urate handling in ADPKD that may lead to hyperuricemia. However, our result needs to be validated through functional study in a large number of samples.
Our study has several limitations. This is a single-center, retrospective cohort study. Therefore, further prospective study is warranted to evaluate the causal relationship between hyperuricemia and renal function decline. In addition, genetic factor, which is the strongest predictor of renal progression, has not been evaluated in this study. In the analysis of the hyperuricemia and annual eGFR decline, mixed effects model or generalized estimating equation model would be best suited. However, they could not be used due to some missing data of sUA and eGFR during follow-up period and the interval of measurement was not regular. Lastly, we failed to show the independent effect of hyperuricemia on renal progression.

Conclusions

In conclusion, we failed to show the independent effect of hyperuricemia on renal progression. Nevertheless, for the first time, we demonstrated the correction of hyperuricemia with uric acid lowering agents may attenuate renal progression in the early CKD stages suggesting that treatment of hyperuricemia would be beneficial for some ADPKD patients to preserve renal function. Further prospective study is needed to verify the impact of hyperuricemia control on disease progression in ADPKD.

Acknowledgments

We greatly appreciate MH Kim and HH Jang for their dedicated efforts in giving patient education at our outpatient clinic and for professional assistance in gathering information. This study was supported in part by Cooperative Research Grant 2009 from the Korean Society of Nephrology and by a grant of the Korean Health Technology R & D Project, Ministry of Health & Welfare, Republic of Korea (A120017).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​ ) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MH collected the data and drafted the manuscript. HP participated in the design of the study and drafted the manuscript. HK participated in the acquisition of data. HC, HH participated in the analysis and interpretation of data. JJ, AK carried out immunohistochemical staining. SK carried out kidney volume measurement. HC, DK participated in critical revising the manuscript. JY, KO participated in the design of the study and interpretation of data. YH helped interpretation of data and drafted the manuscript. CA conceived of the study, interpretation of data and revising the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Weiner DE, Tighiouart H, Elsayed EF, Griffith JL, Salem DN, Levey AS: Uric acid and incident kidney disease in the community. J Am Soc Nephrol. 2008, 19 (6): 1204-1211. 10.1681/ASN.2007101075.CrossRefPubMedPubMedCentral Weiner DE, Tighiouart H, Elsayed EF, Griffith JL, Salem DN, Levey AS: Uric acid and incident kidney disease in the community. J Am Soc Nephrol. 2008, 19 (6): 1204-1211. 10.1681/ASN.2007101075.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Obermayr RP, Temml C, Gutjahr G, Knechtelsdorfer M, Oberbauer R, Klauser-Braun R: Elevated uric acid increases the risk for kidney disease. J Am Soc Nephrol. 2008, 19 (12): 2407-2413. 10.1681/ASN.2008010080.CrossRefPubMedPubMedCentral Obermayr RP, Temml C, Gutjahr G, Knechtelsdorfer M, Oberbauer R, Klauser-Braun R: Elevated uric acid increases the risk for kidney disease. J Am Soc Nephrol. 2008, 19 (12): 2407-2413. 10.1681/ASN.2008010080.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Iseki K, Ikemiya Y, Inoue T, Iseki C, Kinjo K, Takishita S: Significance of hyperuricemia as a risk factor for developing ESRD in a screened cohort. Am J Kidney Dis. 2004, 44 (4): 642-650. 10.1053/j.ajkd.2004.06.006.CrossRefPubMed Iseki K, Ikemiya Y, Inoue T, Iseki C, Kinjo K, Takishita S: Significance of hyperuricemia as a risk factor for developing ESRD in a screened cohort. Am J Kidney Dis. 2004, 44 (4): 642-650. 10.1053/j.ajkd.2004.06.006.CrossRefPubMed
4.
Zurück zum Zitat Hsu CY, Iribarren C, McCulloch CE, Darbinian J, Go AS: Risk factors for end-stage renal disease: 25-year follow-up. Arch Intern Med. 2009, 169 (4): 342-350. 10.1001/archinternmed.2008.605.CrossRefPubMedPubMedCentral Hsu CY, Iribarren C, McCulloch CE, Darbinian J, Go AS: Risk factors for end-stage renal disease: 25-year follow-up. Arch Intern Med. 2009, 169 (4): 342-350. 10.1001/archinternmed.2008.605.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Shi Y, Chen W, Jalal D, Li Z, Chen W, Mao H, Yang Q, Johnson RJ, Yu X: Clinical outcome of hyperuricemia in IgA nephropathy: a retrospective cohort study and randomized controlled trial. Kidney Blood Press Res. 2012, 35 (3): 153-160. 10.1159/000331453.CrossRefPubMed Shi Y, Chen W, Jalal D, Li Z, Chen W, Mao H, Yang Q, Johnson RJ, Yu X: Clinical outcome of hyperuricemia in IgA nephropathy: a retrospective cohort study and randomized controlled trial. Kidney Blood Press Res. 2012, 35 (3): 153-160. 10.1159/000331453.CrossRefPubMed
6.
Zurück zum Zitat Sturm G, Kollerits B, Neyer U, Ritz E, Kronenberg F, Group MS: Uric acid as a risk factor for progression of non-diabetic chronic kidney disease? The Mild to Moderate Kidney Disease (MMKD) Study. Exp Gerontol. 2008, 43 (4): 347-352. 10.1016/j.exger.2008.01.006.CrossRefPubMed Sturm G, Kollerits B, Neyer U, Ritz E, Kronenberg F, Group MS: Uric acid as a risk factor for progression of non-diabetic chronic kidney disease? The Mild to Moderate Kidney Disease (MMKD) Study. Exp Gerontol. 2008, 43 (4): 347-352. 10.1016/j.exger.2008.01.006.CrossRefPubMed
7.
Zurück zum Zitat Madero M, Sarnak MJ, Wang X, Greene T, Beck GJ, Kusek JW, Collins AJ, Levey AS, Menon V: Uric acid and long-term outcomes in CKD. Am J Kidney Dis. 2009, 53 (5): 796-803. 10.1053/j.ajkd.2008.12.021.CrossRefPubMedPubMedCentral Madero M, Sarnak MJ, Wang X, Greene T, Beck GJ, Kusek JW, Collins AJ, Levey AS, Menon V: Uric acid and long-term outcomes in CKD. Am J Kidney Dis. 2009, 53 (5): 796-803. 10.1053/j.ajkd.2008.12.021.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Rivera JV, Martinez Maldonado M, Ramirezdearellano GA, Ehrlich L: Association of Hyperuricemia and Polycystic Kidney Disease. Bol Asoc Med P R. 1965, 57: 251-262.PubMed Rivera JV, Martinez Maldonado M, Ramirezdearellano GA, Ehrlich L: Association of Hyperuricemia and Polycystic Kidney Disease. Bol Asoc Med P R. 1965, 57: 251-262.PubMed
9.
Zurück zum Zitat Newcombe DS: Letter: Gouty arthritis and polycystic kidney disease. Ann Intern Med. 1973, 79 (4): 605-10.7326/0003-4819-79-4-605.CrossRefPubMed Newcombe DS: Letter: Gouty arthritis and polycystic kidney disease. Ann Intern Med. 1973, 79 (4): 605-10.7326/0003-4819-79-4-605.CrossRefPubMed
10.
Zurück zum Zitat Mejias E, Navas J, Lluberes R, Martinez-Maldonado M: Hyperuricemia, gout, and autosomal dominant polycystic kidney disease. Am J Med Sci. 1989, 297 (3): 145-148. 10.1097/00000441-198903000-00002.CrossRefPubMed Mejias E, Navas J, Lluberes R, Martinez-Maldonado M: Hyperuricemia, gout, and autosomal dominant polycystic kidney disease. Am J Med Sci. 1989, 297 (3): 145-148. 10.1097/00000441-198903000-00002.CrossRefPubMed
11.
Zurück zum Zitat Kaehny WD, Tangel DJ, Johnson AM, Kimberling WJ, Schrier RW, Gabow PA: Uric acid handling in autosomal dominant polycystic kidney disease with normal filtration rates. Am J Med. 1990, 89 (1): 49-52. 10.1016/0002-9343(90)90097-W.CrossRefPubMed Kaehny WD, Tangel DJ, Johnson AM, Kimberling WJ, Schrier RW, Gabow PA: Uric acid handling in autosomal dominant polycystic kidney disease with normal filtration rates. Am J Med. 1990, 89 (1): 49-52. 10.1016/0002-9343(90)90097-W.CrossRefPubMed
12.
Zurück zum Zitat Helal I, McFann K, Reed B, Yan XD, Schrier RW, Fick-Brosnahan GM: Serum uric acid, kidney volume and progression in autosomal-dominant polycystic kidney disease. Nephrol Dial Transplant. 2013, 28 (2): 380-385. 10.1093/ndt/gfs417.CrossRefPubMed Helal I, McFann K, Reed B, Yan XD, Schrier RW, Fick-Brosnahan GM: Serum uric acid, kidney volume and progression in autosomal-dominant polycystic kidney disease. Nephrol Dial Transplant. 2013, 28 (2): 380-385. 10.1093/ndt/gfs417.CrossRefPubMed
13.
Zurück zum Zitat Panizo N, Goicoechea M, Garcia de Vinuesa S, Arroyo D, Yuste C, Rincon A, Verdalles U, Ruiz-Caro C, Quiroga B, Luno J: Chronic kidney disease progression in patients with autosomal dominant polycystic kidney disease. Nefrologia. 2012, 32 (2): 197-205.PubMed Panizo N, Goicoechea M, Garcia de Vinuesa S, Arroyo D, Yuste C, Rincon A, Verdalles U, Ruiz-Caro C, Quiroga B, Luno J: Chronic kidney disease progression in patients with autosomal dominant polycystic kidney disease. Nefrologia. 2012, 32 (2): 197-205.PubMed
14.
Zurück zum Zitat Pei Y, Obaji J, Dupuis A, Paterson AD, Magistroni R, Dicks E, Parfrey P, Cramer B, Coto E, Torra R, San Millan JL, Gibson R, Breuning M, Peters D, Ravine D: Unified criteria for ultrasonographic diagnosis of ADPKD. J Am Soc Nephrol. 2009, 20 (1): 205-212. 10.1681/ASN.2008050507.CrossRefPubMedPubMedCentral Pei Y, Obaji J, Dupuis A, Paterson AD, Magistroni R, Dicks E, Parfrey P, Cramer B, Coto E, Torra R, San Millan JL, Gibson R, Breuning M, Peters D, Ravine D: Unified criteria for ultrasonographic diagnosis of ADPKD. J Am Soc Nephrol. 2009, 20 (1): 205-212. 10.1681/ASN.2008050507.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Reed B, Helal I, McFann K, Wang W, Yan XD, Schrier RW: The impact of type II diabetes mellitus in patients with autosomal dominant polycystic kidney disease. Nephrol Dial Transplant. 2012, 27 (7): 2862-2865. 10.1093/ndt/gfr744.CrossRefPubMed Reed B, Helal I, McFann K, Wang W, Yan XD, Schrier RW: The impact of type II diabetes mellitus in patients with autosomal dominant polycystic kidney disease. Nephrol Dial Transplant. 2012, 27 (7): 2862-2865. 10.1093/ndt/gfr744.CrossRefPubMed
16.
Zurück zum Zitat Fick-Brosnahan GM, Belz MM, McFann KK, Johnson AM, Schrier RW: Relationship between renal volume growth and renal function in autosomal dominant polycystic kidney disease: a longitudinal study. Am J Kidney Dis. 2002, 39 (6): 1127-1134. 10.1053/ajkd.2002.33379.CrossRefPubMed Fick-Brosnahan GM, Belz MM, McFann KK, Johnson AM, Schrier RW: Relationship between renal volume growth and renal function in autosomal dominant polycystic kidney disease: a longitudinal study. Am J Kidney Dis. 2002, 39 (6): 1127-1134. 10.1053/ajkd.2002.33379.CrossRefPubMed
17.
Zurück zum Zitat Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group: KDIGO clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl. 2013, 3 (1): 1-150.CrossRef Kidney Disease: Improving Global Outcomes (KDIGO) CKD Work Group: KDIGO clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl. 2013, 3 (1): 1-150.CrossRef
18.
Zurück zum Zitat Siu YP, Leung KT, Tong MK, Kwan TH: Use of allopurinol in slowing the progression of renal disease through its ability to lower serum uric acid level. Am J Kidney Dis. 2006, 47 (1): 51-59. 10.1053/j.ajkd.2005.10.006.CrossRefPubMed Siu YP, Leung KT, Tong MK, Kwan TH: Use of allopurinol in slowing the progression of renal disease through its ability to lower serum uric acid level. Am J Kidney Dis. 2006, 47 (1): 51-59. 10.1053/j.ajkd.2005.10.006.CrossRefPubMed
19.
Zurück zum Zitat Goicoechea M, de Vinuesa SG, Verdalles U, Ruiz-Caro C, Ampuero J, Rincon A, Arroyo D, Luno J: Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin J Am Soc Nephrol. 2010, 5 (8): 1388-1393. 10.2215/CJN.01580210.CrossRefPubMedPubMedCentral Goicoechea M, de Vinuesa SG, Verdalles U, Ruiz-Caro C, Ampuero J, Rincon A, Arroyo D, Luno J: Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin J Am Soc Nephrol. 2010, 5 (8): 1388-1393. 10.2215/CJN.01580210.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Kanbay M, Segal M, Afsar B, Kang DH, Rodriguez-Iturbe B, Johnson RJ: The role of uric acid in the pathogenesis of human cardiovascular disease. Heart. 2013, 99 (11): 759-766. 10.1136/heartjnl-2012-302535.CrossRefPubMed Kanbay M, Segal M, Afsar B, Kang DH, Rodriguez-Iturbe B, Johnson RJ: The role of uric acid in the pathogenesis of human cardiovascular disease. Heart. 2013, 99 (11): 759-766. 10.1136/heartjnl-2012-302535.CrossRefPubMed
21.
Zurück zum Zitat Helal I, Reed B, Mettler P, Mc Fann K, Tkachenko O, Yan XD, Schrier RW: Prevalence of cardiovascular events in patients with autosomal dominant polycystic kidney disease. Am J Nephrol. 2012, 36 (4): 362-370. 10.1159/000343281.CrossRefPubMedPubMedCentral Helal I, Reed B, Mettler P, Mc Fann K, Tkachenko O, Yan XD, Schrier RW: Prevalence of cardiovascular events in patients with autosomal dominant polycystic kidney disease. Am J Nephrol. 2012, 36 (4): 362-370. 10.1159/000343281.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Schrier RW: Renal volume, renin-angiotensin-aldosterone system, hypertension, and left ventricular hypertrophy in patients with autosomal dominant polycystic kidney disease. J Am Soc Nephrol. 2009, 20 (9): 1888-1893. 10.1681/ASN.2008080882.CrossRefPubMed Schrier RW: Renal volume, renin-angiotensin-aldosterone system, hypertension, and left ventricular hypertrophy in patients with autosomal dominant polycystic kidney disease. J Am Soc Nephrol. 2009, 20 (9): 1888-1893. 10.1681/ASN.2008080882.CrossRefPubMed
23.
Zurück zum Zitat Wang D, Iversen J, Wilcox CS, Strandgaard S: Endothelial dysfunction and reduced nitric oxide in resistance arteries in autosomal-dominant polycystic kidney disease. Kidney Int. 2003, 64 (4): 1381-1388. 10.1046/j.1523-1755.2003.00236.x.CrossRefPubMed Wang D, Iversen J, Wilcox CS, Strandgaard S: Endothelial dysfunction and reduced nitric oxide in resistance arteries in autosomal-dominant polycystic kidney disease. Kidney Int. 2003, 64 (4): 1381-1388. 10.1046/j.1523-1755.2003.00236.x.CrossRefPubMed
24.
Zurück zum Zitat Johnson AM, Gabow PA: Identification of patients with autosomal dominant polycystic kidney disease at highest risk for end-stage renal disease. J Am Soc Nephrol. 1997, 8 (10): 1560-1567.PubMed Johnson AM, Gabow PA: Identification of patients with autosomal dominant polycystic kidney disease at highest risk for end-stage renal disease. J Am Soc Nephrol. 1997, 8 (10): 1560-1567.PubMed
25.
Zurück zum Zitat Zhou Y, Fang L, Jiang L, Wen P, Cao H, He W, Dai C, Yang J: Uric acid induces renal inflammation via activating tubular NF-kappaB signaling pathway. PLoS One. 2012, 7 (6): e39738-10.1371/journal.pone.0039738.CrossRefPubMedPubMedCentral Zhou Y, Fang L, Jiang L, Wen P, Cao H, He W, Dai C, Yang J: Uric acid induces renal inflammation via activating tubular NF-kappaB signaling pathway. PLoS One. 2012, 7 (6): e39738-10.1371/journal.pone.0039738.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Kang DH, Park SK, Lee IK, Johnson RJ: Uric acid-induced C-reactive protein expression: implication on cell proliferation and nitric oxide production of human vascular cells. J Am Soc Nephrol. 2005, 16 (12): 3553-3562. 10.1681/ASN.2005050572.CrossRefPubMed Kang DH, Park SK, Lee IK, Johnson RJ: Uric acid-induced C-reactive protein expression: implication on cell proliferation and nitric oxide production of human vascular cells. J Am Soc Nephrol. 2005, 16 (12): 3553-3562. 10.1681/ASN.2005050572.CrossRefPubMed
27.
Zurück zum Zitat Talaat KM, el-Sheikh AR: The effect of mild hyperuricemia on urinary transforming growth factor beta and the progression of chronic kidney disease. Am J Nephrol. 2007, 27 (5): 435-440. 10.1159/000105142.CrossRefPubMed Talaat KM, el-Sheikh AR: The effect of mild hyperuricemia on urinary transforming growth factor beta and the progression of chronic kidney disease. Am J Nephrol. 2007, 27 (5): 435-440. 10.1159/000105142.CrossRefPubMed
28.
Zurück zum Zitat Bobulescu IA, Moe OW: Renal transport of uric acid: evolving concepts and uncertainties. Adv Chronic Kidney Dis. 2012, 19 (6): 358-371. 10.1053/j.ackd.2012.07.009.CrossRefPubMedPubMedCentral Bobulescu IA, Moe OW: Renal transport of uric acid: evolving concepts and uncertainties. Adv Chronic Kidney Dis. 2012, 19 (6): 358-371. 10.1053/j.ackd.2012.07.009.CrossRefPubMedPubMedCentral
Metadaten
Titel
Hyperuricemia and deterioration of renal function in autosomal dominant polycystic kidney disease
verfasst von
Miyeun Han
Hayne Cho Park
Hyunsuk Kim
Hyung Ah Jo
Hyuk Huh
Joon Young Jang
Ah-Young Kang
Seung Hyup Kim
Hae Il Cheong
Duk-Hee Kang
Jaeseok Yang
Kook-Hwan Oh
Young-Hwan Hwang
Curie Ahn
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2014
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/1471-2369-15-63

Weitere Artikel der Ausgabe 1/2014

BMC Nephrology 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.