jurnal ginjal1.pdf

Upload: stdessert-eagle

Post on 03-Apr-2018

212 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/29/2019 jurnal ginjal1.pdf

    1/6

    Uric Acid and Chronic Kidney Disease:New Understanding of an Old Problem

    Duk-Hee Kang, MD, PhD,* and Wei Chen, MD

    Summary: Although an elevation of serum uric acid level is often associated with chronic kidney disease (CKD),

    it remains controversial whether hyperuricemia per se is a true risk factor for the development or aggravation of

    CKD. Recent epidemiologic studies in healthy populations or in subjects with established kidney disease havereported the independent role of uric acid in lowering glomerular filtration rate and increasing the risk for new-onset

    kidney disease. Furthermore, lowering uric acid in patients with established renal disease has been reported to

    stabilize renal function independent of other confounders, suggesting a causative role of elevated uric acid in

    progression of CKD, rather than as an incidental finding related to CKD severity. In this manuscript we will discuss

    the potential role of uric acid in the development and aggravation of CKD based on epidemiologic, clinical and

    experimental studies. Given the worldwide epidemic of CKD, the importance of identifying modifiable risk factors

    of CKD, and the clinical implication of hyperuricemia in CKD, we propose large randomized clinical trials to

    investigate whether uric acid-lowering therapy can slow the progression of CKD.

    Semin Nephrol 31:447-452 2011 Elsevier Inc. All rights reserved.

    Keywords:hyperuricemia, allopurinol, gout, chronic kidney disease

    INTRODUCTION

    Although hyperuricemia and gout have beenknown to be associated with renal dysfunctionsince the late 19th century,1 there has been debate

    over whether uric acid may have a true pathogenic role in

    renal disease. Originally the focus was whether goutmight cause kidney disease via the deposition of crystals

    associated with inflammation, and hence manifest as an

    extra-articular form of gout. Natural history studiesprior to the availability of uric acid-lowering drugs re-

    ported that up to 25% of gouty subjects developed pro-

    teinuria, 50% developed renal insufficiency, and 10% to

    25% developed end-stage renal disease.2,3 Both renalbiopsies and renal tissue at autopsy showed relatively

    nonspecific features consisting of arteriolosclerosis, glo-

    merulosclerosis, and tubulointerstitial fibrosis.3 Interest-

    ingly, many of these biopsies also showed characteristicfocal deposition of monosodium urate crystals in the

    distal collecting duct and the medullary interstitium with

    a secondary inflammatory reaction. This led to this lesionbeing described as chronic uric acid nephropathy (also

    known as chronic urate nephropathyor gout nephrop-

    athy). However, there was a debate of chronic urate

    nephropathy as a true disease entity since focal deposi-tion of uric acid crystals could not be a mechanism to

    explain the diffuse renal injury observed in biopsies of

    gouty patients with CKD.4-6 Urate crystals could also be

    identified in the kidneys of autopsied subjects who didnot have evidence for kidney disease. Hence, gouty ne-

    phropathy was viewed as a non-entity7, and since then

    most nephrologists do not measure uric acid or considerit as a risk factor in the management of CKD.

    FACTORS THAT

    MODULATESERUMURIC ACID LEVELS

    Uric acid is a weak acid trioxypurine (M.W. 168) that is

    composed of a pyrimidine and imidazole substructurewith oxygen molecules, which is produced primarily in

    the liver, muscle, and intestine.8 The immediate precursor

    of uric acid is xanthine, which is degraded into uric acidby xanthine oxidoreductase. Both exogenous (present in

    fatty meat, organ meats, and seafood) and endogenous

    purines are major sources of xanthine and uric acid inhumans. Fructose, such as from added sugars and fruits,

    is another major source of uric acid. Fructose is unique

    among sugars in that its phosphorylation by fructokinase

    results in a transient reduction in ATP levels in the cell.In turn, the AMP generated is acted on by AMP deami-

    nase to form IMP which is then further degraded to uricacid.

    Approximately two thirds of total body urate is pro-

    duced endogenously, while the remaining one third isaccounted for by dietary purines. The primary site of

    excretion of uric acid is the kidney. The normal urinaryurate excretion in the range of 250 to 750 mg per day,

    approximately 70% of the daily urate production.9 The

    classic paradigm of uric acid excretion consists of a

    four-step model with glomerular filtration, reabsorption,

    *Division of Nephrology, Department of Internal Medicine, Ewha

    Womans University School of Medicine, Ewha Medical Research

    Center, Seoul, Korea.

    Division of Nephrology, University of Colorado, CO.

    This work was supported by a National Research Foundation Grant

    funded by the Korean government (MEST) (2010-0019866).

    Address correspondence to Duk-Hee Kang, MD, PhD, Division of

    Nephrology, Ewha University School of Medicine, 911 Mok-dong

    Yangchun-ku, Seoul 158-710, Korea; Tel 82-2-2650-2870; Fax 82-

    2-2655-2076; E-mail: [email protected]/ - see front matter

    2011 Elsevier Inc. All rights reserved.

    doi:10.1016/j.semnephrol.2011.08.009

    Seminars in Nephrology, Vol 31, No 5, September 2011, pp 447-452 447

    mailto:[email protected]:[email protected]:[email protected]
  • 7/29/2019 jurnal ginjal1.pdf

    2/6

    secretion, and postsecretory reabsorption; the latter threeprocesses occur in the proximal convoluted tubule.10

    More recently emphasis has focused on the role of spe-cific transporters, such as URAT1, SLC2A9, and oth-ers.11,12 Although urate (the form of uric acid at blood pHof 7.4) is freely filtered in the glomerulus, the fractionalurate excretion is only 8% to 10% due to reabsorption in

    proximal tubules in the normal adult. Some adaptationoccurs with renal disease, in which the fractional excre-tion of urate will increase to the 10% to 20%. Theremainder of uric acid excretion occurs through the gut,where uric acid is degraded by uricolytic bacteria. Thegastrointestinal tract may eliminate up to one-third of thedaily uric acid load in the setting of CKD.

    Overall, serum uric acid level is determined by the

    balance between generation and excretion of uric acid.Obesity, insulin resistance, hypertension and use of di-uretics are several conditions associated with an increasein urate reabsorption in renal tubules and hyperurice-

    mia.13

    EPIDEMIOLOGICSTUDIES

    Recently, Bellomo et al demonstrated the associationbetween uric acid and change in estimated glomerularfiltration rate (GFR) in a prospective cohort of 900healthy normotensive adult blood donors.14 Higher uric

    acid levels were associated with subsequent worsening ofkidney function, and this association remained significantafter adjustment for theorized confounders such as bodymass index (BMI), blood pressure and urine albumin-creatinine ratio.14 A recent study in 21,475 healthy par-

    ticipants who were followed up prospectively for a me-dian of 7 years also revealed that increased uric acid levelindependently increased the risk for new-onset kidney

    disease.15 In addition, the Atherosclerosis Risks in Com-

    munities and the Cardiovascular Health Study collecteddata from 13,338 participants with intact kidney functionand demonstrated that increased serum uric acid level isa modest, independent risk factor for incidental kidneydisease in the general population.16 A few large epide-miologic studies performed in Asian countries, Austriaand the United States have also shown that uric acid levelwas a major predictor for the development of incident

    kidney disease.17-22

    Studies of subjects with type 1 dia-betes have also found that an elevated uric acid canpredict the development of either overt diabetic nephrop-athy23 or the development of micro- and macroalbumin-uria.24

    The role of uric acid in predicting progression of renaldisease in subjects with established CKD is more con-troversial. For example, some studies have found anelevated uric acid to be an independent risk factor forprogression of kidney disease in kidney transplant pa-

    tients whereas others have not.25-27 Neither the Modifi-cation of Diet in Renal Disease Study28 nor the Mild toModerate Kidney Disease Study29 could identify uricacid as an independent risk factor. In contrast, a recent

    study in middle-aged and old Taiwanese found that ele-

    vated uric acid level increased the risk of renal disease

    only in stage 3 CKD but not with stage 4 or 5 CKD. 30

    These studies suggest that once CKD is advanced that the

    progression of renal disease may be driven by so many

    additional factors that the role of uric acid is not signif-

    icant.

    CLINICAL INTERVENTIONALSTUDIES

    Studies in Chronic Kidney DiseaseThere have been limited number of studies to examine the

    effect of uric acid-lowering in the development or progres-

    sion of CKD. Kanbay et al. reported that treatment of

    asymptomatic hyperuricemia improved renal function.31

    Likewise, Siu et al. reported that the treatment of asymp-

    tomatic hyperuricemia delayed disease progression with a

    lesser increase in blood pressure with 12-month-treatment

    of allopurinol in patient with CKD.32 More recently, Goi-

    coechea et al performed a randomized, prospective study in113 patients with estimated GFR (eGFR) 60 ml/min and

    demonstrated that allopurinol (100 mg/day) is able to slow

    the progression of renal disease after a mean time of 23.4

    7.8 months.33 No changes in blood pressure or in albumin-

    uria induced by allopurinol have been observed. Interest-

    ingly, allopurinol treatment also reduces cardiovascular and

    hospitalization risk in these subjects.

    Interestingly, there is some evidence that the effect of

    allopurinol may mimic the effects of agents that block the

    renin-angiotensin system (RAS). For example, Talaat

    performed an interesting study in which he withdrew

    allopurinol from subjects with CKD, and found that thiswas associated with worsening hypertension, proteinuria

    and loss of eGFR only in those subjects not taking ACE

    inhibitors or other agents that block the RAS.34 Likewise,

    a small clinical trial of allopurinol in Chinese subjects

    with early IgA nephropathy who were not receiving ACE

    inhibitors demonstrated that allopurinol tended to reduce

    GFR acutely, but then was followed by stabilization of

    the slope in GFR. This finding is consistent with exper-

    imental studies suggesting that lowering uric acid may

    lower glomerular pressure via angiotensin II-dependent

    mechanisms (W Chen, unpublished).

    While these studies suggest a potential benefit of low-ering uric acid in subjects with CKD, it is important to

    realize these are small clinical studies and that major

    clinical trials need to be performed prior to routinely

    lowering uric acid in subjects with CKD. This is partic-

    ularly true since allopurinol can induce a hypersensitivity

    syndrome that can be fatal.35 Second, the above studies

    do not separate whether the benefit of lowering uric acid

    with allopurinol is due to the reduction in uric acid levels

    per se or due to other effects of allopurinol. For example,

    allopurinol also blocks the production of oxidants that are

    generated during the conversion of xanthine to uric acid

    by xanthine oxidase. Some studies, especially in the

    cardiovascular literature, suggest the latter xanthine oxi-

    448 D.-H. Kang and W. Chen

  • 7/29/2019 jurnal ginjal1.pdf

    3/6

    dase-induced oxidants as being key in driving the vascu-lar effects associated with uric acid.36

    Studies on VascularFunction and HypertensionVascular and endothelial function are known to have amajor role in driving CKD.37,38 In this regard, an elevatedserum uric acid is strongly associated with endothelialdysfunction.39-41 Zoccali et al demonstrated an inverserelationship between uric acid and acetylcholine-stimu-

    lated vasodilatation in patients with untreated essentialhypertension, even after adjusting for differences in tra-ditional cardiovascular risk factors.41 Endothelial func-tion assessed by flow-mediated vasodilation (FMD) ofbrachial artery or acetylcholine-induced coronary bloodflow was inversely correlated with serum uric acid lev-els.39-41 Furthermore, allopurinol treatment has been re-ported to improve peripheral or cerebrovascular endothe-lial function in patients with chronic heart failure,42,43

    recent ischemic stroke,44 type 2 diabetes45, metabolicsyndrome46 and even subjects with asymptomatic hyper-uricemia.47 Importantly, George et al demonstrated asteep dose-response relationship between allopurinol andits effect on endothelial function in chronic heart failure;however, the uricosuric agent probenecid had no effecton endothelial function despite a comparable reduction inserum uric acid levels.48 One possible explanation is thatsubjects with heart failure have high levels of xanthine

    oxidase in their blood vessels, and hence a xanthineoxidase inhibitor may be more effective at lowering uricacid levels inside the endothelial cell as compared to a

    uricosuric agent such as probenecid. It is also possiblethat the benefit is due to the inhibition of xanthine oxi-dase associated oxidants as opposed to lowering uricacid.

    Hypertension is also a well-established risk factor for

    CKD.49 Hyperuricemia is known to be associated with an

    elevation of blood pressure despite a continuing contro-versy regarding its causative role.50 A recent study sug-gests uric acid may have a causative role in adolescentswith essential hypertension. In particular, a randomizedcontrol trial found that allopurinol treatment could reduceblood pressure in adolescents with newly diagnosed hy-pertension, which resulted in normal blood pressure in

    66% of adolescents with essential hypertension versus3% of controls.50 Thus, there is emerging evidence that

    lowering uric acid with allopurinol may have a variety ofbenefits, including on endothelial function, blood pres-sure, and renal function. However, to date all studieshave been limited and should be viewed as pilot innature.

    EXPERIMENTAL STUDIES

    Establishment of an animal model of hyperuricemia us-ing uricase inhibitors have deepened our understandingregarding uric acid-related renal disease and its mecha-nisms. Hyperuricemic rats showed preglomerular arterial

    disease, renal inflammation and hypertension via an ac-

    tivation of the RAS and COX-2 systems.51,52 Uric acid is

    also a mitogen for vascular smooth muscle cells whereas

    it inhibits a proliferation of vascular endothelial cells. Rat

    aortic vascular smooth muscle cells showed de novo

    expression of COX-2 mRNA after incubation with uric

    acid.52 Incubation of the vascular smooth muscle cells

    with either a COX-2 inhibitor or with a TX-A2 receptor

    inhibitor prevented the proliferative response to uric acid.

    COX-2 was also shown to be expressed de novo in the

    preglomerular vessels of animal model of CKD with

    hyperuricemia, and its expression correlated both with

    the uric acid levels and with the degree of smooth muscle

    cell proliferation. These findings suggest a critical role

    for uric acid-mediated COX-2 generated thromboxane in

    vascular smooth muscle cell proliferation in an animal

    model of CKD. It is also likely that angiotensin II con-

    tributes to uric acid-induced vasculopathy. Preglomerular

    vasculopathy in rats with oxonic acid-induced hyperuri-

    cemia can be largely prevented by blocking the RAS. 51

    Consistent with these in-vivo findings, uric acid mediated

    effects on vascular smooth muscle and endothelial cell

    can be partially inhibited by blocking the angiotensin II

    type 1 receptor.51 Therefore, both angiotensin II and

    COX-2 are involved in the vascular proliferation and

    inflammation observed in in-vitro and in-vivo animal

    studies.

    Once thickening of the afferent arterioles and macro-

    phage infiltration in vessel wall was induced, preglo-

    merular vasculopathy may potentiate renal injury by

    causing ischemia to the postglomerular circulation. The

    reduction in lumen diameter could also provide a stimu-lus for the increase in renin expression we observed, and

    might also contribute to the development of the marked

    hypertension in these rats.51,53,54 Furthermore, there is

    evidence that the arteriolopathy also leads to ineffective

    autoregulation and increased transmission of systemic

    pressures to the glomerulus,55 which can also potentiate

    renal damage.

    Uric acid also induced the proinflammatory cytokine,

    monocyte chemoattractant protein-1 (MCP-1) and de

    novo expression of C-reactive protein (CRP) in vascular

    smooth muscle and endothelial cells, which was further

    shown to be due to direct entry of uric acid into cells withactivation of mitogen activated protein kinase (MAPK)

    and nuclear transcription factor (NF-kB).56,57 In addition,

    uric acid can become pro-oxidative under certain circum-

    stances.58 The prooxidative effects are primarily medi-

    ated by intracellular uric acid, and can be shown in

    endothelial cells, vascular smooth muscle cells, renal

    tubular cells, adipocytes, and cardiac fibroblasts.59-63 On

    the other hand, in the extracellular environment, uric acid

    may function as an antioxidant, particularly as a scaven-

    ger of peroxynitrite.64 The different effects of uric acid

    may depend on the host environment.

    Recent data also suggested the possibility of direct

    effect of uric acid on renal tubular cells. Uric acid per se

    Uric acid and CKD 449

  • 7/29/2019 jurnal ginjal1.pdf

    4/6

    may induce phenotypic transition of cultured renal tubu-

    lar cells, as epithelial-to-mesenchymal transition (EMT)can be demonstrated in the kidneys of hyperuricemic

    rats.65 Given the consideration of EMT as one of the

    earliest phenomena of renal fibrosis, 66 it will be inter-esting to further investigate the mechanism of EMT of

    renal tubular cells as a novel mechanism of uric acid-induced renal disease.

    Taking all into consideration, uric acid may induce

    renal disease via an induction of afferent arteriopathy asa consequence of an altered proliferation and senescence

    of vascular cells, an induction of local oxidative stressand inflammation with an activation of RAS, followed by

    impaired peritubular capillary circulation and renal isch-

    emia. Uric acid-induced phenotypic transition of renaltubular cells also could be important (Figure 1).

    CONCLUSIONS

    There is accumulating epidemiologic, clinical and exper-imental evidence supporting hyperuricemia as a true risk

    factor of CKD rather than an incidental findings relatedto declining glomerular filtration. Nonetheless, there are

    still controversies regarding the causative role of uricacid in the development or aggravation of CKD with

    conflicting results in different studies. There is no con-sensus yet whether we need to treat asymptomatic hy-

    peruricemia in CKD patients or whether a level of serumuric acid should be targeted for renoprotection with uric

    acid-lowering therapy. Given the worldwide epidemic ofCKD population, it is critical to identify modifiable,

    novel risk factors of CKD and treat them adequately.

    Uric acid may be one of the ignored risk factors of CKD.We recommend large randomized clinical trials to eval-

    uate the effect of uric acid reduction on progression of

    renal function, cardiovascular disease and mortality in

    CKD patients.

    REFERENCES

    1. Davis NS: The cardio-vascular and renal relations and manifes-

    tations of gout. JAMA 29: 261-262, 1897.

    2. Brchner-Mortensen K: 100 gouty patients. Acta Medica Scand

    106:81-107, 1941.

    3. Talbott JH, Terplan KL: The Kidney in Gout. Medicine. 39:405-

    467, 1960.4. Linnane JW, Burry AF, Emmerson BT: Urate deposits in the renal

    medulla. Prevalence and association. Nephron 29:216-222, 1981.

    5. Yu TF, Berger L, Dorph DJ, et al: Renal function in gout. V.

    Factors influencing the renal hemodynamics. Am J Med 67:766-

    771, 1979.

    6. Berger L, Y T: Renal Function in Gout: IV. An Analysis of 524

    Gouty Subjects Including Long-Term Follow-Up Studies. Am J

    Med 59:605-613, 1975.

    7. Beck LH: Requiem for gouty nephropathy. Kidney Int 30(2):

    280-7, 1986.

    8. Hediger MA, Johnson RJ, Miyazaki H, et al: Molecular physiol-

    ogy of urate transport. Physiology 20: 125-133, 2005.

    9. Maesaka JK, Fishbane S: Regulation of renal urate excretion: A

    critical review. Am J Kidney Dis 32:917-933, 1998.

    10. Roch-Ramel F, Werner D, Guisan B: Urate transport in brush-

    border membrane of human kidney. Am J Physiol.Renal Physiol

    266:F797-F805, 1994.

    11. Enomoto A, Kimura H, Chairoungdua A, et al: Molecular iden-

    tification of a renal urate anion exchanger that regulates blood

    urate levels. Nature 417(6887):447-52, 2002.

    12. Anzai N, Ichida K, Jutabha P, et al: Plasma urate level is

    directly regulated by a voltage-driven urate efflux transporter

    URATv1 (SLC2A9) in humans. J Biol Chem 283(40):26834-8,

    2008.

    13. Wyngaarden, JB, Kelley, WN. Gout and hyperuricemia. Grune

    and Stratton, New York, 1976.

    14. Bellomo G, Venanzi S, Verdura C, et al: Association of uric acid

    with change in kidney function in healthy normotensive individ-

    uals. Am J Kidney Dis 56:264-272, 2010.15. Obermayr RP, Temml C, Gutjahr G, et al: Elevated uric acid

    Uric Acid

    ro erat on o

    Inhibition of proliferation of VEC with

    cell senescence

    Activation of local COX-2 & RAS

    Induction of inflammatory reaction

    Decrease in NO production

    Induction of oxidative stress

    Preglomerular ArteriopathyEMT of Renal

    Tubular Cells

    Production of ECMImpairment of peritubular

    circulation

    Renal ischemia

    Ineffective

    autoregulation of

    glomerular pressure

    Glomerular

    HypertensionActivation of RAS

    Renal fibrosis

    Tubulointerstitial

    Inflammation

    Figure 1. Summary of potential mechanisms of uric acid-induced kidney disease proposed by experimental data from hyperuricemic rats.

    VSMC, vascular smooth muscle cells, VEC, vascular endothelial cells, COX-2, cyclooxygenase-2, RAS, renin-angiotensin system, NO, nitricoxide, EMT, epithelial-to-mesenchymal transition, ECM, extracellular matrix.

    450 D.-H. Kang and W. Chen

  • 7/29/2019 jurnal ginjal1.pdf

    5/6

    increases the risk for kidney disease. J Am Soc Nephrol 19:

    2407-2413, 2008.

    16. Weiner DE, Tighiouart H, Elsayed EF, et al: Uric acid and

    incident kidney disease in the community. J Am Soc Nephrol

    19:1204-11, 2008.

    17. Lee JE, Kim YG, Choi YH, et al: Serum uric acid is associated

    with microalbuminuria in prehypertension. Hypertension 47: 962-

    967, 2006.

    18. Rosolowski ET, Ficociello LH, Maselli NJ, et al: High-normalserum uric acid is associated with impaired glomerular filtration

    rate in nonproteinuric patients with type 1 diabetes. Clin J Am Soc

    Nephrol 3:706-713, 2008.

    19. Iseki K, Oshiro S, Tozawa M, et al: Significance of hyperuricemia

    on the early detection of renal failure in a cohort of screened

    subjects. Hypertens Res 24:691-697, 2001.

    20. Iseki K, Ikemiya Y, Inoue T, et al: Significance of hyperuricemia

    as a risk factor for developing ESRD in a screened cohort. Am J

    Kidney Dis 44: 642-650, 2004.

    21. Domrongkitchaiporn S, Sritara P, Kitiyakara C, et al: Risk factors

    for developing decreased kidney function in a Southeast Asian

    population: a 12-year cohort study. J Am Soc Nephrol 16: 791-

    799, 2005.

    22. Hsu CY, Iribarren C, McCulloch CE, et al: Risk factors for

    end-stage renal disease. Arch Intern Med 169: 342-350, 2009.

    23. Hovind P, Rossing P, Tarnow L, et al: Serum uric acid as a

    predictor for development of diabetic nephropathy in type 1

    diabetes: an inception cohort study. Diabetes 58(7):1668-71,

    2009.

    24. Jalal DI, Rivard CJ, Johnson RJ, et al: Serum uric acid levels

    predict the development of albuminuria over 6 years in patients

    with type 1 diabetes: findings from the Coronary Artery Calcifi-

    cation in Type 1 Diabetes study. Nephrol Dial Transplant 25(6):

    1865-9, 2010.

    25. Gores PF, Fryd DS, Sutherland DE, et al: Hyperuricemia after

    renal transplantation. Am J Surg 156: 397-400, 1988.

    26. Chonchol M, Shlipak MG, Katz R, et al: Relationship of uric acid

    with progression of kidney disease. Am J Kidney Dis 50: 239-247,

    2007.27. Madero M, Sarnak MJ., Wang X, et al: Uric acid and long-term

    outcomes in CKD. Am J Kidney Dis 53: 796-803, 2009.

    28. Hunsicker LG, Adler S, Caggiula A, et al: Predictors of the

    progression of renal disease in the Modification of Diet in Renal

    Disease Study. Kidney Int 51:1908-19, 1997.

    29. Sturm G, Kollerits B, Neyer U, et al: MMKD Study Group. Uric

    acid as a risk factor for progression of non-diabetic chronic

    kidney disease? The Mild to Moderate Kidney Disease (MMKD)

    Study. Exp Gerontol 43:347-52, 2008.

    30. 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. 339(6):509-15, 2010.

    31. Kanbay M, Ozkara A, Selcoki Y, et al: Effect of treatment of

    hiperuricemia with allopurinl on blood pressure, creatinine clear-

    ance, and proteinuria in patients with normal renal function. IntUrol Nephrol 39: 1227-1233, 2007.

    32. Siu YP, Leung KT, Tong MK, et al: Use of allopurinol in slowing

    the progression of renal disease through its ability to lower serum

    uric acid level. Am J Kidney Dis 47. (1): 51-59, 2006.

    33. Goicoechea M, de Vinuesa SG, Verdalles U, et al: Effect of

    allopurinol in chronic kidney disease progression and cardiovas-

    cular risk. Clin J Am Soc Nephrol. 5(8):1388-93, 2010.

    34. Talaat KM, El-Sheikh AR: The Effect of Mild Hyperuricemia

    on Urinary Transforming Growth Factor Beta and the Progres-

    sion of Chronic Kidney Disease. Am J Nephrol 27(5):435-440,

    2007.

    35. Anderson BE, Adams DR: Allopurinol hypersensitivity syn-

    drome. J Drugs Dermatol 1(1):60-2, 2002.

    36. George J, Carr E, Davies J, et al: High-dose allopurinol improvesendothelial function by profoundly reducing vascular oxidative

    stress and not by lowering uric acid. Circulation 114(23):2508-16,

    2006.

    37. Baylis C: Nitric oxide deficiency in chronic kidney disease. Am J

    Physiol Renal Physiol 294(1):F1-9, 2008.

    38. Kang DH, Kanellis J, Hugo C, et al: Role of endothelium in

    progressive renal disease. J Am Soc Nephrol 13(3):806-816,

    2002.

    39. Ho WJ, Tsai WP, Yu KH, et al: Association between endothelial

    dysfunction and hyperuricaemia. Rheumatology (Oxford).49(10):1929-34, 2010.

    40. Kuwahata S, Hamasaki S, Ishida S, et al: Effect of uric acid on

    coronary microvascular endothelial function in women: associa-

    tion with eGFR and ADMA. J Atheroscler Thromb. 17(3):259-69,

    2010.

    41. Zoccali C, Maio R, Mallamaci F, et al: Uric acid and endothelial

    dysfunction in essential hypertension. J Am Soc Nephrol 17:

    1466-71, 2006.

    42. Doehner W, Schoene N, Rauchhaus M, et al: Effects of xanthine

    oxidase inhibition with allopurinol on endothelial function and

    peripheral blood flow in hyperuricemic patients with chronic heart

    failure: results from 2 placebo-controlled studies. Circulation.

    105:2619-24, 2002.

    43. Farquharson CA, Butler R, Hill A, et al: Allopurinol improves

    endothelial dysfunction in chronic heart failure. Circulation.106(2):221-6, 2002.

    44. Khan F, George J, Wong K, et al: Allopurinol treatment reduces

    arterial wave reflection in stroke survivors. Cardiovasc Ther 26:

    247-52, 2008.

    45. Dawson J, Quinn TJ, Harrow C, et al: Allopurinol and nitric oxide

    activity in the cerebral circulation of those with diabetes. Diabetes

    Care 32: 135-7, 2009.

    46. Yiginer O, Ozcelik F, Inanc T, et al: Allopurinol improves endo-

    thelial function and reduces oxidant-inflammatory enzyme of

    myeloperoxidase in metabolic syndrome. Clin Res Cardiol. 97(5):

    334-40, 2008.

    47. Mercuro G, Vitale C, Cerquetani E, et al: Effect of hyperuricemia

    upon endothelial function in patients at increased cardiovascular

    risk. Am J Cardiol 94(7):932-5, 2004.

    48. George J, Struthers AD: The role of urate and xanthine oxidaseinhibitors in cardiovascular disease. Cardiovasc Ther. 26(1):59-

    64, 2008.

    49. Fogo AB: Hypertensive risk factors in kidney disease in African

    Americans. Kidney Int Suppl 83:S17-21, 2003.

    50. Feig DI, Soletsky B, Johnson RJ: Effect of allopurinol on blood

    pressure of adolescents with newly diagnosed essential hyperten-

    sion: a randomized trial. JAMA 300: 924-932, 2008.

    51. Mazzali M, Kanellis J, Han L, et al: Hyperuricemia induces a

    primary arteriolopathy in rats by a blood pressure-independent

    mechanism. Am J Physiol Renal Physiol 282:F991-F997, 2002.

    52. Kang DH, Nakagawa T, Feng L, et al: A Role for Uric Acid in the

    Progression of Renal Disease. J Am Soc Nephrol 13: 2888-2897,

    2002.

    53. Mazzali M, Hughes J, Kim YG, et al: Elevated uric acid increases

    blood pressure in the rat by a novel crystal-independent mecha-

    nism. Hypertension 38:1101-1106, 2001.

    54. Mazzali M, Jefferson JA, Ni Z, et al: Microvascular and tubulo-

    interstitial injury associated with chronic hypoxia-induced hyper-

    tension. Kidney Int 63:2088-93, 2003.

    55. Sanchez-Lozada LG, Tapia E, Avila-Casado C, et al: Mild hy-

    peruricemia induces glomerular hypertension in normal rats. Am J

    Physiol Renal Physiol. 283(5):F1105-10, 2002.

    56. Kanellis J, Watanabe S, Li JH, et al: Uric acid stimulates mono-

    cyte chemoattractant protein-1 production in vascular smooth

    muscle cells via mitogen-activated protein kinase and cyclooxy-

    genase-2. Hypertension. 41(6):1287-93, 2003.

    57. Kang DH, Park SK, Lee IK et al: Uric acid-induced C-reactive

    protein expression: implication on cell proliferation and nitric

    oxide production of human vascular cells. J Am Soc Nephrol.6(12):3553-62, 2005.

    Uric acid and CKD 451

  • 7/29/2019 jurnal ginjal1.pdf

    6/6

    58. Bagnati M, Perugini C, Cau C, et al: When and why a water-

    soluble antioxidant becomes pro-oxidant during copper-induced

    low-density lipoprotein oxidation: a study using uric acid.

    J. Biochem. 340:143-152, 1999.

    59. Yu MA, Sanchez-Lozada LG, Johnson RJ, Kang DH: Oxidative

    stress with an activation of the renin-angiotensin system in human

    vascular endothelial cells as a novel mechanism of uric acid-

    induced endothelial dysfunction. J Hypertens 28:1234-42, 2010.

    60. Corry DB, Eslami P, Yamamoto K, et al: Uric acid stimulatesvascular smooth muscle cell proliferation and oxidative stress via

    the vascular renin-angiotensin system. J Hypertens 26(2):269-75,

    2008.

    61. Sautin YY, Nakagawa T, Zharikov S, Johnson RJ: Adverse ef-

    fects of the classic antioxidant uric acid in adipocytes: NADPH

    oxidase-mediated oxidative/nitrosative stress. Am J Physiol Cell

    Physiol 293(2):C584-96, 2007.

    62. Cirillo P, Gersch MS, Mu W, et al: Ketohexokinase-dependent

    metabolism of fructose induces proinflammatory mediators in

    proximal tubular cells. J Am Soc Nephrol 20(3):545-53, 2009.

    63. Cheng TH, Lin JW, Chao HH, et al: Uric acid activates extracel-

    lular signal-regulated kinases and thereafter endothelin-1 expres-

    sion in rat cardiac fibroblasts. Int J Cardiol 139:42-49, 2010.

    64. Ames BN, Cathcart R, Schwiers E, et al: Uric acid provides an

    antioxidant defense in humans against oxidant- and radical-

    caused aging and cancer: a hypothesis. Proc Natl Acad Sci U S A.

    78(11):6858-62, 1981.

    65. Yu, M Park, EY KimSJ, et al: Uric acid-induced EMT of renal

    tubular cells as a novel mechanism of kidney disease. J Am Soc

    Nephrol 19:177A, 2008 (abstract).

    66. Zeisberg M, Kalluri R: The role of epithelial-to-mesenchymal

    transition in renal fibrosis. J Mol Med 82:175-81, 2004.

    452 D.-H. Kang and W. Chen