Abstract

HomeCirculationVol. 107, No. 15Uric Acid Predicts Clinical Outcomes in Heart Failure Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBUric Acid Predicts Clinical Outcomes in Heart FailureInsights Regarding the Role of Xanthine Oxidase and Uric Acid in Disease Pathophysiology Joshua M. Hare, MD and Richard J. Johnson, MD Joshua M. HareJoshua M. Hare From the Department of Medicine, Cardiology Division, Johns Hopkins Hospital, Baltimore, Md (J.M.H.); and the Nephrology Division, Baylor College of Medicine, Houston, Tex (R.J.J.). Search for more papers by this author and Richard J. JohnsonRichard J. Johnson From the Department of Medicine, Cardiology Division, Johns Hopkins Hospital, Baltimore, Md (J.M.H.); and the Nephrology Division, Baylor College of Medicine, Houston, Tex (R.J.J.). Search for more papers by this author Originally published22 Apr 2003https://doi.org/10.1161/01.CIR.0000066420.36123.35Circulation. 2003;107:1951–1953In the current issue of Circulation, Anker and colleagues1 report that elevated levels of uric acid (UA) predict mortality and the need for heart transplantation in patients with congestive heart failure (HF). Serum concentrations of UA added important prognostic information alone and when combined with measures of cardiac function (ejection fraction) and patient functional status (maximal oxygen consumption with exercise) and were independent of renal function, serum sodium, serum urea, diuretic usage, and patient age. Receiver operating curve analysis identified a cutoff of 585 μmol/L (9.8 mg/dL) as the best mortality predictor. This finding is not only potentially of value in patient management but also raises extremely interesting questions regarding the pathophysiological underpinnings of this finding.See p 1991A consideration of the mechanism of UA production and metabolism offers insight into the relationship between UA levels and HF outcomes. Indeed, accumulating data support the idea that UA, in addition to being a potentially valuable prognostic marker, possesses specific toxic or other properties that could contribute to HF pathophysiology. Moreover, UA levels may reflect xanthine oxidase (XO) pathway activity, which has the potential to contribute to the progression of left ventricular dysfunction by interfering with myocardial energetics2 and myofilament calcium sensitivity.3Potential Mechanisms for Increased Uric Acid in Heart FailureUA is a metabolic byproduct of purine metabolism (Figure). Serum UA may increase in the failing circulation because of increased generation, decreased excretion, or a combination of the 2 factors. There are several possible contributors to increased UA production in HF, including increased abundance and activity of XO,4 increased conversion of xanthine dehydrogenase (XDH) to XO,5 or increased XO substrate resulting from enhanced ATP breakdown to adenosine and hypoxanthine. As UA is excreted primarily by the kidney, decreased renal perfusion could lead to increased UA levels. To the extent that HF leads to tissue ischemia (in advanced HF) and a rise in serum lactate, renal UA excretion can be further impaired as lactate competes with urate via an organic anion exchanger in the proximal tubule.6Download figureDownload PowerPointPurine metabolism. UA is produced in the terminal step of purine metabolism catalyzed by XO. Hominoid urate oxidase has undergone inactivation because of gene mutation, and thus UA loss is primarily via renal and gastrointestinal excretion.7 XO pathway activity also results in the production of superoxide. XO may be inhibited by allopurinol or oxypurinol (allopurinol is a prodrug converted to oxypurinol by XO).Pathophysiological Role of the Xanthine Oxidase Pathway in Heart FailureThere is increasing evidence that strongly supports a direct pathophysiological role for the metabolic pathway leading to UA production in the failing circulation.2,7 In this regard, the 2 terminal steps in urate production are catalyzed by XO, which also produces a molecule of superoxide for each reaction5 (Figure). XO is the product of the xanthine oxidoreductase gene that encodes XDH, an 150 Kda protein, which functions as a homodimer. XDH is converted to XO by proteolytic cleavage or sulfhydryl modification.5The elevation in serum UA may reflect increased XO pathway activity and in turn the generation of superoxide and resultant oxidative stress via the XO system.8 XO is upregulated within the heart in both experimental4,9 and human2 heart failure. Much had previously been made of the difficulty in identifying XO within the hearts of certain mammalian species, including humans;10 nevertheless, it is clear that XO, which is produced in highest abundance in the liver and gut, may circulate in the blood and adhere to endothelium in distant sites.11 Moreover, XO is expressed in cardiac myocytes, as shown by immunohistochemistry and may participate in intracrine signaling.12From a functional standpoint, XO activity participates in both mechanoenergetic uncoupling and vascular dysfunction in the failing circulation. Mechanoenergetic uncoupling is the process whereby cardiac energy consumption remains the same or increases while cardiac work falls dramatically, and is increasingly being perceived as a potential key lesion in the failing heart. Inhibition of XO with allopurinol restores depressed myocardial energetics toward normal, and this effect can be mimicked by the antioxidant ascorbate.12 Furthermore, several recent studies have demonstrated that XO inhibition improves endothelial dysfunction in patients with congestive heart failure in association with reduction in circulating markers of oxidative stress,13,14 thereby providing evidence that XO inhibition reduces oxidant generation.Pathophysiological Role of Uric Acid in Heart FailureBeyond XO activity, recent experimental studies suggest that UA itself may have a role in cardiovascular and renal pathophysiology. This might seem surprising, as UA can function as an antioxidant, both by itself and by promoting superoxide dismutase activity,15,16 and might therefore be considered potentially protective. However, UA potently stimulates vascular smooth muscle cell proliferation in vitro, an effect mediated by stimulation of mitogen-activated protein kinases, cyclooxygenase-2, and platelet-derived growth factor.7,17,18 Furthermore, rats with mild experimentally-induced hyperuricemia develop intrarenal vascular disease with increased renin expression, systemic and glomerular hypertension, and renal injury in the absence of intrarenal crystal deposition.19,20 These hemodynamic and structural changes can be prevented if UA elevation is prevented by allopurinol.19,20Interaction of Xanthine Oxidase and Uric Acid With Nitric Oxide PathwaysBoth XO activity and UA may also affect cardiac and renal nitric oxide signaling,12,19 which exerts key cardiac and vascular effects. The impact of XO inhibition to restore depressed myocardial energetics requires intact NO pathway activity.12 UA may also impair NO production directly, as suggested by the finding that UA infusion into forearm veins of humans attenuates acetylcholine-stimulated vasodilation.21 Likewise, the hypertension associated with hyperuricemia in rats is associated with reduced expression of macula densa neuronal nitric oxide synthase (NOS) and can be partially reversed by the NOS substrate l-arginine.19 This finding has interesting implications for cardiac function, as neuronal NOS plays a key role in modulating cardiac excitation-contraction coupling by facilitating sarcoplasmic reticulum calcium release.22Clinical Utility of Uric Acid MeasurementsFrom a clinical perspective, the current study raises the issue of whether serum UA levels should be routinely measured in HF patients. Indeed this is likely to be a controversial issue, and one which will require evaluation in the context of measurement of brain natriuretic peptide (BNP), a serum marker that also possesses prognostic and diagnostic value in HF patients.23 Much in the same way as BNP has been evaluated, it will be of great value to assess whether UA levels change in response to HF therapy in a manner that predicts clinical outcome.Whether or not UA levels are ready for clinical use, the observation that UA levels possess prognostic information adds an extremely intriguing finding to mounting evidence that XO and UA play pathophysiological roles in HF and its precursor, hypertension. Indeed, the amassing data have led to the planning of a clinical trial entitled A Phase II-III Prospective, Randomized, Double-Blind, Placebo-Controlled Efficacy and Safety Study of Oxypurinol Added to Standard Therapy in Patients with NYHA Class III-IV Congestive Heart Failure (OPT-CHF), initiated in 2003, which will test clinical outcomes using a composite endpoint comprising measures of heart failure morbidity, exercise capacity, and mortality. The findings of Anker and colleagues,1 therefore, not only bring to light a potentially new diagnostic test but also provide a novel line of evidence that the XO pathway and/or UA itself may be of pathophysiological importance in heart failure progression.The opinions expressed in this editorial are not necessarily those of the editors or of the American Heart Association.Dr Hare serves as a consultant to Cardiome Pharma Corp. Dr Johnson is listed as an inventor of a patent application placed by Merck and the University of Washington that proposes the use of agents to lower uric acid as a means to prevent or treat hypertension.This work was supported by National Institutes of Health grants RO1 HL-65455 (to Dr Hare) and RO1 HL68607 (to Dr Johnson). Dr Hare is the recipient of a Paul Beeson Physician Faculty Scholars in Aging Research Award.FootnotesCorrespondence to Joshua M. Hare, MD, The Johns Hopkins Hospital School of Medicine, Cardiology Division, 600 N Wolfe St, Carnegie 568, Baltimore, MD 21287 (e-mail [email protected]); or Richard J. Johnson, MD, Division of Nephrology, Baylor College of Medicine, 6550 Fannin, SM 1273, Houston, TX 77030 (e-mail [email protected]). References 1 Anker S, Doehner W, Rauchaus M, et al. Uric acid and survival in chronic heart failure: validation and application in metabolic, functional, and hemodynamic staging. Circulation. 2003; 107: 1991–1997.LinkGoogle Scholar2 Cappola TP, Kass DA, Nelson GS, et al. Allopurinol improves myocardial efficiency in patients with idiopathic dilated cardiomyopathy. Circulation. 2001; 104: 2407–2411.CrossrefMedlineGoogle Scholar3 Pérez NG, Gao WD, Marbán E. Novel myofilament calcium-sensitizing property of xanthine oxidase inhibitors. Circ Res. 1998; 83: 423–430.CrossrefMedlineGoogle Scholar4 Ekelund UEG, Harrison RW, Shokek O, et al. Intravenous allopurinol decreases myocardial oxygen consumption and increases mechanical efficiency in dogs with pacing-induced heart failure. Circ Res. 1999; 85: 437–445.CrossrefMedlineGoogle Scholar5 Saugstad OD. Role of xanthine oxidase and its inhibitor in hypoxia: reoxygenation injury. Pediatrics. 1996; 98: 103–107.CrossrefMedlineGoogle Scholar6 Roch-Ramel F, Guisan B, Diezi J. Effects of uricosuric and antiuricosuric agents on urate transport in human brush-border membrane vesicles. J Pharmacol Exp Ther. 1997; 280: 839–845.MedlineGoogle Scholar7 Watanabe S, Kang DH, Feng L, et al. Uric acid, hominoid evolution, and the pathogenesis of salt-sensitivity. Hypertension. 2002; 40: 355–360.LinkGoogle Scholar8 McCord JM. Oxygen-derived free radicals in postischemic tissue injury. N Eng J Med. 1985; 312: 159–163.CrossrefMedlineGoogle Scholar9 de Jong JW, Schoemaker RG, de Jonge R, et al. Enhanced expression and activity of xanthine oxidoreductase in the failing heart. J Mol Cell Cardiol. 2000; 32: 2083–2089.CrossrefMedlineGoogle Scholar10 de Jong JW. Xanthine oxidoreductase activity in perfused hearts of various species, including humans. Circ Res. 1990; 67: 770–773.CrossrefMedlineGoogle Scholar11 White CR, Darley-Usmar V, Berrington WR, et al. Circulating plasma xanthine oxidase contributes to vascular dysfunction in hypercholesterolemic rabbits. Proc Natl Acad Sci U S A. 1996; 93: 8745–8749.CrossrefMedlineGoogle Scholar12 Saavedra WF, Paolocci N, St John ME, et al. Imbalance between xanthine oxidase and nitric oxide synthase signaling pathways underlies mechanoenergetic uncoupling in the failing heart. Circ Res. 2002; 90: 297–304.CrossrefMedlineGoogle Scholar13 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. 2002; 105: 2619–2624.LinkGoogle Scholar14 Farquharson CA, Butler R, Hill A, et al. Allopurinol improves endothelial dysfunction in chronic heart failure. Circulation. 2002; 106: 221–226.LinkGoogle Scholar15 Waring WS, Webb DJ, Maxwell SR. Systemic uric acid administration increases serum antioxidant capacity in healthy volunteers. J Cardiovasc Pharmacol. 2001; 38: 365–371.CrossrefMedlineGoogle Scholar16 Hink HU, Santanam N, Dikalov S, et al. Peroxidase properties of extracellular superoxide dismutase: role of uric acid in modulating in vivo activity. Arterioscler Thromb Vasc Biol. 2002; 22: 1402–1408.LinkGoogle Scholar17 Rao GN, Corson MA, Berk BC. Uric acid stimulates vascular smooth muscle cell proliferation by increasing platelet-derived growth factor A-chain expression. J Biol Chem. 1991; 266: 8604–8608.CrossrefMedlineGoogle Scholar18 Kang D-H, Nakagawa T, Feng L, et al. A role for uric acid in the progression of renal disease. J Am Soc Nephrol. 2002; 13: 2888–2897.CrossrefMedlineGoogle Scholar19 Mazzali M, Hughes J, Kim YG, et al. Elevated uric acid increases blood pressure in the rat by a novel crystal-independent mechanism. Hypertension. 2001; 38: 1101–1106.CrossrefMedlineGoogle Scholar20 Sanchez-Lozada LG, Tapia E, Avila-Casado C, et al. Mild hyperuricemia induces glomerular hypertension in normal rats. Am J Physiol Renal Physiol. 2002; 283: F1105–F1110.CrossrefMedlineGoogle Scholar21 Waring WS, Webb DJ, Maxwell SRJ. Effect of local hyperuricemia on endothelial function in the human forearm vascular bed. Br J Pharmacol. 2000; 49: 511.Google Scholar22 Barouch LA, Harrison RW, Skaf MW, et al. Nitric oxide regulates the heart by spatial confinement of nitric oxide synthase isoforms. Nature. 2002; 416: 337–340.CrossrefMedlineGoogle Scholar23 Maisel AS, Krishnaswamy P, Nowak RM, et al. Rapid measurement of B-type natriuretic peptide in the emergency diagnosis of heart failure. N Engl J Med. 2002; 347: 161–167.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Yılmaz Öztekin G, Genç A, Çağırcı G and Arslan Ş (2022) Prognostic value of the combination of uric acid and NT-proBNP in patients with chronic heart failure, Hellenic Journal of Cardiology, 10.1016/j.hjc.2022.03.009, 65, (35-41), Online publication date: 1-May-2022. Ahsan F, Talukdar M, Qureshi N, Samreen S and Kukreti S (2022) Study of Hs Troponin I & uric acid in patients of myocardial infarction, International Journal of Clinical Biochemistry and Research, 10.18231/j.ijcbr.2021.060, 8:4, (281-284), Online publication date: 28-Jan-2022. Martins C, Bau A, Silva L and Coelho O (2021)(2021)(2021)(2021) Possible Mechanisms of Action of SGLT2 Inhibitors in Heart Failure, ABC: Heart Failure & Cardiomyopathy, 10.36660/abchf.20210007, 1:1, (33-43), Online publication date: 6-Jul-2021., Online publication date: 6-Jul-2021., ., . Подпалова О, Колядко М, Бабенко А, Тимошкова О, Русских И, Генералов И and Мрочек А (2021) Telomere Length and Hyperuricemia According to the 10-Year Prospective Research, Кардиология в Беларуси, 10.34883/PI.2020.12.6.006:6, (834-844), Online publication date: 12-Jan-2021. Maciejczyk M, Bielas M, Zalewska A, Gerreth K and Tu W (2021) Salivary Biomarkers of Oxidative Stress and Inflammation in Stroke Patients: From Basic Research to Clinical Practice, Oxidative Medicine and Cellular Longevity, 10.1155/2021/5545330, 2021, (1-22), Online publication date: 7-Apr-2021. Huddam B, Alp A, Genek D, Azak A and Karakus V (2021) Increased QT Dispersion and High Risk of Ventricular Arrhythmias is Associated with Hyperuricemia in Individuals with Normal Renal Function, Journal Of Cardiovascular Emergencies, 10.2478/jce-2021-0011, 7:3, (77-83), Online publication date: 1-Sep-2021., Online publication date: 1-Sep-2021. Bupparenoo P, Pakchotanon R, Narongroeknawin P, Asavatanabodee P and Chaiamnuay S (2020) Effect of Curcumin on Serum Urate in Asymptomatic Hyperuricemia: A Randomized Placebo-Controlled Trial, Journal of Dietary Supplements, 10.1080/19390211.2020.1757798, 18:3, (248-260), Online publication date: 4-May-2021. Rotkvić P, Berković M, Bulj N, Rotkvić L and Ćelap I (2020) Sodium-glucose cotransporter 2 inhibitors’ mechanisms of action in heart failure, World Journal of Diabetes, 10.4239/wjd.v11.i7.269, 11:7, (269-279), Online publication date: 15-Jul-2020. Shi Y, Zhang X, Du J, Chen S, Zhang H, Yang L and Zheng Z (2020) Elevated postoperative serum uric acid is associated with major adverse events following coronary artery bypass grafting, Journal of Cardiac Surgery, 10.1111/jocs.14845, 35:10, (2559-2566), Online publication date: 1-Oct-2020. Mineiro M, Patricio E, Peixoto Á, Araujo T, da Silva R, Moretti A, Lima F, Laurindo F and Meotti F (2020) Urate hydroperoxide oxidizes endothelial cell surface protein disulfide isomerase-A1 and impairs adherence, Biochimica et Biophysica Acta (BBA) - General Subjects, 10.1016/j.bbagen.2019.129481, 1864:3, (129481), Online publication date: 1-Mar-2020. Lee S, Oh B and Sung K (2020) Uric acid and cardiometabolic diseases, Clinical Hypertension, 10.1186/s40885-020-00146-y, 26:1, Online publication date: 1-Dec-2020. Kang D and Johnson R (2020) Uric Acid Metabolism and the Kidney Chronic Renal Disease, 10.1016/B978-0-12-815876-0.00043-7, (689-701), . Gonzalez-Martin G, Cano J, Carriazo S, Kanbay M, Perez-Gomez M, Fernandez-Prado R and Ortiz A (2020) The dirty little secret of urate-lowering therapy: useless to stop chronic kidney disease progression and may increase mortality, Clinical Kidney Journal, 10.1093/ckj/sfaa236, 13:6, (936-947), Online publication date: 28-Dec-2021. Lopaschuk G and Verma S (2020) Mechanisms of Cardiovascular Benefits of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors, JACC: Basic to Translational Science, 10.1016/j.jacbts.2020.02.004, 5:6, (632-644), Online publication date: 1-Jun-2020. Grubić Rotkvić P, Cigrovski Berković M, Bulj N and Rotkvić L (2019) Minireview: are SGLT2 inhibitors heart savers in diabetes?, Heart Failure Reviews, 10.1007/s10741-019-09849-3, 25:6, (899-905), Online publication date: 1-Nov-2020. Wijnker P, Sequeira V, Kuster D and Velden J (2019) Hypertrophic Cardiomyopathy: A Vicious Cycle Triggered by Sarcomere Mutations and Secondary Disease Hits, Antioxidants & Redox Signaling, 10.1089/ars.2017.7236, 31:4, (318-358), Online publication date: 1-Aug-2019. Cicero A, Cosentino E, Kuwabara M, Degli Esposti D and Borghi C (2019) Effects of allopurinol and febuxostat on cardiovascular mortality in elderly heart failure patients, Internal and Emergency Medicine, 10.1007/s11739-019-02070-y, 14:6, (949-956), Online publication date: 1-Sep-2019. Oki Y, Kawai M, Minai K, Ogawa K, Inoue Y, Morimoto S, Tanaka T, Nagoshi T, Ogawa T and Yoshimura M (2019) High Serum Uric Acid is Highly Associated with a Reduced Left Ventricular Ejection Fraction Rather than Increased Plasma B-type Natriuretic Peptide in Patients with Cardiovascular Diseases, Scientific Reports, 10.1038/s41598-018-37053-0, 9:1, Online publication date: 1-Dec-2019. SABAKA P, DUKAT A, GAJDOSIK J, CAPRNDA M, BENDZALA M and SIMKO F (2019) Uric Acid Level is Positively Associated With NT-proBNP Concentration in Slovak Heart Failure Patients, Physiological Research, 10.33549/physiolres.934186, (767-774), Online publication date: 21-Oct-2019. Huang G, Qin J, Deng X, Luo G, Yu D, Zhang M, Zhou S and Wang L (2019) Prognostic value of serum uric acid in patients with acute heart failure, Medicine, 10.1097/MD.0000000000014525, 98:8, (e14525), Online publication date: 1-Feb-2019. Wang H, Yang J, Sao J, Zhang J and Pang X The prediction of cardiac events in patients with acute ST segment elevation myocardial infarction: A meta–analysis of serum uric acid, Open Life Sciences, 10.1515/biol-2018-0050, 13:1, (413-421) Kumar R, Joshi G, Kler H, Kalra S, Kaur M and Arya R (2017) Toward an Understanding of Structural Insights of Xanthine and Aldehyde Oxidases: An Overview of their Inhibitors and Role in Various Diseases, Medicinal Research Reviews, 10.1002/med.21457, 38:4, (1073-1125), Online publication date: 1-Jul-2018. Mirończuk-Chodakowska I, Witkowska A and Zujko M (2018) Endogenous non-enzymatic antioxidants in the human body, Advances in Medical Sciences, 10.1016/j.advms.2017.05.005, 63:1, (68-78), Online publication date: 1-Mar-2018. Mogensen U, Køber L, Jhund P, Desai A, Senni M, Kristensen S, Dukát A, Chen C, Ramires F, Lefkowitz M, Prescott M, Shi V, Rouleau J, Solomon S, Swedberg K, Packer M and McMurray J (2017) Sacubitril/valsartan reduces serum uric acid concentration, an independent predictor of adverse outcomes in PARADIGM-HF, European Journal of Heart Failure, 10.1002/ejhf.1056, 20:3, (514-522), Online publication date: 1-Mar-2018. Chen Y, Wu S, Li W, Wang B, Lv H, Yang X, Khalid B, Yin X and Xia Y (2018) Gender-Related Association of Serum Uric Acid Levels with Premature Ventricular Contraction, International Heart Journal, 10.1536/ihj.17-474, 59:6, (1246-1252), Online publication date: 30-Nov-2018. Liu J, Zhang G, Chen D, Chen J, Yuan Z, Zhang E, Gao Y, Xu G, Sun B, Liao W and Gao Y (2017)(2017) UPLC-QTOFMS-based metabolomic analysis of the serum of hypoxic preconditioning mice, Molecular Medicine Reports, 10.3892/mmr.2017.7493, 16:5, (6828-6836), Online publication date: 1-May-2017. Wu X, Xing Y, Pierce D and Zhao J (2017) One-Pot Synthesis of Reduced Graphene Oxide/Metal (Oxide) Composites, ACS Applied Materials & Interfaces, 10.1021/acsami.7b12539, 9:43, (37962-37971), Online publication date: 1-Nov-2017. La Frano M, Fahrmann J, Grapov D, Fiehn O, Pedersen T, Newman J, Underwood M, Steinhorn R and Wedgwood S (2017) Metabolic perturbations of postnatal growth restriction and hyperoxia-induced pulmonary hypertension in a bronchopulmonary dysplasia model, Metabolomics, 10.1007/s11306-017-1170-6, 13:4, Online publication date: 1-Apr-2017. Tanaka Y, Nagoshi T, Kawai M, Uno G, Ito S, Yoshii A, Kimura H, Inoue Y, Ogawa K, Tanaka T, Minai K, Ogawa T and Yoshimura M (2017) Close linkage between serum uric acid and cardiac dysfunction in patients with ischemic heart disease according to covariance structure analysis, Scientific Reports, 10.1038/s41598-017-02707-y, 7:1, Online publication date: 1-Dec-2017. Martínez-Ramírez M, Flores-Castillo C, Sánchez-Lozada L, Bautista-Pérez R, Carreón-Torres E, Fragoso J, Rodriguez-Pérez J, García-Arroyo F, López-Olmos V, Luna-Luna M, Vargas-Alarcón G, Franco M and Pérez-Méndez O (2017) Hyperuricemia is Associated with Increased Apo AI Fractional Catabolic Rates and Dysfunctional HDL in New Zealand Rabbits, Lipids, 10.1007/s11745-017-4301-y, 52:12, (999-1006), Online publication date: 1-Dec-2017. Liao W, Liu B, Chen J, Cui J, Gao Y, Liu F, Xu G, Sun B, Zhang E, Yuan Z, Zhang G and Gao Y (2016) Metabolite Modulation in Human Plasma in the Early Phase of Acclimatization to Hypobaric Hypoxia, Scientific Reports, 10.1038/srep22589, 6:1, Online publication date: 1-Mar-2016. Qin T, Zhou X, Wang J, Wu X, Li Y, Wang L, Huang H and Li J (2016) Hyperuricemia and the Prognosis of Hypertensive Patients: A Systematic Review and Meta-Analysis, The Journal of Clinical Hypertension, 10.1111/jch.12855, 18:12, (1268-1278), Online publication date: 1-Dec-2016. Wu A, Gladden J, Ahmed M, Ahmed A and Filippatos G (2016) Relation of serum uric acid to cardiovascular disease, International Journal of Cardiology, 10.1016/j.ijcard.2015.08.110, 213, (4-7), Online publication date: 1-Jun-2016. Huang W, Hsu P, Cheng H, Lu D, Cheng Y, Guo C, Sung S, Yu W and Chen C (2016) Determinants and Prognostic Impact of Hyperuricemia in Hospitalization for Acute Heart Failure, Circulation Journal, 10.1253/circj.CJ-15-0964, 80:2, (404-410), . Nagula P, Reddy Yerrabandi V, Otikunta A and Karumuri S (2016) ROLE OF SERUM BILIRUBIN AS A MARKER OF CORONARY ARTERY DISEASE, Journal of Evidence Based Medicine and Healthcare, 10.18410/jebmh/2016/1043, 3:91, (4954-4958), Online publication date: 11-Nov-2016. Kelley E (2015) Dispelling dogma and misconceptions regarding the most pharmacologically targetable source of reactive species in inflammatory disease, xanthine oxidoreductase, Archives of Toxicology, 10.1007/s00204-015-1523-8, 89:8, (1193-1207), Online publication date: 1-Aug-2015. Zhu L, Wang J, Wang Y, Jia L, Sun K, Wang H, Zou Y, Tian T, Liu Y, Zou J, Hui R, Yuan Z and Song L (2015) Plasma Uric Acid as a Prognostic Marker in Patients With Hypertrophic Cardiomyopathy, Canadian Journal of Cardiology, 10.1016/j.cjca.2015.02.018, 31:10, (1252-1258), Online publication date: 1-Oct-2015. Zhang Y, Xu R, Li S, Zhu C, Guo Y, Sun J and Li J (2015) Lipoprotein subfractions partly mediate the association between serum uric acid and coronary artery disease, Clinica Chimica Acta, 10.1016/j.cca.2014.12.030, 441, (109-114), Online publication date: 1-Feb-2015. Kang D and Johnson R (2015) Uric Acid Metabolism and the Kidney Chronic Renal Disease, 10.1016/B978-0-12-411602-3.00035-4, (418-428), . Nakagomi A, Saiki Y, Noma S, Kohashi K, Morisawa T, Kosugi M, Kusama Y, Atarashi H and Shimizu W (2015) Effects of febuxostat and allopurinol on the inflammation and cardiac function in chronic heart failure patients with hyperuricemia, IJC Metabolic & Endocrine, 10.1016/j.ijcme.2015.07.001, 8, (46-55), Online publication date: 1-Sep-2015. Turker Y, Ekinozu I, Turker Y and Akkaya M (2014) High levels of high-sensitivity C-reactive protein and uric acid can predict disease severity in patients with mitral regurgitation, Revista Portuguesa de Cardiologia, 10.1016/j.repc.2014.03.014, 33:11, (699-706), Online publication date: 1-Nov-2014. Huang H, Huang B, Li Y, Huang Y, Li J, Yao H, Jing X, Chen J and Wang J (2013) Uric acid and risk of heart failure: a systematic review and meta-analysis, European Journal of Heart Failure, 10.1093/eurjhf/hft132, 16:1, (15-24), Online publication date: 1-Jan-2014. Yan L, Liu Z and Zhang C (2014) Uric Acid as a Predictor of In-Hospital Mortality in Acute Myocardial Infarction: A Meta-analysis, Cell Biochemistry and Biophysics, 10.1007/s12013-014-0101-7, 70:3, (1597-1601), Online publication date: 1-Dec-2014. Battelli M, Bolognesi A and Polito L (2014) Pathophysiology of circulating xanthine oxidoreductase: New emerging roles for a multi-tasking enzyme, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 10.1016/j.bbadis.2014.05.022, 1842:9, (1502-1517), Online publication date: 1-Sep-2014. Oberbach A, Neuhaus J, Schlichting N, Kugler J, Baumann S and Till H (2014) Sleeve gastrectomy reduces xanthine oxidase and uric acid in a rat model of morbid obesity, Surgery for Obesity and Related Diseases, 10.1016/j.soard.2013.12.010, 10:4, (684-690), Online publication date: 1-Jul-2014. Turker Y, Ekinozu I, Turker Y and Akkaya M (2014) High levels of high-sensitivity C-reactive protein and uric acid can predict disease severity in patients with mitral regurgitation, Revista Portuguesa de Cardiologia (English Edition), 10.1016/j.repce.2014.03.014, 33:11, (699-706), Online publication date: 1-Nov-2014. Borghi C, Cosentino E, Rinaldi E and Cicero A (2014) Uricaemia and ejection fraction in elderly heart failure outpatients, European Journal of Clinical Investigation, 10.1111/eci.12273, 44:6, (573-577), Online publication date: 1-Jun-2014. Borghi C, Verardi F, Pareo I, Bentivenga C and Cicero A (2014) Hyperuricemia and cardiovascular disease risk, Expert Review of Cardiovascular Therapy, 10.1586/14779072.2014.957675, 12:10, (1219-1225), Online publication date: 1-Oct-2014. Kivity S, Kopel E, Maor E, Abu-Bachar F, Segev S, Sidi Y and Olchovsky D (2013) Association of Serum Uric Acid and Cardiovascular Disease in Healthy Adults, The American Journal of Cardiology, 10.1016/j.amjcard.2012.12.034, 111:8, (1146-1151), Online publication date: 1-Apr-2013. Kalogeropoulos A, Georgiopoulou V and Butler J (2012) Biomarkers in Acute Heart Failure: From Risk Markers to Therapeutic Targets, Congestive Heart Failure, 10.1111/chf.12019, 19:2, (51-52), Online publication date: 1-Mar-2013. Kaufman M and Guglin M (2012) Uric acid in heart failure: a biomarker or therapeutic target?, Heart Failure Reviews, 10.1007/s10741-012-9322-2, 18:2, (177-186), Online publication date: 1-Mar-2013. Kanbay M, Segal M, Afsar B, Kang D, Rodriguez-Iturbe B and Johnson R (2013) The role of uric acid in the pathogenesis of human cardiovascular disease, Heart, 10.1136/heartjnl-2012-302535, 99:11, (759-766), Online publication date: 1-Jun-2013. N S, Hemalatha H, K. C S, Shwetha S and T.M. R (2013) ANTIOXIDANT STATUS, OXIDATIVE STRESS AND LIPID PROFILE IN ESSENTIAL HYPERTENSIVE MEN, Journal of Evolution of Medical and Dental Sciences, 10.14260/jemds/640, 2:17, (2950-2955), Online publication date: 29-Apr-2013. Timóteo A, Lousinha A, Labandeiro J, Miranda F, Papoila A, Oliveira J, Ferreira M and Ferreira R (2013) Serum uric ac

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call