Horm Metab Res 2022; 54(05): 325-334
DOI: 10.1055/a-1815-7559
Original Article: Endocrine Care

The Negative Association Between NAFLD Severity and CKD in a Non-Diabetic Gouty Population

Yajing Huang
1   Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
,
Yue Zhou
2   Endocrinology and Metabolism, Fudan University, Shanghai, China
,
Yahao Wang
2   Endocrinology and Metabolism, Fudan University, Shanghai, China
,
Jingwei Chi
1   Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
,
Wenshan Lv
1   Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
,
Yangang Wang
1   Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
,
Ying Chen
3   Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
› Author Affiliations

Abstract

Abstract Nonalcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD) share common pathogenic mechanisms and risk factors. We aim to evaluate the association between NAFLD and CKD in a non-diabetic gouty population. The retrospective cross sectional study was performed on 1049 non-diabetic gouty participants, who were hospitalized between 2014 and 2020, across 4 districts in Shandong, China. Demographic and clinical characteristics of the study population were collected. The odds ratios (OR) and corresponding 95% confidence intervals (CI) about the NAFLD severity determined by ultrasonography were obtained by multiple logistic regression analysis. An unexpectedly inverse relationship was found between NAFLD severity and the risk of CKD in people with gout. Multivariate logistic regression analysis demonstrated that a higher degree of NAFLD severity is independently associated with a lower risk of CKD in people with gout, after adjusted for age, sex, smoking, gout duration, and metabolic risk factors including obesity, hypertension, hyperglycemia, hyperuricemia, and dyslipidemia, with OR 0.392 (95% CI 0.248–0.619, p<0.001), 0.379 (95% CI 0.233–0.616, p<0.001) and 0.148 (95% CI 0.043–0.512, p=0.003) in participants with mild, moderate, and severe NAFLD, respectively, compared to those without NAFLD. We also observed a weakened association of serum uric acid (SUA) with metabolic risk factors and NAFLD under circumstances of CKD in people with gout (r=–0.054, p=0.466). In conclusion, the presence and severity of NAFLD were negatively associated with the risk of CKD in the non-diabetic gouty population.



Publication History

Received: 05 January 2022

Accepted after revision: 04 April 2022

Accepted Manuscript online:
04 April 2022

Article published online:
09 May 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Stamp LK, Chapman PT. Gout and its comorbidities: implications for therapy. Rheumatology (Oxford) 2013; 52: 34-44
  • 2 Kuo CF, Grainge MJ, Zhang W. et al. Global epidemiology of gout: prevalence, incidence and risk factors. Nat Rev Rheumatol 2015; 11: 649-662
  • 3 Ryu ES, Kim MJ, Shin HS. et al. Uric acid-induced phenotypic transition of renal tubular cells as a novel mechanism of chronic kidney disease. Am J Physiol Renal Physiol 2013; 304: F471-F480
  • 4 Perlstein TS, Gumieniak O, Hopkins PN. et al. Uric acid and the state of the intrarenal renin-angiotensin system in humans. Kidney Int 2004; 66: 1465-1470
  • 5 Weiner DE, Tighiouart H, Elsayed EF. et al. Uric acid and incident kidney disease in the community. J Am Soc Nephrol 2008; 19: 1204-1211
  • 6 Roughley M, Belcher J, Mallen C. et al. Gout and risk of chronic kidney disease and nephrolithiasis: meta-analysis of observational studies. Arthritis Res Ther 2015; 17: 90
  • 7 Yu K, Kuo C, Luo S. et al. Risk of end-stage renal disease associated with gout: a nationwide population study. Arthritis Res Ther 2012; 14: R83
  • 8 Lusco MA, Fogo AB, Najafian B. et al. AJKD atlas of renal pathology: gouty nephropathy. Am J Kidney Dis 2017; 69: e5-e6
  • 9 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
  • 10 Kanellis J, Watanabe S, Li JH. et al. Uric acid stimulates monocyte chemoattractant protein-1 production in vascular smooth muscle cells via mitogen-activated protein kinase and cyclooxygenase-2. Hypertension 2003; 41: 1287-1293
  • 11 Gul A, Zager P. Does Altered uric acid metabolism contribute to diabetic kidney disease pathophysiology?. Curr Diab Rep 2018; 18: 18
  • 12 Targher G, Chonchol MB, Byrne CD. CKD and nonalcoholic fatty liver disease. Am J Kidney Dis 2014; 64: 638-652
  • 13 Musso G, Cassader M, Cohney S. et al. Fatty liver and chronic kidney disease: novel mechanistic insights and therapeutic opportunities. Diabetes Care 2016; 39: 1830-1845
  • 14 Byrne CD, Targher G. NAFLD as a driver of chronic kidney disease. J Hepatol 2020; 72: 785-801
  • 15 Kanbay M, Bulbul MC, Copur S. et al. Therapeutic implications of shared mechanisms in non-alcoholic fatty liver disease and chronic kidney disease. J Nephrol 2020; 34: 649-659
  • 16 Zhang M, Lin S, Wang MF. et al. Association between NAFLD and risk of prevalent chronic kidney disease: why there is a difference between east and west?. BMC Gastroenterol 2020; 20: 139
  • 17 Chen PC, Kao WY, Cheng YL. et al. The correlation between fatty liver disease and chronic kidney disease. J Formos Med Assoc 2020; 119: 42-50
  • 18 Akahane T, Akahane M, Namisaki T. Association between non-alcoholic fatty liver disease and chronic kidney disease: a cross-sectional study. J Clin Med 2020; 9: 1635
  • 19 Targher G, Bertolini L, Rodella S. et al. Non-alcoholic fatty liver disease is independently associated with an increased prevalence of chronic kidney disease and proliferative/laser-treated retinopathy in type 2 diabetic patients. Diabetologia 2008; 51: 444-450
  • 20 Hwang ST, Cho YK, Yun JW. et al. Impact of non-alcoholic fatty liver disease on microalbuminuria in patients with prediabetes and diabetes. Intern Med J 2010; 40: 437-442
  • 21 Richette P, Doherty M, Pascual E. et al. 2016 updated EULAR evidence-based recommendations for the management of gout 2017; 76: 29-42
  • 22 WHO. Obesity: preventing and managing the global epidemic. Report of a WHO consultation. World Health Organ Tech Rep Ser 2000; 894 i–xii 1-253
  • 23 Matthews DR, Hosker JP, Rudenski AS. et al. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 1985; 28: 412-419
  • 24 Stevens LA, Claybon MA, Schmid CH. et al. Evaluation of the chronic kidney disease epidemiology collaboration equation for estimating the glomerular filtration rate in multiple ethnicities. Kidney Int 2011; 79: 555-562
  • 25 Inker LA, Astor BC, Fox CH. et al. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for the evaluation and management of CKD. Am J Kidney Dis 2014; 63: 713-735
  • 26 Sinn DH, Kang D, Jang HR. et al. Development of chronic kidney disease in patients with non-alcoholic fatty liver disease: a cohort study. J Hepatol 2017; 67: 1274-1280
  • 27 Musso G, Cassader M, Cohney S. et al. Emerging liver-kidney interactions in nonalcoholic fatty liver disease. Trends Mol Med 2015; 21: 645-662
  • 28 Lee YJ, Wang CP, Hung WC. et al. Common and unique factors and the bidirectional relationship between chronic kidney disease and nonalcoholic fatty liver in type 2 diabetes patients. 2020; 13: 1203-1214
  • 29 Khukhlina O, Antoniv A, Kanovska L. et al. Intensity of the antioxidant protection system and oxidative stress factors in patients with non-alcoholic steatohepatitis depending on the form of chronic kidney disease. Georgian Med News 2018: 71–76
  • 30 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 1981; 78: 6858-6862
  • 31 Frei B, Stocker R, Ames BN. Antioxidant defenses and lipid peroxidation in human blood plasma[J]. Proc Natl Acad Sci U S A 1988; 85: 9748-9752
  • 32 Yeum KJ, Russell RM, Krinsky NI. et al. Biomarkers of antioxidant capacity in the hydrophilic and lipophilic compartments of human plasma. Arch Biochem Biophys 2004; 430: 97-103
  • 33 Murea M, Tucker BM. The physiology of uric acid and the impact of end-stage kidney disease and dialysis. Semin Dial 2019; 32: 47-57
  • 34 Stinefelt B, Leonard SS, Blemings KP. et al. Free radical scavenging, DNA protection, and inhibition of lipid peroxidation mediated by uric acid. Ann Clin Lab Sci 2005; 35: 37-45
  • 35 Becker BF. Towards the physiological function of uric acid. Free Radic Biol Med 1993; 14: 615-631
  • 36 Vukovic J, Modun D, Budimir D. et al. Acute, food-induced moderate elevation of plasma uric acid protects against hyperoxia-induced oxidative stress and increase in arterial stiffness in healthy humans. Atherosclerosis 2009; 207: 255-260
  • 37 Davies KJ, Sevanian A, Muakkassah-Kelly SF. et al. Uric acid-iron ion complexes. A new aspect of the antioxidant functions of uric acid. Biochem J 1986; 235: 747-754
  • 38 Strazzullo P, Puig JG. Uric acid and oxidative stress: relative impact on cardiovascular risk?. Nutr Metab Cardiovasc Dis 2007; 17: 409-414
  • 39 Cheong E, Ryu S, Lee JY. et al. Association between serum uric acid and cardiovascular mortality and all-cause mortality: a cohort study. J Hypertens 2017; 35: S3-S9
  • 40 Gluba-Brzozka A, Franczyk B, Bartnicki P. et al. Lipoprotein subfractions, uric acid and cardiovascular risk in end-stage renal disease (ESRD) patients. Curr Vasc Pharmacol 2017; 15: 123-134
  • 41 Li Y, Xu C, Yu C. et al. Association of serum uric acid level with non-alcoholic fatty liver disease: a cross-sectional study. J Hepatol 2009; 50: 1029-1034
  • 42 Xia MF, Lin HD, Li XM. et al. Renal function-dependent association of serum uric acid with metabolic syndrome and hepatic fat content in a middle-aged and elderly Chinese population. Clin Exp Pharmacol Physiol 2012; 39: 930-937
  • 43 Baker JF, Krishnan E, Chen L. et al. Serum uric acid and cardiovascular disease: recent developments, and where do they leave us?. Am J Med 2005; 118: 816-826
  • 44 George J, Carr E, Davies J. et al. High-dose allopurinol improves endothelial function by profoundly reducing vascular oxidative stress and not by lowering uric acid. Circulation 2006; 114: 2508-2516
  • 45 Ifudu O, Tan CC, Dulin AL. et al. Gouty arthritis in end-stage renal disease: clinical course and rarity of new cases. Am J Kidney Dis 1994; 23: 347-351
  • 46 Ohno I, Ichida K, Okabe H. et al. Frequency of gouty arthritis in patients with end-stage renal disease in Japan. Intern Med 2005; 44: 706-709
  • 47 Schreiner O, Wandel E, Himmelsbach F. et al. Reduced secretion of proinflammatory cytokines of monosodium urate crystal-stimulated monocytes in chronic renal failure: an explanation for infrequent gout episodes in chronic renal failure patients?. Nephrol Dial Transplant 2000; 15: 644-649
  • 48 Nishikawa T, Nagata N. Xanthine oxidase inhibition attenuates insulin resistance and diet-induced steatohepatitis in mice. 2020; 10: 815
  • 49 Nakatsu Y, Seno Y, Kushiyama A. et al. The xanthine oxidase inhibitor febuxostat suppresses development of nonalcoholic steatohepatitis in a rodent model. Am J Physiol Gastrointest Liver Physiol 2015; 309: G42-G51
  • 50 Takano Y, Hase-Aoki K, Horiuchi H. et al. Selectivity of febuxostat, a novel non-purine inhibitor of xanthine oxidase/xanthine dehydrogenase. Life Sci 2005; 76: 1835-1847
  • 51 Fishbane S, Spinowitz B. Update on anemia in ESRD and earlier stages of CKD: core curriculum 2018. Am J Kidney Dis 2008; 71: 423-435
  • 52 Kim C, Chang Y, Sung E. et al. Serum ferritin levels predict incident non-alcoholic fatty liver disease in healthy Korean men 2012; 61: 1182-1188
  • 53 Facchini F, Hua N, Stoohs RJG. Effect of iron depletion in carbohydrate-intolerant patients with clinical evidence of nonalcoholic fatty liver disease 2002; 122: 931-939
  • 54 Valenti L, Fracanzani A, Dongiovanni P. et al. A randomized trial of iron depletion in patients with nonalcoholic fatty liver disease and hyperferritinemia 2014; 20: 3002-3010
  • 55 Adams L, Crawford D, Stuart K. et al. The impact of phlebotomy in nonalcoholic fatty liver disease: a prospective, randomized, controlled trial 2015; 61: 1555-1564
  • 56 Tarantino G, Vinciguerra M, Ragosta A. et al. Do transferrin levels predict haemodialysis adequacy in patients with end-stage renal disease?. Nutrients 2019; 11: 1123
  • 57 Chen L, Xiong S, She H. et al. Iron causes interactions of TAK1, p21ras, and phosphatidylinositol 3-kinase in caveolae to activate IkappaB kinase in hepatic macrophages 2007; 282: 5582-5588
  • 58 Ruddell R, Hoang-Le D, Barwood J. et al. Ferritin functions as a proinflammatory cytokine via iron-independent protein kinase C zeta/nuclear factor kappaB-regulated signaling in rat hepatic stellate cells. 2009; 49: 887-900