Association Between Serum Uric Acid Levels and Metabolic-Associated Fatty Liver Disease in Southeast China: A Cross-Sectional Study

Background

Non-alcoholic fatty liver disease (NAFLD) refers to the excessive deposition of fat in hepatocytes, which is diagnosed after excluding excessive alcohol consumption and other definite factors that lead to liver damage.1 However, with the increasing prevalence of hepatic steatosis, the diagnostic criteria for NAFLD are gradually unable to meet the needs of clinical work. In 2020, the International Panel of Liver Experts renamed NAFLD to metabolic-associated fatty liver disease (MAFLD),2 which is a more appropriate disease designation for liver diseases associated with metabolic dysfunction.3 MAFLD is one of the most prevalent chronic liver diseases worldwide, affecting at least 25% of the world’s adult population and 29–46% of the Chinese population.4–6 MAFLD not only cause cirrhosis and hepatocellular carcinoma, but also increases the risk of type 2 diabetes, cardiovascular disease (CVD), chronic kidney disease, and metabolic syndrome.7–9 Therefore, early detection and intervention of risk factors for MAFLD is extremely important.

Uric Acid (UA), the end product of purine metabolism, is continuously produced, excreted, and maintained at a certain concentration in the blood. Overproduction or reduced UA excretion increases serum Uric Acid (sUA) levels. Several studies have shown that sUA was a risk factor of CVD, type 2 diabetes mellitus, and metabolic syndrome.10,11 Worldwide, high level of sUA is also associated with an increased prevalence of metabolic syndrome.12–14 Many studies have demonstrated that sUA is a risk factor for NAFLD, a study in US found that sUA was independently associated with NAFLD, increasing sUA was associated with increasing severity of NAFLD.15 A cross-sectional study also showed serum uric acid/creatinine ratio was significantly higher in subjects with NAFLD than those without NAFLD.16 Meta-analysis showed positive correlations between sUA and the risk of NAFLD.17–19 But the different definitions of NAFLD and MAFLD inevitably lead to differences between the affected populations. The association between sUA levels and MAFLD remained unclear. The participants included in our study were permanent residents of Nanping City, where the prevalence of NAFLD was 32.8%, which was higher than other cities in Fujian province.20 This cross-sectional study aimed to explore the association between sUA and the risk of MAFLD in Southeast China.

Materials and Methods Study Design and Subjects

This cross-sectional study, involving 2605 subjects who underwent physical examination and completed abdominal ultrasonography between April 2015 and August 2017 at the Physical Examination Center of Nanping First Hospital Affiliated to Fujian Medical University. All participants provided informed consent before the start of the study.

The inclusion criterion for participants was permanent residents of Nanping, aged between 18–74 years old. The exclusion criteria were as follows: (A) presence of acute and chronic infections; (B) participants with missing information on abdominal ultrasound, blood tests, and physical measurements; (C) participants with chronic hepatitis and cirrhosis, coronary heart disease, stroke, and cancer; and (D) pregnant or lactating mothers.

Diagnostic Criteria for MAFLD

The diagnosis of MAFLD is based on ultrasound showing hepatic steatosis using one of the following three criteria: (A) overweight or obesity (BMI≥ 23.0 kg/m2), (B) type 2 Diabetes Mellitus, and (C) metabolic dysfunction among non-overweight individuals (BMI< 23.0 kg/m2). Metabolic dysfunction is defined as meeting two of the following indicators: (a) Waist circumference: Asian male/female≥ 90/80 cm; (b) Blood pressure≥ 130/85 mmHg or receiving specific medication; (c) Plasma triglycerides≥ 150 mg/dL (≥ 1.7 mmol/L) or on specific drug treatment; (d) HDL-c< 40 mg/dL(1.0 mmol/L) for men and < 50 mg/dL (1.3 mmol/L) for women or on specific drug treatment; (e) Pre-diabetes; (f) HOMA-IR≥ 2.5; (g) C-reactive protein> 2 mg/L.

Data Collection and Measurement

Information on MAFLD risk factors was obtained from face-to-face interviews with trained investigators. Risk factors included gender, age, smoking status, drinking status, tea drinking status, lifestyle, dietary habits, disease history and treatment status. Clinical variables were collected after overnight fasting, including height (m), weight (kg), body mass index (BMI, kg/m2), hip circumference (HC, cm), waist circumference (WC, cm), Waist-hip Ratio (WHR), diastolic blood pressure (DBP, mmHg), systolic blood pressure (SBP, mmHg), serum triglyceride (TG, mmol/L), total cholesterol (TC, mmol/L), low-density lipoprotein (LDL, mmol/L), high-density lipoprotein (HDL, mmol/L), fasting plasma glucose (FPG, mmol/L), gamma-glutamyl transferase (GGT, U/L), blood urea nitrogen (BUN, mmol/L), creatinine (CR, umol/L) and serum Uric Acid (sUA, mg/dL). Hypertension was defined as SBP≥ 140 mmHg and/or DBP≥ 90 mmHg or current use of antihypertensive medication. Diabetes was defined as FPG≥ 7.0 mmol/L or current use of hypoglycaemic agents.

Statistical Analysis

The baseline characteristics of the subjects were analyzed using the Nonparametric Kruskal–Wallis test for non-normal continuous variables and the chi-square test for nominal variables. Continuous variables were expressed as median (interquartile range, IQR). Nominal variables are expressed as frequencies (n) and constitutive ratios (%).

Univariate and multivariate logistic regression analysis were performed to analyze the association between sUA and MAFLD risk. A RCS plot was used to present the dose–response relationship between sUA and MAFLD risk. Subgroups analysis and multiplicative interaction analysis were used to examine the relationship of sUA with MAFLD risk by the following subgroups: gender (male or female), age (≤ 60 years or> 60 years), BMI (<24 kg/m2 or≥24 kg/m2), drinking status (yes or no), smoking status (yes or no), tea drinking status (yes or no). I2 and Q tests were used to assess heterogeneity. A P value < 0.05 for the Q statistic or I2 more than 50% suggests notable heterogeneity. Multiplicative interaction analysis was performed based on the heterogeneity. Statistical analysis were performed in SPSS version 26.0, Stata version 17.0, and R version 4.3.0. P significance was set at P< 0.05.

Results Baseline Characteristics

The demographics, lifestyle habits, and clinical characteristics of subjects are shown in Tables 1 and 2. Of the 2605 participants, 726 had MAFLD, with a prevalence of 27.9%. Compared with subjects without MAFLD, subjects with MAFLD were more likely to be male, smokers, drinkers, tea drinkers, have higher BMI, prefer salty foods, eat fast, and have diabetes or hypertension (all P< 0.001). Additionally, there were significant differences in clinical detection indicators (WC, HC, WHR, SBP, DBP, HDL, LDL, GGT, TC, FPG, TG, sUA, BUN, and CR) between the two groups (all P < 0.001).

Table 1 Comparison of Demographic and Lifestyle Habits Characteristics

Table 2 Comparison of Biochemical Indices

Association of sUA with MAFLD

sUA levels were categorized into two groups according to the normal reference range of sUA (2.5–7.0 mg/dL for males; 1.5–6 mg/dL for females). A logistic regression model was used to analyze the association between sUA and MAFLD. As shown in Table 3, in the crude model, sUA was positively correlated with MAFLD in total population, males and females. After adjusting for age, BMI, smoking status, drinking status, tea drinking status, taste, eating speed, hypertension, hypertension treatment status, diabetes, diabetes treatment status, HDL, LDL, TC, BUN and CR, the positive association between sUA and MAFLD remained unchanged in total population and males.

Table 3 Univariate and Multivariate Logistic Analysis of sUA and MAFLD

The dose-response relationship between sUA and MAFLD risk was interpreted by RCS analysis. There was a significant nonlinear correlation between sUA and MAFLD risk (P nonlinear< 0.001), and the risk of MAFLD increased significantly when sUA> 5.56 mg/dL (Figure 1).

Figure 1 Restrictive cubic spline modelling of the association between sUA levels and MAFLD risk.

Notes: Red area, 95% CI. The RCS model was adjusted for age, gender, BMI, smoking status, drinking status, tea drinking status, taste, eating speed, hypertension, hypertension treatment status, diabetes, diabetes treatment status, HDL, LDL, TC, BUN and CR.

Subgroups Analysis

Subgroup analysis was performed to investigate the robustness of relationship between sUA and MAFLD risk. The positive association between sUA and MAFLD risk was consistent across strata of gender, age, BMI, drinking status, smoking status and tea drinking status. Significant associations between sUA and MAFLD risk were not only found in males but also existed in subjects whose age≤ 60, BMI≥ 24 kg/m2, drinkers, smokers and tea-drinkers. Adjusted ORs were estimated to be 2.01, 1.95, 2.11, 2.29, 2.64 and 2.20, respectively (Figure 2).

Figure 2 Forest plot of subgroups analysis of the relationship between sUA and MAFLD risk.

Notes: Adjusted for gender, age, BMI, smoking status, drinking status, tea drinking status, taste, eating speed, hypertension, hypertension treatment status, diabetes, diabetes treatment status, HDL, LDL, TC, BUN and CR.

Interaction Analysis

According to the subgroups analysis, a large heterogeneity was observed in the subgroup of gender, age, drinking status, smoking status and tea drinking status (I2gender=70.2%, I2age=53.9%, I2drinking status=68.1%, I2smoking status=60.6% and I2tea drinking status=81.8%), indicating possible interactions between gender, age, drinking status, smoking status, tea drinking status and sUA. Multiplicative interactions analysis were performed to further explore the robustness of association between sUA and MALFD risk. The results showed multiplicative interactions were not observed (P interaction all > 0.05) (Tables 4–8).

Table 4 Multiplicative Interaction Analysis Between Gender and sUA

Table 5 Multiplicative Interaction Analysis Between Age and sUA

Table 6 Multiplicative Interaction Analysis Between Drinking Status and sUA

Table 7 Multiplicative Interaction Analysis Between Smoking Status and sUA

Table 8 Multiplicative Interaction Analysis Between Tea Drinking Status and sUA

Discussion

In this cross-sectional study, we explore the association between sUA levels and MALFD risk in Southeast China. Our results highlighted that sUA was positively associated with the risk of MAFLD. This association persisted after adjustment for potential confounding factors. The RCS plot demonstrated that there was a significant nonlinear correlation between sUA and MAFLD risk, and the risk of MAFLD increased significantly when sUA> 5.56 mg/dL. Subgroup and multiplicative interaction analysis were performed to investigate the robustness of association between sUA and MAFLD risk. The results revealed that the positive association between sUA and MAFLD risk persisted in all subgroups, indicating strong robustness.

Previous studies have investigated the association between sUA and NAFLD, and the results showed that high sUA levels may be associated with NAFLD.21–24 However, there are differences in diagnostic criteria and epidemiological characteristics between NAFLD and MAFLD.25 Evidence on association between sUA and MAFLD are limited. Several studies revealed that serum uric acid to serum creatinine ratio was positively associated with the risk of MAFLD. But the diagnosis of MAFLD in these studies was based on abdominal computed tomography rather than ultrasound,26,27 and these studies did not explore the dose-response relationship between sUA and MAFLD risk. In our study, univariate logistic regression analysis revealed a strong association between sUA and MAFLD risk, OR (95% CI) =3.48 (2.87–4.22). SUA is also associated with hypertension,28 obesity,29 metabolic syndrome,30 type 2 diabetes mellitus,31 chronic kidney disease,32 dyslipidemia,33 and CVD.34 The treatment received by patients with diabetes and hypertension, particularly those involving diuretics,35 has also been identified as an important factor influencing sUA levels. Therefore, we further included relevant variables in the multivariate model. After adjusting for potential confounders, the positive association between sUA and MAFLD risk remained unchanged, OR (95% CI)= 1.90 (1.49 ~ 2.42).

Despite the lack of consensus on the optimal range of sUA levels, a widely accepted therapeutic goal for patients with hyperuricemia is to maintain sUA levels of< 6.0 mg/dL for females and< 7.0 mg/dL for males.36 In our study, the RCS plot demonstrated that there was a significant nonlinear correlation between sUA and MAFLD risk, and the risk of MAFLD increased significantly when sUA> 5.56 mg/dL, a threshold lower than the aforementioned therapeutic goal. Similar findings have also been observed in the relationship between sUA and CVD.37 A large-scale cohort study conducted in Italy has revealed a significant association elevated sUA levels and the risk of fatal myocardial infarction, with a clear cutoff value established at SUA levels exceeding 5.70 mg/dL.38 Another study demonstrated U-shaped curve relationship between sUA levels and CVD. Specifically, sUA levels> 370.5 μmol/L (6.2mg/dL) in males and 327.65 μmol/L (5.5mg/dL) in females were associated with increased CVD mortality.39 The similarity between the sUA cut-off values in MAFLD patients and the sUA levels predictive of cardiovascular (CV) events could be attributed to the similar pathophysiological roles that sUA plays in both MAFLD and CVD, serving as a promoter of oxidative stress, inflammatory response, and endothelial dysfunction.40 Furthermore, hypouricemic agents have already shown to reduce CV outcomes in people with metabolic syndrome.41–43 Hence, controlling sUA levels in patients with MAFLD may help identify those at higher risk of CV events, enabling earlier intervention and potentially improving outcomes.

Subgroups analysis revealed that the positive association between sUA and MAFLD persisted significantly in males but not in females, aOR male (95% CI)= 2.01 (1.54 ~ 2.62), aOR female (95% CI)= 1.15 (0.62 ~ 2.11), respectively. Our findings are consistent with previous studies. He and Ye found sUA levels were positively associated with the severity of steatosis in male MAFLD patients. However, these associations were not found for females.44 Similar gender differences have been found in other related studies involving sUA and NAFLD.45,46 We also found the prevalence of MAFLD was higher in males (39.7%) than females (12.5%), which was consistent with previous studies.25,47,48 In general, males have higher sUA levels than females because the estrogen in females can promote UA excretion.49 In addition, gender-specific differences in genetic factors and gene expression may also lead to differential associations between sUA and MAFLD.50,51

However, the specific mechanism of the association between sUA and MAFLD has not been confirmed. Studies have shown that there exists a bidirectional relationship between sUA and metabolic syndrome. Metabolic syndrome is characterized by a cluster of metabolic abnormalities, including hypertension, dyslipidemia, and abdominal obesity, all of which can contribute to increased sUA production and decreased excretion.52 On the other hand, sUA has emerged as a definitive role for metabolic syndrome. High level of sUA is associated with hypertension, NAFLD, chronic kidney disease, and CVD.40 MAFLD is generally considered to be the hepatic manifestation of metabolic syndrome. Studies have shown that multiple components of metabolic syndrome, such as dyslipidemia, and central obesity, are potential pathophysiologic mechanisms and risk factors for the development of MAFLD.53–55 These risk factors shared by MAFLD and metabolic syndrome contribute to increased sUA production and decreased excretion, and increased the risk of developing NAFLD.56 Our findings are consistent with this argument. In our study, compared to subjects without MAFLD, subjects with MAFLD had higher BMI, SBP, DBP, LDL, TG, TC levels and lower HDL levels. Furthermore, after adjustment for components of metabolic syndrome and other potential confounders, elevated sUA is independently associated with increased risk of MAFLD, with the adjusted OR of 1.90 (1.49 ~ 2.42) (P < 0.01). Insulin resistance (IR) is the basis of the occurrence and development of metabolic dysfunction and MAFLD.57 Both IR and the triglyceride–glucose (TyG) index were positively associated with sUA.58 It has been well documented that high concentration of UA can induce oxidative stress and promote the generation of reactive oxygen species, which cause IR and lead to the occurrence of MAFLD.59,60 Our previous findings are in favor of this argument. We found the TyG index can be used as an alternative maker for IR and effectively identify MAFLD, and the AUC of the TyG index for predicting MAFLD was up to 0.793.61 The energy metabolism of hepatocytes is mainly mediated by mitochondria. UA can induce mitochondrial morphological changes and oxidative stress, which promote the development of MAFLD.62,63 Basic studies have shown that sUA may induce hepatic fat accumulation through ROS/JNK/AP-1 signaling pathway, thus promoting MAFLD progression.64 High levels of sUA activates the NLRP3 inflammasome, which may be positively correlated with the progression of MAFLD.65–67 The effect of UA on lipid accumulation in hepatocytes may be related to microRNA,68 and the abnormal expression of microRNA is involved in the pathogenesis of MAFLD.69 Overall, sUA may affect the progression and development of MAFLD through several mechanisms, but the exact mechanism remains to be further investigated.

Compared to NAFLD, MAFLD is a more appropriate disease definition for liver diseases associated with metabolic dysfunction. Our study demonstrates a significant positive association between sUA levels and MAFLD risk. Notably, we also found the risk of MAFLD increased significantly when sUA levels exceed 5.56 mg/dL, a threshold lower than the traditional used cut-off value for the diagnosis of hyperuricemia. It is meaningful that our findings may promote the further consideration of the underestimated diagnostic role of sUA in MAFLD. However, our study has several limitations. First, this was a cross-sectional study, and cause-effect inferences could not be made. Second, due to the limitations of the research region and population, the representativeness of the results is limited, and multi-region and multi-center research should be performed to confirm the results. Third, as an observational study, the presence of unmeasured confounders is possible. For example, as diuretics were reported to be associated with increased sUA levels,33 related data were lack in our study. Hence, the possible interference of diuretics may exist. However, we have collected the information on whether subjects with hypertension and diabetes have received treatment. After further adjustment for hypertension, hypertension treatment status, diabetes, and diabetes treatment status, the positive association between sUA and MAFLD remained statistically significant in the total population and in males. Moreover, we have performed subgroups and multiplicative interactions analysis to examine the relationship of sUA levels and MAFLD risk. This suggests that the variable of not having a diuretic did not have a large impact on our results. Last, in our study, MAFLD was diagnosed by ultrasonic examination, which is not sufficiently sensitive to detect the severity of hepatic steatosis. However, this non-invasive method has a specificity of 84% and is still widely used in population-based studies.70

Conclusion

According to our study, sUA was positively associated with the risk of MAFLD. Additionally, the risk of MAFLD increased significantly when sUA levels exceeded 5.56 mg/dL. Our study may help clarify whether sUA plays a diagnostic role in MAFLD.

Data Sharing Statement

Data are available upon reasonable request. Data are stored in the Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China. Data are available upon request from Xian-E Peng; Email address: [email protected].

Ethics Approval and Consent to Participate

The study protocol conformed to the ethical guidelines of the 1975 Declaration of Helsinki (6th revision, 2008) and was approved by Ethics Committee of Fujian Medical University (ethics number 2014096). Participants gave informed consent to participate in the study before taking part.

Acknowledgments

The authors express their gratitude to all the participants for their cooperation.

Funding

This work was supported by the National Natural Science Foundation of China (No. 81473047), the Natural Science Foundation of Fujian Province (No. 2019J01316), and the Natural Science Foundation of Fujian Province (No. 2023J01628).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Brown GT, Kleiner DE. Histopathology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Metabolism. 2016;65(8):1080–1086. doi:10.1016/j.metabol.2015.11.008

2. Liu Q, Zhao G, Li Q, Wu W, Zhang Y, Bian H. A comparison of NAFLD and MAFLD diagnostic criteria in contemporary urban healthy adults in China: a cross-sectional study. BMC Gastroenterol. 2022;22(1):471. doi:10.1186/s12876-022-02576-4

3. Sarin SK, Eslam M, Fan JG, Lin HC, George J, Omata M. MAFLD, patient-centred care, and APASL. Hepatol Int. 2022;16(5):1032–1034. doi:10.1007/s12072-022-10408-6

4. Eslam M, Sanyal AJ, George J; International Consensus P. MAFLD: a consensus-driven proposed nomenclature for metabolic associated fatty liver disease. Gastroenterology. 2020;158(7):1999–2014e1991. doi:10.1053/j.gastro.2019.11.312

5. Liang Y, Chen H, Liu Y, et al. Association of MAFLD with diabetes, chronic kidney disease, and cardiovascular disease: a 4.6-year cohort study in China. J Clin Endocrinol Metab. 2022;107(1):88–97. doi:10.1210/clinem/dgab641

6. Li H, Guo M, An Z, et al. Prevalence and Risk Factors of Metabolic Associated Fatty Liver disease in Xinxiang, China. Int J Environ Res Public Health. 2020;17(6):1.

7. Cordova-Gallardo J, Keaveny AP, Qi X, Mendez-Sanchez N. Metabolic associated fatty liver disease and acute-on-chronic liver failure: common themes for common problems. Eur J Gastroenterol Hepatol. 2021;33(1S Suppl 1):e84–e93. doi:10.1097/MEG.0000000000002335

8. Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol. 2015;62(1 Suppl):S47–64. doi:10.1016/j.jhep.2014.12.012

9. Kim D, Kim WR. Nonobese fatty liver disease. Clin Gastroenterol Hepatol. 2017;15(4):474–485. doi:10.1016/j.cgh.2016.08.028

10. Wu AH, Gladden JD, Ahmed M, Ahmed A, Filippatos G. Relation of serum uric acid to cardiovascular disease. Int J Cardiol. 2016;213:4–7. doi:10.1016/j.ijcard.2015.08.110

11. Li C, Hsieh MC, Chang SJ. Metabolic syndrome, diabetes, and hyperuricemia. Curr Opin Rheumatol. 2013;25(2):210–216. doi:10.1097/BOR.0b013e32835d951e

12. Chen LY, Zhu WH, Chen ZW, et al. Relationship between hyperuricemia and metabolic syndrome. J Zhejiang Univ Sci B. 2007;8(8):593–598. doi:10.1631/jzus.2007.B0593

13. Ishizaka N, Ishizaka Y, Toda E, Nagai R, Yamakado M. Association between serum uric acid, metabolic syndrome, and carotid atherosclerosis in Japanese individuals. Arterioscler Thromb Vasc Biol. 2005;25(5):1038–1044. doi:10.1161/01.ATV.0000161274.87407.26

14. Choi HK, Ford ES. Prevalence of the metabolic syndrome in individuals with hyperuricemia. Am J Med. 2007;120(5):442–447. doi:10.1016/j.amjmed.2006.06.040

15. Sirota JC, McFann K, Targher G, Johnson RJ, Chonchol M, Jalal DI. Elevated serum uric acid levels are associated with non-alcoholic fatty liver disease independently of metabolic syndrome features in the United States: liver ultrasound data from the national health and nutrition examination survey. Metabolism. 2013;62(3):392–399. doi:10.1016/j.metabol.2012.08.013

16. Sookoian S, Pirola CJ. The serum uric acid/creatinine ratio is associated with nonalcoholic fatty liver disease in the general population. J Physiol Biochem. 2023;79(4):891–899. doi:10.1007/s13105-022-00893-6

17. Yuan H, Yu C, Li X, et al. Serum uric acid levels and risk of metabolic syndrome: a dose-response meta-analysis of prospective studies. J Clin Endocrinol Metab. 2015;100(11):4198–4207. doi:10.1210/jc.2015-2527

18. Darmawan G, Hamijoyo L, Hasan I. Association between serum uric acid and non-alcoholic fatty liver disease: a meta-analysis. Acta Med Indones. 2017;49(2):136–147.

19. Huang F, Liu A, Fang H, Geng X. Serum uric acid levels in non-alcoholic steatosis patients: a meta-analysis. Asia Pac J Clin Nutr. 2017;26(2):334–342. doi:10.6133/apjcn.092016.04

20. Peng H, Xie X, Pan X, et al. Association of meat consumption with NAFLD risk and liver-related biochemical indexes in older Chinese: a cross-sectional study. BMC Gastroenterol. 2021;21(1):221. doi:10.1186/s12876-021-01688-7

21. Lee YJ, Lee HR, Lee JH, Shin YH, Shim JY. Association between serum uric acid and non-alcoholic fatty liver disease in Korean adults. Clin Chem Lab Med. 2010;48(2):175–180. doi:10.1515/CCLM.2010.037

22. Shih MH, Lazo M, Liu SH, Bonekamp S, Hernaez R, Clark JM. Association between serum uric acid and nonalcoholic fatty liver disease in the US population. J. Formos Med Assoc. 2015;114(4):314–320. doi:10.1016/j.jfma.2012.11.014

23. Lee JM, Kim HW, Heo SY, et al. Associations of serum uric acid level with liver enzymes, nonalcoholic fatty liver disease, and liver fibrosis in Korean men and women: a cross-sectional study using nationally representative data. J Korean Med Sci. 2023;38(34):e267. doi:10.3346/jkms.2023.38.e267

24. Choi J, Joe H, Oh JE, Cho YJ, Shin HS, Heo NH. The correlation between NAFLD and serum uric acid to serum creatinine ratio. PLoS One. 2023;18(7):e0288666. doi:10.1371/journal.pone.0288666

25. Huang XJ, Yin M, Zhou BQ, et al. Impact renaming non-alcoholic fatty liver disease to metabolic associated fatty liver disease in prevalence, characteristics and risk factors. World J Hepatol. 2023;15(8):985–1000. doi:10.4254/wjh.v15.i8.985

26. Liu J, Peng H, Wang C, et al. Correlation between the severity of metabolic dysfunction-associated fatty liver disease and serum uric acid to serum creatinine ratio. Int J Endocrinol. 2023;2023:6928117. doi:10.1155/2023/6928117

27. Han AL, Lee HK. Association of the metabolic dysfunction-associated fatty liver disease with serum uric acid-to-creatinine ratio. Metab Syndr Relat Disord. 2022;20(7):370–376. doi:10.1089/met.2022.0013

28. Sun HL, Pei D, Lue KH, et al. Uric acid levels can predict metabolic syndrome and hypertension in adolescents: a 10-year longitudinal study. PLoS One. 2015;10:e0143786. doi:10.1371/journal.pone.0143786

29. Ogura T, Matsuura K, Matsumoto Y, et al. Recent trends of hyperuricemia and obesity in Japanese male adolescents, 1991 through 2002. Metabolism. 2004;53:448–453. doi:10.1016/j.metabol.2003.11.017

30. King C, Lanaspa MA, Jensen T, et al. Uric acid as a cause of the metabolic syndrome. Contrib Nephrol. 2018;192:88–102.

31. Johnson RJ, Nakagawa T, Sanchez-Lozada LG, et al. Sugar, uric acid, and the etiology of diabetes and obesity. Diabetes. 2013;62:3307–3315. doi:10.2337/db12-1814

32. D’Elia L, Masulli M, Cirillo P, et al. Serum uric acid/serum creatinine ratio and cardiovascular mortality in diabetic individuals-the uric acid right for heart health (urrah) project. Metabolites. 2024;14(3):164. doi:10.3390/metabo14030164

33. Maloberti A, Vanoli J, Finotto A, et al. Uric acid relationships with lipid profile and adiposity indices: impact of different hyperuricemic thresholds. J Clin Hypertens (Greenwich). 2023;25(1):78–85. doi:10.1111/jch.14613

34. Johnson RJ, Bakris GL, Borghi C, et al. Hyperuricemia, acute and chronic kidney disease, hypertension, and cardiovascular disease: report of a scientific workshop organized by the national kidney foundation. Am J Kidney Dis. 2018;71:851–865. doi:10.1053/j.ajkd.2017.12.009

35. Maloberti A, Bombelli M, Facchetti R, et al. Relationships between diuretic-related hyperuricemia and cardiovascular events: data from the URic acid Right for heArt Health study. J Hypertens. 2021;39(2):333–340. doi:10.1097/HJH.0000000000002600

36. Demiray A, Afsar B, Covic A, et al. The role of uric acid in the acute myocardial infarction: a narrative review. Angiology. 2022;73:9–17. doi:10.1177/00033197211012546

37. Maloberti A, Mengozzi A, Russo E, et al. The Results of the URRAH (Uric Acid Right for Heart Health) project: a focus on hyperuricemia in relation to cardiovascular and kidney disease and its role in metabolic dysregulation. High Blood Press Cardiovasc Prev. 2023;30(5):411–425. doi:10.1007/s40292-023-00602-4

38. Casiglia E, Tikhonoff V, Virdis A, et al. Serum uric acid and fatal myocardial infarction: detection of prognostic cut-off values: the URRAH (Uric Acid Right for Heart Health) study. J Hypertens. 2020;38:412–419. doi:10.1097/HJH.0000000000002287

39. Virdis A, Masi S, Casiglia E, et al. Identification of the uric acid thresholds predicting an increased total and cardiovascular mortality over 20 years. Hypertension. 2020;75:302–308. doi:10.1161/HYPERTENSIONAHA.119.13643

40. Copur S, Demiray A, Kanbay M. Uric acid in metabolic syndrome: does uric acid have a definitive role? Eur J Int Med. 2022;103:4–12. doi:10.1016/j.ejim.2022.04.022

41. Nadwa EH, Morcos GNB, Salama NM, et al. Comparing the effects of febuxostat and allopurinol in an animal model of metabolic syndrome. Pharmacology. 2021;106(9–10):564–572. doi:10.1159/000516495

42. Piani F, Agnoletti D, Borghi C. Advances in pharmacotherapies for hyperuricemia. Expert Opin Pharmacother. 2023;24(6):737–745. doi:10.1080/14656566.2023.2197591

43. Suzuki I, Yamauchi T, Onuma M, et al. Allopurinol, an inhibitor of uric acid synthesis--can it be used for the treatment of metabolic syndrome and related disorders? Drugs Today (Barc). 2009;45(5):363–378. doi:10.1358/dot.2009.45.5.1377598

44. He J, Ye J, Sun Y, et al. The additive values of the classification of higher serum uric acid levels as a diagnostic criteria for metabolic-associated fatty liver disease. Nutrients. 2022;14(17):3587. doi:10.3390/nu14173587

45. Yu XL, Shu L, Shen XM, Zhang XY, Zheng PF. Gender difference on the relationship between hyperuricemia and nonalcoholic fatty liver disease among Chinese: an observational study. Medicine (Baltimore). 2017;96(39):e8164. doi:10.1097/MD.0000000000008164

46. Fan N, Zhang L, Xia Z, Peng L, Wang Y, Peng Y. Sex-Specific Association between Serum Uric Acid and Nonalcoholic Fatty Liver Disease in Type 2 Diabetic Patients. J Diabetes Res. 2016;2016:3805372. doi:10.1155/2016/3805372

47. Ito T, Ishigami M, Zou B, et al. The epidemiology of NAFLD and lean NAFLD in Japan: a meta-analysis with individual and forecasting analysis, 1995–2040. Hepatol Int. 2021;15(2):366–379. doi:10.1007/s12072-021-10143-4

48. Guan L, Zhang X, Tian H, et al. Prevalence and risk factors of metabolic-associated fatty liver disease during 2014–2018 from three cities of Liaoning Province: an epidemiological survey. BMJ Open. 2022;12(2):e047588. doi:10.1136/bmjopen-2020-047588

49. DiStefano JK. NAFLD and NASH in postmenopausal women: implications for diagnosis and treatment. Endocrinology. 2020;161:10. doi:10.1210/endocr/bqaa134

50. Döring A, Gieger C, Mehta D, et al. Slc2a9 influences uric acid concentrations with pronounced sex-specific effects. Nat Genet. 2008;40:430–436. doi:10.1038/ng.107

51. Kolz M, Johnson T, Sanna S. et al. Meta-Analysis of 28,141 individuals identifies common variants within five new loci that influence uric acid concentrations. PLoS Genet. 2009;5:e1000504. doi:10.1371/journal.pgen.1000504

52. Borghi C, Fogacci F, Piani F. Not all the eggs and the chickens are the same: the case of uric acid and metabolic syndrome. Eur J Intern Med. 2022;103:36–37. doi:10.1016/j.ejim.2022.07.006

53. Eslam M, Newsome PN, Sarin SK, et al. A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J Hepatol. 2020;73(1):202–209. doi:10.1016/j.jhep.2020.03.039

54. Bansal SK, Bansal MB. Pathogenesis of MASLD and MASH - role of insulin resistance and lipotoxicity. Aliment Pharmacol Ther. 2024;59:S10–S22.

55. Mayén AL, Sabra M, Aglago EK, et al. Hepatic steatosis, metabolic dysfunction and risk of mortality: findings from a multinational prospective cohort study. BMC Med. 2024;22(1):221. doi:10.1186/s12916-024-03366-3

56. Liu CQ, He CM, Chen N, et al. Serum uric acid is independently and linearly associated with risk of nonalcoholic fatty liver disease in obese Chinese adults. Sci Rep. 2016;6:38605. doi:10.1038/srep38605

57. Luukkonen PK, Qadri S, Ahlholm N, et al. Distinct contributions of metabolic dysfunction and genetic risk factors in the pathogenesis of non-alcoholic fatty liver disease. J Hepatol. 2022;76(3):526–535. doi:10.1016/j.jhep.2021.10.013

58. Mazidi M, Katsiki N, Mikhailidis DP, et al. The link between insulin resistance parameters and serum uric acid is mediated by adiposity. Atherosclerosis. 2018;270:180–186. doi:10.1016/j.atherosclerosis.2017.12.033

59. Zhu Y, Hu Y, Huang T, et al. High uric acid directly inhibits insulin signalling and induces insulin resistance. Biochem Biophys Res Commun. 2014;447(4):707–714. doi:10.1016/j.bbrc.2014.04.080

60. Wan X, Xu C, Lin Y, et al. Uric acid regulates hepatic steatosis and insulin resistance through the NLRP3 inflammasome-dependent mechanism. J Hepatol. 2016;64(4):925–932. doi:10.1016/j.jhep.2015.11.022

61. Yu R, Xie W, Peng H, et al. Diagnostic value of triglyceride-glucose index and related parameters in metabolism-associated fatty liver disease in a Chinese population: a cross-sectional study. BMJ Open. 2023;13(9):e075413. doi:10.1136/bmjopen-2023-075413

62. Lanaspa MA, Sanchez-Lozada LG, Choi YJ, et al. Uric acid induces hepatic steatosis by generation of mitochondrial oxidative stress: potential role in fructose-dependent and -independent fatty liver. J Biol Chem. 2012;287(48):40732–40744. doi:10.1074/jbc.M112.399899

63. Castro L, Tórtora V, Mansilla S, Radi R. Aconitases: non-redox Iron-Sulfur Proteins Sensitive to Reactive Species. Acc Chem Res. 2019;52(9):2609–2619. doi:10.1021/acs.accounts.9b00150

64. Xie D, Zhao H, Lu J, et al. High uric acid induces liver fat accumulation via ROS/JNK/AP-1 signaling. Am J Physiol Endocrinol Metab. 2021;320(6):E1032–E1043. doi:10.1152/ajpendo.00518.2020

65. Xu C, Wan X, Xu L, et al. Xanthine oxidase in non-alcoholic fatty liver disease and hyperuricemia: one stone hits two birds. J Hepatol. 2015;62(6):1412–1419. doi:10.1016/j.jhep.2015.01.019

66. Lv Y, Gao X, Luo Y, et al. Apigenin ameliorates HFD-induced NAFLD through regulation of the XO/NLRP3 pathways. J Nutr Biochem. 2019;71:110–121. doi:10.1016/j.jnutbio.2019.05.015

67. Shaker ME. The contribution of sterile inflammation to the fatty liver disease and the potential therapies. Biomed Pharmacother. 2022;148:112789. doi:10.1016/j.biopha.2022.112789

68. Chen S, Chen D, Yang H, et al. Uric acid induced hepatocytes lipid accumulation through regulation of miR-149-5p/FGF21 axis. BMC Gastroenterol. 2020;20(1):39. doi:10.1186/s12876-020-01189-z

69. Su Q, Kumar V, Sud N, Mahato RI. MicroRNAs in the pathogenesis and treatment of progressive liver injury in NAFLD and liver fibrosis. Adv Drug Deliv Rev. 2018;129:54–63. doi:10.1016/j.addr.2018.01.009

70. Mendler MH, Bouillet P, Le Sidaner A, et al. Dual-energy CT in the diagnosis and quantification of fatty liver: limited clinical value in comparison to ultrasound scan and single-energy CT, with special reference to iron overload. J Hepatol. 1998;28(5):785–794. doi:10.1016/S0168-8278(98)80228-6

留言 (0)

沒有登入
gif