Next Article in Journal
Probiotics and Polycystic Ovary Syndrome: A Perspective for Management in Adolescents with Obesity
Previous Article in Journal
Diet Plus Inositols, α-Lactalbumin and Gymnema sylvestre: The Successful Combo to Restore Body Weight and Metabolic Profile in Obese and Dysmetabolic Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Analysis of Metabolites in Gout: A Systematic Review and Meta-Analysis

1
Medical Experimental Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
2
Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2023, 15(14), 3143; https://doi.org/10.3390/nu15143143
Submission received: 21 June 2023 / Revised: 4 July 2023 / Accepted: 11 July 2023 / Published: 14 July 2023
(This article belongs to the Section Clinical Nutrition)

Abstract

:
(1) Background: Many studies have attempted to explore potential biomarkers for the early detection of gout, but consistent and high levels of evidence are lacking. In this study, metabolomics was used to summarize the changes of metabolites in the literature and explore the potential value of metabolites in predicting the occurrence and development of gout. (2) Methods: We searched the databases including the EMBASE, the Cochrane Library, PubMed, Web of Science, VIP Date, Wanfang Data, and CNKI, and the screening was fulfilled on 30 July 2022. The records were screened according to the inclusion criteria and the risk of bias was assessed. Qualitative analysis was performed for all metabolites, and meta-analysis was performed for metabolite concentrations using random effects to calculate the Std mean difference and 95% confidence interval. (3) Results: A total of 2738 records were identified, 33 studies with 3422 participants were included, and 701 metabolites were identified. The qualitative analysis results showed that compared with the healthy control group, the concentration of 56 metabolites increased, and 22 metabolites decreased. The results of the meta-analysis indicated that 17 metabolites were statistically significant. (4) Conclusions: Metabolites are associated with gout. Some specific metabolites such as uric acid, hypoxanthine, xanthine, KYNA, guanosine, adenosine, creatinine, LB4, and DL-2-Aminoadipic acid have been highlighted in the development of gout.

1. Introduction

Gout is a metabolic immune disease characterized by disorders of purine and uric acid (UA) metabolism. It is caused by the super-saturation of UA, which crystallizes in joints, leading to peripheral joint synovitis and severe pain [1,2]. With the increasing number of acute gout attacks, the patient gradually progresses to joint damage, deformity, chronic use-related pain, subcutaneous tophi deposits, and renal damage. Additionally, gout is often associated with metabolic syndrome [3], cerebrovascular diseases [4], cardiovascular diseases [5,6], and renal damage [7]. The prevalence of gout ranges from approximately 1% to 6.8%, with an incidence rate of 0.58 to 2.89 per 1000 persons per year [8]. Moreover, it has doubled in less than 30 years [9]. Studies have shown that gout patients have high healthcare costs due to frequent emergency room visits, hospitalizations, reduced productivity, and disability [10,11,12], which range from USD 172 to USD 6179 [13].
The clinical signs of gout are usually severe pain, edema, redness, and inflammation of the joints, which occur suddenly during the night and then gradually diminish and disappear over a period of days or weeks. Currently, the clinical diagnosis of gout is based on serum uric acid detection, joint synovial fluid examination, and imaging examination [14]. As gout is often acute and easily confused with other forms of arthritis, these diagnostic methods do not seem to be adequately timely and accurate [15], and thus cause patients to suffer from delayed intervention and treatment [16]. UA is a common and important biochemical indicator for the diagnosis of gout, yet not all individuals with hyperuricemia have or will ever develop gout [17,18]. Treatment of gout is divided into acute arthritis treatment and uric acid management. Acute gout is usually treated with high doses of NSAIDs, glucocorticoids, colchicine, and biologic agents over a short period of time. However, NSAIDs may increase the risk of hypertension and acute heart failure [19,20]. Glucocorticoids are associated with potentially serious systemic side effects, including electrolyte disturbances, cardiovascular effects, diabetes, and osteoporosis [21]. Colchicine is widely used in the acute phase of gout [22,23], but the evidence shows that medication adherence to Urate-Lowering Therapy among gout patients was poor worldwide [24,25]. The prevention of acute and chronic gout not only relieves the pain but also reduces associated healthcare costs [26]. Therefore, it is increasingly recognized that early identification and intervention of gout is the most cost-effective path [27].
Gout has a long-term basis of metabolic disorders before the onset. The correlation between metabolism and immunity in the pathogenesis of gout is gradually being noticed [3]. Attention has shifted from simple uric acid metabolites to a variety of other potential metabolites, and exploring changes in metabolites becomes a direction for finding biomarkers [28]. Currently, metabolites have been showing great advantages in disease diagnosis and pathogenesis exploration, providing a new idea for predicting gout [29]. Metabolomics, a platform with powerful spectroscopy and separation techniques, is considered ideal for metabolite determination due to its comprehensiveness and accuracy in metabolite identification [30]. This technique has been used to find biomarkers that predict the development of diseases such as liver cancer, diabetes, osteoarthritis, sepsis, etc. [31,32].
At present, many scholars have attempted to explore the predictive value of metabolites during gout development [33], and some reliable qualitative and quantitative results have been obtained [34]. Some specific metabolites have been confirmed to be related to gout [35,36]. Hypoxanthine and xanthine, metabolic precursors of UA in purine metabolism, have been found to be biomarkers of gout, regardless of whether the patient has hyperuricemia [37,38]. Creatinine clearance, an indicator of kidney function, has been shown to be associated with the early development of subcutaneous urate deposits in gout patients [39]. Methods for the simultaneous measuring of purine, amino acid, creatinine, and other metabolites have been widely developed [40,41]. Thus, metabolites have promising prospects for gout prediction. However, a consistent and comprehensive conclusion is still missing. The aim of this study is to summarize existing studies using metabolomics techniques to detect metabolites in gout patients and to provide comprehensive and reliable conclusions for the prediction of potential biomarkers of gout occurrence.

2. Materials and Methods

Systematic reviews and meta-analyses are reported in accordance with the regulations of the Preferred Reporting Program for Systematic Reviews and Meta-analyses (PRISMA-P) [42]. This study is registered in the International Prospective Register of Systematic Reviews (PROPSERO CRD4022366809).

2.1. Selection Criteria

The inclusion criteria were: (1) gout patients; (2) the samples (blood, urine, feces, and saliva) from patients and healthy controls were measured and analyzed by metabolomics technique; (3) reported the metabolites profile; (4) cohort study, case–control study, and randomized controlled trial based on human. Exclusion criteria were as follows: (1) obtaining blood samples from patients who are taking gout medication or urate-lowering medication; (2) repeated records; (3) lack of information; (4) and no full text.

2.2. Search Strategy

All records were identified by the Cochrane Library, EMBASE, PubMed, Web of Science, VIP Date, Wanfang Data, and CNKI up to 31 July 2022 with the keywords and synonyms combining “gout”, “metabolites”, and “metabolomics” (Supplemental Table S1). No restrictions were placed on the language or date of publication.

2.3. Study Selection

All identified records were downloaded to Endnote X9, and then the duplicates were removed by computer and manual deletion. Two independent researchers (LYY and TJL) screened the studies by title and abstract, respectively, and then the studies meeting our criteria were found in the full text for further screening. Any disagreements were discussed with the third researcher (TJL) until the team reached a consensus. A senior researcher (ZHY) gave the guidance and supervised.

2.4. Risk of Bias Assessment

Two researchers (LYY and HX) assessed the risk of bias in the included records independently by the Newcastle–Ottawa Scale (NOS), a tool for assessing the quality of observational studies [43]. It evaluates research through a “star system” based on selectivity, comparability, and exposure.

2.5. Data Collection

The extracted research data included the following items: first author’s name, publication year, country of origin, study design type, language, age, sample size, diagnostic criteria of patients, statistical methods, detection techniques, metabolomics techniques, and trends and concentrations of metabolites in gout and healthy controls. Data were obtained from the article and the Supplementary Material. When the data is presented graphically only and the original author cannot be contacted for details of the data, we use Web Plot Digitizer (Web Plot Digitizer, V.4.2, San Francisco, California: Ankit Rohatgi, 2019) to extract data from the graph of the article. For those articles with validation cohorts, data from the validation cohorts were also included.
We use PubChem (https://pubchem.ncbi.nlm.nih.gov/ (accessed on 20 January 2023)) to search the metabolites which need to convert their units, and the molecular weight (g/mol) of them are shown in Supplementary Table S2. The quartile and median or interquartile range (IQR) were converted into mean and standard deviation (SD). For median and quartile, we tested the skewness first and applied a new piecewise function based on the size of the sample [36,44,45,46,47,48]. For median and IQR, formula 1 was applied. When a study matches two or more experimental groups to one control group, the mean and standard deviation of the experimental group are combined according to formula 2. All formulas we applied were derived from the Cochrane Handbook of Systematic Reviews of Interventions and are shown in Supplementary Table S3.

2.6. Data Synthesis

A qualitative analysis was performed for changing the direction of metabolites by counting the frequency across the studies. Afterward, the concentrations of metabolites performed the meta-analysis across studies using the standardized mean difference of 46 metabolites (SMD) and confidence intervals of 95% (95% CI). Heterogeneity was deemed substantial if I2 was greater than 60%. While in this study, a random effects model was chosen due to the heterogeneity among the records. Funnel plots and Egger’s test were used to assess publication bias when feasible (10 or more studies) [49,50]. Finally, all data synthesis was performed using R software with a meta package (version 3.6.2).

3. Results

3.1. Literature Search

A total of 2738 records were eventually identified; 902 duplicate records were deleted. Based on the title and abstract screening, 1733 unrelated studies were removed. In the remaining 103 studies, 71 studies were removed for reasons including not providing full text, insufficient data, and failure to meet our criteria. Ultimately, we included 33 studies [28,33,34,35,36,37,44,45,46,47,48,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72] and 3422 participants (2149 gout patients and 1273 healthy controls). The flow diagram for the assessment of studies identified in the systematic review is shown in Figure 1.

3.2. Characteristics of Included Studies

The included records were published between 1999 and 2021 and all of them were case–control designs, among which 18 studies and 15 studies were reported in English and Chinese, respectively. Participants were from the Slovak Republic, the United States, and China. Age and sex were well matched in all studies, except that 4 studies did not report age, and 7 studies only reported age in the gout group. The studies were classified according to different sample types, including 23 blood samples, 2 urine samples, 2 stool samples, 1 saliva study, and 5 hematuria studies [33,36,47,66,71]. All the included studies measured metabolites mostly using liquid chromatography-mass spectrometry (LC-MS) or gouts chromatography-mass spectrometry (GC-MS) techniques. The specific characteristics of each study are shown in Table 1.
A total of 701 metabolites or their ratios in 33 studies were extracted. Of these, 104 metabolites occur twice or more and can be qualitatively synthesized. The 46 metabolites were distributed across 11 studies with accurate concentration values and units for meta-analysis, among which 9 studies [33,36,37,48,54,67,68,71,72] were directly obtained from the article, and 2 studies [44,46] were extracted by software from the box plots. Median and quartiles were transformed to mean and SD in 3 studies [44,46,69], and median and IQR were transformed in 3 studies [44,46,69]. Six studies [36,44,45,46,47,48] combined the mean and SD. The characteristics of these metabolites are shown in Supplementary Table S4.

3.3. Risk of Bias of Included Studies

The risk of bias in all included studies was assessed using the NOS scale. The quality of the included studies was generally high with 22 studies (66.7%) receiving over 7 stars. Five studies received 6 stars due to low exposure scores [28,48,51,53,54]. The remaining 4 studies scored less than 6 stars due to inadequate case definition [45,55,56,62]. Results on the risk of bias in the included studies and details of the evaluation of the included studies are presented in Table 1 and Supplementary Table S5, respectively.

3.4. Qualitative Synthesis

A total of 701 metabolites were identified in 33 studies, of which 567 were in blood samples, 65 were in urine samples, 49 were in fecal samples, and 20 were in saliva samples. Qualitative synthesis was performed on 104 metabolites that were reported more than twice, including 94 blood samples, 5 urine samples, and 1 fecal sample. Based on the sample type classification, 45 metabolites showed increased concentrations, 20 showed decreased concentrations, and 33 showed inconsistent concentrations in blood samples. In urine samples, 2 metabolites showed a decrease in concentration and 3 metabolites showed inconsistent concentrations. In fecal samples, only the concentrations of taurine increased.
In total, 45 metabolites had increased concentrations, 23 had decreased concentrations, and 36 showed inconsistent trends. The details of the qualitative synthesis are given in Table 2. Information about metabolites with inconsistent trends in blood (A) and urine (B) samples is shown in Figure 2.

3.5. Meta-Analysis

A total of 46 metabolites were available for meta-analysis, all of them were derived from blood samples. Among these, 17 metabolites were found to be statistically significant by meta-analysis, while 26 were not. The results showed that patients with gout have increased concentrations of UA, hypoxanthine, xanthine, KYNA, LB4, guanosine, 2-Deoxyadenosine, creatinine, 13(S)-HODE, 9(S)-HODE, 5-oxo-ETE, decreased concentration of 12-HETE, 20-carboxy-ARA, 19,20-DHDPA, 11,12-DHET, DL-2-Aminoadipic acid, and adenosine compared to healthy people. However, high-density lipoprotein, low-density lipoprotein, blood urea nitrogen, 11-HETE, 8-HETE, 14(15)EET, 11(12)EET, 8(9)EET, 5(6)EET, 19(20)EDP, 17,18-DHETE, thromboxinB2, inosine, uracil, linoleic acid, 15-HETE, 5-HETE, 13(S)-HOTrE, 9(S)-HOTrE, 13-oxo-ODE, 9-oxo-ODE, 12(13)EpOME, 9(10)EpOME, 12,13-DHOME, 9,10-DHOME, 14,15-DHET, 8,9-DHET, 5,6-DHET, and thromboxin B3 showed no difference between gout and healthy controls. The forest plots for different metabolites between gout and health and the results of the meta-analysis for different metabolites between gout and health are shown in Figure 3 and Table 3.
Effect estimates were relatively consistent across studies, although substantial heterogeneity was found in some metabolite comparisons. We conducted sensitivity analyses for studies with a high risk of bias. However, the results did not change significantly from previous results. At the same time, due to the omissions in the order of individual studies, no study has greater sensitivity.

4. Discussion

This paper summarized all studies on metabolite changes in gout patients and identified a characteristic metabolite profile for gout. The meta-analysis found 10 metabolites with significant differences and almost all of them were associated with inflammation.
Since hyperuricemia is the largest risk factor for gout, exploring the production and excretion of UA is fundamental to understanding gout. As the degradation product of purine, the main sources of UA are cell turnover, dietary intake, and de novo synthesis [73], and the primary excretion mode for UA is renal excretion [74,75,76]. Previous studies have shown that decreased renal excretion is the main cause of elevated UA, and high levels of UA are strongly associated with kidney disease [77], yet the mechanism is not clear. We aimed to investigate the role of UA and kidney in gout so that we could find reliable evidence to explain the sequence and pathological mechanism. UA induces vascular lesions characterized by endothelial dysfunction, medial wall thickening, and macrophage infiltration, reducing the blood supply to the renal tubular area and causing ischemic injury in the renal tubular area [7,78]. In addition, studies have shown that the cellular communication between renal tubules and immune cells in the tubulointerstitium plays a key role in the progression of kidney disease [79]. In this process, UA seems to accelerate the interaction between these cells, leading to persistent inflammation and progression of tubulointerstitial fibrosis.
In order to better understand the relationship between UA and the kidney, we need to understand the relationship between the two major systems of metabolism and immunity beforehand. From a survival and evolutionary perspective, the most important ability of a species is to resist starvation and mount an effective immune response to pathogens. To maximize energy efficiency, metabolism, and immunity use the same or overlapping key regulatory molecules and signaling systems [80]. In fact, functional units that control critical metabolic and immune functions in higher organisms evolved from a common ancestral structure, meaning that nutrients can act via the pathogen-sensing system to elicit metabolic or nutrient-induced inflammatory responses. In the long history of human existence, faced with a chronically unstable food supply, the human body has automatically evolved a mechanism to store excess calories. However, with the improvement of human living standards, this once beneficial bond leads to excessive accumulation of nutrients and inflammation in the absence of pathogen infection, this activation is clearly harmful, and the disorder of UA metabolism is one of them. Excessive metabolism produces more UA and activates inflammation, which is the main biological process of kidney disease [81,82,83], and meta-inflammation is the mediator to understanding the close relationship between UA and kidney disease [77,84,85]. In summary, we argue that metabolic excesses of energy and lipids lead to hyperuricemia and activate chronic metabolic inflammation. The excretion of high UA from the kidneys is the result of metabolic inflammation, and UA is both a consequence and a precipitant of renal impairment.
Confirmation of the existence of the same signaling pathway associated with UA and inflammation in gout patients is a prerequisite to proving this conjecture. Our study supports this hypothesis by showing creatinine and DL-2-aminoadipic acid were significantly different between gout and healthy controls. As an intermediate metabolite of lysine metabolism, DL-2-aminoadipic acid plays an important role in the regulation of glucose and lipid metabolism, and studies have shown that DL-2-aminoadipic acid enhances the body’s metabolism and associates with damage to renal function in gout patients [46]. Therefore, the increase in DL-2-aminoadipic acid may act as a compensatory mechanism to promote the body to better carry out excess energy and fat consumption. Hypoxanthine has been shown to have the potential to cause renal inflammation in addition to its various pathophysiological consequences in humans [86,87]. As a purine metabolite, adenosine acts primarily on G protein-coupled receptors and plays an anti-inflammatory role in regulating the onset and remission of gout [88,89]. Specifically, it binds to different purinergic receptors to regulate IL-1β secretion, which is involved in the pathogenesis of gout attack [88]. Kynurenic acid (KYNA), as a branch product of the tryptophan metabolism pathway, is involved in the regulation of the cells of the immune system and a variety of immune-mediated diseases [90]. According to the newest research, chronic stress or mild inflammation can promote the production of KYNA and promote various immunomodulatory actions due to KYNA-mediated signaling pathways [91].
Having explored the meta-inflammatory basis of gout flares, we then turn to inflammation in the conventional sense caused by sodium urate crystals. In a vicious cycle of metabolic surplus and inflammation, eventually crystals precipitate and gout arthritis occurs. Sodium urate can promote the secretion of pro-inflammatory cytokines, which act on the kidneys, leading to a further decline in kidney function [66,83]. At the same time, monosodium urate crystals precipitate in the less-perfused joints and then induce an inflammatory response in the immune system [92,93]. Studies by RAE SA et al. have shown that LB4 is an important chemical mediator in acute gout attacks [94]. LB4 stimulates the production of multiple pro-inflammatory cytokines and mediators, and pharmacological data suggest that it could enhance and prolong tissue inflammation [95].
Metabolic inflammation and inflammation induced by sodium urate crystals are the basis for understanding the pathology of gout. Our study also confirmed the importance of inflammation in the pathogenesis of gout. The present study suggests that the signaling pathways used by metabolic inflammation and traditional inflammation are relatively consistent [96], starting from the root of energy metabolism and finding related biomarkers. It may be used as a new idea for early detection and prevention of gout attacks.

5. Limitations of the Study

Our study does have some limitations. (1) The main limitation of the study is that, while the direction of effect estimates is relatively consistent, the results of the meta-analysis do have a high degree of heterogeneity. Heterogeneity is inevitable due to different study populations, different body conditions, and different methods of metabolite detection. (2) Although we use a new method of partition functions in converting sample sizes to means and standard deviations, the distribution of the data is still problematic. In further studies, we hope to better address this issue. (3) There was no search of the clinical trial registry database, which may have resulted in an inadequate comprehensive search.

6. Conclusions

Changes in metabolites in humans do shed light on the underlying mechanisms of gout development. Some specific metabolites such as UA, hypoxanthine, xanthine, KYNA, guanosine, adenosine, creatinine, LB4, DL-2-Aminoadipic acid have been highlighted in the development of gout, establishing the potential of metabolites as predictive biomarkers of gout. However, these findings require further investigation and validation in larger prospective cohort studies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nu15143143/s1, Table S1: Search strategy; Table S2: The molar mass of each bile acid for transforming unit; Table S3: (A) Formula 1 for converting median and interquartile range (IQR) into mean and standard deviation (SD); (B) Formula 2 for combining mean and SD; Table S4: The characteristic for each bile acids; Table S5: The NOS assessment scale for every study.

Author Contributions

Conceptualization: Y.L.; investigation: X.H. and J.T.; methodology: M.J.; project administration: H.Z.; supervision: M.J. and Y.W.; writing—original draft: Y.L. and Z.L.; writing—review and editing: J.T. and X.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Natural Science Foundation of China (Grant number: 82074299) and the China Academy of Chinese Medical Sciences (CACMS) Innovation Fund (Grant number: CI2021A00603).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data reported in this review are all freely available through a database search using the included search terms.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dalbeth, N.; Merriman, T.R.; Stamp, L.K. Gout. Lancet 2016, 388, 2039–2052. [Google Scholar]
  2. Bardin, T.; Richette, P. Definition of hyperuricemia and gouty conditions. Curr. Opin. Rheumatol. 2014, 26, 186–191. [Google Scholar] [CrossRef] [PubMed]
  3. Thottam, G.E.; Krasnokutsky, S.; Pillinger, M.H. Gout and Metabolic Syndrome: A Tangled Web. Curr. Rheumatol. Rep. 2017, 19, 60. [Google Scholar]
  4. Wiseman, S.J.; Ralston, S.H.; Wardlaw, J.M. Cerebrovascular Disease in Rheumatic Diseases: A Systematic Review and Meta-Analysis. Stroke 2016, 47, 943–950. [Google Scholar] [CrossRef]
  5. Cox, P.; Gupta, S.; Zhao, S.S.; Hughes, D.M. The incidence and prevalence of cardiovascular diseases in gout: A systematic review and meta-analysis. Rheumatol. Int. 2021, 41, 1209–1219. [Google Scholar] [CrossRef] [PubMed]
  6. Mhanna, M.; Jabri, A.; Omar, Y.A.; AI-Abdouh, A.; Beran, A.; Ramahi, A.; Alrifai, N.; Almahameed, S.; Altorok, N. The Burden of Cardiac Arrhythmias in Gout: A National Representative Database Study. Curr. Probl. Cardiol. 2023, 48, 101437. [Google Scholar] [CrossRef] [PubMed]
  7. Fan, S.; Zhang, P.; Wang, A.Y.; Wang, X.; Wang, L.; Li, G.; Hong, D.Q. Hyperuricemia and its related histopathological features on renal biopsy. BMC Nephrol. 2019, 20, 95. [Google Scholar] [CrossRef] [Green Version]
  8. Dehlin, M.; Jacobsson, L.; Roddy, E. Global epidemiology of gout: Prevalence, incidence, treatment patterns and risk factors. Nat. Rev. Rheumatol. 2020, 16, 380–390. [Google Scholar] [CrossRef]
  9. Xia, Y.; Wu, Q.; Wang, H.; Zhang, S.; Jiang, Y.T.; Gong, T.T.; Xu, X.; Chang, Q.; Niu, K.J.; Zhao, Y.H. Global, regional and national burden of gout, 1990–2017: A systematic analysis of the Global Burden of Disease Study. Rheumatology 2020, 59, 1529–1538. [Google Scholar] [CrossRef]
  10. Danve, A.; Neogi, T. Rising Global Burden of Gout: Time to Act. Arthritis Rheumatol. 2020, 72, 1786–1788. [Google Scholar] [CrossRef]
  11. Kuo, C.F.; Grainge, M.J.; Mallen, C.; Zhang, W.Y.; Doherty, M. Rising burden of gout in the UK but continuing suboptimal management: A nationwide population study. Ann. Rheum. Dis. 2015, 74, 661–667. [Google Scholar] [CrossRef] [PubMed]
  12. Smith, E.; Hoy, D.; Cross, M.; Merriman, T.R.; Vos, T.; Buchbinder, R.; Woolf, A.; March, L. The global burden of gout: Estimates from the Global Burden of Disease 2010 study. Ann. Rheum. Dis. 2014, 73, 1470–1476. [Google Scholar] [PubMed]
  13. Rai, S.K.; Burns, L.C.; De Vera, M.A.; Haji, A.; Giustini, D.; Choi, H.K. The economic burden of gout: A systematic review. Semin. Arthritis Rheum. 2015, 45, 75–80. [Google Scholar] [CrossRef]
  14. Richette, P.; Doherty, M.; Pascual, E.; Barskova, V.; Becce, F.; Castaneda, J.; Coyfish, M.; Guillo, S.; Jansen, T.; Janssens, H.; et al. 2018 updated European League Agoutinst Rheumatism evidence-based recommendations for the diagnosis of gout. Ann. Rheum. Dis. 2020, 79, 31–38. [Google Scholar] [CrossRef] [Green Version]
  15. Neilson, J.; Bonnon, A.; Dickson, A.; Roddy, E. Gout: Diagnosis and management-summary of NICE guidance. BMJ 2022, 378, o1754. [Google Scholar] [CrossRef]
  16. Zhang, W.Z. Why Does Hyperuricemia Not Necessarily Induce Gout? Biomolecules 2021, 11, 280. [Google Scholar]
  17. Lin, K.C.; Lin, H.Y.; Chou, P. The interaction between uric acid level and other risk factors on the development of gout among asymptomatic hyperuricemic men in a prospective study. J. Rheumatol. 2000, 27, 1501–1505. [Google Scholar]
  18. Schlesinger, N.; Norquist, J.M.; Watson, D.J. Serum urate during acute gout. J. Rheumatol. 2009, 36, 1287–1289. [Google Scholar] [CrossRef]
  19. Theken, K.N. Variability in analgesic response to non-steroidal anti-inflammatory drugs. Prostaglandins Other Lipid Mediat. 2018, 139, 63–70. [Google Scholar] [PubMed]
  20. Whelton, A.; Hamilton, C.W. Nonsteroidal anti-inflammatory drugs: Effects on kidney function. J. Clin. Pharmacol. 1991, 31, 588–598. [Google Scholar] [CrossRef]
  21. Whitehouse, M.W. Anti-inflammatory glucocorticoid drugs: Reflections after 60 years. Inflammopharmacology 2011, 19, 1–19. [Google Scholar] [PubMed]
  22. Marinaki, S.; Skalioti, C.; Boletis, J.N. Colchicine in Renal Diseases: Present and Future. Curr. Pharm. Des. 2018, 24, 675–683. [Google Scholar] [CrossRef] [PubMed]
  23. Pascart, T.; Richette, P. Colchicine in Gout: An Update. Curr. Pharm. Des. 2018, 24, 684–689. [Google Scholar] [CrossRef] [PubMed]
  24. Scheepers, L.; Van Onna, M.; Stehouwer, C.D.A.; Singh, J.A.; Arts, L.C.W.; Boonen, A. Medication adherence among patients with gout: A systematic review and meta-analysis. Semin. Arthritis Rheum. 2018, 47, 689–702. [Google Scholar] [CrossRef]
  25. Pillinger, M.H.; Mandell, B.F. Therapeutic approaches in the treatment of gout. Semin. Arthritis Rheum. 2020, 50, S24–S30. [Google Scholar]
  26. Singh, J.A.; Reddy, S.G.; Kundukulam, J. Risk factors for gout and prevention: A systematic review of the literature. Curr. Opin. Rheumatol. 2011, 23, 192–202. [Google Scholar] [CrossRef] [Green Version]
  27. Wilson, L.; Saseen, J.J. Gouty Arthritis: A Review of Acute Management and Prevention. Pharmacotherapy 2016, 36, 906–922. [Google Scholar] [CrossRef]
  28. Liu, Y.; Yu, P.; Sun, X.; Di, D.L. Metabolite target analysis of human urine combined with pattern recognition techniques for the study of symptomatic gout. Mol. Biosyst. 2012, 8, 2956–2963. [Google Scholar] [CrossRef]
  29. Muthubharathi, B.C.; Gowripriya, T.; Balamurugoutn, K. Metabolomics: Small molecules that matter more. Mol. Omics 2021, 17, 210–229. [Google Scholar]
  30. Bujak, R.; Struck-Lewicka, W.; Markuszewski, M.J.; Kaliszan, R. Metabolomics for laboratory diagnostics. J. Pharm. Biomed. Anal. 2015, 113, 108–120. [Google Scholar] [CrossRef]
  31. Guijas, C.; Montenegro-Burke, J.R.; Warth, B.; Spilker, M.E.; Siuzdak, G. Metabolomics activity screening for identifying metabolites that modulate phenotype. Nat. Biotechnol. 2018, 36, 316–320. [Google Scholar] [CrossRef]
  32. Jin, Q.; Ma, R.C.W. Metabolomics in Diabetes and Diabetic Complications: Insights from Epidemiological Studies. Cells 2021, 10, 2832. [Google Scholar] [CrossRef] [PubMed]
  33. Huang, Y.; Xiao, M.; Ou, J.; Lv, Q.; Wei, Q.J.; Chen, Z.N.; Wu, J.L.; Tu, L.D.; Jiang, Y.T.; Zhang, X.; et al. Identification of the urine and serum metabolomics signature of gout. Rheumatology 2020, 59, 2960–2969. [Google Scholar] [CrossRef] [PubMed]
  34. Shen, X.; Wang, C.; Liang, N.; Liu, Z.; Li, X.D.; Zhu, Z.J.; Merriman, T.R.; Dalbeth, N.; Terkeltaub, R.; Li, C.J.; et al. Serum Metabolomics Identifies Dysregulated Pathways and Potential Metabolic Biomarkers for Hyperuricemia and Gout. Arthritis Rheumatol. 2021, 73, 1738–1748. [Google Scholar] [CrossRef]
  35. Zhong, Z. Preliminary Study on Serum Metabolic Prifiling of Gout Patients Based on UPLC-Q-TOF/MS. Master’s Thesis, Southern Medical University, Guangzhou, China, 2020. [Google Scholar]
  36. Liu, Y. Metabolomics Method Based on High Performance Liquid Chromatography Was Used to Study Biomarkers of Gout. Ph.D. Thesis, University of Chinese Academy of Sciences, Beijing, China, 2013. [Google Scholar]
  37. Zhao, J.-Y. Biomakers Study of Errors of Metabolism by Coupling Chromatography with MS. Master’s Thesis, Tsinghua University, Beijing, China, 2005. [Google Scholar]
  38. Wang, Y.; Deng, M.; Deng, B.; Ye, L.M.; Fei, X.M.; Huang, Z.G. Study on the diagnosis of gout with xanthine and hypoxanthine. J. Clin. Lab. Anal. 2019, 33, e22868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Tarng, D.C.; Lin, H.Y.; Shyong, M.L.; Wang, J.S.; Yang, W.C.; Huang, T.P. Renal Function in Gout Patients. Am. J. Nephrol. 1995, 15, 31–37. [Google Scholar] [CrossRef]
  40. Yuan, Y.; Jiang, M.; Zhang, H.; Liu, J.; Zhang, M.; Hu, P. Simultaneous quantification of urinary purines and creatinine by ultrahigh performance liquid chromatography with ultraviolet spectroscopy and quadrupole time-of-flight mass spectrometry: Method development, validation, and application to gout study. J. Sep. Sci. 2019, 42, 2523–2533. [Google Scholar] [CrossRef] [PubMed]
  41. Feng, J.D.; Yeung, P.K. A simple high-performance liquid chromatography assay for simultaneous measurement of adenosine, guanosine, and the oxypurine metabolites in plasma. Ther. Drug Monit. 2000, 22, 177–183. [Google Scholar] [CrossRef]
  42. Page, M.J.; Mckenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  43. Stang, A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur. J. Epidemiol. 2010, 25, 603–605. [Google Scholar] [CrossRef] [Green Version]
  44. Kang, L.; Liu, J.; Zhang, H.; Jiang, M.; Jin, Y.D.; Zhang, M.; Hu, P. Improved ultra-high performance liquid chromatographic method for simultaneous determination of five gout-related metabolites in human serum. J. Sep. Sci. 2021, 44, 954–962. [Google Scholar] [CrossRef] [PubMed]
  45. Lv, S. Study on the Mechanism of Pulchinenoside b4 Anty-Gouty Arthritis (GOUT) Based on Metabolomics. Ph.D. Thesis, Jiangxi University of Chinese Medicine, Nanchang, China, 2020. [Google Scholar]
  46. Lyu, S.; Rao, Y.; Liu, P.; Yang, S.L.; Chen, W.Y.; Yang, H.J.; Ke, S.K.; Yang, H.O.; He, M.Z.; Feng, Y.L. Metabolomics analysis reveals four biomarkers associated with the gouty arthritis progression in patients with sequential stages. Semin. Arthritis Rheum. 2022, 55, 152022. [Google Scholar] [CrossRef] [PubMed]
  47. Huang, Q.-L. Pricking Blood Therapy of Zhuang Medicine Combined with Medicated Thread Moxibustion in the Treatment of Acute Gouty Arthritis and Metabonomics Study. Master’s Thesis, Guangxi University of Chinese Medicine, Nanning, China, 2014. [Google Scholar]
  48. Chen, J.; Zhou, J.; Wei, S.-S.; Li, H.-C.; Wen, C.-P.; Xu, G.-W. Analysis of Serum Metabolic Characteristics in Patients with Gout Using Gouts Chromatography—Mass Spectrometry. J. Instrum. Anal. 2016, 35, 137–142. [Google Scholar]
  49. Sterne, J.A.; Sutton, A.J.; Ioannidis, J.P.; Terrin, N.; Jones, D.R.; Lau, J.; Carpenter, J.; Rücker, G.; Harbord, R.M.; Schmid, C.H.; et al. Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials. BMJ 2011, 343, d4002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Egger, M.; Davey Smith, G.; Schneider, M.; Minder, C. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997, 315, 629–634. [Google Scholar] [CrossRef] [Green Version]
  51. Qiu, F.-H.; Zhang, H.-Z. Effect of pricking blood therapy on acute gouty arthritis and its effect on serum metabolites. China Medical. Herald. 2018, 15, 135–139+81. [Google Scholar]
  52. Yin, T.-T.; Zhao, C.-J.; Zhang, Y.-Z. Study on substance of damp-heat syndrome of rheumatic disease based on lipidomics. China J. Tradit. Chin. Med. 2013, 28, 210–213. [Google Scholar]
  53. Yin, T.-T.; Zhao, T. Study of gouty arthritis on lipidomics of the human serum. J. Shenyang Pharm. Univ. 2021, 38, 815–820. [Google Scholar]
  54. Yu, X.-F.; Chen, K.-Y.; Tong, Y. Lipid metabolism study of hyperuricemia and gout arthritis based on lipidomics technology. World Latest Med. Inf. 2016, 16, 30–31. [Google Scholar]
  55. Li, Y.-T.; Sang, C.; Liang, D.-D.; Chen, T.-L. A metabolic profiling study on typical arthritis based on Bayesian network. Transl. Med. J. 2019, 8, 232–236. [Google Scholar]
  56. Zhao, T.; Yin, T.-T.; Zhang, Y.-Z.; Lu, J.; Li, P.; Wang, G.-C.; Zu, N.; Yan, X.-P.; Zhang, X.-L. Study on Common Features of Damp-Heat Syndrome in Rheumatism Based on Metabolomics. J. Tradit. Chin. Med. 2013, 54, 592–596. [Google Scholar]
  57. Sun, F.-S. Clinical Obserwation of Zhibai Bixie Dihuang Decoction in the Treatment of Acute Gouty Arthritis and Metabonomics Study. Master’s Thesis, Nanjing University of Chinese Medicine, Nanjing, China, 2019. [Google Scholar]
  58. Wang, M.-J. Biology Characteristics of Spleen-Deficiency Syndrome Gout Based on Faces Metabonomics and Genomics. Master’s Thesis, Zhejiang Chinese Medicine University, Hangzhou, China, 2013. [Google Scholar]
  59. Zhang, M.-M. Serum Metabonomic of Patients with Gout and Hyperuricemia Based on UPLC-MC. Master’s Thesis, Southwest Jiaotong University, Chengdu, China, 2021. [Google Scholar]
  60. Chen, Y.; Ma, H.; Du, Y.; Dong, J.; Jin, C.; Tan, L.; Wei, R. Functions of 1,25-dihydroxy vitamin D3, vitamin D3 receptor and interleukin-22 involved in pathogenesis of gout arthritis through altering metabolic pattern and inflammatory responses. PeerJ 2021, 9, e12585. [Google Scholar] [CrossRef] [PubMed]
  61. Cui, L.; Liu, J.; Yan, X.; Hu, S. Identification of Metabolite Biomarkers for Gout Using Capillary Ion Chromatography with Mass Spectrometry. Anal. Chem. 2017, 89, 11737–11743. [Google Scholar] [CrossRef] [PubMed]
  62. Istok, R.; Kovalancík, M.; Rovenský, J. Total plasma homocysteine in patients with gout. J. Rheumatol. 1999, 26, 2068–2069. [Google Scholar] [PubMed]
  63. Jiang, M.; Chen, T.; Feng, H.; Zhang, Y.; Li, L.; Zhao, A.; Niu, X.; Liang, F.; Wang, M.; Zhan, J.; et al. Serum metabolic signatures of four types of human arthritis. J. Proteome Res. 2013, 12, 3769–3779. [Google Scholar] [CrossRef]
  64. Li, Q.; Wei, S.; Wu, D.; Wen, C.; Zhou, J. Urinary Metabolomics Study of Patients with Gout Using Gouts Chromatography-Mass Spectrometry. Biomed. Res. Int. 2018, 2018, 3461572. [Google Scholar] [CrossRef] [Green Version]
  65. Liu, S.; Wang, Y.; Liu, H.; Xu, T.; Wang, M.; Lu, J.; Guo, Y.; Chen, W.; Ke, M.; Zhou, G.; et al. Serum lipidomics reveals distinct metabolic profiles for asymptomatic hyperuricemic and gout patients. Rheumatology 2022, 61, 2644–2651. [Google Scholar] [CrossRef]
  66. Liu, Y.; Sun, X.; Di, D.; Quan, J.; Zhang, J.; Yang, X. A metabolic profiling analysis of symptomatic gout in human serum and urine using high performance liquid chromatography-diode array detector technique. Clin. Chim. Acta 2011, 412, 2132–2140. [Google Scholar] [CrossRef]
  67. Luo, Y.; Wang, L.; Peng, A.; Liu, J. Metabolic profiling of human plasma reveals the activation of 5-lipoxygenase in the acute attack of gouty arthritis. Rheumatology 2019, 58, 345–351. [Google Scholar] [CrossRef]
  68. Shao, T.; Shao, L.; Li, H.; Xie, Z.; He, Z.; Wen, C. Combined Signature of the Fecal Microbiome and Metabolome in Patients with Gout. Front. Microbiol. 2017, 8, 268. [Google Scholar] [CrossRef] [Green Version]
  69. Stamp, L.K.; Turner, R.; Khalilova, I.S.; Zhang, M.; Drake, J.; Forbes, L.; Kettle, A. Myeloperoxidase and oxidation of uric acid in gout: Implications for the clinical consequences of hyperuricaemia. Rheumatology 2014, 53, 1958–1965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Zhang, Y.; Zhang, H.; Chang, D.; Guo, F.; Pan, H.; Yang, Y. Metabolomics approach by (1) H NMR spectroscopy of serum reveals progression axes for asymptomatic hyperuricemia and gout. Arthritis Res. Ther. 2018, 20, 111. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, Y.Z.; Sui, X.L.; Xu, Y.P.; Gu, F.J.; Zhang, A.S.; Chen, J.H. NLRP3 inflammasome and lipid metabolism analysis based on UPLC-Q-TOF-MS in gouty nephropathy. Int. J. Mol. Med. 2019, 44, 172–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhao, J.; Liang, Q.; Luo, G.; Wang, Y.; Zuo, Y.; Jiang, M.; Yu, G.; Zhang, T. Purine metabolites in gout and asymptomatic hyperuricemia: Analysis by HPLC-electrospray tandem mass spectrometry. Clin. Chem. 2005, 51, 1742–1744. [Google Scholar] [CrossRef] [PubMed]
  73. Dalbeth, N.; Choi, H.K.; Joosten LA, B.; Khanna, P.P.; Matsuo, H.; Perez-Ruiz, F.; Stamp, L.K. Gout. Nat. Rev. Dis. Primers 2019, 5, 69. [Google Scholar] [CrossRef] [PubMed]
  74. Rees, F.; Hui, M.; Doherty, M. Optimizing current treatment of gout. Nat. Rev. Rheumatol. 2014, 10, 271–283. [Google Scholar]
  75. So, A.; Thorens, B. Uric acid transport and disease. J. Clin. Investig. 2010, 120, 1791–1799. [Google Scholar] [CrossRef] [Green Version]
  76. Goldfarb, D.S.; Macdonald, P.A.; Hunt, B.; Gunawardhana, L. Febuxostat in gout: Serum urate response in uric acid overproducers and underexcretors. J. Rheumatol. 2011, 38, 1385–1389. [Google Scholar] [CrossRef]
  77. Jung, S.W.; Kim, S.M.; Kim, Y.G.; Lee, S.H.; Moon, J.Y. Uric acid and inflammation in kidney disease. Am. J. Physiol. Renal. Physiol. 2020, 318, F1327–F1340. [Google Scholar] [CrossRef]
  78. Kang, D.H.; Nakagoutwa, T.; Feng, L.; Watanabe, S.; Han, L.; Mazzali, M.; Truong, L.; Harris, R.; Johnson, R.J. A role for uric acid in the progression of renal disease. J. Am. Soc. Nephrol. 2002, 13, 2888–2897. [Google Scholar] [CrossRef] [Green Version]
  79. Kamijo-Ikemori, A.; Sugoutya, T.; Hibi, C.; Nakamura, T.; Murase, T.; Oikawa, T.; Hoshino, S.; Hisamichi, M.; Hirata, K.; Kimura, K.; et al. Renoprotective effect of the xanthine oxidoreductase inhibitor topiroxostat on adenine-induced renal injury. Am. J. Physiol. Renal. Physiol. 2016, 310, F1366–F1376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Cai, W.; Duan, X.M.; Liu, Y.; Yu, J.; Tang, Y.L.; Liu, Z.L.; Jiang, S.; Zhang, C.P.; Liu, J.Y.; Xu, J.X. Uric Acid Induces Endothelial Dysfunction by Activating the HMGB1/RAGE Signaling Pathway. Biomed. Res. Int. 2017, 2017, 4391920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Kim, S.M.; Lee, S.H.; Kim, Y.G.; Kim, S.Y.; Seo, J.W.; Choi, Y.W.; Kim, D.J.; Jeong, K.H.; Lee, T.W.; Ihm, C.G.; et al. Hyperuricemia-induced NLRP3 activation of macrophages contributes to the progression of diabetic nephropathy. Am. J. Physiol. Renal. Physiol. 2015, 308, F993–F1003. [Google Scholar] [CrossRef] [PubMed]
  82. Xiao, J.; Fu, C.; Zhang, X.; Zhu, D.Y.; Chen, W.J.; Lu, Y.J.; Ye, Z.B. Soluble monosodium urate, but not its crystal, induces toll like receptor 4-dependent immune activation in renal mesangial cells. Mol. Immunol. 2015, 66, 310–318. [Google Scholar] [CrossRef]
  83. Martinon, F.; Petrilli, V.; Mayor, A.; Tardivel, A.; Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006, 440, 237–241. [Google Scholar] [CrossRef] [Green Version]
  84. Fathallah-Shaykh, S.A.; Cramer, M.T. Uric acid and the kidney. Pediatr. Nephrol. 2014, 29, 999–1008. [Google Scholar] [CrossRef]
  85. Ramos, G.K.; Goldfarb, D.S. Update on Uric Acid and the Kidney. Curr. Rheumatol. Rep. 2022, 24, 132–138. [Google Scholar] [CrossRef]
  86. Zhuang, J.; Zhou, X.; Liu, T.; Zhang, S.; Yuan, F.; Zhang, L.F.; Yang, Z.S.; Chen, Y. Astaxanthin attenuated hyperuricemia and kidney inflammation by inhibiting uric acid synthesis and the NF-kappa B/NLRP3 signaling pathways in potassium oxonate and hypoxanthine-induced hyperuricemia mice. Pharmazie 2021, 76, 551–558. [Google Scholar]
  87. Devito, S.; Woodrick, J.; Song, L.; Roy, R. Mutagenic potential of hypoxanthine in live human cells. Mutat. Res. 2017, 803–805, 9–16. [Google Scholar] [CrossRef]
  88. Li, X.; Gouto, J.; Tao, J. Purinergic Signaling in the Regulation of Gout Flare and Resolution. Front. Immunol. 2021, 12, 785425. [Google Scholar] [CrossRef]
  89. Linden, J.; Cekic, C. Regulation of lymphocyte function by adenosine. Arterioscler Thromb. Vasc. Biol. 2012, 32, 2097–2103. [Google Scholar] [CrossRef] [Green Version]
  90. Wirthgen, E.; Hoeflich, A.; Rebl, A.; Günther, J. Kynurenic Acid: The Janus-Faced Role of an Immunomodulatory Tryptophan Metabolite and Its Link to Pathological Conditions. Front. Immunol. 2017, 8, 1957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Kiank, C.; Zeden, J.P.; Drude, S.; Domanska, G.; Fusch, G.; Otten, W.; Schuett, C. Psychological stress-induced, IDO1-dependent tryptophan catabolism: Implications on immunosuppression in mice and humans. PLoS ONE 2010, 5, e11825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Oh, Y.J.; Moon, K.W. Presence of tophi is associated with a rapid decline in the renal function in patients with gout. Sci. Rep. 2021, 11, 5684. [Google Scholar] [CrossRef] [PubMed]
  93. Narang, R.K.; Dalbeth, N. Pathophysiology of Gout. Semin. Nephrol. 2020, 40, 550–563. [Google Scholar] [CrossRef] [PubMed]
  94. Rae, S.A.; Davidson, E.M.; Smith, M.J. Leukotriene B4, an inflammatory mediator in gout. Lancet 1982, 2, 1122–1124. [Google Scholar] [CrossRef]
  95. Crookss, S.W.; Stockley, R.A. Molecules in focus. Leukotriene B4. Int. J. Biochem. Cell Biol. 1998, 30, 173–178. [Google Scholar] [CrossRef]
  96. Hotamisligil, G.S. Inflammation, metaflammation and immunometabolic disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef]
Figure 1. Flow diagram for the assessment of studies identified in the systematic review.
Figure 1. Flow diagram for the assessment of studies identified in the systematic review.
Nutrients 15 03143 g001
Figure 2. The occurrence frequency of metabolites with different concentration trends between gout and health: (a) blood sample; (b) urine sample.
Figure 2. The occurrence frequency of metabolites with different concentration trends between gout and health: (a) blood sample; (b) urine sample.
Nutrients 15 03143 g002
Figure 3. Forest plot for different metabolites between gout and health. Data from references [7,28,33,34,35,36,37,44,45,46,47,48,51,52,53,54,55,56,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72].
Figure 3. Forest plot for different metabolites between gout and health. Data from references [7,28,33,34,35,36,37,44,45,46,47,48,51,52,53,54,55,56,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72].
Nutrients 15 03143 g003aNutrients 15 03143 g003bNutrients 15 03143 g003c
Table 1. The characteristics and NOS rate of the included studies.
Table 1. The characteristics and NOS rate of the included studies.
Author (Year) CountrySample (Gout/Health)Age (Gout/Health) Metabolomics TechniqueNOS
Fanghui Qiu et al. (2018) [51]China10/1046.70 ± 8.69/NGLC-MS6
Tingting Yin et al. (2013) [52] China29/2245.6 ± 13.0/45.7 ± 12.2LC-GC7
Zheng Zhong et al. (2020) [35] China31/3134.72 ± 10.62/34.83 ± 8.16UPLC-Q-TOF/MS7
Yitao Li et al. (2019) [55]China34/6051 ± 12/26 ± 12LC-GC5
Shang Lv et al. (2020) [45]China69/8049.23 ± 17.90/NGUPLC-Hqtof-MS8
Tie Zhao et al. (2013) [56]China29/2245.6 ± 13.0/45.7 ± 12.2UPLC-Q-TOF/MS5
Meijiao Wang et al. (2013) [58] China53/4033.1 ± 8.6/30.6 ± 6.6NMR7
Yun Liu et al. (2013) [36] China21/2048.3 ± 16.7/44.8 ± 13.12HPLC7
Jiao Chen et al. (2016) [48] China29/2650.3 ± 11.4/NGGC-MS6
Mingmei Zhang et al. (2021) [59]China50/5038.61 ± 12.6/NGUPLC-MS7
Xuefeng Yu et al. (2016) [54] China20/15NGUPLC6
Jiyuan Zhao et al. (2005) [37] China49/35NGHPLC-MS-MS4
Tingting Yin et al. (2021) [53] China29/2245.6/18–70UPLC/MS/MS6
Fanshu Sun et al. (2019) [7] China57/9248.26 ± 14.21/46.85 ± 12.50UPLC-MS7
Qilin Huang et al. (2014) [47] China60/3044. 8 ± 8.2/NGGC-MS7
Tiejuan Shao et al. (2017) [68] China26/2643.60 ± 1.98/39.42 ± 2.331H NMR7
Yuqi Chen et al. (2021) [60] China58/2043 ± 15.68/43.0 ± 8.6GC-TOF-MS7
Li Cui et al. (2017) [61] China8/15NGMS/MS/MS7
Yefei Huang et al. (2020) [33] China30/3044.27 ± 12.69/39.93 ± 9.57UPLC-MS7
Kang Lu et al. (2021) [44] China50/15NGUPLC3
Yun Liu et al. (2011) [66] China21/2048.3 ± 16.70/47.4 ± 14.24HPLC-DAD6
Ying Luo et al. (2019) [67] China26/2648.9 (12.8)/51.3 (9.3)LC-MS/MS7
Yun Liu et al. (2012) [28] China45/4143 (20–74) HPLC-DAD7
Shang Lyu et al. (2022) [46] China295/8046.5 ± 15.8/52.1 ± 9.3UHPLC-QTOF-MS-MS8
Yannan Zhang et al. (2018) [70] China49/5045.6 ± 7.3/43.8 ± 11.5H NMR7
Lisa K. Stamp et al. (2014) [69] New Zealand31/2760.6 (40~91)/58.1 (39~79) HPLC7
Zizhang Yan et al. (2019) [71] China30/3049.56 ± 11.78/44.32 ± 11.51UPLC-Q-TOF-MS4
Jiyuan Zhao et al. (2005) [72] China12/35NGHPLC-UV-MS/MS8
Shijia Liu et al. (2022) [65] China183/8851.3 ± 13.8/46.3 ± 15.8UHPLC-Q8
Jiang Miao et al. (2013) [63] China33/6051 (30–69)/34 (25–74) (GC−TOF MS) and (UPLC−QTOF MS) 8
Shen Xia et al. (2021) [34] China109/11943.94 ± 11.88/46.77 ± 10.14LC-MS8
Richard et al. (1999) [62] Slovak28/1850.2 ± 10.3HPLC4
Qianqian Li et al. (2018) [64]China35/2945.3 ± 1.8/43.1 ± 1.6GC-MS8
Notes: LC-MS: liquid chromatography-mass spectrometry; LC-GC: liquid chromatography-gas chromatography; UPLC-Q-TOF/MS: ultra performance liquid chromatography quadrupole time-of-flight mass spectrometry; UPLC-Hqtof-MS: ultra performance liquid chromatography quadrupole time-of-flight mass spectrometry; NMR: nuclear magnetic resonance imaging; HPLC: high performance liquid chromatography; GC-MS: gas chromatograohy mass spectrometry; UPLC-MS: ultra performance liquid chromatography/tandem mass spectrometry; UPLC: ultra performance liquid chromatography; HPLC-MS-MS: high performance liquid chromatography–tandem mass spectrometry; UPLC/MS/MS: ultra performance liquid chromatography/tandem mass spectrometry; 1H NMR: nuclear magnetic resonance; GC-TOF-MS: gas chromatography time-of-flight mass spectrometry; HPLC-DAD: high performance liquid chromatography with diode array detector; UHPLC-QTOF-MS-MS: ultra performance liquid chromatography quadrupole time-of-flight mass spectrometry; GC−TOF MS: gas chromatography–time-of-flight mass spectrometry.
Table 2. The details of the qualitative synthesis.
Table 2. The details of the qualitative synthesis.
Concentration TrendSmall Molecule Metabolite Name
Blood SamplesUrine SamplesFecal Samples
UpwardUric acid, Phenylalanine, Hypoxanthine, Xanthine, Creatinine, Kynurenic acid, Mannose, Mannitol, Leukotriene B4, Leucine, Guanosine, Gluconic acid, Creatine, 13(S)-HODE, 2-deoxyadenosine, 2PY, 5-oxo-ETE, 9(S)-HODE, Acetylornithine, Alanine, Arabitol, Aspartate, Aspartic acid, Blood urea nitrogen, Cis-5,8,11,14,17-eicosapentaenoic acid, Cysteine, D-Gluconic acid, Dihydroxyfumaric acid, Glyceraldehyde, Homoserine, Indoleacetic acid, Isoleucine, Lactic acid, L-Ornithine, Low density lipoprotein, LPC14:0, LPC20:3, LPE18:0, LysoPC(16:0), Malic acid, PE16:0-18:2, PE18:0-18:1, Succinic acid, Thromboxin B2, Valine Taurine
DownwardArachidonic acid, LysoPC(18:2(9Z,12Z)), Lauric acid, Threonate, Stearic acid, High-density lipoprotein, 11-HETE, 8-HETE, 20-carboxy-ARA, 14(15)EET, 11(12)EET, 8(9)EET, 5(6)EET, 19(20)EDP, 19,20-DHDPA, 17,18-DHETE, 11,12-DHET, TAG18:0-18:1-22:1, TAG18:1-20:0-22:1, 12-HETETryptophan, Creatinine
InconsistentInosine, Linoleic acid, Glycerol, Uracil, Tryptophan, Adenosine, Taurine, Dehydroepiandrosterone sulfate, Myristic acid, DL-2-Aminoadipic acid, Urea, Tyramine, Glycine, Oleic acid, Triglyceride, Glycoursodeoxycholic acid, 15-HETE, 5-HETE, 13(S)-HOTrE, 9(S)-HOTrE, 13-oxo-ODE, 9-oxo-ODE, 12(13)EpOME, 9(10)EpOME, 12(13)EpOME, 9(10)EpOME, 12,13-DHOME, 9,10-DHOME, 14,15-DHET, 8,9-DHET, 5,6-DHET, Thromboxin B3, L-Phenylalanine, TAG16:0-16:1-18:1, Choline phosphateUric acid, Hypoxanthine, Guanosine
Notes: HODE: hydroxyoctadecadienoic acid; 2PY: N1-Methyl-2-pyridone-5-carboxamide; oxo-ETE: oxo-eicosatetraenoic acid; LPC: lysophosphatidylcholine; LPE: lysophosphatidylethanolamine; LysoPC: lysophosphatidylcholine; PE: phosphatidylethanolamine; HETE: hydroxyeicosatetrasanoic acid; EET: epoxyeicosatrienoic acid; EDP: epoxydocosapentaenoic acid; DHDPA: dihydroxydocosapentaenoic acid; DHETE: dihydroxyeicosateteaenoic acid; TAG: triacylglycerol; HOTrE: hydroxyoctadecadienoic acid; oxo-ODE: oxo-octadecadienoic acid; EpOME: epoxyoctadecamonoeneoic acid; DHOME: dihydroxyoctadecamonoeneoic acid; DHET: dihydroxyeicosatrienoic acid. The following articles and tables are the same.
Table 3. The results of meta-analysis for different metabolites between gout and health.
Table 3. The results of meta-analysis for different metabolites between gout and health.
Small Molecule MetabolitesStudies for SynthesisSMD95% CII2p-Value
Uric acidJiyuan Zhao et al. (2005) [37]; Jiyuan Zhao et al. (2005) [72]; Yun Liu et al. (2013) [36]; Jiao Chen et al. (2016) [48]; Xuefeng Yu et al. (2016) [54]; Tiejuan Shao et al. (2017) [68]; Zizhang Yan et al. (2019) [71]; Yefei Huang et al. (2020) [33]; Kang Lu et al. (2021) [44]2.27[1.55, 2.99]93%p < 0.00001
Kynurenic acidShang Lv et al. (2020) [45]; Shang Lyu et al. (2022) [46]0.580.36–0.790%p < 0.00001
GuanosineJiyuan Zhao et al. (2005) [37]; Jiyuan Zhao et al. (2005) [72]0.9[0.58, 1.23]0%p < 0.00001
CreatinineTiejuan Shao et al. (2017) [68]; Zizhang Yan et al. (2019) [71]; Yefei Huang et al. (2020) [33]; Kang Lu et al. (2021) [44]1.4[0.96, 1.84]67%p < 0.00001
DL-2-Aminoadipic acidShang Lv et al. (2020) [45]; Shang Lyu et al. (2022) [46]1.45[1.21, 1.69]0%p < 0.00001
AdenosineJiyuan Zhao et al. (2005) [37]; Jiyuan Zhao et al. (2005) [72]; Yun Liu et al. (2013) [36]1.17[0.89, 1.44]0%p < 0.00001
19,20-DHDPAYing Luo et al. (2019) [67]−0.92[−1.35, −0.49]0%p < 0.0001
XanthineJiyuan Zhao et al. (2005) [37]; Jiyuan Zhao et al. (2005) [72]7.27[3.35–11.8]99%p = 0.0003
5-oxo-ETEYing Luo et al. (2019) [67]0.57[0.15, 0.99]0%p = 0.008
Leukotriene B4Ying Luo et al. (2019) [67]0.59[0.17, 1.01]0%p = 0.005
HypoxanthineJiyuan Zhao et al. (2005) [37]; Jiyuan Zhao et al. (2005) [72]; Yun Liu et al. (2013) [36]; Kang Lu et al. (2021) [44]1.02[0.36, 1.69]86%p = 0.002
2-DeoxyadenosineJiyuan Zhao et al. (2005) [37]; Jiyuan Zhao et al. (2005) [72]0.38[0.07, 0.69]0%p = 0.02
13(S)-HODEYing Luo et al. (2019) [67]0.51[0.09, 0.92]0%p = 0.02
9(S)-HODEYing Luo et al. (2019) [67]0.52[0.10, 0.93]0%p = 0.02
11,12-DHETYing Luo et al. (2019) [67]−0.5[−0.91, −0.08]0%p = 0.02
12-HETEYing Luo et al. (2019) [67]−0.73[−1.38, −0.08]56%p = 0.03
20-carboxy-ARAYing Luo et al. (2019) [67]−0.46[−0.87, −0.04]0%p = 0.03
High-density lipoproteinXuefeng Yu et al. (2016) [54]; Zizhang Yan et al. (2019) [71]−1.28[−3.05, 0.48]93%p = 0.15
Low-density lipoproteinXuefeng Yu et al. (2016) [54]; Zizhang Yan et al. (2019) [71]2.37[−0.02, 4.75]95%p = 0.05
Blood urea nitrogenTiejuan Shao et al. (2017) [68]; Zizhang Yan et al. (2019) [71]2.47[−0.63, 5.57]97%p = 0.12
11-HETEYing Luo et al. (2019) [67]−0.49[−1.18, 0.20]62%p = 0.16
8-HETEYing Luo et al. (2019) [67]−0.2[−0.61, 0.21]0%p = 0.34
14(15)EETYing Luo et al. (2019) [67]−0.93[−2.07, 0.22]85%p = 0.11
11(12)EETYing Luo et al. (2019) [67]−0.84[−2.07, 0.39]87%p = 0.18
8(9)EETYing Luo et al. (2019) [67]0.91[−1.95, 0.13]82%p = 0.09
5(6)EETYing Luo et al. (2019) [67]−1.04[−2.53, 0.45]9%p = 0.17
19(20)EDPYing Luo et al. (2019) [67]−0.59[−1.47, 0.28]76%p = 0.18
17,18- DHETEYing Luo et al. (2019) [67]−0.42[−0.83, −0.00]0%p = 0.05
Thromboxin B2Ying Luo et al. (2019) [67]0.25[−0.16, 0.66]0%p = 0.23
InosineJiyuan Zhao et al. (2005) [37]; Jiyuan Zhao et al. (2005) [72]; Yun Liu et al. (2013) [36]; Kang Lu et al. (2021) [44]0.09[−1.07, 1.26]95%p = 0.87
UracilYun Liu et al. (2013) [36]; Shang Lyu et al. (2022) [46]−5.14[−15.12, 4.84]99%p = 0.31
Linoleic acidQilin Huang et al. (2014) [47]; Xuefeng Yu et al. (2016) [54]−0.36[−3.69, 2.97]98%p = 0.83
15-HETEYing Luo et al. (2019) [67]−0.27[−0.91, 0.37]57%p = 0.40
5-HETEYing Luo et al. (2019) [67]−0.28[−2.37, 1.80]85%p = 0.79
13(S)-HOTrEYing Luo et al. (2019) [67]0.95[−0.15, 2.05]66%p = 0.09
9(S)-HOTrEYing Luo et al. (2019) [67]0.42[−0.65, 1.48]84%p = 0.44
13-oxo-ODEYing Luo et al. (2019) [67]−0.31[−2.55, 1.94]96%p = 0.79
9-oxo-ODEYing Luo et al. (2019) [67]−0.15[−2.28, 1.97]96%p = 0.89
12(13)EpOMEYing Luo et al. (2019) [67]0.47[−0.75, 1.70]88%p = 0.45
9(10)EpOMEYing Luo et al. (2019) [67]0.06[−0.92, 1.05]82%p = 0.90
12,13-DHOMEYing Luo et al. (2019) [67]0.26[−0.63, 1.15]78%p = 0.57
9,10-DHOMEYing Luo et al. (2019) [67]0.00[−1.09, 1.09]85%p = 1.00
14,15-DHETYing Luo et al. (2019) [67]−0.04[−1.07, 0.98]83%p = 0.93
8,9-DHETYing Luo et al. (2019) [67]0.42[−0.63, 1.47]83%p = 0.44
5,6-DHETYing Luo et al. (2019) [67]−0.47[−2.42, 1.49]95%p = 0.64
Thromboxin B3 Ying Luo et al. (2019) [67]0.25[−0.41, 0.41]0%p = 0.98
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, Y.; Han, X.; Tong, J.; Wang, Y.; Liu, X.; Liao, Z.; Jiang, M.; Zhao, H. Analysis of Metabolites in Gout: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 3143. https://doi.org/10.3390/nu15143143

AMA Style

Li Y, Han X, Tong J, Wang Y, Liu X, Liao Z, Jiang M, Zhao H. Analysis of Metabolites in Gout: A Systematic Review and Meta-Analysis. Nutrients. 2023; 15(14):3143. https://doi.org/10.3390/nu15143143

Chicago/Turabian Style

Li, Yuanyuan, Xu Han, Jinlin Tong, Yuhe Wang, Xin Liu, Zeqi Liao, Miao Jiang, and Hongyan Zhao. 2023. "Analysis of Metabolites in Gout: A Systematic Review and Meta-Analysis" Nutrients 15, no. 14: 3143. https://doi.org/10.3390/nu15143143

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop