Skip to main content

Association of serum uric acid with prognosis in patients with myocardial infarction: an update systematic review and meta-analysis

A Correction to this article was published on 11 December 2023

This article has been updated

Abstract

Background

The prognostic significance of serum uric acid (SUA) in individuals who have experienced myocardial infarction (MI) remains a subject of academic debate. Thus, the aim of this study was to examine the occurrence of immediate and long-term adverse outcomes in individuals with elevated levels of uric acid (UA) following a diagnosis of MI.

Method

This study conducted a literature search from PubMed, Embase, Web of Science, Medline, Cochrane Library, Emcrae, and Scopus to perform a systematic review and meta-analysis of the prognostic impact of MI with a hyper SUA to assess short-term (30-day or in-hospital) and long-term mortality, the incidence of major adverse cardiovascular events (MACE), and its adverse event rate in relation to SUA. The literature search was conducted up until April 2023. A random effects model and risk ratio (RR) were used as epidemiological indicators. For indicators with low disease rates, treatment intensity was reduced and RR was considered equivalent to odds ratio (OR). Hazard Ratio (HR), RR, and OR extracted from the data were simultaneously subjected to multivariable adjustment for confounding factors. In addition, P values for all original hypotheses were extracted and a meta-analysis was conducted. High SUA was defined as SUA levels equal to or greater than 420 μmol/L (7.0 mg/dL) for males and equal to or greater than 357 μmol/L (6.0 mg/dL) for females. The quality of the literature was evaluated using the Newcastle–Ottawa Scale (NOS).

Results

This comprehensive study included a total of 41 investigations, involving a large sample size of 225,600 individuals who had experienced MI. The findings from the meta-analysis reveal that patients diagnosed with hyperuricemia have significantly increased rates of short-term mortality (RR = 2.14, 95% CI = 1.86, 2.48) and short-term incidence of MACE (RR = 1.94, 95% CI = 1.65–2.11). Furthermore, long-term adverse outcomes, including all-cause mortality (RR = 1.46, 95% CI = 1.40–1.51) and incidence of MACE (RR = 1.43, 95% CI = 1.35–1.52), were also found to be higher in this specific patient population.

Conclusion

Patients diagnosed with MI and elevated SUA levels exhibit a heightened incidence of MACE during their hospital stay. Furthermore, these individuals also experience elevated rates of in-hospital mortality and mortality within one year of hospitalization. However, it is important to note that further randomized controlled trials are necessary to validate and authenticate these findings.

Peer Review reports

Introduction

UA serves as the end product of purine metabolism in thehuman body. Its production is facilitated by the enzyme xanthine oxidase (XO), which not only plays a role in UA synthesis but also acts as a source of reactive oxygen species (ROS) in the cardiovascular system [1, 2]. Xanthine oxidase is an enzyme widely distributed throughout different human body organs [3]. Additionally, the enzyme uricase has undergone genetic mutations in humans, resulting in circulating UA levels 3 to 10 times higher than in other mammals [4].

A correlation has been observed between heightened levels of SUA and various pathological processes, including but not limited to heightened oxidative stress, inflammation, and endothelial dysfunction [5]. Consequently, elevated SUA levels are frequently linked to unfavourable outcomes in diverse cardiovascular conditions [6]. UA undergoes a swift escalation within the tissues, subsequently discharged into the vascular lumen. Upon arrival, a reduction in intracellular pH and reversing negative membrane potential transpires [7]. Studies indicate an elevation in the synthesis of UA and an increase in the activity of xanthine oxidase during instances of myocardial ischemia [8]. Hyperuricemia frequently co-occurs with metabolic syndrome, comprising hypertension, diabetes, dyslipidemia, and obesity, all of which are established risk factors for coronary heart disease (CHD) [9]. Epidemiological studies have provided evidence that elevated UA levels pose a significant risk factor for the development of cardiovascular disease. Moreover, UA has been associated with various forms of cardiovascular disease-related mortality, including acute, subacute, and chronic presentations. As such, heightened levels of UA may serve as a predictive indicator for acute myocardial infarction (AMI) [10]. The relationship between SUA levels and prognostic outcomes in patients with MI remains a topic of debate within the academic community. The uncertain generalizability of existing studies can be attributed to variations in demographics, temporal factors, and spatial-geographical considerations across different research endeavors. In light of this, the current investigation aimed to address this gap by conducting a comprehensive evaluation and statistical analysis to examine the differences in short- and long-term prognostic consequences between patients with MI who have hyperuricemia and those with normouricemia..

Methods

In accordance with the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-analyses) guidelines, a systematic review and meta-analysis were conducted [11]. This study was initially registered on the “PROSPERO” platform (Official Register Number: CRD42023415647).

Eligibility criteria

We incorporated the PECOs (Population, Exposure, Comparator, Outcome, and Study) criteria to ensure clarity and appropriate study selection. As this study does not involve a randomized controlled trial (RCT), it is crucial to establish well-defined and detailed inclusion criteria to guide the analysis effectively.

The population under investigation encompassed individuals with MI, specifically AMI, ST-segment elevation MI (STEMI), non-ST-segment elevation MI (NSTEMI), silent MI (SMI), and prior chronic ischemic myocardial injury (PCIMI). The included studies examined individuals with varying levels of SUA as the exposure of interest. Comparative analyses were conducted among different SUA levels within the specified population.

The focus of the analysis was on short- and long-term prognostic outcomes associated with MI. To ensure the inclusion of appropriate studies, cohort studies or other similar designs were considered as the preferred study design. By utilizing the PECOs framework, the revised approach provides clear and organized inclusion criteria, addressing any ambiguity regarding eligible study designs.

It is important to note that the inclusion criteria did not impose temporal or spatial constraints, allowing for a comprehensive evaluation of relevant literature without limitation to specific regions or time periods. The findings were based on previously published studies, selected based on predefined ethical considerations to protect participant identity.

This study aims to offer valuable insights to medical professionals, helping them improve patient outcomes and enhance overall quality of life. To maintain methodological rigor, systematic reviews, case reports, editorials, and policy documents were excluded from the analysis. Furthermore, only English-language papers were considered for inclusion in this review, ensuring consistency and facilitating a thorough examination of the selected literature.

Information sources

To conduct a comprehensive search, we examined electronic bibliographic databases including PubMed, Embase, Web of Science, Medline, Cochrane Library, Emcrae, and Scopus. Additionally, we reviewed the reference sections of relevant publications to identify any supplementary studies that were not initially captured through the aforementioned search approach.

Search strategy

On 1st Mar. 2023, we searched for medical literature using MeSH in PubMed and Emtree in Embase, and free-text phrases. We adapted headings and wildcards to meet search requirements of different databases. On 1st Apr. 2023, we launched another search for new literature. And we used the “Covidence” for reference management, removing duplicates automatically and manually. The detailed search strategy is in Supplementary Information 1.

Study selection

Author Jiacheng-Rong and Lei-Huang screened the article titles and abstracts and submitted any content that could not be determined to author Cheng-Fang and Xudong-Chen for joint discussion. For full-text screening, author Lei-Huang recorded any article with uncertain eligibility; the ultimate inclusion was decided jointly with authors Jiacheng-Rong and Cheng-Fang.

Data extraction

Data from all included studies were extracted into standardized tables in Microsoft Excel Mac Edition (Microsoft Corp., Redmond, WA) (Supplementary Information 2). The following evaluation indicators were extracted from the included studies: 1. publication data: (i) first author's surname and (ii) year of publication; 2. primary statistical data: (i) study site, (ii) study design, (iii) sample size, (iv) patient type, (v) year of follow-up; 3. epidemiological data: a (i) number of exposed versus unexposed, (i i) statistical parameters of outcome: OR, HR, RR, P value, 95% confidence interval (CI). The original authors were contacted for additional data for articles without precise results or with some missing results. Data extraction tables and their contents were confirmed by three authors (Jiacheng-Rong, Lei-Huang and Cheng-Fang).

Definition of the various outcome indicators

  1. (1)

    Short-term outcome indicators:

Outcome indicators that occur within 30 days of diagnosis of MI and admission to treatment, including:

  1. i.

    death within 30 days, including in-hospital and cardiac deaths.

  2. ii.

    MACE within 30 days, including non-fatal stroke, non-fatal myocardial infarction (NFMI), and cardiovascular death (CD) [12].

  3. iii.

    Other adverse indicators occurring within 30 days, including atrial fibrillation (AF), acute kidney injury (AKI), myocardial infarction recurrence (RMI), complete atrioventricular (AV) block, coronary artery disease (CAD), coronary prevalence (CP), cardiogenic shock (CS), heart failure (HF), impaired renal function (IRF), major bleeding, renal failure (RF), target vessel revascularization (TVR), ventricular arrhythmia (VR) and Angina pectoris (AP).

  4. iv.

    Long-term outcome indicators:

Indicators of adverse outcomes occurring more than 30 days after diagnosis of MI and admission to follow-up, including.

  1. i.

    Death after more than 30 days of follow-up: including all-cause death, cardiac death.

  2. ii.

    MACE after more than 30 days of follow-up.

  3. iii.

    other adverse indicators after 30 days of follow-up, AF, AKI, RMI, complete AV block, CAD, CP (refers to the occurrence or prevalence of CAD within a population), CS, HF, IRF, major bleeding, RF, TVR, VR and AP.

Definition of high SUA

Hyperuricemia was defined as a SUA level equal to or greater than 420 μmol/L (7.0 mg/dL) in males and equal to or greater than 357 μmol/L (6.0 mg/dL) in females [13].

Data Extraction for cohort studies

This review scrutinized the availability of independent data for various cohort time points in the studies under consideration. Following the exclusion of data from duplicate reports, wherein reports of the same cohort in multiple studies were eliminated, the distinct cohort time points were regarded as independent studies in this review.

Study quality assessment

In this study, the quality of literature for the included studies was assessed using The NOS. As all the literature included in this study consisted of cohort studies, the NOS Cohort Study Scale was employed. The scale encompasses three components: population selection, comparability, and exposure assessment (also known as outcome assessment). Overall, a total of eight entries achieved a score of nine on the scale, indicating high quality based on the assessment criteria [14].

Synthesis of results

We divide prognostic indicators into short- and long-term indicators based on follow-up duration. To combine data on outcome indicators for hyperuricaemia and normouricaemia, we use a random effects model and RR as the epidemiological indicator. When the articles provide direct epidemiological effect measures, we use a logarithmic approach. To address the varying levels of evidence, HR, RR, and OR are analyzed separately in this study. This approach is adopted considering their distinct interpretations and applications. In instances where the indicators have low prevalence, treatment effects are downscaled, and RR is equated to OR to ensure comparability.

The extracted HR, RR, and OR values were subjected to a multifactorial treatment of confounders. This approach involves considering and adjusting for various factors that may influence the association between the exposure and outcome variables. By accounting for these confounders, the analysis aims to control for their potential impact on the observed associations, thus enhancing the validity and reliability of the results.

In order to address the original hypothesis, all P-values pertaining to the same hypothesis were extracted and subjected to a meta-analysis.. The binomial data model, which falls under the generalized linear mixed model, necessitates a substantial amount of computations due to the high-dimensional integration of random effects [15,16,17], so DerSimonian and Laird random effects models [18] were utilized in a meta-analysis was conducted to determine the combined prevalence and corresponding 95%CI. The statistical heterogeneity between trials was assessed through employment of the X2 test, while the variance resulting from statistical heterogeneity was determined using the I2 statistic. The I2 values corresponding to 25%, 50%, and 75% were categorized as low, medium, and high estimates, respectively. The statistical analysis was conducted utilizing the Stata Statistical Software: Release 17 MP version (StataCorp LP, College Station, TX, USA).

Assessing publication or reporting bias

We employed funnel plots and conducted Begg's and Egger's tests to evaluate publication bias. However, it is crucial to consider that Cochrane guidelines caution against using funnel plots when the number of included studies is less than 10. In this analysis, the limited number of studies poses a challenge in accurately determining the true effect beyond random chance.

To address the potential publication bias, the research implemented trimming and padding methodologies. These approaches were applied specifically in cases where the results of Begg's or Egger's tests indicated significance with a p-value less than 0.05. By utilizing these techniques, the study aimed to mitigate the impact of publication bias on the overall findings and enhance the reliability of the results.

Results

Study selection

The initial database search yielded a total of 1002 articles, along with an additional 33 cited literature sources. After the initial review, 605 articles were identified as potentially relevant and underwent further evaluation. Out of these, 97 articles were deemed eligible for full-text analysis. Following a comprehensive screening process, 55 articles were excluded based on predefined criteria. Ultimately, a total of 35 papers (referenced as papers [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]) containing 41 cohorts were included in the study (see Fig. 1). For a detailed list of the excluded studies and the rationale behind their exclusion, please refer to Supplementary Information 3. The study encompassed a total of 41 analyses conducted on a collective sample size of 225,600 patients who had experienced MI. These analyses included two cohort studies, which further contributed to the comprehensive examination of the research question at hand. The substantial number of patients included in these analyses provides a robust foundation for drawing valuable insights and conclusions related to MI.

Fig. 1
figure 1

A flow chart depicting the process of study selection along with the rationales for exclusion

The study encompasses a diverse range of geographic locations, with contributions from 18 distinct countries and regions, such as China, Croatia, the Czech Republic, Greece, India, Indonesia, Italy, Japan, Mexico, Norway, Poland, South Africa, and South Korea. The countries under consideration are Switzerland, Taiwan (region), Turkey, and the United States of America. All individuals who took part in the research were diagnosed patients with MI. The subjects were categorized into four distinct groups based on the type of MI they experienced, namely AMI, STEMI, NSTEMI, and SMI (Fig. 2).

Fig. 2
figure 2

Regional and population characteristics of the included studies

NOS study quality assessment results

A total of 25 articles had an overall NOS score of 5 or more, which is of medium and above literature quality. An additional 16 articles were below the quality that falls under the average quality score. Overall, the quality of the included literature is in ideal condition (Fig. 3).

Fig. 3
figure 3

NOS study quality assessment and overall score heat plot

Basic information about the included literature

Two hundred twenty-five thousand six hundred MI patients from 18 different nations and regions were enrolled in 41 studies that made up this review (Supplementary Information 4). The prognostic markers for heart attacks were divided into short-term prognosis and long-term prognosis in this study. There was a total of 19 studies that offered short-term (follow-up 30 days or follow-up outcome occurred in the hospital) outcome indicators, and 37 research that provided long-term (follow-up > 30 days or follow-up outcome happened outside the hospital) outcome data. The outcome signs that occurred most frequently were ACM, MACE, Death, Stroke, and HF.

Meta-analysis of short-term adverse events

The present study aims to investigate the mortality rate in patients diagnosed with MI and hyperuricemia

A meta-analysis was conducted to compare the 30-day mortality rates between patients with MI and high blood UA levels versus those with normal levels. The study yielded noteworthy results, indicating that patients diagnosed with both MI and hyperuricemia had a mortality rate 2.14 times higher (RR = 2.14, 95% CI = 1.86–2.48) than individuals with normouricemia who also experienced MI. This finding is visually represented in Fig. 4a. Separate meta-analytic procedures were employed to analyze HR and RR reported by multiple studies. The research outcomes revealed that individuals with MI and hyperuricemia had a higher frequency of fatal events during the post-treatment phase compared to those with normouricemia and MI. Specifically, the HR for this group was found to be 1.27 (95% CI = 1.01–1.45), as depicted in Fig. 4b. Moreover, the study identified an OR of 1.41 (95% CI = 0.97–1.86) for mortality among patients diagnosed with both MI and hyperuricemia, in comparison to those with normal UA levels and MI, illustrated in Fig. 4c.

Fig. 4
figure 4

Forest plot of survival in patients with MI with hyperuricemia and normocytic acid blood levels

The incidence of MACE in individuals diagnosed with hyperuricemia and MI has been investigated

In this study, a meta-analysis was conducted to compare the occurrence of MACE in patients diagnosed with hyperuricemia and MI, as opposed to those with normal UA levels and MI. The results revealed that patients diagnosed with both MI and hyperuricaemia had a 1.91-fold increased risk of experiencing MACE, in comparison to individuals diagnosed with MI and normouricemia. The calculated relative risk (RR) was 1.91, accompanied by a 95% CI ranging from 1.62 to 2.11. These findings are visually represented in Fig. 5.

Fig. 5
figure 5

Forest plot of MACE occurrence in patients with MI with hyperuricemia and normocytic acid blood levels

Meta-analysis of patients with MI presenting with other cardiovascular adverse outcomes at different blood UA levels

In order to examine the frequency of short-term cardiovascular adverse events, a meta-analysis was conducted. The findings indicate that patients diagnosed with both MI and hyperuricemia are at a heightened risk of experiencing adverse events such as unstable AP compared to those with normal UA levels. The RR was determined to be 1.66, with a 95% CI of 1.29–2.15. Additionally, individuals with MI and hyperuricemia have a significantly higher susceptibility to developing cardiac surgery (CS) in comparison to those with normal UA levels (RR = 2.27, 95% CI = 2.27–4.29). The study also found that patients diagnosed with both hyperuricemia and MI had a significantly increased likelihood of experiencing HF within a 30-day observation period when compared to individuals without hyperuricemia. The relative risk for this group was 2.20, with a 95% CI ranging from 1.93 to 2.51. Furthermore, compared to individuals with normal UA levels during MI, those with hyperuricemia were more prone to experiencing recurrent MI (RR = 1.49, 95% CI = 1.06–2.10). Moreover, the patient cohort under consideration demonstrated a 3.20-fold increase in the likelihood of encountering a stroke (95% CI = 1.90–5.37) compared to patients with normal UA levels during MI (refer to Fig. 6 for detailed illustration).

Fig. 6
figure 6

Forest plot of other short-term cardiovascular adverse outcomes

Meta-analysis of patients with MI presenting with non-cardiovascular adverse outcomes at different blood UA levels

The study findings indicate that individuals diagnosed with MI and hyperuricemia exhibited an increased likelihood of experiencing MB (RR = 1.67, 95% CI. 36–20.5), RF (RR = 1.61, 95% CI = 1.29–2.02), and AKI (RR = 21.44, 95% CI = 6.02–76.36) (detailed in Fig. 7).

Fig. 7
figure 7

Forest plot of non-cardiovascular adverse outcomes at different blood UA levels

Meta-analysis of Long-term adverse events

Association of Hyperuricemia with MI and ACM

A meta-analysis was conducted to evaluate the association between UA levels and ACM outcomes in patients diagnosed with MI. The study's findings indicate that patients with hyperuricemia have a higher likelihood of experiencing an ACM death compared to those with normal blood UA levels who have suffered from MI (Fig. 8a). A meta-analysis of studies that provided HRs also supports this conclusion, showing that the probability of experiencing an ACM outcome in hyperuricemia with MI is significantly greater than in those with normal UA levels (Fig. 8b).

Fig. 8
figure 8

a ACM at different blood UA levels. b ACM at different blood UA levels (combine HR)

Association of Hyperuricemia with MI and MACE

The study findings indicate that the likelihood of MACE in individuals with hyperuricemia and MI was 1.43 times greater than in those with MI and normal blood UA levels (RR = 1.43, 95% CI = 1.35–1.52) (as illustrated in Fig. 9).

Fig. 9
figure 9

MACE at different blood UA levels

Meta-analysis of patients with MI presenting with other cardiovascular adverse outcomes at different blood UA levels

A meta-analysis was carried out with the aim of examining further long-term negative cardiovascular event outcomes. The research results suggest that there is a greater propensity for the onset of HF in individuals afflicted with hyperuricemia and MI, as opposed to those with normal levels of UA. The study found a RR of 1.87 and 95% CI of 1.66–2.10 for MI patients with hyperuricemia, showing a 1.23-fold increase in subsequent MI risk compared to those with normal UA levels (RR 1.23, 95% CI 1.11–1.37). These patients also had a 1.55-fold increase in stroke risk (RR 1.55, 95% CI 1.28–1.87). Patients who have been diagnosed with both MI and hyperuricemia are at a slightly higher risk of experiencing a TVR compared to those with normal UA levels. Specifically, there is a 1.04-fold increase in likelihood, with a 95%CI ranging from 0.94 to 1.14. Individuals who have been diagnosed with both MI and hyperuricemia have a higher probability of experiencing additional unfavorable cardiovascular events, with a 1.29-fold increase in comparison to those who have MI and normal UA levels. This conclusion is based on the statistical analysis of data, with a relative risk of 1.29 and a 95%CI ranging from 1.22 to 1.37, as illustrated in Fig. 10.

Fig. 10
figure 10

Forest plot of other long-term cardiovascular adverse outcomes at different blood UA levels

Sensitivity and publication bias analysis

After sensitivity analysis using an exclusion-by-exclusion approach, we found no significant change in the long-term and short-term adverse outcomes. We also excluded articles with NOS quality scores below 5; no significant differences were observed. These minor changes suggest that the meta-analysis is robust and reliable. We also performed tests for publication bias with all studies having a p-value of > 0.05 for Egger test. For some studies with results close to 0.05, we found only weak changes in the results after using trimming and padding methods, indicating that the meta-analysis was robust (Supplementary information 5).

Discussion

The existing epidemiological research indicates that elevated levels of SUA pose a noteworthy risk for cardiovascular disease, wherein oxidative stress assumes a quite critical pathophysiological function. In addition, research has also demonstrated that the utilization of a xanthine oxidoreductase inhibitor can effectively reduce SUA concentrations and provide safeguarding advantages in scenarios characterized by oxidative stress, such as instances of ischemia–reperfusion injury and cardiovascular disease [54,55,56]. The detection of UA may precede that of other cardiac markers, including cardiac troponins. Spectral unipolar atrial electrograms (SUA) have been identified as a viable early indicator of myocardial ischemia [57, 58]. As such, it has been deemed an efficacious approach for forecasting the concomitant occurrence of MI and troponins.

Prior research has investigated the prognostic properties of relevant biomarkers associated with atherosclerotic cardiovascular disease [59,60,61]. The clinical utility of SUA in patient assessment is apparent, however, the impact of SUA on drug therapy and prognosis enhancement is yet to be fully elucidated [57]. Following a comprehensive examination and statistical analysis of 41 studies, our findings indicate that hyperuricemia is associated with a higher likelihood of short-term adverse outcomes compared to long-term adverse outcomes in individuals diagnosed with MI. The study found a significant increase in risk of short-term (2.14x) and long-term all-cause mortality (1.47x) in MI patients with elevated SUA levels. These patients also exhibited a higher frequency of MACE over both short and long-term periods. Hyperuricemia impacts cardiovascular prognosis and risks. The meta-analysis RR's 95%CI do not encompass non-significant values. The elevated SUA levels have an impact on future mortality rates, MACE occurrence, and other unfavorable cardiovascular outcomes in MI patients.

The meta-analysis exhibits some constraints. Initially, it should be noted the absence of a specialized randomized controlled trial for this investigation necessitated the utilization of solely observational studies for data extraction, which may potentially introduce a risk of associated bias. Despite the extraction of adjusted epidemiological effect values for RR and HR and the subsequent meta-analysis of these values, it is crucial to recognise that a limited quantity of residual confounding factors may exist. Furthermore, it is improbable that the methodology employed in every study was entirely uniform. The studies exhibited variability with regards to factors such as age range, gender distribution, and medical history of the participants. Thus, it should be noted that this meta-analysis has an inherent limitation due to the possibility of heterogeneity among the included studies. And the HSUA cut-off values exhibited variability across the studies, ranging from 5.4 mg/dL to 7.5 mg/dL. However, the majority of the studies (10 out of 13) employed HSUA cut-off levels within the range of 6 mg/dL to 7 mg/dL. All possible endeavours were undertaken to evaluate the cumulative impact by excluding research studies where the HSUA threshold varied significantly from that of other studies.

The present investigation exhibits certain notable strengths. The meta-analysis incorporated studies with considerable sample sizes. A systematic evaluation is required to appraise the quality of individual studies. Nonetheless, there exists a lack of consensus regarding the most suitable approach for evaluating the quality of a study. The decision was made to evaluate the aforementioned studies utilising the Newcastle–Ottawa Scale (NOS), a tool that encompasses three crucial components for the evaluation of cohort studies. The adverse events were classified into short-term and long-term categories, and an effort was made to encompass nearly all conceivable adverse events, rendering this investigation a preeminent and all-encompassing reference.

The study has several notable strengths. Firstly, a comprehensive and extensive search strategy was employed, encompassing both primary articles and relevant cited literature, thus ensuring a thorough exploration of the topic. Additionally, the inclusion of a large sample size comprising 225,600 patients enhances the statistical power and generalizability of the findings. The incorporation of cohort studies further contributes valuable longitudinal data, providing insights into the long-term effects and outcomes related to MI.

However, there are also certain limitations to consider. Despite efforts to address publication bias through methods such as funnel plots and statistical tests, the limited number of included studies may still introduce some uncertainty regarding the true effect beyond random chance. Heterogeneity among the included studies is another potential limitation that could impact the overall results. Conducting subgroup analyses or sensitivity analyses could have helped explore this further. Additionally, variations in data quality across different studies could influence the reliability and consistency of the results.

Comparing our study to other similar meta-analyses, we stand out due to our significantly large sample size and the inclusion of cohort studies. These factors enhance the robustness and reliability of our assessment of MI. However, it is important to acknowledge that other meta-analyses may have addressed certain limitations, such as publication bias, in more comprehensive ways by including a greater number of studies. Moreover, differences in the research question, inclusion criteria, and methodology across studies can lead to variations in results and conclusions. Therefore, a collective consideration of the findings from multiple meta-analyses is crucial for a comprehensive understanding of the topic.

Conclusion

The study provides evidence that patients with MI and elevated SUA levels have higher rates of short and long-term mortality, MACE, and other unfavorable outcomes. These findings suggest that SUA may play a role in the progression of atherosclerosis and could serve as a novel prognostic indicator for monitoring individuals following AMI. Incorporating SUA as a prognostic metric in clinical practice has the potential to significantly improve risk stratification and enhance the precision of existing techniques. This approach could lead to more effective medical interventions, reduced healthcare costs, and improved patient well-being by lowering readmission rates and medical expenses. It is important to note that ethical approval or patient consent was not required for this study as it relied on previously published research.

Availability of data and materials

All data generated or analyzed during this study are extracted from published papers. And all data generated or analyzed during this study are included in this published article [and its supplementary information files].

Change history

References

  1. Demiray A, et al. The role of uric acid in the acute myocardial infarction: a narrative review. Angiology. 2022;73:9–17.

    CAS  PubMed  Google Scholar 

  2. Kanbay M, et al. Uric acid in metabolic syndrome: From an innocent bystander to a central player. Eur J Intern Med. 2016;29:3–8.

    CAS  PubMed  Google Scholar 

  3. Kanbay M, Solak Y, Dogan E, Lanaspa MA, Covic A. Uric acid in hypertension and renal disease: the chicken or the egg. Blood Purif. 2010;30:288–95.

    CAS  PubMed  Google Scholar 

  4. Mazzali M, et al. Uric Acid and Hypertension: Cause or Effect? Curr Rheumatol Rep. 2010;12:108–17.

    CAS  PubMed  Google Scholar 

  5. Kelkar A, Kuo A, Frishman WH. Allopurinol as a cardiovascular drug. Cardiol Rev. 2011;19:265.

    PubMed  Google Scholar 

  6. Wang H, Yang J, Sao J, Zhang J, Pang X. The prediction of cardiac events in patients with acute ST segment elevation myocardial infarction: a meta–analysis of serum uric acid. Open Life Sci. 2018;13:413–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Improvement of Cardiac Function by Allopurinol in Patients... : Journal of Cardiovascular Pharmacology. LWW. 2023. [cited 2023 Sep 30]. Available from: https://journals.lww.com/cardiovascularpharm/abstract/1995/01000/improvement_of_cardiac_function_by_allopurinol_in.19.aspx.

  8. Kroll K, Bukowski TR, Schwartz LM, Knoepfler D, Bassingthwaighte JB. Capillary endothelial transport of uric acid in guinea pig heart. Am J Physiol Heart Circ Physiol. 1992;262:H420–31.

    CAS  Google Scholar 

  9. Car S, Trkulja V. Higher serum uric acid on admission is associated with higher short-term mortality and poorer long-term survival after myocardial infarction: retrospective prognostic study. Croat Med J. 2009;50:559–66.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang J, Chen Y, Wang C, Zhu X. Correlation of serum uric acid levels with coronary flow in patients with ST-segment elevation myocardial infarction undergoing primary coronary intervention. Zhonghua Yi Xue Za Zhi. 2012;92:3100–3.

    CAS  PubMed  Google Scholar 

  11. Moher D, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev. 2015;4:1.

    PubMed  PubMed Central  Google Scholar 

  12. Major Adverse Cardiac Event - an overview | ScienceDirect Topics. https://www.sciencedirect.com/topics/nursing-and-health-professions/major-adverse-cardiac-event.

  13. Barkas F, Elisaf M, Liberopoulos E, Kalaitzidis R, Liamis G. Uric acid and incident chronic kidney disease in dyslipidemic individuals. Curr Med Res Opin. 2018;34:1193–9.

    CAS  PubMed  Google Scholar 

  14. Ottawa Hospital Research Institute. https://www.ohri.ca/programs/clinical_epidemiology/oxford.asp.

  15. Geert Molenberghs, Verbeke G, Garcia C, Vieira C. A family of generalized linear models for repeated measures with normal and conjugate random effects. Stat Sci. 2010;25(3). [cited 2023 Sep 30] Available from:https://projecteuclid.org/journals/statistical-science/volume-25/issue-3/A-Family-of-Generalized-Linear-Models-for-Repeated-Measures-with/10.1214/10-STS328.full.

  16. Engel B, Keen A. A simple approach for the analysis of generalizea linear mixed models. Stat Neerl. 1994;48:1–22.

    Google Scholar 

  17. Breslow NE, Clayton DG. Approximate inference in generalized linear mixed models. J Am Stat Assoc. 1993;88:9–25.

    Google Scholar 

  18. Freeman MF, Tukey JW. Transformations related to the angular and the square root. Ann Math Stat. 1950;21:607–11.

    Google Scholar 

  19. Tian X, et al. High serum uric acid trajectories are associated with risk of myocardial infarction and all-cause mortality in general Chinese population. Arthritis Res Ther. 2022;24:149.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hajizadeh R, Ghaffari S, Salehi R, Mazani S, Aghavali S. Association of serum uric acid level with mortality and morbidity of patients with acute ST-elevation myocardial infarction. J Cardiovasc Thorac Res. 2016;8:56–60.

    PubMed  PubMed Central  Google Scholar 

  21. Akgul O, Uyarel H, Pusuroglu H, Gul M, Isiksacan N, Turen S, et al. Predictive value of elevated uric acid in Turkish patients undergoing primary angioplasty for ST elevation myocardial infarction. Acta Cardiologica Sinica [Internet]. 2014;30(2):119–27. [cited 2023 Sep 30] Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4805017/.

  22. Gazi E. The association between serum uric acid level and heart failure and mortality in the early period of ST-elevation acute myocardial infarction. Arch Turk Soc Cardiol. 2014;42:501–8.

    Google Scholar 

  23. Mora-Ramírez M, et al. Hyperuricemia on admission predicts short-term mortality due to myocardial infarction in a population with high prevalence of cardiovascular risk factors. RIC. 2017;69:349.

    Google Scholar 

  24. Kojima S, et al. Prognostic usefulness of serum uric acid after acute myocardial infarction (The Japanese Acute Coronary Syndrome Study). Am J Cardiol. 2005;96:489–95.

    CAS  PubMed  Google Scholar 

  25. Kowalczyk J, et al. Prognostic significance of hyperuricemia in patients with different types of renal dysfunction and acute myocardial infarction treated with percutaneous coronary intervention. Nephron Clin Pract. 2010;116:c114–22.

    PubMed  Google Scholar 

  26. Sartorius B, Mayise C. Association between hyperuricemia and major adverse cardiac events in patients with acute myocardial infarction. Metab Syndr Relat Disord. 2017;15:18–25.

    PubMed  Google Scholar 

  27. Li L, et al. Hyperuricemia is associated with short-term outcomes in elderly patients with acute myocardial infarction. Aging Clin Exp Res. 2018;30:1211–5.

    PubMed  Google Scholar 

  28. Kaya MG, et al. Prognostic value of uric acid in patients with ST-elevated myocardial infarction undergoing primary coronary intervention. Am J Cardiol. 2012;109:486–91.

    CAS  PubMed  Google Scholar 

  29. Basar N, et al. Elevated serum uric acid predicts angiographic impaired reperfusion and 1-year mortality in ST-segment elevation myocardial infarction patients undergoing percutaneous coronary intervention. J Investig Med. 2011;59:931–7.

    CAS  PubMed  Google Scholar 

  30. Akpek M, et al. The association of serum uric acid levels on coronary flow in patients with STEMI undergoing primary PCI. Atherosclerosis. 2011;219:334–41.

    CAS  PubMed  Google Scholar 

  31. Dyrbuś, M. et al. Serum uric acid is an independent risk factor of worse mid- and long-term outcomes in patients with non-ST-segment elevation acute coronary syndromes. Cardiol J.  2021: VM/OJS/J/84645. https://doi.org/10.5603/CJ.a2021.0156.

  32. Bae MH, et al. Serum uric acid as an Independent and incremental prognostic marker in addition to N-Terminal Pro-B-type natriuretic peptide in patients with acute myocardial infarction. Circ J. 2011;75:1440–7.

    CAS  PubMed  Google Scholar 

  33. Mandurino-Mirizzi A, et al. Elevated serum uric acid is associated with a greater inflammatory response and with short- and long-term mortality in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. Nutr Metab Cardiovasc Dis. 2021;31:608–14.

    CAS  PubMed  Google Scholar 

  34. Çanga Y, et al. Prognostic value of serum uric acid levels in patients with non-STEMI undergoing percutaneous coronary intervention. Herz. 2020;45:389–96.

    PubMed  Google Scholar 

  35. Omidvar B, Ayatollahi F, Alasti M. The prognostic role of serum uric acid level in patients with acute ST elevation myocardial infarction. J Saudi Heart Assoc. 2012;24:73–8.

    PubMed  PubMed Central  Google Scholar 

  36. Ma W, Gao S, Huang S, Yuan J, Yu M. Hyperuricemia as a prognostic marker for long-term outcomes in patients with myocardial infarction with nonobstructive coronary arteries. Nutr Metab (Lond). 2021;18:107.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Chen L, et al. Serum uric acid in patients with acute ST-elevation myocardial infarction. World J Emerg Med. 2012;3:35.

    PubMed  PubMed Central  Google Scholar 

  38. Wildi K, et al. Uric acid for diagnosis and risk stratification in suspected myocardial infarction. Eur J Clin Invest. 2013;43:174–82.

    CAS  PubMed  Google Scholar 

  39. von Lueder TG, et al. Serum uric acid is associated with mortality and heart failure hospitalizations in patients with complicated myocardial infarction: findings from the High-Risk Myocardial Infarction Database Initiative: Serum uric acid in high-risk MI. Eur J Heart Fail. 2015;17:1144–51.

    Google Scholar 

  40. Tian X, et al. Cumulative serum uric acid and its time course are associated with risk of myocardial infarction and all-cause mortality. JAHA. 2021;10:e020180.

    PubMed  PubMed Central  Google Scholar 

  41. Tang X-F, et al. Hyperuricemia is associated with 2- and 5-year adverse outcomes in patients with ST-segment elevation myocardial infarction undergoing percutaneous coronary intervention. Front Endocrinol. 2022;13:852247.

    Google Scholar 

  42. Nakahashi T, et al. Impact of hyperuricemia on coronary blood flow and in-hospital mortality in patients with acute myocardial infarction undergoing percutaneous coronary intervention. J Cardiol. 2022;80:268–74.

    PubMed  Google Scholar 

  43. Moon J, et al. Serum transaminase determined in the emergency room predicts outcomes in patients with acute ST-segment elevation myocardial infarction who undergo primary percutaneous coronary intervention. Int J Cardiol. 2014;177:442–7.

    PubMed  Google Scholar 

  44. Mandurino-Mirizzi A, et al. Elevated serum uric acid is a predictor of contrast associated acute kidney injury in patient with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention. Nutr Metab Cardiovasc Dis. 2021;31:2140–3.

    CAS  PubMed  Google Scholar 

  45. Maliawan RP, Idarto D, Yasmin ADA, Gunadhi IP. Hyperuricemia as predictor of in-hospital major adverse cardiac events in patients with acute myocardial infarction treated at Sanglah General Hospital Denpasar-Bali. Bali Med J. 2017;6:395.

    Google Scholar 

  46. Liang D, et al. The interaction between hyperuricemia and low-density lipoprotein cholesterol increases the risk of 1-year post-discharge all-cause mortality in ST-segment elevation myocardial infarction patients. Nutr Metab Cardiovasc Dis. 2023;33:133–40.

    CAS  PubMed  Google Scholar 

  47. Levantesi G, et al. Uric acid: A cardiovascular risk factor in patients with recent myocardial infarction. Int J Cardiol. 2013;167:262–9.

    PubMed  Google Scholar 

  48. Lazzeri C, Valente S, Chiostri M, Gensini GF. Long-term prognostic role of uric acid in patients with ST-elevation myocardial infarction and renal dysfunction. J Cardiovasc Med. 2015;16:790–4.

    CAS  Google Scholar 

  49. Lazaros G, et al. Uric acid in-hospital changes predict mortality in patients with acute myocardial infarction. Nutr Metab Cardiovasc Dis. 2013;23:1202–9.

    CAS  PubMed  Google Scholar 

  50. Kuźma L, et al. The effect of serum uric acid levels on the long-term prognosis of patients with non-ST-elevation myocardial infarction. Adv Clin Exp Med. 2020;29:1255–63.

    PubMed  Google Scholar 

  51. Krishnan E, Pandya BJ, Lingala B, Hariri A, Dabbous O. Hyperuricemia and untreated gout are poor prognostic markers among those with a recent acute myocardial infarction. Arthritis Res Ther. 2012;14:R10.

    PubMed  PubMed Central  Google Scholar 

  52. Kim S, et al. High uric acid levels in acute myocardial infarction provide better long-term prognosis predictive power when combined with traditional risk factors. JCM. 2022;11:5531.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kalkan S, et al. A New predictor of mortality in ST-elevation myocardial infarction: the uric acid albumin ratio. Angiology. 2022;73:461–9.

    CAS  PubMed  Google Scholar 

  54. Qin T, et al. Hyperuricemia and the Prognosis of Hypertensive Patients: A Systematic Review and Meta-Analysis. J Clin Hypertens. 2016;18:1268–78.

    Google Scholar 

  55. Gagliardi ACM, Miname MH, Santos RD. Uric acid: a marker of increased cardiovascular risk. Atherosclerosis. 2009;202:11–7.

    CAS  PubMed  Google Scholar 

  56. Hare JM, Johnson RJ. Uric acid predicts clinical outcomes in heart failure: insights regarding the role of xanthine oxidase and uric acid in disease pathophysiology. Circulation. 2003;107:1951–3.

    PubMed  Google Scholar 

  57. Liu C-W, et al. Relationship of serum uric acid and Killip class on mortality after acute ST-segment elevation myocardial infarction and primary percutaneous coronary intervention. Int J Cardiol. 2017;226:26–33.

    PubMed  Google Scholar 

  58. Lazzeri C, Valente S, Chiostri M, Picariello C, Gensini GF. Uric acid in the early risk stratification of ST-elevation myocardial infarction. Intern Emerg Med. 2012;7:33–9.

    PubMed  Google Scholar 

  59. Bos MJ, Koudstaal PJ, Hofman A, Witteman JCM, Breteler MMB. Uric acid is a risk factor for myocardial infarction and stroke: the Rotterdam study. Stroke. 2006;37:1503–7.

    CAS  PubMed  Google Scholar 

  60. Festa A, Haffner SM. Inflammation and cardiovascular disease in patients with diabetes: lessons from the diabetes control and complications trial. Circulation. 2005;111:2414–5.

    PubMed  Google Scholar 

  61. Niskanen L, Laaksonen DE, Kristiina Nyyssönen, Georg Alfthan, Hanna‐Maaria Lakka, Lakka TA, et al. Uric Acid Level as a Risk Factor for Cardiovascular and All-Cause Mortality in Middle-aged Men. Archives of internal medicine. 2004;164(14):1546–6. [cited 2023 Sep 30] Available from: https://jamanetwork.com/journals/jamainternalmedicine/fullarticle/217245.

Download references

Acknowledgements

The authors thank the financial support from Medical Health Science and Technology Project of Zhejiang Provincial Health Commission (Grant Number: 2021KY1072) for checking the whole process of our entire meta-analysis.

Funding

This paper work was supported and funded by grants from the general project of Medical Health Science and Technology Project of Zhejiang Provincial Health Commission (Grant Number: 2021KY1072).

Author information

Authors and Affiliations

Authors

Contributions

Author Jiacheng-Rong and Lei-Huang screened the article titles and abstracts and submitted any content that could not be determined to author Cheng-Fang and Xudong-Chen for joint discussion. For full-text screening, author Lei-Huang recorded any article with uncertain eligibility; the ultimate inclusion was decided jointly with authors Jiacheng-Rong and Cheng-Fang. Besides, Chaokun-Hong supervised and participated in the final revision.

Corresponding author

Correspondence to Lei Huang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The original online version of this article was revised: order of the author names been updated.

Supplementary Information

Additional file 1:

 Figure SA. Funnel plot of publication bias for short-term adverse effects. Note: The Egger test p-value was 0.053. We adjusted for this using the trim and fill method and the results showed that, after adjustment, the Egger test p-value was 0.055.There was only a weak change, suggesting that the results of the meta-analysis on short-term mortality were robust. Figure SB. Funnel plot of publication bias for MACE.Note: The p-value for the Egger's test was 0.079, indicating that our results for the MACE meta-analysis were reliable and robust. Figure SC. Funnel plot of publication bias for other cardiovascular adverse outcomes. Note: The Egger test p-value was 0.0501. We adjusted for this using the trim and fill method and the results showed that, after adjustment, the Egger test p-value was 0.0512. There was only a weak change, suggesting that the results of meta-analysis on other cardiovascular adverse outcome were robust. Figure SD.Funnel plot of publication bias for long-term adverse effects. Note:The p-value for the Egger's test was 0.059, indicating that our results for the MACE meta-analysis were reliable and robust. S1 Table 1. Mesh Term & Entry Term. S1 Table 2. Scopus Serach Strategy. S1 Table 3. Cochrane Serach Strategy. S1 Table 4. MEDLINE Serach Strategy. S1 Table 5. Embase Serach Strategy. S1 Table 6. Global Health Serach Strategy. S2 Table 1. Data extraction form. S2 Table 2. Abbreviation. S3 Table 1. Article exlude reason. S4 Table 1. Characteristics of all literature included in this study and the outcome variables included.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rong, J., Fang, C., Chen, X. et al. Association of serum uric acid with prognosis in patients with myocardial infarction: an update systematic review and meta-analysis. BMC Cardiovasc Disord 23, 512 (2023). https://doi.org/10.1186/s12872-023-03523-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12872-023-03523-1

Keywords