Skip to main content

Urine and Serum Biomarkers of Cardiovascular Status and Early Vascular Ageing in Children and Adults

Abstract

Given that cardiovascular disease remains the leading cause of morbidity and mortality worldwide, there is a need to identify biomarkers that are accurate and reproducible to be able to identify which individuals are most at risk of early vascular ageing (EVA) to then allow for prioritisation of interventions to reduce this risk. To date, a myriad of different urine and blood biomarkers have been reported in studies looking at cardiovascular risk and EVA. These biomarkers primarily focus on oxidative stress, inflammation, haemostasis and thrombosis, metabolic markers, cardiovascular injury and epigenetic changes. As such, this review seeks to summarise the most common blood and urine markers reported in the literature and their current reported uses. Reference data in both adult and paediatric populations remain elusive for many of these biomarkers and may also be dependent on the assays used for analysis. It is possible that multi-marker risk scores may be of increased utility in the diagnosis of EVA. In addition, advances in technology may change the landscape of biomarker discovery in future years, with a need to prioritise research in the field of EVA to reduce the worldwide cardiovascular disease burden.

1 Introduction

Cardiovascular disease (CVD) is the leading cause of mortality worldwide, accounting for 38% of premature deaths below the age of 70 years [1]. Over the last 30 years, premature death due to CVD has been increasing, highlighting the ongoing importance of research in the field to understand how to determine cardiovascular status as well as the study of underlying pathophysiological mechanisms to improve outcomes through the development of prevention and treatment strategies [2].

Early vascular ageing (EVA) is defined as the presence of accelerated age-related changes in blood vessels, whereby the structure or function of the vessels resemble a more advanced biologic age than chronologic age, due to a series of characteristic changes (Table 1) [3]. EVA is a recognised independent risk factor for CVD and can be responsible for premature mortality with cardiovascular aetiology [4]. EVA can develop secondary to environmental and social exposures (air pollution, poverty, smoking, obesity, lack of exercise) as well as personal medical risk factors (hypertension, diabetes, genetic factors). Accumulation of subclinical vascular changes directs an individual towards a trajectory of EVA, with the potential to occur even in early fetal life, influencing future adult disease risk [5, 6]. Importantly, EVA changes can be reversible if interventions are implemented at an early stage, thus identifying people at risk and developing strategies for timely detection is crucial [7, 8].

Table 1 Features which characterise early vascular ageing (EVA)

The pathognomonic changes of EVA in the vasculature have been studied on an experimental basis in blood vessels, however, these specimens are obtained by invasive procedures and thus are not practical for routine use in clinical practice to determine those at risk of EVA and premature CVD. As such, it is clear that there is a need to determine which other biomarkers are useful in the initial detection of EVA. In particular, urine and serum biomarkers are relatively easy to obtain through minimally invasive procedures and can be used in clinical practice to identify individuals with EVA before manifestations of CVD, to focus on primary prevention in this patient group.

There is increasing interest in the field of EVA and large international groups of experts in the field of EVA have produced extensive reviews on the matter, for example from the Aging Biomarker Consortium [9] and the European Union Vascagenet [5, 10, 11] and some have now been endorsed in the clinical guideline for the management of CVD [12]. This field, however, is rapidly evolving, with a need to continually synthesise the most up-to-date literature. In addition, there remains little published information regarding current applications in both paediatric and adult populations. Whilst these reviews also consider other clinical biomarkers of EVA, the current review will focus on providing a concise introduction to urine and serum biomarkers of EVA only and will aim to highlight where these have been used in paediatric and adult populations and whether any sex differences have previously been reported to date. A large number of biomarkers have been described in the literature and it is out with the scope of this review to make a comprehensive list of all available and researched biomarkers. Rather we aim to describe the biomarkers of oxidative stress, inflammation, haemostasis and thrombosis, and metabolic and epigenetic markers associated with EVA and CVD that are currently most used.

2 Methods

Literature searches were conducted by 3 independent authors between October 2024 and December 2024 using Pubmed, Science Direct and Google Scholar with no date limitations on the searches. Search terms included ‘biomarker’, ‘vascular’, ‘early vascular ageing’ and ‘premature ageing’. Reference lists of included articles were also included. Biomarkers were included where they had been measured in blood or urine in human subjects with the aim to identify changes in vascular structure or function associated with EVA. For inclusion, biomarkers had to have been described in more than one study including more than one patient and with a clear description of methodology to allow replication. Table 2 summarises the biomarkers discussed in this review.

Table 2 Summary of biomarkers

2.1 What is a Biomarker?

A biomarker is any objective biological measure that can be used as an indicator of the body’s physiological state, pathological processes, or its response to therapeutic interventions [13]. For a biomarker to be clinically useful, it must possess certain additional characteristics which are summarised in Fig. 1. Although the term often refers to substances circulating in the body’s tissues or fluids, it also encompasses measurements of anatomical structures and functions. The difficulty with the use of these clinical biomarkers is that reference data remain sparse particularly in paediatric populations; they are operator-dependent and require cooperation from the patient as well as specialist expertise and equipment. As such their routine use to detect EVA remains limited. Urine and serum are commonly used for biomarker studies due to their relatively convenient accessibility and cost-effectiveness, and as such, this review will focus solely on these [14].

Fig. 1
figure 1

Key characteristics of an ideal biomarker

Urine samples can be collected non-invasively in large volumes, are relatively uncomplex in terms of proteins and cellular components allowing for simplified analysis, and contain certain metabolites in higher concentrations than blood, making them easier to detect [15]. Urine is, however, limited in its biomarker range as large proteins or cells may be absent, and its composition is dependent on factors such as hydration and diet, potentially affecting biomarker stability and reliability [16].

Serum, on the other hand, contains a broad array of proteins, hormones, enzymes and metabolites allowing for comprehensive analysis, and its composition is much more consistent making it potentially more reliable for longitudinal studies [16]. However, the serum collection process, while routine in clinical settings, is invasive, albeit minimally, and samples may be affected by haemolysis which can allow for biomarker measurement errors [17].

2.2 Biomarker Validation

A further potential challenge to the effective use of biomarkers is biomarker validation. This multi-step process assesses a biomarker’s measurement performance characteristics and evidence of the test results’ clinical significance, thereby ensuring the biomarker is reliable, accurate, and meaningful for its intended use [18].

2.2.1 Analytical Validation

The process begins with analytical validation, which assesses the biomarker measurement’s accuracy and reproducibility across different settings and methodologies [13]. Biomarker accuracy is generally expressed via its sensitivity (ability to detect disease when the disease is present e.g. true positives) and specificity (ability to recognise the true absence of disease e.g. true negatives). Receiver-operating curves are often used to visualise and study the relationship between sensitivity and specificity against the clinical gold standard [19], although other statistical models may be appropriate depending on the analyses in question. Changes associated with EVA are often subtle and subclinical, so any assay used to measure or monitor vascular status is likely to need to be measurable at small quantities, adding to the complexity of validation.

2.2.2 Clinical Validation

Clinical validation then establishes how the biomarker correlates with a clinical outcome—its ability to correctly identify individuals with a disease and predict disease risk or progression. This can be done by retrospective use of clinical trial data or via prospective population sampling [20]. Afterwards, the clinical utility of the biomarker must be validated by establishing whether the use of the biomarker in clinical care results in improved patient outcomes via the improved diagnosis, risk stratification or treatment decisions [20]. This requires the availability of long-term longitudinal data and may, therefore, take many years to generate.

2.2.3 Regulatory Validation

Regulatory validation ensures compliance with standards for safety and efficacy, allowing it to be used routinely in clinical practice [21]. Different countries have different regulatory authorities, with their own individual conditions for final approval and validation. Each regulatory board, however, will seek to confirm the data reviewed in earlier stages and to determine the safety and efficacy of the biomarker to be studied.

2.3 Biomarkers of Oxidative Stress

Biomarkers of oxidative stress are strongly associated with cardiovascular diseases and EVA [22]. Whether in urine or serum, they are each consistently elevated in those diagnosed with CVDs compared to healthy controls, evidencing their potential as diagnostic tools [6, 23,24,25]. Reactive oxygen species (ROS) have been implicated in the pathophysiology of EVA and CVD [10]. ROS are highly reactive oxygen-derived molecules with one or more unbound electrons and an excess of ROS results in oxidative stress, which may develop secondary to excessive ROS production, depleted supply of antioxidants, inactivation or impaired production of antioxidant enzymes or a combination of these. Due to their short half-lives, it can be difficult to accurately assess ROS levels directly, therefore the most frequently employed urine and/or serum markers measure post-translational modifications induced by ROS including indicators of oxidative DNA damage (e.g. 8-Hydroxy-2’-deoxyguanosine (8-OHdG)), markers of lipid peroxidation (F2-Isoprostanes and malondialdehyde (MDA)); ROS-producing enzymes (e.g. myeloperoxidase [MPO]); antioxidant capacity (e.g. total antioxidant capacity (TAOC), and total ROS [26] (Fig. 2), each of which will be discussed in this section (Table 2). Several other markers are available and have been reviewed elsewhere [27,28,29,30].

Fig. 2
figure 2

Pathways of oxidative stress and the biomarkers produced. Reactive oxygen species (ROS) are produced from mitochondrial respiration, myeloperoxidases (MPO), NADPH oxidases (NOX), and xanthine oxidases (OX), and are removed by superoxide dismutases (SOD) and glutathione peroxidases (GPx). ROS oxidise protein, lipids and DNA resulting in protein carbonyls, oxidised low-density lipoprotein (oxLDL), F2-isoprostanes, malondialdehyde (MDA), and 8-Hydroxy-2’-deoxyguanosine (8-OHdG) being released into the blood and subsequently urine

2.3.1 8-OHdG

8-OHdG is commonly measured in blood and urine as a marker of oxidative stress and DNA damage and its associations to cardiovascular diseases have been recently reviewed elsewhere [31, 32]. It can be measured in spot urine samples, with the best results with first-passed urine [33]. Higher levels have been associated with a number of CV diseases and there is evidence it can be used to prognosticate and risk stratify individuals with conditions such as heart failure [34]. Data are available in different cohorts of children, including in preterm neonates [35]. The significance of sex differences remains unclear with studies suggesting conflicting results as to whether men or women have higher levels of MDA, 8-OHdG, and F2-isoprostane levels [36,37,38,39,40]. In male children with early onset hypogonadism, levels are higher than in those with normal sex steroid levels [6].

2.3.2 F2-isoprostanes

F2-isoprostane levels are higher in women [36,37,38]. Additionally, these may aid in evaluating cardiovascular therapies, with lowered urinary F2-isoprostanes in men receiving anti-hypertensive treatment [41]. They are also valuable for risk stratification as elevated F2-isoprostanes predict cIMT and the risk of atherosclerosis, coronary artery disease, and cardiovascular events, including myocardial infarction and ischemic stroke [42,43,44,45], as a positive relationship has been reported between F2-isoprostane levels and the number of diseased vessels [46]. In addition, F2-isoprostanes seem to independently predict CVD mortality [44, 45, 47] and have been measured in children to monitor vascular status [48,49,50].

2.3.3 MDA and MPO

MDA and MPO can be measured in serum or urine with the Thiobarbituric Acid Reactive Substances (TBARs) assay most commonly used to measure MDA and ELISA used for MPO. MDA, and MPO independently predict CVD mortality [44, 45, 47] and have been reported in paediatric studies [6, 51]. Again, sex-aggregated data are conflicting with higher levels of both MDA and MPO being reported in men compared to women, in some studies [52,53,54] and the opposite in others [36,37,38].

2.3.4 Total Antioxidant Capacity (TAOC)

TAOC is inversely associated with the incidence of cardiovascular events and can be measured in blood, saliva or urine via ELISA [55]. These have been measured in paediatric populations, including in trials to determine whether antioxidants can improve vascular status in children at risk of EVA [56]. Women have been reported to have lower levels than men [57].

2.3.5 Total ROS

Total ROS can be analysed via measurement of superoxide, hydrogen peroxide or overall ROS levels. Guidelines exist regarding optimal methods of analysis including the use of electron paramagnetic resonance (EPR) for specific measurement of superoxide levels [58, 59]. Total ROS levels have been measured using a variety of probes after the isolation of cells from blood samples (mostly leukocytes and neutrophils) derived from adults [60]. Reactive oxygen metabolites (d-ROMs) have also been measured in the serum of adults and linked to CVD mortality [61]. ROS and their association with CVD have been described in different paediatric populations as reviewed elsewhere [62]. Overall men are reported to have increased levels of ROS compared to women [63]. This may be in part because SIRT1 is an inhibitor of ROS-producing pathways within the vasculature and is necessary for modulating oestrogen receptor signalling [64].

2.4 Biomarkers of Inflammation

Inflammation has long been hypothesised to be a feature of the ageing process, and studies of in vitro and in vivo models have shown its involvement in EVA [28, 65, 66]. In particular, the inflammatory pathways involved in vascular ageing have been shown to be mediated by several cytokines, and these are usually measured in human serum using immunoassays [67, 68]. To date, several biomarkers of inflammation in vascular ageing have been described and shown to correlate with cardiovascular risk. This section will consider 3 of the most commonly reported biomarkers of inflammation: high-sensitivity C-reactive protein (hs-CRP); interleukin (IL)-6 and tumour necrosis factor-alpha (TNF-alpha) (Table 2, Fig. 2).

2.4.1 hs-CRP

High sensitivity C-reactive protein (hs-CRP) is a biomarker used in clinical practice, measured in serum by nephelometry or turbidimetry [69], and has been shown to correlate with increased risk of cardiovascular events including stroke, acute coronary syndrome, and peripheral arterial disease [70]. In a cohort of 3,166 adults, more adverse cardiovascular events occurred in patients with elevated hs-CRP [71]. A further study by Guran et al. 2007 showed that hs-CRP was elevated in children at risk of CVD compared to healthy children with no risk factors [72]. Another study found that hs-CRP levels were significantly higher in male compared to female patients in their cohort of 6060 healthy people and correlated with lower levels of high-density lipoprotein [73].

2.4.2 TNF-alpha

TNF-alpha is usually measured via immunoassay. It has been found to be more abundantly expressed in veins of more advanced age, and it was shown by in vitro models to accelerate endothelial cell death [74, 75]. Bernardi et al. report a higher level of TNF-alpha in healthy men compared to women [76]. Yuan et al. 2020 found that single nucleotide polymorphisms that are associated with increased TNF levels led to an increase in coronary artery disease and ischaemic stroke [77]. A study by Liberale et al. 2021 showed that a higher TNF-alpha level was associated with more advanced age and worse outcomes in ischaemic stroke [78]. A study in children with obesity also demonstrated raised TNF-alpha, which correlated with levels of endothelin 1, a marker of endothelial dysfunction [79]. Other studies have investigated TNF-alpha in children and found this to be raised in other cohorts at risk of EVA [80, 81].

2.4.3 IL-6

Interleukin (IL)-6 is another pro-inflammatory cytokine with a role in stimulating the production of fibrinogen. It has been found to be more expressed in the vascular smooth muscle cells (VSMCs) of ageing blood vessels in an ex-vivo mouse model, and IL-6 levels correlate with long-term poorer prognosis due to cardiovascular events [82, 83]. The reference range of IL-6 levels has been studied and reported in a meta-analysis [84]. We could not identify any papers linking IL-6 to vascular ageing in children, although one study showed that IL-6 could predict the presence of pulmonary arterial hypertension (PAH) [85]. Different studies reported different effects of sex on IL-6 levels, with two groups reporting a correlation of IL-6 complexes with cardiovascular events in men but not women, while Bermudez and colleagues reported a relationship between IL-6 levels in women and the presence of hypertension, high cholesterol and diabetes [86,87,88].

2.5 Biomarkers of Haemostasis and Thrombosis

Ageing has long been associated with increased rates of both venous and arterial thrombotic events [89, 90]. The following biomarkers will be discussed: d-dimer; fibrinogen; plasminogen activator inhibitor-1 (PAI-1) and Von Willebrand factor (vWF) (Table 2, Fig. 3).

Fig. 3
figure 3

Pathways of inflammation and the biomarkers involved. Triggers such as oxidative stress, hypertension and endothelial dysfunction initiate inflammation resulting in the activation of immune cells and endothelial cells. These cells, particularly macrophages and monocytes, release the pro-inflammatory cytokines interleukin-6 (IL-6) and tissue necrosis factor-alpha (TNF-alpha). IL-6 stimulates hepatocytes in the liver to produce C-reactive protein (CRP), which is then released into circulation. Positive feedback loops occur where TNF-alpha and IL-6 further stimulate their own production

2.5.1 d-dimer

d-dimers represent clot degradation products, and have also been shown to rise with increasing age [91]. The LIPID study investigated baseline d-dimer levels in 7,863 individuals, and showed a positive correlation between d-dimer level and age, female sex, hs-CRP, and was a predictor of cardiovascular events and mortality [92]. d-dimer is used clinically and reference data are available for adults and children according to local laboratory assays [93, 94]. A study by Reagh et al. found that women had higher d-dimer levels than men in the absence of thromboembolic events [95] (see Fig. 4).

Fig. 4
figure 4

Pathways of haemostasis/thrombosis and the biomarkers involved. Von Willebrand factor (vWF) is produced by endothelial cells and mediates platelet aggregation and adhesion to form a platelet plug after vascular injury. Fibrinogen is produced by the liver and released into the circulation where the coagulation cascade results in its conversion to fibrin. Fibrin forms a mesh to stabilise the forming blood clot. The enzyme plasmin degrades fibrin, producing D-dimers as clot breakdown products. Plasmin is converted from its inactive precursor by tissue plasminogen activator (tPA), which is inhibited by plasminogen activator inhibitor-1 (PAI-1), thereby inhibiting clot breakdown

2.5.2 Fibrinogen

Fibrinogen levels have been shown to rise with age and in association with ischaemic heart disease [96]. Fibrinogen is considered an acute-phase protein as it increases during acute inflammation [97], and gene expression has been shown to be mediated by higher IL-6 [98]. Again, fibrinogen is often used for haematological investigations clinically with assay-specific reference data available for adults and children [99, 100]. Fibrinogen has been reported as higher in women and to be of increased risk to women when raised [101].

2.5.3 PAI-1

Concentrations of PAI-1 increase and plasminogen activator activity is known to decrease with age [102]. PAI-1 is overexpressed in senescent human umbilical vein endothelial cells, while expression of sirtuin-1 (SIRT-1) leads to epigenetic modifications that led to downregulation of PAI-1 and reduced senescence [103]. In patients, rising PAI-1, measurable by immunoassay of blood specimens, was associated with acute myocardial infarction and with recurrence of coronary events [104]. PAI-1 has been measured in children and adolescents at risk of EVA [105, 106]. Data are conflicting as to whether PAI-1 is higher in males or females [101, 107].

2.5.4 vWF

Von Willebrand factor is a protein associated with platelet aggregation and vessel wall adherence and as a carrier protein to factor VIII to the area of clot formation. On immunohistochemistry in vitro, vWF has been shown to be increased in endothelial cells of more advanced age [108]. In a genome-wide association study, mendelian randomisation demonstrated that plasma vWF levels were associated with an increased ischaemic stroke risk, and of a causative effect of factor VIII on venous thrombosis and coronary artery disease [109]. A further case–control study further investigated markers of thrombosis and haemostasis in patients with kidney failure and found that prothrombin I and II, d-dimer and vWF were downregulated, while antithrombotic factors such as protein C were downregulated [110]. Reference data for vWF are available and this assay has been measured clinically in different paediatric populations [111, 112]. Levels of vWF are generally higher in females but also differ according to ethnicity [113]. That said, the ristocetin assay, which can be used to measure vWF function reports higher activity in males compared to females [114].

2.6 Epigenetic Markers

With increasing age, cells are more prone to mutations and loss of genomic stability and altered epigenetic signatures, which can be quantified in a number of different ways. This section will consider: chromatin; DNA methylation; histone modification; micro-RNA and telomere length (Table 2).

2.6.1 Chromatin

Chromatin measurements have been associated with atherogenic changes in vascular smooth muscle cells [115]. Chromatin can be measured in blood or urine where there is sufficient genetic material. Single-cell analysis of chromatin in the human heart has shown sexual dimorphism and it is clear there are sex differences associated with epigenetics within the cardiovascular system [116].

2.6.2 DNA Methylation

DNA methylation is an epigenetic change that happens usually on CpG regions (cytosines preceding guanine), and interferes with gene transcription, downregulating expression of coded regions. Increasing DNA methylation is associated with increased atherosclerotic cardiovascular disease [117]. DNA methylation can affect the transcription of genes coding for proteins involved in other pathways of vascular ageing, for example fibrinogen and Klotho gene [118]. DNA methylation is best measured using serum due to increased quantities of DNA although urine can be used. A great deal of recent research has focused on DNA methylation and its association with CVD, with a DNA methylation map of atherosclerosis being published in 2015 [119]. DNA methylation has been measured in individuals from newborn age throughout the lifespan with sex differences noted at all ages. For example, a meta-analysis of newborn blood identified lower methylation levels at CpG regions in boys than girls [120] and multiple differences have been noted between the sexes from samples from placenta, cord blood and newborn blood [121]. DNA methylation differences in males and females for CVDs have been reviewed extensively elsewhere [122, 123].

2.6.3 Histone Modification

Histone modifications are epigenetic changes that underpin gene regulation and chromatin structure in eukaryotic nuclei. The significance of modifications have been reviewed elsewhere and can be found in those with evidence of EVA as well as congenital cardiovascular disease with some sex differences noted [124].

2.6.4 Micro-RNA

Micro-RNAs are non-coding RNA sequences that have a regulatory role in gene expression. There has been increasing interest in the field of micro-RNA research, and there is a rapid growth of the list of identified significant micro-RNAs in human physiological processes, with continuing expansion in their role as biomarkers in disease. There are several methods for quantification of micro-RNAs, including RNA sequencing and Northern blot, quantitative real-time reverse transcriptase PCR (qRT-PCR), and microarray. Serum or plasma samples can be used with qRT-PCR in clinical practice, and several studies have been carried out to determine intra- and pre-analytic standardisation and normalisation of results, including for use in cardiovascular events. Specifically, miR-133a/b, miR-122 miR-124, miR-34a and miR-134 were found to be significantly increased in patients with coronary artery disease [125, 126], while miR-145 and miR-155 were inversely related to development of coronary artery disease [127, 128]. miR-208b and miR-133a were associated with increased mortality [129]. miR-1 has also been implicated in vascular ageing: rising levels of miR-1 correlate with lower mean cardio-ankle vascular index and positively correlated with carotid artery stiffness [130]. Multiple studies have looked at different micro-RNAs in paediatric populations as recently reviewed by Paslawska and colleagues [131]. Sex influences levels of micro-RNAs in different ways depending on the miR type [132,133,134].

2.6.5 Telomere Length

A hallmark of cellular ageing is a reduction in telomere length [135]. Telomere length can be measured by quantitative polymerase chain reaction (PCR) or by Southern blot [136], and has been found to be longer in females during early childhood [137, 138]. Telomere length has also been demonstrated to be associated with advanced and not early arteriosclerosis [139], but preceding atherosclerosis [140]. Mean telomere length is also related to the clinical parameters of systolic and diastolic blood pressure and with odds of hypertension [141]. In a cohort of 308 adult patients requiring haemodialysis, a longer telomere length was more frequently observed in patients with a vascular cause of nephropathy and end stage renal failure, and importantly was associated with an increased risk of adverse cardiovascular outcomes [142].

2.7 Biomarkers of Cardiovascular Injury

Various biomarkers of CV injury have been studied to date. This section will consider: cell adhesion molecules (ICAM-1, ICAM-3, VCAM-1); N-terminal fragment of pro-brain (B-type) natriuretic peptide (NT-proBNP) and troponin (Table 2).

2.7.1 Cell Adhesion Molecules

Intracellular adhesion molecules (ICAM) 1 and 3 are glycoproteins that have potential as biomarkers associated with vascular injury. ICAM-3 has been associated with increased stroke risk [143], while soluble ICAM-1 has been found to be associated with vascular cognitive impairment in an elderly population [144], carotid plaque formation, and risk of stroke and stroke recurrence [145, 146]. Vascular cell adhesion molecule 1 (VCAM-1) is another glycoprotein that has been suggested as a biomarker of vascular injury, having been shown to associate with early atherosclerosis in a mouse model [147]. ICAM and VCAM can be measured by ELISA immunoassay, and both ICAM-1 and VCAM-1 can be used to predict the risk of major adverse cardiovascular events in patients who have had ST-elevation myocardial infarctions [148] and in haemodialysis patients [149]. Circulating VCAM-1 levels are also independently associated with more advanced chronological age [150]. In paediatric patients with metabolic syndrome, VCAM-1 has been found to be more elevated in boys than girls [151].

2.7.2 NT-proBNP

NT-proBNP is measured in blood samples using immunoassays [152]. It can be used to prognosticate mortality in heart failure and ischaemic heart disease [153,154,155]. In children with congenital heart disease, NT-proBNP levels that have been adjusted for age can be predictive of major adverse cardiovascular events [156]. Ethnic differences have been noted for NT-proBNP [157] but reference data are available, even for specific populations such as pregnant women [158]. Overall women have higher levels than men [159].

2.7.3 Troponin

Troponin I (cTnI) and troponin T (cTnT) are serum tests with high sensitivity to myocardial injury, routinely used for the detection of acute coronary syndromes and myocardial injury in clinical practice. High-sensitivity cTnI can be used as a predictive biomarker for future cardiovascular events in the non-acute setting [160]. Trajectory analysis of hs-cTnI showed that a trend of rising troponin occurs over decades, and years before cardiovascular events manifest [161]. Other applications for cTnI as a biomarker show that it can be used to predict the risk of death following trauma and admission to intensive care [162]. In paediatric populations, the level is highest in healthy infants and neonates and then reduces with progression to childhood and adolescence, and boys have higher baseline troponin levels than girls [163,164,165,166,167,168,169,170,171,172,173,174,175]. In children, hs-cTnI does not show an elevation on serial measurements as in adults, however, if the baseline result is elevated, it may point towards a cardiac aetiology [176]. Women have lower troponin levels compared to men [159].

2.8 Additional Biomarkers

Additional biomarkers that have been studied and will be discussed in this section are: albumin; alpha-klotho protein; endothelin-1; insulin-like growth factor 1 (IGF-1); matrix metalloproteinases; lactate; and omics technologies.

2.8.1 Albumin

The simple measurement of albumin in the urine (microalbuminuria) has been demonstrated consistently to be associated with all-cause mortality and cardiovascular mortality independent of glomerular filtration rate via data analysis of large population datasets including the Hunt-II study and the Dutch Prevention of renal and Vascular Endstage Disease (PREVEND) study [177] among others. As such, the measurement of microalbuminuria has been postulated as a useful tool to track the development of EVA in paediatric populations at risk [178, 179]. Sex differences have been noted with stronger associations between albumin and blood pressure identified in women [180]. Currently, albumin is one of the few clinically validated tools that can be utilised, however, the use of 24 h urine samples to do so is costly and can be considered inconvenient and difficult to comply with in some patients.

2.8.2 Alpha-klotho Protein

Alpha-Klotho is a protein that has been associated with anti-ageing effects and has been suggested as another potential biomarker of CVD and EVA. Suppressed levels of alpha-Klotho are associated with an increased risk of congestive heart failure and stroke [181]. In a mouse model with Klotho deficiency, which demonstrates features of hypertension, obesity, hypertriglyceridaemia, a viral delivery system of klotho gene delivery improved endothelial dysfunction and remodelling, and reduced hypertension [182]. Cohort studies in populations of children and adolescents at risk of EVA have shown detectable levels, which correlate to their disease group [183, 184]. Males have been reported as having lower alpha-klotho levels [181], with a U-shaped association between klotho levels and all-cause mortality in females, but no significant pattern in males [185].

2.8.3 Endothelin-1

Endothelin-1 (ET-1) and relating peptides have been detected in the circulating plasma and have been widely studied as potential biomarkers of CVD, with associations reported between endothelin-1 and blood pressure and age-related cardiac remodelling and cardiac fibrosis [186]. In addition, studies have investigated its use as a prognostic marker of coronary artery disease, myocardial infarction and heart failure [187]. ET-1 can be measured in children although data remain conflicting regarding whether levels increase in individuals at risk of EVA, such as those with hypertension and more research in this area is therefore required [188]. Sex differences in the vascular endothelium are well described, with young women having greater endothelium-dependent dilation compared to men, with age-related declines in endothelial function being delayed by approximately 10 years in women compared to men [189]. Plasma ET-1 levels are higher in men and increase with age [190].

2.8.4 IGF-1

Insulin-like growth factor 1 (IGF-1) is another potential serum marker of vascular ageing. Studies in individuals with hypopituitarism and in older adults with falling levels of IGF-1 suggest that cardiovascular aetiology for mortality increases in individuals with lower IGF-1 levels, and life expectancy decreases [191,192,193,194,195,196]. Studies in Laron syndrome (growth hormone resistance leading to IGF-1 deficiency) demonstrate impairments in left ventricular mass, stroke volume and cardiac output [197], and improvement in ventricular mass following initiation of treatment [198]. IGF-1 has been used clinically for a long time in the assessment of growth in children, with reference data available and no differences between the sexes, although IGF-1 peaks earlier in girls than boys [199]. Of note, a recent UK Biobank study identified a U-shaped association between serum IGF-1 and incident CVD, CVD mortality, myocardial infarction and heart failure, suggesting that monitoring of circulating IGF-1 status may be important for CV screening [200].

2.8.5 MMPs

Matrix metalloproteinases (MMPs) are a group of enzymes that are involved in the degradation and remodelling of the extracellular matrix. Declining levels of MMP following an ischaemic stroke in patients was predictive of a better outcome [201]. Individuals with obesity and smokers who are also at increased risk of cardiovascular events, were found to have increased MMP-9 levels [202, 203]. In a study by Cancemi et al., elderly individuals found a direct relationship between cholesterol and low-density lipoprotein concentrations and MMP-9 levels, and a negative correlation between MMP-2 and CRP [204]. MMPs have been successfully measured in paediatric populations assessing vascular function [205, 206].

The role of sex and gender remain conflicting. Zhong et al. showed an increased level of MMP-9 in males with ischaemic stroke, which was associated with worse prognosis and higher rates of long-term disability and mortality [207], however, Tegeler et al. found no association between sex and MMP levels [208].

2.8.6 Lactate

Lactate is a metabolic product of glycolysis that is commonly measured in the blood in acute clinical practice, including by point of care quantification. It is a non-specific marker elevated in multiple conditions. In the context of cardiovascular disease and vascular ageing, lactate has a role in multiple conditions including pulmonary arterial hypertension and heart failure [209]. Aside from its function as a metabolite and substrate in myocardial tissue, lactate is also a signalling molecule, promoting proliferation and migration of VSMCs [210], promoting angiogenesis [211], electrophysiological changes and vasodilation of vessels leading to vascular resistance [212]. Lactate has been used clinically in the assessment of sepsis and other conditions for decades, with reference data available in both child and adult populations at local biochemical laboratories [213] and no significant sex differences in lactate ranges have been noted.

2.8.7 Omics Technologies

One difficulty with biomarker development for CVD is that its development is multifactorial, with multiple different mechanisms predisposing an individual to pathology. As such, a multidisciplinary approach to CVD biomarker discovery may be required, which the advent of emerging technologies may be best placed to study. Plasma and urine metabolomics, peptidomics, proteomics and even genomics have shown promise in the detection of EVA, with evidence of alterations in individuals with ageing syndromes such as progeria [214] and good correlation to clinical vascular phenotype in those with hypertension [215,216,217]. These technologies offer potential for new biomarker discovery with the aim of precision medicine and treatments catered to the individual patient for maximum benefit [218]. Currently, however, omic analysis is expensive and results in the production of large volumes of data, of which much is currently of unknown clinical significance.

3 Future Directions

3.1 New Biomarker Discovery

Table 2 summarises the markers discussed in this review. As advances in technologies such as proteomics, metabolomics and genomics develop, more urine and blood biomarkers are likely to be reported in the coming years, although the clinical significance of these will require investigation. Given the significant number of potentially measurable targets, it is possible that multi-marker risk scores, combining the results of multiple different markers may be of increased utility in the diagnosis of EVA. Large-scale population data studies incorporating machine learning will be required to determine which are most useful and future studies should focus not only on the reporting of novel biomarkers but also on consideration regarding which are most accurate and accessible to researchers and clinicians alike.

3.2 Studies in More Diverse Populations

Particular research gaps have been noted within this review. Firstly, there is a paucity of data on the use of biomarkers associated with EVA in children. This is disappointing given the fact that where EVA is detected early, there is potential for vascular plasticity to reverse the process of biological ageing and reduce future cardiovascular disease risk [4, 180]. As previously discussed, sex and gender clearly influence CVD pathophysiology but many of the reported biomarkers do not segregate data and may not consider that sex and gender have different definitions. In addition, the majority of studies do not report whether ethnicity alters biomarker results, an area which has become increasingly apparent as racial disparities in CVD have been reported. Finally, could any of these biomarkers be of clinical significance in other important at-risk populations, such as during pregnancy or the post-partum period to detect individuals who would benefit from increased lifestyle or other intervention to improve future CV health, or indeed the CV health of the offspring? To address these considerations, panels of markers should be measured in the urine and serum of large groups of healthy individuals from different populations and compared on an individual basis, and also in different combinations. Reporting of these studies requires that data be segregated to determine if different populations have different normative values. Comparisons should be made with clinical data such as BP, PWV and population data such as hospital admissions or primary care diagnoses to determine which are most likely to be clinically reliable.

3.3 Reference Data and Clinical Applicability

The challenge of interpreting biomarker results also remains. Many studies do report reference data, but as above, these need to be considered in the context of the population included in the study and the assay used. In addition, comparison with clinical biomarkers such as BP, PWV and BMI is required as well as longitudinal studies to determine the clinical significance of elevated biomarkers. Where biomarkers become endorsed in clinical practice guidelines, consideration also needs to be given regarding service delivery of CVD prevention strategies to ensure equitable access to biomarker detection and risk stratification, particularly given the preponderance of CVD in low-middle income countries.

4 Conclusions

A large number of different urine and blood markers of EVA have been reported in the literature to date, with a particular focus on markers associated with oxidative stress, inflammation, haemostasis and thrombosis, metabolic markers, cardiovascular injury and epigenetic changes. In conclusion, there remains a significant need for future research into biomarkers for CVD to confirm which are most useful in different clinical settings, all with a view to reduce the current burden of CVD throughout the world.

Availability of Data and Materials

No datasets were generated or analysed during the current study.

References

  1. Timmis A, Vardas P, Torbica A, Katus H, De Smedt D, Gale CP, et al. European Society of Cardiology: cardiovascular disease statistics 2021. Eur Heart J. 2022;43(8):716–99.

    Article  PubMed  Google Scholar 

  2. Giovanni A, Enrico A, Aime B, Michael B, Marianne B, Jonathan C, et al. Global Burden of cardiovascular diseases and risk factors, 1990–2019: update from the GBD 2019 study. J Am Coll Cardiol. 2020;76(25):2982–3021.

    Article  Google Scholar 

  3. Currie G, Nilsson PM. Healthy Vascular Ageing and Early Vascular Ageing. Textbook of Vascular Medicine. 2019:307–18.

  4. Climie RE, Park C, Avolio A, Mynard JP, Kruger R, Bruno R-M. Vascular ageing in youth: a call to action. Heart Lung Circ. 2021;30(11):1613–26.

    Article  PubMed  Google Scholar 

  5. Climie RE, Alastruey J, Mayer CC, Schwarz A, Laucyte-Cibulskiene A, Voicehovska J, et al. Vascular ageing: moving from bench towards bedside. Eur J Prev Cardiol. 2023;30(11):1101–17.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Lucas-Herald AK, Montezano AC, Alves-Lopes R, Haddow L, Alimussina M, O’Toole S, et al. Vascular dysfunction and increased cardiovascular risk in hypospadias. Eur Heart J. 2022;43(19):1832–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Saz-Lara A, Cavero-Redondo I, Pascual-Morena C, Martínez-García I, Rodríguez-Gutiérrez E, Lucerón-Lucas-Torres M, et al. Early vascular aging as an index of cardiovascular risk in healthy adults: confirmatory factor analysis from the EVasCu study. Cardiovasc Diabetol. 2023;22(1):209.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Najjar SS, Scuteri A, Lakatta EG. Arterial aging: is it an immutable cardiovascular risk factor? Hypertension. 2005;46(3):454–62.

    Article  CAS  PubMed  Google Scholar 

  9. Aging Biomarker C, Zhang L, Guo J, Liu Y, Sun S, Liu B, et al. A framework of biomarkers for vascular aging: a consensus statement by the Aging Biomarker Consortium. Life Medicine. 2023;2(4):lnad033.

    Article  Google Scholar 

  10. Gopcevic KR, Gkaliagkousi E, Nemcsik J, Acet Ö, Bernal-Lopez MR, Bruno RM, et al. Pathophysiology of circulating biomarkers and relationship with vascular aging: a review of the literature from VascAgeNet Group on circulating biomarkers, European cooperation in science and technology action 18216. Front Physiol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphys.2021.789690.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Climie RE, Mayer CC, Bruno RM, Hametner B. Addressing the unmet needs of measuring vascular ageing in clinical practice—European cooperation in science and technology action VascAgeNet. Artery Res. 2020;26(2):71–5.

    Article  Google Scholar 

  12. McEvoy JW, McCarthy CP, Bruno RM, Brouwers S, Canavan MD, Ceconi C, et al. 2024 ESC Guidelines for the management of elevated blood pressure and hypertension: developed by the task force on the management of elevated blood pressure and hypertension of the European Society of Cardiology (ESC) and endorsed by the European Society of Endocrinology (ESE) and the European Stroke Organisation (ESO). Eur Heart J. 2024;45(38):3912–4018.

    Article  PubMed  Google Scholar 

  13. Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS. 2010;5(6):463–6.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Shama A, Soni T, Jawanda IK, Upadhyay G, Sharma A, Prabha V. The latest developments in using proteomic biomarkers from urine and serum for non-invasive disease diagnosis and prognosis. Biomark Insights. 2023;18:11772719231190218.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Jing J, Gao Y. Urine biomarkers in the early stages of diseases: current status and perspective. Discov Med. 2018;25(136):57–65.

    PubMed  Google Scholar 

  16. Xue C, Yang B, Fu L, Hou H, Qiang J, Zhou C, et al. Urine biomarkers can outperform serum biomarkers in certain diseases. Urine. 2023;5:57–64.

    Article  Google Scholar 

  17. Tóth J, Oláh AV, Petercsák T, Kovács T, Kappelmayer J. Detection of haemolysis, a frequent preanalytical problem in the serum of newborns and adults. Ejifcc. 2020;31(1):6–14.

    PubMed  PubMed Central  Google Scholar 

  18. Hunter DJ, Losina E, Guermazi A, Burstein D, Lassere MN, Kraus V. A pathway and approach to biomarker validation and qualification for osteoarthritis clinical trials. Curr Drug Targets. 2010;11(5):536–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Puntmann VO. How-to guide on biomarkers: biomarker definitions, validation and applications with examples from cardiovascular disease. Postgrad Med J. 2009;85(1008):538–45.

    Article  CAS  PubMed  Google Scholar 

  20. Ou FS, Michiels S, Shyr Y, Adjei AA, Oberg AL. Biomarker discovery and validation: statistical considerations. J Thorac Oncol. 2021;16(4):537–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kraus VB. Biomarkers as drug development tools: discovery, validation, qualification and use. Nat Rev Rheumatol. 2018;14(6):354–62.

    Article  CAS  PubMed  Google Scholar 

  22. Valaitienė J, Laučytė-Cibulskienė A. Oxidative stress and its biomarkers in cardiovascular diseases. Artery Res. 2024;30(1):18.

    Article  Google Scholar 

  23. Shishehbor MH, Zhang R, Medina H, Brennan M-L, Brennan DM, Ellis SG, et al. Systemic elevations of free radical oxidation products of arachidonic acid are associated with angiographic evidence of coronary artery disease. Free Radical Biol Med. 2006;41(11):1678–83.

    Article  CAS  Google Scholar 

  24. Moronkeji A, Olaniyan M. Evaluation of 8-hydroxy-2’-deoxyguanosine, interleukin-8 and malondialdehyde among patients with cardiovascular diseases in Ibadan, Oyo State. Sokoto J Med Lab Sci. 2024;9(2):160–9.

    Article  Google Scholar 

  25. Binti NN, Ferdausi N, Anik MEK, Islam LN. Association of albumin, fibrinogen, and modified proteins with acute coronary syndrome. PLoS ONE. 2022;17(7): e0271882.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Panda P, Verma HK, Lakkakula S, Merchant N, Kadir F, Rahman S, et al. Biomarkers of oxidative stress tethered to cardiovascular diseases. Oxid Med Cell Longev. 2022;2022:9154295.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Martínez de Toda I, González-Sánchez M, Díaz-Del Cerro E, Valera G, Carracedo J, Guerra-Pérez N. Sex differences in markers of oxidation and inflammation Implications for ageing. Mech Ageing Dev. 2023;211:111797.

    Article  PubMed  Google Scholar 

  28. Guzik TJ, Touyz RM. Oxidative stress, inflammation, and vascular aging in hypertension. Hypertension. 2017;70(4):660–7.

    Article  CAS  PubMed  Google Scholar 

  29. Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol. 2023;20(11):754–77.

    Article  PubMed  Google Scholar 

  30. Izzo C, Vitillo P, Di Pietro P, Visco V, Strianese A, Virtuoso N, et al. The role of oxidative stress in cardiovascular aging and cardiovascular diseases. Life. 2021;11(1):60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Graille M, Wild P, Sauvain JJ, Hemmendinger M, Guseva Canu I, Hopf NB. Urinary 8-OHdG as a biomarker for oxidative stress: a systematic literature review and meta-analysis. Int J Mol Sci. 2020;21(11):3743.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Di Minno A, Turnu L, Porro B, Squellerio I, Cavalca V, Tremoli E, et al. 8-Hydroxy-2-deoxyguanosine levels and cardiovascular disease: a systematic review and meta-analysis of the literature. Antioxid Redox Signal. 2016;24(10):548–55.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Zanolin ME, Girardi P, Degan P, Rava M, Olivieri M, Di Gennaro G, et al. Measurement of a urinary marker (8-hydroxydeoxyguanosine, 8-OHdG) of DNA oxidative stress in epidemiological surveys: a pilot study. Int J Biol Markers. 2015;30(3):341–5.

    Article  Google Scholar 

  34. Suzuki S, Shishido T, Ishino M, Katoh S, Sasaki T, Nishiyama S, et al. 8-Hydroxy-2′-deoxyguanosine is a prognostic mediator for cardiac event. Eur J Clin Invest. 2011;41(7):759–66.

    Article  CAS  PubMed  Google Scholar 

  35. Matsubasa T, Uchino T, Karashima S, Tanimura M, Endo F. Oxidative stress in very low birth weight infants as measured by urinary 8-OHdG. Free Radical Res. 2002;36(2):189–93.

    Article  CAS  Google Scholar 

  36. Wu LL, Chiou CC, Chang PY, Wu JT. Urinary 8-OHdG: a marker of oxidative stress to DNA and a risk factor for cancer, atherosclerosis and diabetics. Clin Chim Acta. 2004;339(1–2):1–9.

    Article  CAS  PubMed  Google Scholar 

  37. Block G, Dietrich M, Norkus EP, Morrow JD, Hudes M, Caan B, et al. Factors associated with oxidative stress in human populations. Epidemiol Rev. 2002;156(3):274–85.

    Google Scholar 

  38. Ma E, Ingram KH, Milne GL, Garvey WT. F2-isoprostanes reflect oxidative stress correlated with lean mass and bone density but not insulin resistance. J Endocr Soc. 2017;1(5):436–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Collins AR, Gedik CM, Olmedilla B, Southon S, Bellizzi M. Oxidative DNA damage measured in human lymphocytes: Large differences between sexes and between countries, and correlations with heart disease mortality rates. FASEB J. 1998;12(13):1397–400.

    Article  CAS  PubMed  Google Scholar 

  40. Proteggente AR, England TG, Rehman A, Rice-Evans CA, Halliwell B. Gender differences in steady-state levels of oxidative damage to DNA in healthy individuals. Free Radical Res. 2002;36(2):157–62.

    Article  CAS  Google Scholar 

  41. Ward NC, Hodgson JM, Puddey IB, Mori TA, Beilin LJ, Croft KD. Oxidative stress in human hypertension: association with antihypertensive treatment, gender, nutrition, and lifestyle. Free Radic Biol Med. 2004;36(2):226–32.

    Article  CAS  PubMed  Google Scholar 

  42. Yoon J-H, Kim J-Y, Park J-K, Ko S-B. Oxidative damage markers are significantly associated with the carotid artery intima-media thickness after controlling for conventional risk factors of atherosclerosis in men. PLoS ONE. 2015;10(3): e0119731.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Meuwese MC, Stroes ES, Hazen SL, van Miert JN, Kuivenhoven JA, Schaub RG, et al. Serum myeloperoxidase levels are associated with the future risk of coronary artery disease in apparently healthy individuals: the EPIC-Norfolk Prospective Population Study. J Am Coll Cardiol. 2007;50(2):159–65.

    Article  CAS  PubMed  Google Scholar 

  44. Baldus S, Heeschen C, Meinertz T, Zeiher AM, Eiserich JP, Münzel T, et al. Myeloperoxidase serum levels predict risk in patients with acute coronary syndromes. Circulation. 2003;108(12):1440–5.

    Article  CAS  PubMed  Google Scholar 

  45. Xuan Y, Gào X, Holleczek B, Brenner H, Schöttker B. Prediction of myocardial infarction, stroke and cardiovascular mortality with urinary biomarkers of oxidative stress: Results from a large cohort study. Int J Cardiol. 2018;273:223–9.

    Article  PubMed  Google Scholar 

  46. Vassalle C, Petrozzi L, Botto N, Andreassi M, Zucchelli G. Oxidative stress and its association with coronary artery disease and different atherogenic risk factors. J Intern Med. 2004;256(4):308–15.

    Article  CAS  PubMed  Google Scholar 

  47. Lorente L, Martín MM, Abreu-González P, Ramos L, Argueso M, Solé-Violán J, et al. Serum malondialdehyde levels in patients with malignant middle cerebral artery infarction are associated with mortality. PLoS ONE. 2015;10(5): e0125893.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Engler MM, Engler MB, Malloy MJ, Chiu EY, Schloetter MC, Paul SM, et al. Antioxidant vitamins C and E improve endothelial function in children with hyperlipidemia: Endothelial Assessment of Risk from Lipids in Youth (EARLY) Trial. Circulation. 2003;108(9):1059–63.

    Article  CAS  PubMed  Google Scholar 

  49. Engler MM, Engler MB, Malloy M, Chiu E, Besio D, Paul S, et al. Docosahexaenoic acid restores endothelial function in children with hyperlipidemia: results from the EARLY study. Int J Clin Pharmacol Ther. 2004;42(12):672–9.

    Article  CAS  PubMed  Google Scholar 

  50. Butani L, Dharmar M, Devaraj S, Jialal I. Preliminary report of inflammatory markers, oxidative stress, and insulin resistance in adolescents of different ethnicities. Metab Syndr Relat Disord. 2016;14(3):182–6.

    Article  CAS  PubMed  Google Scholar 

  51. Olza J, Aguilera CM, Gil-Campos M, Leis R, Bueno G, Martínez-Jiménez MD, et al. Myeloperoxidase is an early biomarker of inflammation and cardiovascular risk in prepubertal obese children. Diabetes Care. 2012;35(11):2373–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hoy A, Tregouet D, Leininger-Muller B, Poirier O, Maurice M, Sass C, et al. Serum myeloperoxidase concentration in a healthy population: biological variations, familial resemblance and new genetic polymorphisms. Eur J Hum Genet. 2001;9(10):780–6.

    Article  CAS  PubMed  Google Scholar 

  53. Gungor ZB, Sipahioglu N, Sonmez H, Ekmekci H, Toprak S, Ayaz G, et al. Endothelial dysfunction markers in low cardiovascular risk individuals: comparison of males and females. J Med Biochem. 2017;36(1):62–72.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Pinchuk I, Weber D, Kochlik B, Stuetz W, Toussaint O, Debacq-Chainiaux F, et al. Gender- and age-dependencies of oxidative stress, as detected based on the steady state concentrations of different biomarkers in the MARK-AGE study. Redox Biol. 2019;24: 101204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. da Silva A, Caldas APS, Pinto SL, Hermsdorff HHM, Marcadenti A, Bersch-Ferreira ÂC, et al. Dietary total antioxidant capacity is inversely associated with cardiovascular events and cardiometabolic risk factors: a cross-sectional study. Nutrition. 2021;89: 111140.

    Article  PubMed  Google Scholar 

  56. Rimoldi SF, Sartori C, Rexhaj E, Bailey DM, Marchi SFD, McEneny J, et al. Antioxidants improve vascular function in children conceived by assisted reproductive technologies: a randomized double-blind placebo-controlled trial. Eur J Prevent Cardiol. 2015;22(11):1399–407.

    Article  Google Scholar 

  57. Lettrichová I, Tóthová Lu, Hodosy J, Behuliak M, Celec P. Variability of salivary markers of oxidative stress and antioxidant status in young healthy individuals. Redox Rep. 2016;21(1):24–30.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Murphy MP, Bayir H, Belousov V, Chang CJ, Davies KJ, Davies MJ, et al. Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat Metab. 2022;4(6):651–62.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Griendling KK, Touyz RM, Zweier JL, Dikalov S, Chilian W, Chen Y-R, et al. Measurement of reactive oxygen species, reactive nitrogen species, and redox-dependent signaling in the cardiovascular system: a scientific statement from the American Heart Association. Circ Res. 2016;119(5):e39–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Daiber A, August M, Baldus S, Wendt M, Oelze M, Sydow K, et al. Measurement of NAD (P) H oxidase-derived superoxide with the luminol analogue L-012. Free Radical Biol Med. 2004;36(1):101–11.

    Article  CAS  Google Scholar 

  61. Schöttker B, Brenner H, Jansen EHJM, Gardiner J, Peasey A, Kubínová R, et al. Evidence for the free radical/oxidative stress theory of ageing from the CHANCES consortium: a meta-analysis of individual participant data. BMC Med. 2015;13(1):300.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Hertiš Petek T, Petek T, Močnik M, Marčun VN. Systemic inflammation, oxidative stress and cardiovascular health in children and adolescents: a systematic review. Antioxidants. 2022;11(5):894.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Kander MC, Cui Y, Liu Z. Gender difference in oxidative stress: a new look at the mechanisms for cardiovascular diseases. J Cell Mol Med. 2017;21(5):1024–32.

    Article  PubMed  Google Scholar 

  64. Hajializadeh Z, Khaksari M. The protective effects of 17-β estradiol and SIRT1 against cardiac hypertrophy: a review. Heart Fail Rev. 2022;27(2):725–38.

    Article  CAS  PubMed  Google Scholar 

  65. Barros PR, Costa TJ, Akamine EH, Tostes RC. Vascular aging in rodent models: contrasting mechanisms driving the female and male vascular senescence. Front Aging. 2021;2: 727604.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Costa TJ, De Oliveira JC, Giachini FR, Lima VV, Tostes RC, Bomfim GF. Programming of vascular dysfunction by maternal stress: immune system implications. Front Physiol. 2022;13: 787617.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Valaperti A, Li Z, Vonow-Eisenring M, Probst-Müller E. Diagnostic methods for the measurement of human TNF-alpha in clinical laboratory. J Pharm Biomed Anal. 2020;179: 113010.

    Article  CAS  PubMed  Google Scholar 

  68. Choi YJ, Roh J, Kim S, Lee KA, Park Y. Comparison of IL-6 measurement methods with a special emphasis on COVID-19 patients according to equipment and sample type. J Clin Lab Anal. 2022;36(1): e24182.

    Article  CAS  PubMed  Google Scholar 

  69. Dominici R, Luraschi P, Franzini C. Measurement of C-reactive protein: two high sensitivity methods compared. J Clin Lab Anal. 2004;18(5):280–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ridker PM. Clinical application of C-reactive protein for cardiovascular disease detection and prevention. Circulation. 2003;107(3):363–9.

    Article  PubMed  Google Scholar 

  71. Jalal D, Chonchol M, Etgen T, Sander D. C-reactive protein as a predictor of cardiovascular events in elderly patients with chronic kidney disease. J Nephrol. 2012;25(5):719.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Guran O, Akalin F, Ayabakan C, Dereli FY, Haklar G. High-sensitivity C-reactive protein in children at risk for coronary artery disease. Acta Paediatr. 2007;96(8):1214–9.

    Article  PubMed  Google Scholar 

  73. Hiura M, Kikuchi T, Nagasaki K, Uchiyama M. Elevation of serum C-reactive protein levels is associated with obesity in boys. Hypertens Res. 2003;26(7):541–6.

    Article  CAS  PubMed  Google Scholar 

  74. Shimada Y, Ito H, Kaji K, Fukuda M. Tumor necrosis factor reduces lifespan of human endothelial cells in vitro. Mech Ageing Dev. 1990;55(3):245–54.

    Article  CAS  PubMed  Google Scholar 

  75. Shimada Y, Kaji K, Ito H, Noda K, Matsuo M. Growth-inhibiting effect of tumor necrosis factor on human umbilical vein endothelial cells is enhanced with advancing age in vitro. J Cell Physiol. 1990;142(1):31–8.

    Article  CAS  PubMed  Google Scholar 

  76. Bernardi S, Toffoli B, Tonon F, Francica M, Campagnolo E, Ferretti T, et al. Sex differences in proatherogenic cytokine levels. Int J Mol Sci. 2020;21(11):3861.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Yuan S, Carter P, Bruzelius M, Vithayathil M, Kar S, Mason AM, et al. Effects of tumour necrosis factor on cardiovascular disease and cancer: a two-sample Mendelian randomization study. EBioMedicine. 2020;59:102956.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Liberale L, Bonetti NR, Puspitasari YM, Vukolic A, Akhmedov A, Diaz-Cañestro C, et al. TNF-α antagonism rescues the effect of ageing on stroke: perspectives for targeting inflamm-ageing. Eur J Clin Invest. 2021;51(11): e13600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Singh R, Verma A, Aljabari S, Vasylyeva TL. Urinary biomarkers as indicator of chronic inflammation and endothelial dysfunction in obese adolescents. BMC obesity. 2017;4:1–8.

    Article  Google Scholar 

  80. Nohynek H, Teppo AM, Laine E, Leinonen M, Eskola J. Serum tumor necrosis factor-alpha concentrations in children hospitalized for acute lower respiratory tract infection. J Infect Dis. 1991;163(5):1029–32.

    Article  CAS  PubMed  Google Scholar 

  81. Carrizo Tdel R, Díaz EI, Velarde MS, Prado MM, Bazán MC, Abregú AV. Tumor necrosis factor-alpha in a children population with overweight. Medicina (B Aires). 2013;73(4):310–4.

    PubMed  Google Scholar 

  82. Song Y, Shen H, Schenten D, Shan P, Lee PJ, Goldstein DR. Aging enhances the basal production of IL-6 and CCL2 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2012;32(1):103–9.

    Article  CAS  PubMed  Google Scholar 

  83. Mossmann M, Wainstein MV, Mariani S, Machado GP, de Araújo GN, Andrades M, et al. Increased serum IL-6 is predictive of long-term cardiovascular events in high-risk patients submitted to coronary angiography: an observational study. Diabetol Metab Syndr. 2022;14(1):125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Said EA, Al-Reesi I, Al-Shizawi N, Jaju S, Al-Balushi MS, Koh CY, et al. Defining IL-6 levels in healthy individuals: a meta-analysis. J Med Virol. 2021;93(6):3915–24.

    Article  CAS  PubMed  Google Scholar 

  85. Megied M, Abouelhassan MA, Hadwa E. Prognostic and diagnostic utility of interleukin-6 in pediatric pulmonary arterial hypertension—a case-control study. Eur J Pediatr. 2024;183(4):1637–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Ziegler L, Frumento P, Wallén H, de Faire U, Gigante B. The predictive role of interleukin 6 trans-signalling in middle-aged men and women at low-intermediate risk of cardiovascular events. Eur J Prev Cardiol. 2020;27(2):122–9.

    Article  PubMed  Google Scholar 

  87. Yasmin M, Karin L, Per E, Bruna G, Louise Z. Interleukin 6 trans-signalling and the risk of future cardiovascular events in men and women. Open Heart. 2021;8(2): e001694.

    Article  Google Scholar 

  88. Bermudez EA, Rifai N, Buring J, Manson JE, Ridker PM. Interrelationships among circulating interleukin-6, C-reactive protein, and traditional cardiovascular risk factors in women. Arterioscler Thromb Vasc Biol. 2002;22(10):1668–73.

    Article  CAS  PubMed  Google Scholar 

  89. Carobbio A, Thiele J, Passamonti F, Rumi E, Ruggeri M, Rodeghiero F, et al. Risk factors for arterial and venous thrombosis in WHO-defined essential thrombocythemia: an international study of 891 patients. Blood. 2011;117(22):5857–9.

    Article  CAS  PubMed  Google Scholar 

  90. Silverstein MD, Heit JA, Mohr DN, Petterson TM, O’Fallon WM, Melton LJ. Trends in the incidence of deep vein thrombosis and pulmonary embolism: a 25-year population-based study. Arch Intern Med. 1998;158(6):585–93.

    Article  CAS  PubMed  Google Scholar 

  91. Hager K, Platt D. Fibrin degeneration product concentrations (D-dimers) in the course of ageing. Gerontology. 1995;41(3):159–65.

    Article  CAS  PubMed  Google Scholar 

  92. Simes J, Robledo KP, White HD, Espinoza D, Stewart RA, Sullivan DR, et al. D-dimer predicts long-term cause-specific mortality, cardiovascular events, and cancer in patients with stable coronary heart disease: LIPID study. Circulation. 2018;138(7):712–23.

    Article  CAS  PubMed  Google Scholar 

  93. Kanis J, Hall CL, Pike J, Kline JA. Diagnostic accuracy of the D-dimer in children. Arch Dis Child. 2018;103(9):832–4.

    Article  PubMed  Google Scholar 

  94. Avila L, Amiri N, Pullenayegum E, Sealey VA, De R, Williams S, et al. Diagnostic value of D-dimers for limb deep vein thrombosis in children: a prospective study. Am J Hematol. 2021;96(8):954–60.

    Article  CAS  PubMed  Google Scholar 

  95. Reagh JJ, Zheng H, Stolz U, Parry BA, Chang AM, House SL, et al. Sex-related differences in D-dimer levels for venous thromboembolism screening. Acad Emerg Med. 2021;28(8):873–81.

    Article  PubMed  Google Scholar 

  96. Hager K, Felicetti M, Seefried G, Platt D. Fibrinogen and aging. Aging Clin Exp Res. 1994;6:133–8.

    Article  CAS  Google Scholar 

  97. Vilar R, Fish RJ, Casini A, Neerman-Arbez M. Fibrin (ogen) in human disease: both friend and foe. Haematologica. 2020;105(2):284.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Ray A. A SAF binding site in the promoter region of human γ-fibrinogen gene functions as an IL-6 response element. J Immunol. 2000;165(6):3411–7.

    Article  CAS  PubMed  Google Scholar 

  99. Bao W, Srinivasan SR, Berenson GS. Plasma fibrinogen and its correlates in children from a biracial community: the Bogalusa Heart Study. Pediatr Res. 1993;33(4):323–6.

    CAS  PubMed  Google Scholar 

  100. Appel IM, Grimminck B, Geerts J, Stigter R, Cnossen MH, Beishuizen A. Age dependency of coagulation parameters during childhood and puberty. J Thromb Haemost. 2012;10(11):2254–63.

    Article  CAS  PubMed  Google Scholar 

  101. van der Weerd N, van Os HJ, Ali M, Schoones JW, van den Maagdenberg AM, Kruyt ND, et al. Sex differences in hemostatic factors in patients with ischemic stroke and the relation with migraine—a systematic review. Front Cell Neurosci. 2021;15: 711604.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Hashimoto Y, Kobayashi A, Yamazaki N, Sugawara Y, Takada Y, Takada A. Relationship between age and plasma t-PA, PA-inhibitor, and PA activity. Thromb Res. 1987;46(5):625–33.

    Article  CAS  PubMed  Google Scholar 

  103. Wan YZ, Gao P, Zhou S, Zhang ZQ, Hao DL, Lian LS, et al. SIRT 1-mediated epigenetic downregulation of plasminogen activator inhibitor-1 prevents vascular endothelial replicative senescence. Aging Cell. 2014;13(5):890–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Wiman B, Andersson T, Hallqvist J, Reuterwall C, Ahlbom A, deFaire U. Plasma levels of tissue plasminogen activator/plasminogen activator inhibitor-1 complex and von Willebrand factor are significant risk markers for recurrent myocardial infarction in the Stockholm Heart Epidemiology Program (SHEEP) study. Arteriosc Thromb Vasc Biol. 2000;20(8):2019–23.

    Article  CAS  Google Scholar 

  105. Kudinova AY, Kulak MJ, Daniels TE, de Lewis Los Angeles W, de la Monte S, Mathis KJ, et al. Increased plasminogen activator inhibitor-1 (PAI-1) and its associations with metabolic risk in healthy young adults with early life stress. Psychoneuroendocrinology. 2024;166:107071.

    Article  CAS  PubMed  Google Scholar 

  106. Nocuń-Wasilewska K, Zwolińska D, Zubkiewicz-Kucharska A, Polak-Jonkisz D. Evaluation of vascular endothelial function in children with type 1 diabetes mellitus. J Clin Med. 2021;10(21):5065.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Asselbergs FW, Williams SM, Hebert PR, Coffey CS, Hillege HL, Navis G, et al. The gender-specific role of polymorphisms from the fibrinolytic, renin-angiotensin, and bradykinin systems in determining plasma t-PA and PAI-1 levels. Thromb Haemost. 2006;96(4):471–7.

    Article  CAS  PubMed  Google Scholar 

  108. Müller AM, Skrzynski C, Nesslinger M, Skipka G, Müller K-M. Correlation of age with in vivo expression of endothelial markers. Exp Gerontol. 2002;37(5):713–9.

    Article  PubMed  Google Scholar 

  109. Sabater-Lleal M, Huffman JE, De Vries PS, Marten J, Mastrangelo MA, Song C, et al. Genome-wide association transethnic meta-analyses identifies novel associations regulating coagulation factor VIII and von Willebrand factor plasma levels. Circulation. 2019;139(5):620–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Tran L, Pannier B, Lacolley P, Serrato T, Benetos A, London GM, et al. A case-control study indicates that coagulation imbalance is associated with arteriosclerosis and markers of endothelial dysfunction in kidney failure. Kidney Int. 2021;99(5):1162–72.

    Article  CAS  PubMed  Google Scholar 

  111. Gill JC, Conley SF, Johnson VP, Christopherson PA, Haberichter SL, Diaz CD, et al. Low VWF levels in children and lack of association with bleeding in children undergoing tonsillectomy. Blood Adv. 2020;4(1):100–5.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Dean JA, Blanchette VS, Carcao MD, Stain AM, Sparling CR, Siekmann J, et al. von Willebrand disease in a pediatric-based population–comparison of type 1 diagnostic criteria and use of the PFA-100 and a von Willebrand factor/collagen-binding assay. Thromb Haemost. 2000;84(3):401–9.

    Article  CAS  PubMed  Google Scholar 

  113. Conlan MG, Folsom AR, Finch A, Davis C, Sorlie P, Marcucci G, et al. Associations of factor VIII and von Willebrand factor with age, race, sex, and risk factors for atherosclerosis. Thromb Haemost. 1993;70(09):380–5.

    CAS  PubMed  Google Scholar 

  114. Sarji K, Graves J, Colwell J. Von Willebrand factor activity in normal subjects: sex difference and variability. Thromb Res. 1975;7(6):885–95.

    Article  CAS  PubMed  Google Scholar 

  115. Wang Y, Gao H, Wang F, Ye Z, Mokry M, Turner AW, et al. Dynamic changes in chromatin accessibility are associated with the atherogenic transitioning of vascular smooth muscle cells. Cardiovasc Res. 2022;118(13):2792–804.

    Article  CAS  PubMed  Google Scholar 

  116. Read DF, Booth GT, Daza RM, Jackson DL, Gladden RG, Srivatsan SR, et al. Single-cell analysis of chromatin and expression reveals age-and sex-associated alterations in the human heart. Communications Biology. 2024;7(1):1052.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Si J, Ma Y, Yu C, Sun D, Pang Y, Pei P, et al. DNA Methylation Age Mediates Effect of Metabolic Profile on Cardiovascular and General Aging. Circ Res. 2024;135(9):954–66.

    Article  CAS  PubMed  Google Scholar 

  118. Fujita T. Recent Advances in Hypertension: Epigenetic Mechanism Involved in Development of Salt-Sensitive Hypertension. Hypertension. 2023;80(4):711–8.

    Article  CAS  PubMed  Google Scholar 

  119. Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S, Condom E, et al. DNA methylation map of human atherosclerosis. Circulation: Cardiovascular Genetics. 2014;7(5):692–700.

  120. Solomon O, Huen K, Yousefi P, Küpers LK, González JR, Suderman M, et al. Meta-analysis of epigenome-wide association studies in newborns and children show widespread sex differences in blood DNA methylation. Mutat Res Rev Mutat Res. 2022;789: 108415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Bozack AK, Colicino E, Just AC, Wright RO, Baccarelli AA, Wright RJ, et al. Associations between infant sex and DNA methylation across umbilical cord blood, artery, and placenta samples. Epigenetics. 2022;17(10):1080–97.

    Article  PubMed  Google Scholar 

  122. Xu H, Li S, Liu Y-S. Roles and mechanisms of DNA methylation in vascular aging and related diseases. Front Cell Dev Biol. 2021;9: 699374.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Sakkers TR, Mokry M, Civelek M, Erdmann J, Pasterkamp G, Benavente ED, et al. Sex differences in the genetic and molecular mechanisms of coronary artery disease. Atherosclerosis. 2023;384:117279.

    Article  CAS  PubMed  Google Scholar 

  124. Robinson EL. Chapter 5 - Histone modifications in cardiovascular disease initiation and progression. In: Devaux Y, Robinson EL, editors. Epigenetics in cardiovascular disease, vol. 24. Academic Press: New York; 2021. p. 77–112.

    Chapter  Google Scholar 

  125. Gacoń J, Kabłak-Ziembicka A, Stępień E, Enguita FJ, Karch I, Derlaga B, et al. Decision-making microRNAs (miR-124, -133a/b, -34a and -134) in patients with occluded target vessel in acute coronary syndrome. Kardiol Pol. 2016;74(3):280–8.

    Article  PubMed  Google Scholar 

  126. Gao W, He HW, Wang ZM, Zhao H, Lian XQ, Wang YS, et al. Plasma levels of lipometabolism-related miR-122 and miR-370 are increased in patients with hyperlipidemia and associated with coronary artery disease. Lipids Health Dis. 2012;11:55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Zhu GF, Yang LX, Guo RW, Liu H, Shi YK, Ye JS, et al. microRNA-155 is inversely associated with severity of coronary stenotic lesions calculated by the Gensini score. Coron Artery Dis. 2014;25(4):304–10.

    Article  PubMed  Google Scholar 

  128. Gao H, Guddeti RR, Matsuzawa Y, Liu LP, Su LX, Guo D, et al. Plasma levels of microRNA-145 are associated with severity of coronary artery disease. PLoS ONE. 2015;10(5): e0123477.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Widera C, Gupta SK, Lorenzen JM, Bang C, Bauersachs J, Bethmann K, et al. Diagnostic and prognostic impact of six circulating microRNAs in acute coronary syndrome. J Mol Cell Cardiol. 2011;51(5):872–5.

    Article  CAS  PubMed  Google Scholar 

  130. Šatrauskienė A, Navickas R, Laucevičius A, Krilavičius T, Užupytė R, Zdanytė M, et al. Mir-1, miR-122, miR-132, and miR-133 are related to subclinical aortic atherosclerosis associated with metabolic syndrome. Int J Environ Res Public Health. 2021;18(4):1483.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Pasławska M, Grodzka A, Peczyńska J, Sawicka B, Bossowski AT. Role of miRNA in cardiovascular diseases in children—systematic review. Int J Mol Sci. 2024;25(2):956.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Sharma S, Eghbali M. Influence of sex differences on microRNA gene regulation in disease. Biol Sex Differ. 2014;5:1–8.

    Article  Google Scholar 

  133. Ji H, Kwan AC, Chen MT, Ouyang D, Ebinger JE, Bell SP, et al. Sex differences in myocardial and vascular aging. Circ Res. 2022;130(4):566–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Meder B, Backes C, Haas J, Leidinger P, Stähler C, Großmann T, et al. Influence of the confounding factors age and sex on microRNA profiles from peripheral blood. Clin Chem. 2014;60(9):1200–8.

    Article  CAS  PubMed  Google Scholar 

  135. Müezzinler A, Zaineddin AK, Brenner H. A systematic review of leukocyte telomere length and age in adults. Ageing Res Rev. 2013;12(2):509–19.

    Article  PubMed  Google Scholar 

  136. Aviv A, Hunt SC, Lin J, Cao X, Kimura M, Blackburn E. Impartial comparative analysis of measurement of leukocyte telomere length/DNA content by Southern blots and qPCR. Nucleic Acids Res. 2011;39(20): e134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Ly K, Walker C, Berry S, Snell R, Marks E, Thayer Z, et al. Telomere length in early childhood is associated with sex and ethnicity. Sci Rep. 2019;9(1):10359.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Bosquet Enlow M, Kane-Grade F, De Vivo I, Petty CR, Nelson CA. Patterns of change in telomere length over the first three years of life in healthy children. Psychoneuroendocrinology. 2020;115: 104602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Willeit P, Willeit J, Brandstätter A, Ehrlenbach S, Mayr A, Gasperi A, et al. Cellular aging reflected by leukocyte telomere length predicts advanced atherosclerosis and cardiovascular disease risk. Arterioscler Thromb Vasc Biol. 2010;30(8):1649–56.

    Article  CAS  PubMed  Google Scholar 

  140. Benetos A, Okuda K, Lajemi M, Kimura M, Thomas F, Skurnick J, et al. Telomere length as an indicator of biological aging: the gender effect and relation with pulse pressure and pulse wave velocity. Hypertension. 2001;37(2):381–5.

    Article  CAS  PubMed  Google Scholar 

  141. Huang YQ, Liu L, Lo K, Huang JY, Zhang B, Feng YQ. The relationship between mean telomere length and blood pressure: results from the National Health and Nutrition Examination Surveys. Ann Transl Med. 2020;8(8):535.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Vostatek R, Hohensinner P, Schmaldienst S, Lorenz M, Klauser-Braun R, Pabinger I, et al. Telomere length is associated with increased risk of cardiovascular events in patients with end-stage kidney disease on hemodialysis. Cardiorenal Med. 2024;14(1):524–32.

    Article  PubMed  Google Scholar 

  143. Pletsch-Borba L, Grafetstätter M, Hüsing A, Johnson T, González Maldonado S, Groß M-L, et al. Vascular injury biomarkers and stroke risk: a population-based study. Neurology. 2020;94(22):e2337–45.

    Article  CAS  PubMed  Google Scholar 

  144. Gregory MA, Manuel-Apolinar L, Sánchez-Garcia S, Villa Romero AR, de JesúsIuitRivera J, Basurto Acevedo L, et al. Soluble intercellular adhesion molecule-1 (sICAM-1) as a biomarker of vascular cognitive impairment in older adults. Dementia Geriatr Cogn Disord. 2019;47(4–6):243–53.

    Article  CAS  Google Scholar 

  145. Wang L, Chen Y, Feng D, Wang X. Serum ICAM-1 as a predictor of prognosis in patients with acute ischemic stroke. Biomed Res Int. 2021;2021(1):5539304.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Puig N, Camps-Renom P, Camacho M, Aguilera-Simón A, Jiménez-Altayó F, Fernández-León A, et al. Plasma sICAM-1 as a biomarker of carotid plaque inflammation in patients with a recent ischemic stroke. Transl Stroke Res. 2022;13(5):745–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, et al. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J Clin Investig. 2001;107(10):1255–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Yu J, Liu Y, Peng W, Xu Z. Serum VCAM-1 and ICAM-1 measurement assists for MACE risk estimation in ST-segment elevation myocardial infarction patients. J Clin Lab Anal. 2022;36(10): e24685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Papayianni A, Alexopoulos E, Giamalis P, Gionanlis L, Belechri AM, Koukoudis P, et al. Circulating levels of ICAM-1, VCAM-1, and MCP-1 are increased in haemodialysis patients: association with inflammation, dyslipidaemia, and vascular events. Nephrol Dial Transplant. 2002;17(3):435–41.

    Article  CAS  PubMed  Google Scholar 

  150. Richter V, Rassoul F, Purschwitz K, Hentschel B, Reuter W, Kuntze T. Circulating vascular cell adhesion molecules VCAM-1, ICAM-1, and E-selectin in dependence on aging. Gerontology. 2003;49(5):293–300.

    Article  CAS  PubMed  Google Scholar 

  151. Widjaja NA, Caesar LA, Nova S, Ardianah E. Beyond the scale: investigating adiponectin, ICAM-1, and VCAM-1 as metabolic markers in obese adolescents with metabolic syndrome. J Obes. 2023;2023(1):4574042.

    PubMed  PubMed Central  Google Scholar 

  152. Li L, Semenov AG, Feygina EE, Yang C, Wang N, Chen C, et al. Diagnostic utility of total NT-proBNP testing by immunoassay based on antibodies targeting glycosylation-free regions of NT-proBNP. Clin Chem Lab Med (CCLM). 2023;61(3):485–93.

    Article  CAS  PubMed  Google Scholar 

  153. De Lemos JA, Morrow DA, Bentley JH, Omland T, Sabatine MS, McCabe CH, et al. The prognostic value of B-type natriuretic peptide in patients with acute coronary syndromes. N Engl J Med. 2001;345(14):1014–21.

    Article  PubMed  Google Scholar 

  154. Kragelund C, Grønning B, Køber L, Hildebrandt P, Steffensen R. N-terminal pro–B-type natriuretic peptide and long-term mortality in stable coronary heart disease. N Engl J Med. 2005;352(7):666–75.

    Article  CAS  PubMed  Google Scholar 

  155. Luers C, Sutcliffe A, Binder L, Irle S, Pieske B. NT-proANP and NT-proBNP as prognostic markers in patients with acute decompensated heart failure of different etiologies. Clin Biochem. 2013;46(12):1013–9.

    Article  CAS  PubMed  Google Scholar 

  156. Palm J, Holdenrieder S, Hoffmann G, Hörer J, Shi R, Klawonn F, et al. Predicting major adverse cardiovascular events in children with age-adjusted NT-proBNP. J Am Coll Cardiol. 2021;78(19):1890–900.

    Article  CAS  PubMed  Google Scholar 

  157. Whitman IR, Vittinghoff E, DeFilippi CR, Gottdiener JS, Alonso A, Psaty BM, et al. NT-pro BNP as a mediator of the racial difference in incident atrial fibrillation and heart failure. J Am Heart Assoc. 2019;8(7): e010868.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Dockree S, Brook J, Shine B, James T, Vatish M. Pregnancy-specific reference intervals for BNP and NT-pro BNP—changes in natriuretic peptides related to pregnancy. J Endocr Soc. 2021;5(7):bvab091.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Rao RA, Bhardwaj A, Munagala M, Abraham S, Adig S, Shen A, et al. Sex differences in circulating biomarkers of heart failure. Curr Heart Fail Rep. 2024;21(1):11–21.

    Article  CAS  PubMed  Google Scholar 

  160. Haller PM, Goßling A, Magnussen C, Brenner H, Schöttker B, Iacoviello L, et al. Biomarker-based prediction of fatal and non-fatal cardiovascular outcomes in individuals with diabetes mellitus. Eur J Prev Cardiol. 2023;30(12):1218–26.

    Article  PubMed  Google Scholar 

  161. Kimenai DM, Anand A, De Bakker M, Shipley M, Fujisawa T, Lyngbakken MN, et al. Trajectories of cardiac troponin in the decades before cardiovascular death: a longitudinal cohort study. BMC Med. 2023;21(1):216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Decavèle M, Gault N, Gauss T, Pease S, Moyer J, Paugam-Burtz C, et al. Cardiac troponin I as an early prognosis biomarker after trauma: a retrospective cohort study. Br J Anaesth. 2018;120(6):1158–64.

    Article  PubMed  Google Scholar 

  163. Trevisanuto D, Pitton M, Altinier S, Zaninotto M, Plebani M, Zanardo V. Cardiac troponin I, cardiac troponin T and creatine kinase MB concentrations in umbilical cord blood of healthy term neonates. Acta Paediatr. 2003;92(12):1463–7.

    Article  CAS  PubMed  Google Scholar 

  164. Shelton SD, Fouse BL, Holleman CM, Sedor FA, Herbert WN. Cardiac troponin T levels in umbilical cord blood. Am J Obstet Gynecol. 1999;181(5):1259–62.

    Article  CAS  PubMed  Google Scholar 

  165. Engin Y, Üstün Y, Kurtay G. Cardiac troponin I levels in umbilical cord blood. Int J Gynecol Obstet. 2002;77(3):239–41.

    Article  CAS  Google Scholar 

  166. Soldin SJ, Murthy JN, Agarwalla PK, Ojeifo O, Chea J. Pediatric reference ranges for creatine kinase, CKMB, Troponin I, iron, and cortisol. Clin Biochem. 1999;32(1):77–80.

    Article  CAS  PubMed  Google Scholar 

  167. Mondal T, Ryan PM, Gupta K, Radovanovic G, Pugh E, Chan AK, et al. Cord-blood high-sensitivity troponin-I reference interval and association with early neonatal outcomes. Am J Perinatol. 2022;29(14):1548–54.

    Article  PubMed  Google Scholar 

  168. Lam E, Higgins V, Zhang L, Chan MK, Bohn MK, Trajcevski K, et al. Normative values of high-sensitivity cardiac troponin T and N-terminal pro-B-type natriuretic peptide in children and adolescents: a study from the CALIPER cohort. The Journal of Applied Laboratory Medicine. 2021;6(2):344–53.

    Article  PubMed  Google Scholar 

  169. Jehlicka P, Rajdl D, Sladkova E, Sykorova A, Sykora J. Dynamic changes of high-sensitivity troponin T concentration during infancy: clinical implications. Physiol Res. 2021;70(1):27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Karlén J, Karlsson M, Eliasson H, Bonamy A-KE, Halvorsen CP. Cardiac troponin T in healthy full-term infants. Pediatric Cardiology. 2019;40:1645–54.

  171. Bohn MK, Higgins V, Kavsak P, Hoffman B, Adeli K. High-sensitivity generation 5 cardiac troponin T sex-and age-specific 99th percentiles in the CALIPER cohort of healthy children and adolescents. Clin Chem. 2019;65(4):589–91.

    Article  CAS  PubMed  Google Scholar 

  172. Caselli C, Ragusa R, Prontera C, Cabiati M, Cantinotti M, Federico G, et al. Distribution of circulating cardiac biomarkers in healthy children: from birth through adulthood. Biomark Med. 2016;10(4):357–65.

    Article  CAS  PubMed  Google Scholar 

  173. Caselli C, Cangemi G, Masotti S, Ragusa R, Gennai I, Del Ry S, et al. Plasma cardiac troponin I concentrations in healthy neonates, children and adolescents measured with a high sensitive immunoassay method: high sensitive troponin I in pediatric age. Clin Chim Acta. 2016;458:68–71.

    Article  CAS  PubMed  Google Scholar 

  174. Franzini M, Lorenzoni V, Masotti S, Prontera C, Chiappino D, Della Latta D, et al. The calculation of the cardiac troponin T 99th percentile of the reference population is affected by age, gender, and population selection: a multicenter study in Italy. Clin Chim Acta. 2015;438:376–81.

    Article  CAS  PubMed  Google Scholar 

  175. Koerbin G, Potter JM, Abhayaratna WP, Telford RD, Badrick T, Apple FS, et al. Longitudinal studies of cardiac troponin I in a large cohort of healthy children. Clin Chem. 2012;58(12):1665–72.

    Article  CAS  PubMed  Google Scholar 

  176. Dionne A, Kheir JN, Sleeper LA, Esch JJ, Breitbart RE. Value of troponin testing for detection of heart disease in previously healthy children. J Am Heart Assoc. 2020;9(4): e012897.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Smink PA, Lambers Heerspink HJ, Gansevoort RT, de Jong PE, Hillege HL, Bakker SJ, et al. Albuminuria, estimated GFR, traditional risk factors, and incident cardiovascular disease: the PREVEND (prevention of renal and vascular endstage disease) study. Am J Kidney Dis. 2012;60(5):804–11.

    Article  PubMed  Google Scholar 

  178. Du MF, Wang Y, Hu GL, Wang D, Man ZY, Chu C, et al. Association of high-normal albuminuria and vascular aging: Hanzhong adolescent hypertension study. J Clin Hypertension. 2023;25(12):1096–104.

    Article  Google Scholar 

  179. Sellers EA, Dart AB, McGavock J, Wicklow BA. Cardiovascular comorbidity associated with albuminuria in youth-onset type 2 diabetes: analyses from the iCARE study. Can J Diabetes. 2021;45(5):458–65.

    Article  PubMed  Google Scholar 

  180. Wang Y, Wang J, Zheng X-W, Du M-F, Zhang X, Chu C, et al. Early-life cardiovascular risk factor trajectories and vascular aging in midlife: a 30-year prospective cohort study. Hypertension. 2023;80(5):1057–66.

    Article  CAS  PubMed  Google Scholar 

  181. Xu J-P, Zeng R-X, He M-H, Lin S-S, Guo L-H, Zhang M-Z. Associations between serum soluble α-Klotho and the prevalence of specific cardiovascular disease. Front Cardiovasc Med. 2022;9: 899307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Saito Y, Nakamura T, Ohyama Y, Suzuki T, Iida A, Shiraki-Iida T, et al. In vivo klotho gene delivery protects against endothelial dysfunction in multiple risk factor syndrome. Biochem Biophys Res Commun. 2000;276(2):767–72.

    Article  CAS  PubMed  Google Scholar 

  183. Karava V, Kondou A, Dotis J, Christoforidis A, Taparkou A, Farmaki E, et al. Fibroblast growth-factor 23-Klotho axis is associated with systemic inflammation and myokine profile in children with chronic kidney disease. Hormones. 2024;23(3):517–26.

    Article  PubMed  Google Scholar 

  184. Awad MM, Elshorbagy EA, Nasr AM, Hamed MG. Study of plasma klotho and plasma copeptin level in adolescents with type 1 diabetes mellitus and relation to microvascular complications. Egypt J Obes Diabetes Endocrinol. 2021;6(1).

  185. Wang J, Bai L, Ye Y, Chen X, Hu X, Peng Y. Sex differences in mortality risk and U-shaped relationship with klotho levels: a long-term cohort study. Exp Gerontol. 2024;198: 112643.

    Article  CAS  PubMed  Google Scholar 

  186. Wang X, Guo Z, Ding Z, Khaidakov M, Lin J, Xu Z, et al. Endothelin-1 upregulation mediates aging-related cardiac fibrosis. J Mol Cell Cardiol. 2015;80:101–9.

    Article  CAS  PubMed  Google Scholar 

  187. Castiglione V, Aimo A, Vergaro G, Saccaro L, Passino C, Emdin M. Biomarkers for the diagnosis and management of heart failure. Heart Fail Rev. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10741-021-10105-w.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Genovesi S, Giussani M, Orlando A, Lieti G, Viazzi F, Parati G. Relationship between endothelin and nitric oxide pathways in the onset and maintenance of hypertension in children and adolescents. Pediatr Nephrol. 2022;37(3):537–45.

    Article  PubMed  Google Scholar 

  189. Celermajer DS, Sorensen KE, Spiegelhalter DJ, Georgakopoulos D, Robinson J, Deanfield JE. Aging is associated with endothelial dysfunction in healthy men years before the age-related decline in women. J Am Coll Cardiol. 1994;24(2):471–6.

    Article  CAS  PubMed  Google Scholar 

  190. Kuczmarski AV, Welti LM, Moreau KL, Wenner MM. ET-1 as a sex-specific mechanism impacting age-related changes in vascular function. Front Aging. 2021;2: 727416.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Laughlin GA, Barrett-Connor E, Criqui MH, Kritz-Silverstein D. The prospective association of serum insulin-like growth factor I (IGF-I) and IGF-binding protein-1 levels with all cause and cardiovascular disease mortality in older adults: the Rancho Bernardo Study. J Clin Endocrinol Metab. 2004;89(1):114–20.

    Article  CAS  PubMed  Google Scholar 

  192. Vasan RS, Sullivan LM, D’Agostino RB, Roubenoff R, Harris T, Sawyer DB, et al. Serum insulin-like growth factor I and risk for heart failure in elderly individuals without a previous myocardial infarction: the Framingham Heart Study. Ann Intern Med. 2003;139(8):642–8.

    Article  CAS  PubMed  Google Scholar 

  193. Bates A, Van’t Hoff W, Jones P, Clayton R. The effect of hypopituitarism on life expectancy. J Clin Endocrinol Metab. 1996;81(3):1169–72.

    CAS  PubMed  Google Scholar 

  194. Tomlinson J, Holden N, Hills RK, Wheatley K, Clayton R, Bates A, et al. Association between premature mortality and hypopituitarism. The Lancet. 2001;357(9254):425–31.

    Article  CAS  Google Scholar 

  195. Rosen T, Bengtsson B-Å. Premature mortality due to cardiovascular disease in hypopituitarism. The Lancet. 1990;336(8710):285–8.

    Article  CAS  Google Scholar 

  196. Bülow B, Hagmar L, Mikoczy Z, Nordström CH, Erfurth EM. Increased cerebrovascular mortality in patients with hypopituitarism. Clin Endocrinol. 1997;46(1):75–81.

    Article  Google Scholar 

  197. Feinberg MS, Scheinowitz M, Laron Z. Echocardiographic dimensions and function in adults with primary growth hormone resistance (Laron syndrome). Am J Cardiol. 2000;85(2):209–13.

    Article  CAS  PubMed  Google Scholar 

  198. Feinberg MS, Scheinowitz M, Laron Z. Cardiac dimension and function in patients with childhood onset growth hormone deficiency, before and after growth hormone retreatment in adult age. Am Heart J. 2003;145(3):549–53.

    Article  CAS  PubMed  Google Scholar 

  199. Xu S, Gu X, Pan H, Zhu H, Gong F, Li Y, et al. Reference ranges for serum IGF-1 and IGFBP-3 levels in Chinese children during childhood and adolescence. Endocr J. 2010;57(3):221–8.

    Article  CAS  PubMed  Google Scholar 

  200. Lin J, Yang L, Huang J, Liu Y, Lei X, Chen R, et al. Insulin-like growth factor 1 and risk of cardiovascular disease: results from the UK biobank cohort study. J Clin Endocrinol Metab. 2023;108(9):e850–60.

    Article  PubMed  Google Scholar 

  201. Li Y, Han X, Luo S, Huang H, Huang X, Li M, et al. Predictive value of longitudinal changes of serum matrix metalloproteinase-9 and brain-derived neurotrophic factor in acute ischemic stroke. Front Aging Neurosci. 2022;14: 952038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Nath D, Shivasekar M, Vinodhini V. Smoking induces the circulating levels of matrix metalloproteinase-9 and its association with cardiovascular risk in young smokers. Medeniyet Med J. 2022;37(4):306–12.

    Article  Google Scholar 

  203. Luizon MR, Belo VA, Fernandes KS, Andrade VL, Tanus-Santos JE, Sandrim VC. Plasma matrix metalloproteinase-9 levels, MMP-9 gene haplotypes, and cardiovascular risk in obese subjects. Mol Biol Rep. 2016;43:463–71.

    Article  CAS  PubMed  Google Scholar 

  204. Cancemi P, Aiello A, Accardi G, Caldarella R, Candore G, Caruso C, et al. The role of matrix metalloproteinases (MMP-2 and MMP-9) in ageing and longevity: Focus on sicilian long-living individuals (LLIs). Mediators Inflamm. 2020;2020(1):8635158.

    PubMed  PubMed Central  Google Scholar 

  205. Pavan Kumar N, Venkataraman A, Varadarjan P, Nancy A, Rajamanickam A, Selladurai E, et al. Role of matrix metalloproteinases in multi-system inflammatory syndrome and acute COVID-19 in children. Front Med. 2022;9:1050804.

    Article  Google Scholar 

  206. Făgărășan A, Săsăran MO. The predictive role of plasma biomarkers in the evolution of aortopathies associated with congenital heart malformations. Int J Mol Sci. 2022;23(9):4993.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Zhong C, Yang J, Xu T, Xu T, Peng Y, Wang A, et al. Serum matrix metalloproteinase-9 levels and prognosis of acute ischemic stroke. Neurology. 2017;89(8):805–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Tegeler C, O’Sullivan JL, Bucholtz N, Goldeck D, Pawelec G, Steinhagen-Thiessen E, et al. The inflammatory markers CRP, IL-6, and IL-10 are associated with cognitive function—data from the Berlin Aging Study II. Neurobiol Aging. 2016;38:112–7.

    Article  CAS  PubMed  Google Scholar 

  209. Rees ML, Subramaniam J, Li Y, Hamilton DJ, Frazier OH, Taegtmeyer H. A PKM2 signature in the failing heart. Biochem Biophys Res Commun. 2015;459(3):430–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Heiss EH, Schachner D, Donati M, Grojer CS, Dirsch VM. Increased aerobic glycolysis is important for the motility of activated VSMC and inhibited by indirubin-3’-monoxime. Vascul Pharmacol. 2016;83:47–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Hunt TK, Aslam RS, Beckert S, Wagner S, Ghani QP, Hussain MZ, et al. Aerobically derived lactate stimulates revascularization and tissue repair via redox mechanisms. Antioxid Redox Signal. 2007;9(8):1115–24.

    Article  CAS  PubMed  Google Scholar 

  212. Montoya JJ, Fernández N, Monge L, Diéguez G, Villalón AL. Nitric oxide-mediated relaxation to lactate of coronary circulation in the isolated perfused rat heart. J Cardiovasc Pharmacol. 2011;58(4):392–8.

    Article  CAS  PubMed  Google Scholar 

  213. Schumacher KR, Reichel RA, Vlasic JR, Yu S, Donohue J, Gajarski RJ, et al. Rate of increase in serum lactate level risk-stratifies infants after surgery for congenital heart disease. J Thorac Cardiovasc Surg. 2014;148(2):589–95.

    Article  CAS  PubMed  Google Scholar 

  214. Lucas-Herald AK, Zürbig P, Mason A, Kinning E, Brown CE, Mansoorian B, et al. Proteomic evidence of biological aging in a child with a compound heterozygous ZMPSTE24 mutation. PROTEOMICS Clin Appl. 2019;13(2):1800135.

    Article  PubMed  Google Scholar 

  215. de Beer D, Mels CMC, Schutte AE, Delles C, Mary S, Mullen W, et al. A urinary peptidomics approach for early stages of cardiovascular disease risk: the African-PREDICT study. Hypertens Res. 2023;46(2):485–94.

    Article  PubMed  Google Scholar 

  216. Padmanabhan S, Dominiczak AF. Genomics of hypertension: the road to precision medicine. Nat Rev Cardiol. 2021;18(4):235–50.

    Article  CAS  PubMed  Google Scholar 

  217. Degenaar A, Jacobs A, Kruger R, Delles C, Mischak H, Mels C. Cardiovascular risk and kidney function profiling using conventional and novel biomarkers in young adults: the African-PREDICT study. BMC Nephrol. 2023;24(1):96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Dominiczak A, Delles C, Padmanabhan S. Genomics and precision medicine for clinicians and scientists in hypertension. Hypertension. 2017;69(4):e10–3.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

No specific funding was received for this work.

Author information

Authors and Affiliations

Authors

Contributions

AKLH conceptualised the review. MF, CF and AKLH wrote the article. All authors approved the final manuscript prior to submission.

Corresponding author

Correspondence to Angela K. Lucas-Herald.

Ethics declarations

Conflict of interest

The authors have no conflicts of interest.

Ethical Approval

Not applicable.

Consent for Publication

Not applicable.

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Frixou, M., Fraser, C. & Lucas-Herald, A.K. Urine and Serum Biomarkers of Cardiovascular Status and Early Vascular Ageing in Children and Adults. Artery Res 31, 7 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s44200-025-00072-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s44200-025-00072-0

Keywords