Kidney Res Clin Pract > Volume 41(Suppl 2); 2022 > Article
Jung and Yoo: Novel biomarkers for diabetic kidney disease

Abstract

Although diabetic kidney disease (DKD) remains one of the leading causes of reduced lifespan in patients with diabetes mellitus; its prevalence has failed to decline over the past 30 years. To identify those at high risk of developing DKD and disease progression at an early stage, extensive research has been ongoing in the search for prognostic and surrogate endpoint biomarkers for DKD. Although biomarkers are not used routinely in clinical practice or prospective clinical trials, many biomarkers have been developed to improve the early identification and prognostication of patients with DKD. Novel biomarkers that capture one specific mechanism of the DKD disease process have been developed, and studies have evaluated the prognostic value of assay-based biomarkers either in small sets or in combinations involving multiple biomarkers. More recently, several studies have assessed the prognostic value of omics-based biomarkers that include proteomics, metabolomics, and transcriptomics. This review will first describe the biomarkers used in current practice and their limitations, and then summarize the current status of novel biomarkers for DKD with respect to assay-based protein biomarkers, proteomics, metabolomics, and transcriptomics.

Introduction

Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease (CKD) and end-stage kidney disease (ESKD) worldwide [1]. Due to its association with a concomitant increase in cardiovascular morbidity and mortality, it remains one of the leading causes of reduced lifespan in people with diabetes mellitus (DM). Given the high risk of progressive deterioration in kidney function eventually leading to ESKD that requires kidney transplantation or chronic kidney replacement therapy, as well as increased risk of cardiovascular morbidity and mortality, early identification and risk stratification of disease is essential. Although our understanding of the pathophysiology of the disease has improved over the years, the prevalence of DKD has not changed significantly over the past 30 years [2].
To reduce disease prevalence and reverse or slow down disease progression, efforts to develop novel drugs have been ongoing, but many phase 3 clinical trials have failed to show any clinically significant findings [1,3]. For example, as oxidative stress is an important pathophysiological process in DKD, antioxidants have been developed to target the pathways of this disease. Although the antioxidant bardoxolone methyl (Reata Pharmaceuticals, Plano, TX, USA) showed promising phase 2 study results [4], administration of this agent in a phase 3 study showed an increased risk of early-onset fluid overload in patients with risk factors for heart failure [5]. Sulodexide, a purified mixture of sulfated glycosaminoglycan polysaccharides, can ameliorate DKD in animal experiments [6]. However, larger randomized controlled trials failed to demonstrate any beneficial clinical outcomes with the use of this agent [7,8]. As a result, efforts have been ongoing to develop novel biomarkers of DKD that can detect DKD at very early stages, as well as identify high-risk patients that are highly likely to eventually develop ESKD. Such novel biomarkers have also been developed to allow for risk stratification in clinical trials, as well as the development of drugs that specifically target these biomarkers.
This review will first describe current biomarkers used to predict the risk of DKD and disease progression, and then describe the current status of multiple novel biomarkers with respect to assay-based biomarkers, as well as omics-based biomarkers that include proteomics, metabolomics, and transcriptomics.

Biomarkers used in current practice

Markers of kidney function

The most commonly used current biomarkers of DKD are albuminuria and estimated glomerular filtration rate (eGFR) [9].
Given that albuminuria is an important component of DKD, as well as the strongest predictor of ESKD and cardiovascular morbidity in patients with type 2 DM [10], it is important to establish the degree of albuminuria in patients with DKD at the time of diagnosis. However, a major limitation of albuminuria as a biomarker of DKD is that not all patients with DKD have albuminuria. For example, approximately 30% of patients with DKD do not have albuminuria [11,12]. There is also a growing body of evidence suggesting that patients with type 1 or 2 DM can progress to ESKD in the absence of albuminuria, even after accounting for renoprotective agents [1318]. In a recent study of 935 patients with type 1 DM and 1,984 patients with type 2 DM, followed for up to 16 years after the development of CKD stage 3, mean annual declines in eGFR for normo-, micro-, and macroalbuminuria for the first 10 years following the development of CKD stage 3 were 1.9, 2.3, and 3.3 mL/min/1.73 m2 in type 1 DM, and 1.9, 2.1, and 3.0 mL/min/1.73 m2 in type 2 DM, respectively [18]. In patients with normoalbuminuria, two distinct eGFR patterns were found, with one displaying an accelerated rate of eGFR decline. Of note, patients displaying this accelerated rate of eGFR decline were associated with less use of lipid-lowering treatment, renin-angiotensin system (RAS) blockers, and other antihypertensive treatments. These findings suggest that albuminuria has its limitations as a prognostic marker for DKD.
Current eGFR and past glomerular filtration rate (GFR) trajectory are well-established predictors of the future risk of ESKD [19]. Thus, eGFR is the most common prognostic biomarker used for predicting ESKD in both clinical practice and clinical trials. However, similar to albuminuria, eGFR also has important limitations as a prognostic biomarker of DKD. Limitations of using eGFR to predict DKD progression include different equations used to estimate eGFR, which include the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI), the Modification of Diet in Renal Disease (MDRD) equation, and eGFR calculated using cystatin C instead of creatinine [14]. The risk of DKD development or progression may be under or overestimated depending on which eGFR equation is used for risk stratification. For instance, in a study to assess population-based incidence rates of CKD in patients with DM depending on the eGFR equation, CKD incidence rates were higher when the MDRD equation, rather than the CKD-EPI equation, was used [20]. Moreover, considering that GFR is the product of the number of nephrons and the mean single nephron GFR, a reduction in the number of nephrons due to the progression of DKD can be compensated by an increase in the single nephron GFRs of surviving nephrons. Such changes may not be accurately reflected by the eGFR, as eGFR decline may only happen when each nephron exceeds its maximal filtration capacity. This is further confounded because the vasodilation of the afferent arteriole in patients with DKD may increase single nephron GFR even in the absence of nephron loss [21]. Thus, patients without actual functional nephron loss and those with actual functional nephron loss, but with compensation in GFR by remnant nephrons, may have identical eGFRs with vastly different prognoses.
As both albuminuria and eGFR have important limitations as prognostic biomarkers of DKD, the identification of novel diagnostic and prognostic biomarkers for the early risk stratification of DKD is much needed. Potential novel biomarkers for DKD are summarized in Table 1.

Novel biomarkers of diabetic kidney disease—assay based

Considering the implications of the delayed identification of DKD progression, many studies have investigated potential predictive and prognostic biomarkers [22]. These biomarkers typically capture one specific aspect of the pathophysiology of the disease process such as tubular damage, inflammation, or oxidative stress [23]. However, given that DM is a heterogeneous disease involving multiple pathophysiological mechanisms, using just a single biomarker for the risk stratification of disease progression has several limitations. Single biomarkers have problems with individual, biological, and analytical variability. For example, novel biomarkers such as tissue necrosis factor receptor (TNFR) 1, TNFR2, fibroblast growth factor-23 (FGF-23), and high-density lipoprotein cholesterol are known to predict kidney outcomes in patients with type 2 DM [2426]. However, TNFR1 and TNFR2 are not specific to type 2 DM, and measurements of FGF-23 may vary according to the choice of the analytical method [27]. Due to such issues, it is more likely that a panel of biomarkers will be needed to predict disease progression.

Single biomarkers or small sets of biomarkers—assay based

Single biomarkers typically capture a specific pathophysiological pathway of the DKD process, such as tubular damage, inflammation, oxidative stress, or endothelial dysfunction, whereas others focus primarily on glomerular features such as glycocalyx abnormalities, podocyte damage, or glomerular fibrosis.

Markers of tubular damage

Markers of tubular damage include kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and liver fatty acid-binding protein (L-FABP). Urinary concentrations of KIM-1, which is a protein expressed on the apical membrane of the proximal tubule cells, increase in response to acute kidney injury [28]. In a nested case-control study and a prospective cohort study, plasma KIM-1 levels were independently associated with a higher risk of eGFR decline in persons with early or advanced DKD [29]. Similarly, in a case-cohort study of 894 participants with DKD from the Chronic Renal Insufficiency Cohort (CRIC), higher plasma levels of KIM-1 were associated with an increased risk of DKD progression [30]. Another tubular marker that has been extensively studied is NGAL. NGAL is a 25-kDa protein from the lipocalin superfamily that was initially found in activated neutrophils but is also produced by kidney tubular cells in response to tubular injury. Higher levels of urinary NGAL have been shown to precede microalbuminuria in patients with type 1 DM [31,32]. In another study of 117 patients with type 2 DM, higher values of urinary NGAL have been observed in normoalbuminuric type 2 DM patients, and rose progressively in those with micro- and macroalbuminuria, suggesting that tubular damage may occur even in the very early stages of DKD [33]. Urinary L-FABP levels have also been shown to be associated with DKD progression. In patients with type 1 DM, high levels of urinary L-FABP predicted the initiation and progression of DKD and all-cause mortality, independent of the severity of albuminuria and other established risk factors [34]. In another cross-sectional and longitudinal study of 140 patients with type 2 DM without DKD and 412 healthy control subjects, urinary L-FABP levels accurately reflected the severity of DKD, and these levels were particularly high in those with normoalbuminuria [35]. High urinary L-FABP levels were found to be a strong and independent predictor of DKD progression [36].

Markers of inflammation

Biomarkers of inflammation such as tumor necrosis factor (TNF)-α and interleukin-1β (IL-1β) were first associated with DM in diabetic mouse models. Macrophages incubated with glomerular basement membranes produced significantly greater levels of both TNF-α and IL-1β than in nondiabetic mice [37]. This has led to further investigations into the use of TNF-α as a predictive marker of DM. Urinary TNF-α excretion and serum TNF-α levels were both found to be elevated in DKD [38]. Further investigations into the receptors that TNF-α bind to, namely TNFR1 and TNFR2, have also suggested that circulating TNFR levels could also be used as good predictors of DKD [24]. For example, in a cohort consisting of 349 patients with type 1 DM and proteinuria, TNFR2 levels were the strongest determinant of eGFR decline [39].
Another biomarker that captures the oxidative stress characteristics of the DKD pathophysiologic process is 8-hydroxy-2′-deoxyguanosine (8-OHdG), which is a product of oxidative DNA damage. It is excreted in the plasma and urine after the repair of DNA by nuclease [40]. This has led to the assessment of 8-OHdG as a biomarker of oxidative stress in patients with DM [41,42]. In two cohorts of patients with type 1 DM, higher plasma concentrations of 8-OHdG were independently associated with increased risk of kidney disease in individuals with type 1 DM, suggesting that this marker may be used to evaluate the progression of DKD [42]. However, in a study of patients with type 2 DM and healthy control subjects, although urine 8-OHdG levels were increased in patients with DM, its ability to predict the development of DKD was inferior to the urine albumin-to-creatinine ratio [43].

Markers of glomerular damage

In contrast to biomarkers that capture specific pathophysiological pathways of the DKD process, some well-known biomarkers, including transferrin, immunoglobulin G (IgG), IgM, and ceruloplasmin, reflect glomerular damage. In theory, as the molecular weight of transferrin is similar to that of albumin, urinary transferrin could also be a biomarker of DKD. In a study of 45 patients with type 2 DM and with normoalbuminuria or microalbuminuria, increased urinary transferrin levels in microalbuminuria patients significantly correlated with kidney biopsy-proven tubulointerstitial injuries, suggesting a potential role of urinary transferrin in the prediction of early tubular damage in patients with DKD [44]. In a more recent study of 60 patients with and without type 2 DM, urine transferrin correlated with subclinical atherogenesis in patients with type 2 DM without kidney dysfunction, suggesting that it could potentially be an early marker of endothelial dysfunction in patients with type 2 DM but without kidney dysfunction [45]. Another marker of glomerular damage that has been investigated is ceruloplasmin. Ceruloplasmin is a copper-carrying metalloenzyme that is more negatively charged than albumin, and thus is more difficult to be filtered by the glomerulus. In a study of 140 patients with type 2 DM with normoalbuminuria, urinary ceruloplasmin levels were found to be elevated in normoalbuminuric patients with DM and were highly predictive of the development of microalbuminuria [46]. Urinary IgM and IgG levels have also been shown to be predictive of DKD. A study of 22 patients with type 1 DM and 20 patients with type 2 DM, all with evidence of DKD, revealed that the increased urine excretion of IgG and IgM accompanied albuminuria in patients with type 2 DM, suggesting a potential role of urinary immunoglobulins in the risk stratification of DKD [47].

Multiple biomarkers—assay based

As DKD is a disease entity that involves multiple pathophysiological pathways, a combination of biomarkers may be required to accurately predict disease progression. For example, most studies that have investigated panels of candidate biomarkers have included TNFR, often in combination with biomarkers of tubular damage such as KIM-1 [29,30]. In a nested case-control of 380 participants, and a prospective cohort study of 1,156 participants with type 2 DM, higher plasma levels of KIM-1, TNFR-1, and TNFR-2 were associated with a higher risk of DKD progression, even after adjustments for age, relevant anthropometric, sociodemographic, and laboratory parameters. Of note, when all three plasma biomarkers were added to the clinical model, the area under the curve (AUC) for DKD progression improved from 0.680 to 0.752 [29]. A case-cohort study consisting of 894 participants with both type 1 and 2 DM reported similar findings, where higher levels of monocyte chemoattractant protein 1 (MCP-1) were also associated with a higher risk of DKD progression [30]. In a study of 260 Pima Indians with type 2 DM, urinary NGAL/creatinine was positively associated with risk of ESKD and mortality, whereas L-FABP/creatinine was inversely associated with ESKD. The addition of NGAL/creatinine and L-FABP/creatinine to models that included albuminuria and eGFR increased the C-statistics for predicting the risk of ESKD [48].
Other studies have explored higher numbers of potential candidate biomarkers to improve the prediction of DKD outcomes. In a study evaluating 17 potential urinary and seven plasma biomarkers in 67 participants with type 2 DM, urinary C-terminal FGF-23 was found to show the strongest association with ESKD, whereas plasma vascular endothelial growth factor (VEGF) was associated with the highest risk of the composite outcome of ESKD and death [49]. Another prospective study followed 83 patients with overt diabetic nephropathy and obtained repeated measurements of proteinuria, IL-1β, IL-6, IL-8, MCP-1, TNF-α, transforming growth factor-beta 1 (TGF-β1), and plasminogen activator inhibitor-1 (PAI-1) [50]. In the study, urinary MCP-1 and TGF-β1 predicted kidney function decline that was independent of albuminuria. In a more recent study involving 345 community-based patients with type 2 DM from the Fremantle Diabetes Study Phase II, eight potential candidate biomarkers were studied after adjustment for clinical parameters. Of these eight biomarkers, apolipoprotein A4 (apoA4), CD5 antigen-like (CD5L), and complement C1q subcomponent subunit B (C1QB) were independently associated with the rapid decline in kidney function, improved predictive performance, fitness, discrimination, and reclassification [51].
However, as evidenced in the SUMMIT (the Surrogate Markers for Micro- and Macro-Vascular Hard Endpoints for Innovative Diabetes Tools) program [52], there are very strong correlations between these biomarkers, and this confounds the interpretation of these biomarkers as predictors of disease progression. Further optimization of a panel of best-reported biomarkers would be needed. Ideally, future studies could explore a panel of biomarkers that show low correlation with each other. Moreover, most studies reported to date are small in sample size, and therefore studies consisting of larger populations would be needed for the validation of the aforementioned biomarkers.

Novel biomarkers of diabetic kidney disease—omics based

The omics platform-based approach

Over the last decade, the use of approaches that measure large sets of lipids, metabolites, amino acids, peptides, and proteins are increasing [53]. These approaches have been called omics-based tests, and are defined as an assay derived from multiple molecular measurements that allow the quantification of all RNAs, proteins, and metabolites present in biological samples, and interpreted by computational models to produce clinically meaningful results. Omics-based approaches have the advantage of not only measuring a full spectrum of peptides or metabolites in a short amount of time but also producing large sets of unbiased data that can be used for diagnosis, prediction of disease progression, and treatment response. As a result, this omics platform-based approach has emerged as a strong tool in biomarker discovery in recent years [53] (Fig. 1).

Proteomics

Proteomics allows the simultaneous quantification of multiple protein markers in a biological sample [54,55]. Although tissue samples provide the most information on protein expression, DKD is often diagnosed clinically, rather than by biopsy, and therefore the number of patients who will undergo kidney biopsy for DKD will not increase. Blood samples can display protein signals that are produced from the kidney and of more generic processes such as fibrosis [56]. However, obtaining large volumes of blood samples in patients is often not feasible. The collection of urine samples overcomes the limitations of blood samples. Urine collection is not only available in large volumes, but it is also noninvasive. This led to urinary proteomics gaining more attention as a tool for the identification of diagnostic and prognostic biomarkers in kidney disease [57].
Early studies using proteomics in DKD used urinary samples to improve early diagnosis and the prediction of kidney-related and other outcomes [5860]. The most studied and well-validated proteomic classifier to date is the capillary electrophoresis-mass spectrometry-based urinary peptide classifier, CKD-273. This mass spectrometry-based method combines the data of 273 urinary peptides into a combined score that has high accuracy in predicting the new onset of albuminuria. Approximately 75% of the peptides are collagen fragments, with uromodulin, clusterin, albumin, β-2 microglobulin, α-1 antitrypsin, and others comprising the remainder. The diagnostic utility of this proteomic classifier was first developed in a cross-sectional study of 3,600 patients with different CKD etiologies, where the classifier showed a sensitivity of 85% and a specificity of 100% for the diagnosis of CKD [61].
This classifier was subsequently validated across several cohorts consisting of patients with type 2 DM, where CKD-273 predicted both the development and progression of albuminuria in patients with DKD. In a prospective study of 35 patients with either type 1 or 2 DM, CKD-273 was able to predict progression to macroalbuminuria 5 years before the actual onset [62]. In another prospective case-control study from the PREVEND (Prevention of Renal and Vascular End-stage Disease) study and from the Steno Diabetes Center (Gentofte, Denmark), the proteomic classifier was independently associated with the transition to micro- or macroalbuminuria. The classifier improved the predictive ability of albuminuria and eGFR in the development and progression of albuminuria [63]. Analyses of both the Effect of Candesartan on Progression of Retinopathy in Type 1 Diabetes (DIRECT-Protect 1) and in Type 2 Diabetes (DIRECT-Protect 2) studies demonstrated that the CKD-273 classifier was able to predict microalbuminuria, independent of treatment, age, sex, systolic blood pressure, albuminuria, eGFR, hemoglobin A1c (HbA1c), and DM duration [60,6466]. More recently, in a study of 1,014 individuals with type 1 or 2 DM, baseline eGFR of ≥70 mL/min/1.73 m2, and normoalbuminuria, CKD-273 was able to identify patients with DM who will progress to eGFR of <60 mL/min/1.73 m2 in the absence of albuminuria, independent of age, blood pressure, and baseline eGFR [67]. The concept that CKD-273 may be useful in determining the risk of disease progression and that it may also stratify treatment response to spironolactone was more definitively tested in the recent PRIORITY (Proteomic Prediction and Renin Angiotensin Aldosterone System Inhibition Prevention of Early Diabetic nephropathy in Type 2 Diabetic Patients with Normoalbuminuria) trial. In 1,775 participants with type 2 DM and normoalbuminuria over a median follow-up time of 2.5 years, high-risk patients defined by CKD-273 were more likely to develop microalbuminuria, even after adjustments for baseline risk factors such as HbA1c, systolic blood pressure, baseline albuminuria, and eGFR [68]. However, spironolactone did not prevent progression to microalbuminuria in high-risk patients. On the contrary, in an exploratory analysis of the MARLINA-T2D (Efficacy, Safety & Modification of Albuminuria in Type 2 Diabetes Subjects With Renal Disease With LINAgliptin) trial, where participants were randomized to receive either linagliptin or placebo for 24 weeks, there was a significant correlation between CKD-273 and clinical renal parameters as well as eGFR decline. Patient stratification using this classifier found that linagliptin had the potential to slow progressive kidney function decline in high-risk CKD patients [69].
Results from urinary proteomic studies have improved our knowledge of the pathophysiology of DKD. Other proteomic methods include the use of collagen fragments. For example, the α1 type 1 collagen chain is significantly altered in urine 3 to 5 years before the onset of macroalbuminuria [62]. It was found that before urinary albumin excretion starts to increase, urinary collagen fragments decreased. In a multicenter study involving 165 patients with type 2 DM, type I and type III α1 collagen and α2-Heremans-Schmid-glycoprotein were found to be prominent collagen markers. Therefore, it is thought that collagen fragments originate from the kidney, and a decrease in these fragments in patients with DKD may be due to the accumulation of extracellular matrix and increased kidney fibrosis [70].
Other urinary peptides that have been studied include uromodulin, progranulin, clusterin, α1 acid glycoprotein, and haptoglobin. In a nested case-control design, a panel including uromodulin, progranulin, clusterin, and α1 acid glycoprotein predicted an early decline in eGFR in a cohort of 465 adults with type 1 DM [71]. Moreover, in a study of patients with DKD from the VADT (Veterans Affairs Diabetes Trial), urinary haptoglobin was identified as a candidate biomarker to predict early kidney functional decline [72].
Regardless of which sample is used for proteomic analysis, the use of proteomics to predict DKD development and progression still has several limitations including the absence of well-validated diagnostic criteria. Prospective validation studies are needed before the widespread implementation of proteomics in DKD.

Metabolomics

Metabolomics is the measurement of low-weight intermediates (<500 Da) and small end products of biochemical processes in biological fluids, and they have emerged as another potential tool in the discovery of novel biomarkers for kidney diseases. The metabolome is often regarded as the final downstream integration of biological information that consists of the genome, proteome, transcriptome, and overall enzymatic reactions of an individual [73]. This enables the detection of short- and long-term physiological and pathological changes that occur in chronic diseases. However, their results are often difficult to interpret due to various confounders that include lifestyle, medications, and nutritional status [74].
Metabolites may originate from blood or urine. Plasma nonesterified and esterified fatty acids were found to differentiate albuminuria stages in patients with type 2 DM [75]. In a study of 78 diabetic patients, a combination of serum metabolites with multivariate analyses enabled accurate discrimination of patients with DKD. In the study, the selection of five metabolites that included γ-butyrobetaine, symmetric dimethylarginine, azelaic acid, and two unknowns yielded an AUC value of 0.927 for diagnosing DKD [76]. Another study that included healthy controls and patients with type 2 DM indicated that serum metabolite levels of leucine, dihydrosphingosine, and phytosphingosine were significantly different in these two patient groups [77]. In the SUMMIT study, a combination of biomarkers of tubular damage such as KIM-1 and β2-microglobulin, and metabolite markers were used to predict the rapid progression of DKD in individuals with type 2 DM. A total of 207 serum biomarkers were measured, of which 12 biomarkers showed significant associations with rapid progression, all adjusted for clinical characteristics. A combination of 14 serum biomarkers increased the predictive ability. The addition of biomarkers to clinical data improved baseline AUC from 0.706 to 0.868. Biomarkers in the predictive model included fibroblast growth factor-21, the symmetric to asymmetric dimethylarginine ratio, β2-microglobulin, C16-acylcarnitine, and KIM-1 [78].
Urine metabolites that have been studied to date include octanol, oxalic acid, phosphoric acid, benzamide, creatinine, 3,5-dimethoxymandelic amide, and N-acetylglutamine [79]. One study combined both plasma and urinary metabolites to improve the predictive utility of metabolites. In a study of 90 patients with type 2 DM, urine hexose, glutamine, tyrosine, plasma butenoylcarnitine, and histidine levels predicted the development of albuminuria, independent of baseline albuminuria levels, eGFR, and use of RAS blockers [80]. In a study from the CRIC consisting of 1,001 participants with DM and CKD, after adjustments for clinical variables, urinary 3-hydroxyisobutyrate (3-HIBA) and 3-methylcrotonyglycine levels had a significant negative association with eGFR slope, whereas aconitic and citric acid levels showed a positive association. Levels of 3-HIBA and aconitic acid were each associated with higher and lower risks of ESKD requiring kidney replacement therapy, respectively [81]. Most recently, in 2,670 patients with type 1 DM from the Finnish Diabetic Nephropathy study, seven urinary metabolites, which included leucine, valine, isoleucine, pseudouridine, threonine, and citrate, were associated with DKD progression after adjustment for baseline albuminuria and CKD stage. Moreover, 2-hydroxyisobutyrate was associated with the progression of DKD in individuals with normoalbuminuria, and six amino acids and pyroglutamate were associated with the progression of DKD in those with macroalbuminuria [82].
Although there have been significant advances in the field of metabolomics for patients with DKD, most of the aforementioned studies are cross-sectional. Not only is there a need for more prospective studies to evaluate the predictive utility of these metabolites, but replication of current findings in other cohorts is also needed to convey therapeutic targets and improve the clinical management of DKD.

Transcriptomics

Transcriptomic studies of DKD use micro RNAs (miRNAs), which are small non-coding RNAs that block protein translation and can induce messenger RNA degradation. Thus, miRNAs are regarded as regulators of gene expression. Like metabolites, miRNAs may also originate from plasma, serum, or urine, and their profiling can be performed using either traditional microarray/real-time polymerase chain reaction (RT-PCR) platforms or RNA sequencing.
One of the earliest studies to indicate that certain plasma miRNAs showed an association with the progression of DKD was performed in patients with type 1 DM with albuminuria but normal kidney function. In participants of the Joslin cohort, baseline circulating TGF-β1-regulated miRNA levels were associated with progression to ESKD requiring kidney replacement therapy. Baseline miRNA levels of let-7c-5p and miR-29a-3p were independently associated with more than a 50% reduction in the risk of progression to ESKD, whereas baseline miRNA levels of let-7b-5p and miR-21-5p were associated with a more than 2.5-fold increase in the risk of ESKD [83]. In another cross-sectional nested case-control study from the EURODIAB Prospective Complications Study of 455 patients with type 1 DM, miR-126 levels were negatively associated with all DM-related complications. Although this association was no longer significant after adjustment for both hyperglycemia and duration of DM, a statistically significant 25% risk reduction of proliferative diabetic retinopathy was observed even after adjustments for HbA1c and DM duration [84].
In addition to miRNAs sampled from the plasma or serum, many studies have reported findings from urine samples. One of the earliest studies to report on the association between urinary miRNAs and the risk of DKD progression was conducted with 83 patients with type 2 DM, where baseline urinary miRNA levels of miR-29a and miR-29b were associated with complications of DM. Higher levels of urinary miR-29a were observed in patients with albuminuria compared to those with normoalbuminuria. Urinary miR-29a levels showed a significant correlation with albuminuria and were also correlated with carotid intima-media thickness [85]. Another study assessed the urinary extracellular vesicle (EV)-miRNA profiles of patients with type 1 DM, where 22 of 377 urinary EV-miRNAs were differentially expressed in patients with normoalbuminuria compared to albuminuric patients. Results showed that miR-130a and miR-145 were enriched, whereas miR-155 and miR-424 were reduced in urinary exosomes for patients with albuminuria [86]. More recently, in a study of 209 patients with biopsy-proven DKD, urinary miR-196a levels correlated positively with proteinuria, duration of DM, and systolic blood pressure, whereas baseline eGFR and hemoglobin levels showed a negative correlation with urinary miR-196a. This suggests that increased urinary miR-196a levels were significantly associated with the progression of DKD and could be a noninvasive prognostic marker of kidney fibrosis in patients with DKD [87].
Although several other studies have investigated transcriptomics in patients with DKD, there is no overlap in the specific miRNAs being reported as being relevant to DKD. It is most likely that a combination of miRNAs may be needed for the early detection of DKD rather than a single miRNA [88]. Therefore, the evidence to support a clinically useful role of miRNAs in the early diagnosis and risk stratification of DKD remains uncertain.

Current practice and conclusion

Current treatment of DKD relies on lifestyle modification, and medication that controls hyperglycemia, hypertension, and proteinuria. However, even the optimal implementation of this strategy often fails to prevent progression to ESKD in a substantial proportion of patients. There is hope that novel biomarkers, both assay-based and omics-based, will help to identify patients at the highest risk and guide the treatment of these patients. In reality, only a few trials use biomarkers other than albuminuria or eGFR to enroll and risk stratify study participants [89], and even fewer studies assess the effect of treatments with novel biomarkers [9092]. In part, this may be because potential novel biomarkers only modestly improve the performance of eGFR and albuminuria, which are the biomarkers currently available. Moreover, considering that the progression of DKD is usually a slow process that may take decades to emerge, the setting of robust clinical endpoints in clinical trials is often not feasible.
Although the novel biomarkers discussed in this review have enormous potential in the field of DKD, future studies should look into using these biomarkers either as enrolment criteria for randomized clinical trials or as surrogates of study endpoints. Larger study cohorts with kidney biopsies and both urine and plasma or serum samples from the same patients would also be needed. To obtain comparable and reproducible data, consensus protocols for sample collection, processing, and analysis should be defined across collaborators. Finally, as we gain a deeper understanding of the DKD pathophysiology, an increasing number of potential novel biomarkers will be available. To improve the prognostication of patients with DKD, it will be essential to integrate these novel findings and biomarkers into the design of future clinical trials.

Notes

Conflicts of interest

Tae-Hyun Yoo is the Editor-in-Chief of Kidney Research and Clinical Practice and was not involved in the review process of this article. All authors have no other conflicts of interest to declare.

Funding

This work was supported by a National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (NRF-2020R1F1A1049799).

Authors’ contributions

Writing–original draft: CYJ

Writing–review & editing: THY

All authors read and approved the final manuscript.

Acknowledgments

The authors thank the Medical Illustration & Design team of the Medical Research Support Services of Yonsei University College of Medicine for all artistic support related to this work.

Figure 1.

The concept of the omics platform-based approach consists of the genome, transcriptome, proteome, and metabolome.

j-krcp-22-084f1.jpg
Table 1.
Summary of studies reporting on novel biomarkers for diabetic kidney disease (DKD)
Biomarker Study  Sample size Study population   Main results Adjustments
Assay-based novel biomarkers
 Single biomarkers or small sets of biomarkers
  Markers of tubular damage
   Plasma KIM-1, TNFR-1, TNFR-2, and MCP-1 Schrauben et al. [30] Case-cohort study (n = 894) Type 1 and 2 DM Higher plasma levels of KIM-1, TNFR-1, TNFR-2, and MCP-1 were associated with a risk of DKD progression Age, sex, race/ethnicity, education, clinical center, systolic and diastolic blood pressure, BMI, hsCRP, HbA1c, antihypertensive medication use, smoking status, baseline eGFR, and UPCR
   Urinary KIM-1 and NGAL de Carvalho et al. [33] Cross-sectional study (n = 117) Type 2 DM Urinary KIM-1 and NGAL were increased in type 2 DM patients with normal or mildly increased albuminuria HbA1c, LDL cholesterol, fasting glucose, and medication
   Urinary L-FABP Nielsen et al. [34] Prospective cohort study (n = 165) Type 1 DM High levels of urinary L-FABP predict the initiation and progression to DKD and all-cause mortality Age, sex, HbA1c, systolic and diastolic blood pressure, albuminuria, serum creatinine, and smoking
   Urinary L-FABP Kamijo-Ikemori et al. [35] Cross-sectional study (n = 552) Type 2 DM Urinary L-FABP was increased in advanced DKD and was also high in patients with normoalbuminuria A high value of L-FABP at entry, albuminuria, systolic and diastolic blood pressure, HbA1c, age, sex, and use of RAS inhibitors
    Prospective cohort study (n = 104)
   Urinary L-FABP Panduru et al. [36] Prospective cohort study (n = 1,549) Type 1 DM High urinary L-FABP levels were found to be a strong and independent predictor of DKD progression Risk factors of DKD and albuminuria
  Markers of inflammation
   Plasma TNF-α, TNFR1, TNFR2 Niewczas et al. [24] Prospective cohort study (n = 410) Type 2 DM Elevated circulating TNFR levels are strong predictors of progression to ESKD in subjects with and without proteinuria Age, HbA1c, albuminuria, and eGFR
   Serum TNFR Skupien et al. [39] Prospective cohort study (n = 349) Type 1 DM Circulating TNFR2 is a major determinant of kidney function decline No adjustments
   Urinary 8-OHdG Xu et al. [41] Cross-sectional study (n = 69) Type 2 DM Individuals with type 2 DM have higher levels of 8-OHdG compared to healthy individuals No adjustments
   Plasma 8-OHdG Sanchez et al. [42] Prospective cohort study (n = 704) Type 1 DM Higher levels of 8-OHdG were associated with an increased risk of kidney disease Age, sex, cohort, duration of DM, HbA1c, insulin therapy, systolic blood pressure, use of antihypertensive drugs, RAS inhibitors, diabetic retinopathy stage, lipid-lowering drugs, eGFR, and albuminuria
   Urinary 8-OHdG Serdar et al. [43] Cross-sectional study (n = 92) Type 2 DM Although urinary 8-OHdG levels increase in diabetic patients, their levels do not improve the prediction of progressive DKD by measuring albuminuria No adjustments
  Markers of glomerular damage
   Urinary transferrin Kanauchi et al. [44] Cross-sectional study (n = 45) Type 2 DM Increased urinary transferrin levels in microalbuminuria patients significantly correlated with kidney biopsy-proven tubulointerstitial injuries No adjustments
   Urinary transferrin Sánchez-Hidalgo et al. [45] Case-control study (n = 60) Type 2 DM Urinary transferrin levels correlated with subclinical atherogenesis in patients with type 2 DM without nephropathy No adjustments
   Urinary ceruloplasmin Narita et al. [46] Prospective cohort study (n = 140) Type 2 DM Urinary ceruloplasmin levels were found to be increased in normoalbuminuric DM patients and were highly predictive of the development of microalbuminuria Age, sex, BMI, DM duration, HbA1c, systolic and diastolic blood pressure, lipid profile, presence of retinopathy, use of antihypertensive drugs, use of insulin, and urinary lab
   Urinary immunoglobulin G and M Bakoush et al. [47] Case-control study (n = 72) Type 1 and 2 DM Increased urine excretion of immunoglobulin G and M accompanied albuminuria in patients with type 2 DM No adjustments
  Multiple biomarkers
   Plasma KIM-1, TNFR-1, and TNFR-2 Coca et al. [29] Nested case-control study (n = 380) Type 2 DM Doubling in KIM-1, TNFR-1, and TNFR-2 levels were associated with a higher risk of eGFR decline. Combination of all three plasma biomarkers improved the AUC for DKD progression from 0.680 to 0.752 Treatment arm, baseline eGFR, albuminuria, age, race, systolic and diastolic blood pressure, and medications
    Prospective cohort study (n = 1,156)
   NGAL/Creatinine ratio, FABP/Creatinine ratio Fufaa et al. [48] Prospective cohort study (n = 260) Type 2 DM NGAL/creatinine was positively associated with the risk of ESKD and mortality, whereas L-FABP/creatinine was inversely associated with ESKD. The addition of NGAL/creatinine and L-FABP/creatinine to albuminuria and eGFR increased risk prediction for ESKD Age, sex, duration of DM, hypertension, HbA1c, and study cohort
   17 potential urinary and 7 plasma biomarkers Agarwal et al. [49] Prospective cohort study (n = 67) Type 2 DM Urinary C-terminal FGF-23 was found to show the strongest association with ESKD, while plasma VEGF was associated with the highest risk of the composite outcome of ESKD and death Baseline eGFR, log urine albumin/creatinine
   Proteinuria, urinary IL-1β, IL-6, IL-8, MCP-1, TNF-α, TGF-β1, PAI-1 Verhave et al. [50] Prospective cohort study (n = 83) Type 1 and type 2 DM Urinary MCP-1 and TGF-β1 predicted kidney function decline that was independent of albuminuria Albuminuria
   Plasma apoA4, CD5L, C1QB, CFHR2, IBP3 Peters et al. [51] Prospective cohort study (n = 345) Type 2 DM Plasma apoA4, CD5L, and C1QB were independently associated with a rapid decline in kidney function Baseline eGFR, albuminuria, sex, hypertension, HbA1c, smoking, lipid profile, BMI, and RAS inhibitors
 Omics-based novel biomarkers
  Proteomics
   Urinary CKD-273 Currie et al. [58] Prospective cohort study (n = 155) Type 2 DM CKD-273 was associated with mortality in individuals with type 2 DM and microalbuminuria Age, sex, systolic blood pressure, smoking status, eGFR, albuminuria, CAC score, and NT-proBNP
   Urinary CKD-273 Lindhardt et al. [60] Prospective cohort study (n = 737) Type 2 DM CKD-273 predicted the development of albuminuria Treatment group, age, sex, systolic blood pressure, albuminuria, eGFR, HbA1c, and DM duration
   Urinary CKD-273, α1 type 1 collagen chain Zürbig et al. [62] Prospective cohort study (n = 35) Type 1 and 2 DM CKD-273 predicted progression to macroalbuminuria 5 years before actual onset Age, sex, DM type, albuminuria, eGFR, systolic and diastolic blood pressure, HbA1c, and glucose
   Urinary CKD-273 Roscioni et al. [63] Prospective cohort study (n = 88) Type 2 DM CKD-273 predicted the development of albuminuria independent of other kidney biomarkers used to predict DKD development or progression Albuminuria, eGFR, and use of RAS inhibitors
   Urinary CKD-273 Zürbig et al. [67] Prospective cohort study (n = 1,014) Type 1 and 2 DM In patients with type 1 or 2 DM, baseline eGFR ≥ 70 mL/min/1.73 m2, and normoalbuminuria, CKD-273 was able to identify progression to eGFR < 60 mL/min/1.73 m2 in the absence of albuminuria Age, baseline eGFR, and systolic and diastolic blood pressure
   Urinary CKD-273 Tofte et al. [68] Prospective cohort study (n = 1,775) Type 2 DM High-risk patients defined by CKD-273 were more likely to develop microalbuminuria Age, sex, HbA1c, systolic blood pressure, retinopathy, albuminuria, eGFR
   Urinary CKD-273 Siwy et al. [69] Prospective cohort study (n = 360) Type 2 DM There was a significant correlation between CKD-273 and baseline eGFR, albuminuria, as well as with eGFR decline No adjustments
   Urinary type I and III α1 collagen, α2-HS-glycoprotein Rossing et al. [70] Prospective cohort study (n = 165) Type 2 DM Type I and type III α1 collagen and α2-HS glycoprotein were found to be prominent collagen markers in patients with diabetic renal damage No adjustments
   Urinary uromodulin, progranulin, clusterin, and α1 acid glycoprotein Schlatzer et al. [71] Nested case-control study (n = 465) Type 1 DM A panel including uromodulin, progranulin, clusterin, and α1 acid glycoprotein predicted early eGFR decline Baseline age, DM duration, albuminuria, HbA1c, cystatin C, and uric acid
   Urinary haptoglobin Bhensdadia et al. [72] Prospective cohort study (n = 204) Type 2 DM The haptoglobin to creatinine ratio may be useful in predicting the risk of DKD before the development of albuminuria or kidney function decline Treatment arm, use of ACEi
  Metabolomics
   35 serum non-esterified and 32 serum esterified fatty acids Han et al. [75] Cross-sectional study (n = 150) Type 2 DM Non-esterified and esterified fatty acid discriminated albuminuria stages No adjustments
   19 serum metabolites Hirayama et al. [76] Cross-sectional study (n = 156) Type 2 DM Combination of 19 serum metabolites enabled accurate discrimination of DKD No adjustments
   Serum leucine, dihydrosphingosine, phytosphingosine Zhang et al. [77] Cross-sectional study (n = 66) Type 2 DM Serum metabolite levels of leucine, dihydrosphingosine, and phytosphingosine were significantly different in patients with type 2 DM and healthy controls No adjustments
   207 serum biomarkers Colombo et al. [78] Nested case-control study (n = 840) Type 2 DM 12 biomarkers showed significant associations with a rapid decline in eGFR, of which serum KIM-1 and B2M showed the most robust association Age, sex, baseline eGFR, albuminuria, HbA1c, calendar time, and treatment allocation
   Urine hexose, glutamine, tyrosine, plasma butenoylcarnitine, histidine Pena et al. [80] Prospective cohort study (n = 90) Type 2 DM Urine hexose, glutamine, tyrosine, plasma butenoylcarnitine, and histidine predicted the development of albuminuria Albuminuria, eGFR, and RAS inhibitors
   Urinary 3-hydroxy-isobutyrate, 3-methyl-crotonyglycine, aconitic acid, citric acid Kwan et al. [81] Prospective cohort study (n = 1,001) Type 1 and 2 DM 3-hydroxyisobutyrate and 3-methylcrotonyglycine had a significant negative association with eGFR slope, while aconitic and citric acid showed a positive association Age, race, sex, smoking, body mass index, HbA1c, mean arterial pressure, albuminuria, and baseline eGFR
   Urinary leucine, valine, isoleucine, pseudouridine, threonine, citrate, 2-hydroxyiso-butyrate, pyroglutamate, tyrosine, alanine Mutter et al. [82] Prospective cohort study (n = 2,670) Type 1 DM 7 urinary metabolites that included leucine, valine, isoleucine, pseudouridine, threonine, and citrate were associated with DKD progression. 6 amino acids and pyroglutamate were associated with DKD progression in those with macroalbuminuria Baseline albuminuria, baseline glycemic control, and CKD stage
  Transcriptomics
   Plasma let-7c-5p, miR-29a-3p, let-7b-5p, miR-21-5p, miR-29c-3p Pezzolesi et al. [83] Prospective cohort study (n = 116) Type 1 DM Baseline miRNA levels of let-7c-5p and miR-29a-3p were independently associated with more than a 50% reduction in the risk of rapid progression to ESKD, while levels of let-7b-5p and miR-21-5p were associated with a higher risk of ESKD Age, sex, HbA1c, and duration of DM
   Plasma miR-126 Barutta et al. [84] Nested case-control study (n = 455) Type 1 DM miR-126 levels were negatively associated with all DM-related complications. A 25% risk reduction of proliferative diabetic retinopathy was observed even after adjustments for HbA1c and DM duration HbA1c, DM duration
   Urinary miR-29a, miR-29b Peng et al. [85] Cross-sectional study (n = 83) Type 2 DM Baseline urinary miR-29a and miR-29b were associated with complications of DM such as albuminuria and carotid intima-media thickness No adjustments
   miR-130a, miR-145, miR-155, miR-424 Barutta et al. [86] Cross-sectional study (n = 24) Type 1 DM 22 of 377 urinary EV-miRNAs were differentially expressed in patients with normoalbuminuria compared to albuminuric patients No adjustments
   Urinary miR-196a An et al. [87] Prospective cohort study (n = 209) Type 2 DM Urinary miR-196ta levels correlated positively with proteinuria, duration of DM, and systolic blood pressure, whereas baseline eGFR and hemoglobin levels showed a negative correlation with urinary miR-196a Age, sex, BMI, mean arterial pressure, and HbA1c

apoA4, apolipoprotein A4; AUC, area under curve; B2M, β2-microglobulin; BMI, body mass index; C1QB, complement C1q subcomponent subunit B; CAC, coronary artery calcium; CD5L, CD5 antigen-like; CFHR2, complement factor H-related protein 2; CKD-273, chronic kidney disease 273; DM, diabetes mellitus; eGFR, estimated glomerular filtration rate; ESKD, end-stage kidney disease; EV, extracellular vesicles; FGF-23, fibroblast growth factor-23; HbA1c, hemoglobin A1c; hsCRP, high-sensitivity C-reactive protein; IBP3, insulin-like growth factor-binding protein 3; IL, interleukin; KIM-1, kidney injury molecule 1; L-FABP, liver fatty acid-binding protein; LDL, low-density lipoprotein; MCP-1, monocyte chemoattractant protein 1; NGAL, neutrophil gelatinase-associated lipocalin; NT-proBNP, N-terminal prohormone of brain natriuretic peptide; PAI-1, plasminogen activator inhibitor-1; RAS, renin-angiotensin system; TGF-β1, transforming growth factor-beta 1; TNF-α, tissue necrosis factor-α; TNFR-1, tissue necrosis factor receptor 1; TNFR-2, tissue necrosis factor receptor 2; UPCR, urine protein-to-creatinine ratio; VEGF, vascular endothelial growth factor; α2-HS, alpha-2-Heremans-Schmid; 8-OHdG, 8-hydroxydeoxyguanosine.

References

1. Alicic RZ, Rooney MT, Tuttle KR. Diabetic kidney disease: challenges, progress, and possibilities. Clin J Am Soc Nephrol 2017;12:2032–2045.
pmid pmc
2. Gregg EW, Li Y, Wang J, et al. Changes in diabetes-related complications in the United States, 1990-2010. N Engl J Med 2014;370:1514–1523.
crossref pmid
3. Warren AM, Knudsen ST, Cooper ME. Diabetic nephropathy: an insight into molecular mechanisms and emerging therapies. Expert Opin Ther Targets 2019;23:579–591.
crossref pmid
4. Nangaku M, Kanda H, Takama H, Ichikawa T, Hase H, Akizawa T. Randomized clinical trial on the effect of bardoxolone methyl on GFR in diabetic kidney disease patients (TSUBAKI Study). Kidney Int Rep 2020;5:879–890.
crossref pmid pmc
5. de Zeeuw D, Akizawa T, Audhya P, et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 2013;369:2492–2503.
crossref pmid pmc
6. Liu YN, Zhou J, Li T, et al. Sulodexide protects renal tubular epithelial cells from oxidative stress-induced injury via upregulating klotho expression at an early stage of diabetic kidney disease. J Diabetes Res 2017;2017:4989847.
crossref pmid pmc pdf
7. Packham DK, Wolfe R, Reutens AT, et al. Sulodexide fails to demonstrate renoprotection in overt type 2 diabetic nephropathy. J Am Soc Nephrol 2012;23:123–130.
crossref pmid pmc
8. Lewis EJ, Lewis JB, Greene T, et al. Sulodexide for kidney protection in type 2 diabetes patients with microalbuminuria: a randomized controlled trial. Am J Kidney Dis 2011;58:729–736.
crossref pmid
9. Stevens PE, Levin A; Kidney Disease: Improving Global Outcomes Chronic Kidney Disease Guideline Development Work Group Members. Evaluation and management of chronic kidney disease: synopsis of the kidney disease: improving global outcomes 2012 clinical practice guideline. Ann Intern Med 2013;158:825–830.
crossref pmid
10. Berhane AM, Weil EJ, Knowler WC, Nelson RG, Hanson RL. Albuminuria and estimated glomerular filtration rate as predictors of diabetic end-stage renal disease and death. Clin J Am Soc Nephrol 2011;6:2444–2451.
crossref pmid pmc
11. Caramori ML, Fioretto P, Mauer M. Low glomerular filtration rate in normoalbuminuric type 1 diabetic patients: an indicator of more advanced glomerular lesions. Diabetes 2003;52:1036–1040.
pmid
12. An JH, Cho YM, Yu HG, et al. The clinical characteristics of normoalbuminuric renal insufficiency in Korean type 2 diabetic patients: a possible early stage renal complication. J Korean Med Sci 2009;24:S75–S81.
crossref pmid pmc
13. Macisaac RJ, Jerums G. Diabetic kidney disease with and without albuminuria. Curr Opin Nephrol Hypertens 2011;20:246–257.
crossref pmid
14. Afghahi H, Miftaraj M, Svensson AM, et al. Ongoing treatment with renin-angiotensin-aldosterone-blocking agents does not predict normoalbuminuric renal impairment in a general type 2 diabetes population. J Diabetes Complications 2013;27:229–234.
crossref pmid
15. Porrini E, Ruggenenti P, Mogensen CE, et al. Non-proteinuric pathways in loss of renal function in patients with type 2 diabetes. Lancet Diabetes Endocrinol 2015;3:382–391.
crossref pmid
16. Molitch ME, Steffes M, Sun W, et al. Development and progression of renal insufficiency with and without albuminuria in adults with type 1 diabetes in the diabetes control and complications trial and the epidemiology of diabetes interventions and complications study. Diabetes Care 2010;33:1536–1543.
crossref pmid pmc pdf
17. Penno G, Russo E, Garofolo M, et al. Evidence for two distinct phenotypes of chronic kidney disease in individuals with type 1 diabetes mellitus. Diabetologia 2017;60:1102–1113.
crossref pmid pdf
18. Vistisen D, Andersen GS, Hulman A, Persson F, Rossing P, Jørgensen ME. Progressive decline in estimated glomerular filtration rate in patients with diabetes after moderate loss in kidney function-even without albuminuria. Diabetes Care 2019;42:1886–1894.
crossref pmid pdf
19. Jones RH, Hayakawa H, Mackay JD, Parsons V, Watkins PJ. Progression of diabetic nephropathy. Lancet 1979;1:1105–1106.
crossref pmid
20. Moazzeni SS, Arani RH, Hasheminia M, Tohidi M, Azizi F, Hadaegh F. High incidence of chronic kidney disease among iranian diabetic adults: using CKD-EPI and MDRD equations for estimated glomerular filtration rate. Diabetes Metab J 2021;45:684–697.
crossref pmid pmc pdf
21. Tonneijck L, Muskiet MH, Smits MM, et al. Glomerular hyperfiltration in diabetes: mechanisms, clinical significance, and treatment. J Am Soc Nephrol 2017;28:1023–1039.
crossref pmid pmc
22. Colhoun HM, Marcovecchio ML. Biomarkers of diabetic kidney disease. Diabetologia 2018;61:996–1011.
crossref pmid pmc pdf
23. Pena MJ, de Zeeuw D, Mischak H, et al. Prognostic clinical and molecular biomarkers of renal disease in type 2 diabetes. Nephrol Dial Transplant 2015;30 Suppl 4:iv86–iv95.
crossref pmid
24. Niewczas MA, Gohda T, Skupien J, et al. Circulating TNF receptors 1 and 2 predict ESRD in type 2 diabetes. J Am Soc Nephrol 2012;23:507–515.
crossref pmid pmc
25. Titan SM, Zatz R, Graciolli FG, et al. FGF-23 as a predictor of renal outcome in diabetic nephropathy. Clin J Am Soc Nephrol 2011;6:241–247.
crossref pmid pmc
26. Russo GT, De Cosmo S, Viazzi F, et al. Plasma triglycerides and HDL-C levels predict the development of diabetic kidney disease in subjects with type 2 diabetes: the AMD Annals Initiative. Diabetes Care 2016;39:2278–2287.
crossref pmid pdf
27. Fauconnier C, Roy T, Gillerot G, Roy C, Pouleur AC, Gruson D. FGF23: clinical usefulness and analytical evolution. Clin Biochem 2019;66:1–12.
crossref pmid
28. Han WK, Bailly V, Abichandani R, Thadhani R, Bonventre JV. Kidney Injury Molecule-1 (KIM-1): a novel biomarker for human renal proximal tubule injury. Kidney Int 2002;62:237–244.
crossref pmid
29. Coca SG, Nadkarni GN, Huang Y, et al. Plasma biomarkers and kidney function decline in early and established diabetic kidney disease. J Am Soc Nephrol 2017;28:2786–2793.
crossref pmid pmc
30. Schrauben SJ, Shou H, Zhang X, et al. Association of multiple plasma biomarker concentrations with progression of prevalent diabetic kidney disease: findings from the Chronic Renal Insufficiency Cohort (CRIC) Study. J Am Soc Nephrol 2021;32:115–126.
crossref pmid pmc
31. Yürük Yıldırım Z, Nayır A, Yılmaz A, Gedikbaşı A, Bundak R. Neutrophil gelatinase-associated lipocalin as an early sign of diabetic kidney injury in children. J Clin Res Pediatr Endocrinol 2015;7:274–279.
crossref pmid pmc
32. Lacquaniti A, Donato V, Pintaudi B, et al. “Normoalbuminuric” diabetic nephropathy: tubular damage and NGAL. Acta Diabetol 2013;50:935–942.
crossref pmid pdf
33. de Carvalho JA, Tatsch E, Hausen BS, et al. Urinary kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin as indicators of tubular damage in normoalbuminuric patients with type 2 diabetes. Clin Biochem 2016;49:232–236.
crossref pmid
34. Nielsen SE, Sugaya T, Hovind P, Baba T, Parving HH, Rossing P. Urinary liver-type fatty acid-binding protein predicts progression to nephropathy in type 1 diabetic patients. Diabetes Care 2010;33:1320–1324.
crossref pmid pmc pdf
35. Kamijo-Ikemori A, Sugaya T, Yasuda T, et al. Clinical significance of urinary liver-type fatty acid-binding protein in diabetic nephropathy of type 2 diabetic patients. Diabetes Care 2011;34:691–696.
crossref pmid pmc pdf
36. Panduru NM, Forsblom C, Saraheimo M, et al. Urinary liver-type fatty acid-binding protein and progression of diabetic nephropathy in type 1 diabetes. Diabetes Care 2013;36:2077–2083.
crossref pmid pmc pdf
37. Hasegawa G, Nakano K, Sawada M, et al. Possible role of tumor necrosis factor and interleukin-1 in the development of diabetic nephropathy. Kidney Int 1991;40:1007–1012.
crossref pmid
38. Kalantarinia K, Awad AS, Siragy HM. Urinary and renal interstitial concentrations of TNF-alpha increase prior to the rise in albuminuria in diabetic rats. Kidney Int 2003;64:1208–1213.
pmid
39. Skupien J, Warram JH, Niewczas MA, et al. Synergism between circulating tumor necrosis factor receptor 2 and HbA(1c) in determining renal decline during 5-18 years of follow-up in patients with type 1 diabetes and proteinuria. Diabetes Care 2014;37:2601–2068.
crossref pmid pmc pdf
40. 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;339:1–9.
crossref pmid
41. Xu GW, Yao QH, Weng QF, Su BL, Zhang X, Xiong JH. Study of urinary 8-hydroxydeoxyguanosine as a biomarker of oxidative DNA damage in diabetic nephropathy patients. J Pharm Biomed Anal 2004;36:101–104.
crossref pmid
42. Sanchez M, Roussel R, Hadjadj S, et al. Plasma concentrations of 8-hydroxy-2'-deoxyguanosine and risk of kidney disease and death in individuals with type 1 diabetes. Diabetologia 2018;61:977–984.
crossref pmid pdf
43. Serdar M, Sertoglu E, Uyanik M, et al. Comparison of 8-hydroxy-2'-deoxyguanosine (8-OHdG) levels using mass spectrometer and urine albumin creatinine ratio as a predictor of development of diabetic nephropathy. Free Radic Res 2012;46:1291–1295.
crossref pmid
44. Kanauchi M, Akai Y, Hashimoto T. Transferrinuria in type 2 diabetic patients with early nephropathy and tubulointerstitial injury. Eur J Intern Med 2002;13:190–193.
crossref pmid
45. Sánchez-Hidalgo JJ, Suárez-Cuenca JA, Lozano-Nuevo JJ, et al. Urine transferrin as an early endothelial dysfunction marker in type 2 diabetic patients without nephropathy: a case control study. Diabetol Metab Syndr 2021;13:128.
pmid pmc
46. Narita T, Hosoba M, Kakei M, Ito S. Increased urinary excretions of immunoglobulin g, ceruloplasmin, and transferrin predict development of microalbuminuria in patients with type 2 diabetes. Diabetes Care 2006;29:142–144.
crossref pmid pdf
47. Bakoush O, Tencer J, Tapia J, Rippe B, Torffvit O. Higher urinary IgM excretion in type 2 diabetic nephropathy compared to type 1 diabetic nephropathy. Kidney Int 2002;61:203–208.
crossref pmid
48. Fufaa GD, Weil EJ, Nelson RG, et al. Association of urinary KIM-1, L-FABP, NAG and NGAL with incident end-stage renal disease and mortality in American Indians with type 2 diabetes mellitus. Diabetologia 2015;58:188–198.
crossref pmid pmc pdf
49. Agarwal R, Duffin KL, Laska DA, Voelker JR, Breyer MD, Mitchell PG. A prospective study of multiple protein biomarkers to predict progression in diabetic chronic kidney disease. Nephrol Dial Transplant 2014;29:2293–2302.
crossref pmid
50. Verhave JC, Bouchard J, Goupil R, et al. Clinical value of inflammatory urinary biomarkers in overt diabetic nephropathy: a prospective study. Diabetes Res Clin Pract 2013;101:333–340.
crossref pmid
51. Peters KE, Davis WA, Ito J, et al. Identification of novel circulating biomarkers predicting rapid decline in renal function in type 2 diabetes: the Fremantle Diabetes Study Phase II. Diabetes Care 2017;40:1548–1555.
crossref pmid pdf
52. Looker HC, Colombo M, Hess S, et al. Biomarkers of rapid chronic kidney disease progression in type 2 diabetes. Kidney Int 2015;88:888–896.
crossref pmid
53. Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol 2017;18:83.
crossref pmid pmc pdf
54. Lindsey ML, Mayr M, Gomes AV, et al. Transformative impact of proteomics on cardiovascular health and disease: a scientific statement from the American Heart Association. Circulation 2015;132:852–872.
crossref pmid
55. Wilkins MR, Pasquali C, Appel RD, et al. From proteins to proteomes: large scale protein identification by two-dimensional electrophoresis and amino acid analysis. Biotechnology (N Y) 1996;14:61–65.
pmid
56. Liu Y. Cellular and molecular mechanisms of renal fibrosis. Nat Rev Nephrol 2011;7:684–696.
crossref pmid pmc pdf
57. Dubin RF, Rhee EP. Proteomics and metabolomics in kidney disease, including insights into etiology, treatment, and prevention. Clin J Am Soc Nephrol 2020;15:404–411.
crossref pmid pmc
58. Currie GE, von Scholten BJ, Mary S, et al. Urinary proteomics for prediction of mortality in patients with type 2 diabetes and microalbuminuria. Cardiovasc Diabetol 2018;17:50.
crossref pmid pmc pdf
59. Magagnotti C, Zerbini G, Fermo I, et al. Identification of nephropathy predictors in urine from children with a recent diagnosis of type 1 diabetes. J Proteomics 2019;193:205–216.
crossref pmid
60. Lindhardt M, Persson F, Zürbig P, et al. Urinary proteomics predict onset of microalbuminuria in normoalbuminuric type 2 diabetic patients, a sub-study of the DIRECT-Protect 2 study. Nephrol Dial Transplant 2017;32:1866–1873.
crossref pmid
61. Good DM, Zürbig P, Argilés A, et al. Naturally occurring human urinary peptides for use in diagnosis of chronic kidney disease. Mol Cell Proteomics 2010;9:2424–2437.
crossref pmid pmc
62. Zürbig P, Jerums G, Hovind P, et al. Urinary proteomics for early diagnosis in diabetic nephropathy. Diabetes 2012;61:3304–3313.
crossref pmid pmc pdf
63. Roscioni SS, de Zeeuw D, Hellemons ME, et al. A urinary peptide biomarker set predicts worsening of albuminuria in type 2 diabetes mellitus. Diabetologia 2013;56:259–267.
crossref pmid pdf
64. Chaturvedi N, Porta M, Klein R, et al. Effect of candesartan on prevention (DIRECT-Prevent 1) and progression (DIRECT-Protect 1) of retinopathy in type 1 diabetes: randomised, placebo-controlled trials. Lancet 2008;372:1394–1402.
crossref pmid
65. Sjølie AK, Klein R, Porta M, et al. Effect of candesartan on progression and regression of retinopathy in type 2 diabetes (DIRECT-Protect 2): a randomised placebo-controlled trial. Lancet 2008;372:1385–1393.
crossref pmid
66. Pontillo C, Zhang ZY, Schanstra JP, et al. Prediction of chronic kidney disease stage 3 by CKD273, a urinary proteomic biomarker. Kidney Int Rep 2017;2:1066–1075.
crossref pmid pmc
67. Zürbig P, Mischak H, Menne J, Haller H. CKD273 enables efficient prediction of diabetic nephropathy in nonalbuminuric patients. Diabetes Care 2019;42:e4.
crossref pmid pdf
68. Tofte N, Lindhardt M, Adamova K, et al. Early detection of diabetic kidney disease by urinary proteomics and subsequent intervention with spironolactone to delay progression (PRIORITY): a prospective observational study and embedded randomised placebo-controlled trial. Lancet Diabetes Endocrinol 2020;8:301–312.
pmid
69. Siwy J, Klein T, Rosler M, von Eynatten M. Urinary proteomics as a tool to identify kidney responders to dipeptidyl peptidase-4 inhibition: a hypothesis-generating analysis from the MARLINA-T2D Trial. Proteomics Clin Appl 2019;13:e1800144.
crossref pmid pdf
70. Rossing K, Mischak H, Parving HH, et al. Impact of diabetic nephropathy and angiotensin II receptor blockade on urinary polypeptide patterns. Kidney Int 2005;68:193–205.
crossref pmid
71. Schlatzer D, Maahs DM, Chance MR, et al. Novel urinary protein biomarkers predicting the development of microalbuminuria and renal function decline in type 1 diabetes. Diabetes Care 2012;35:549–555.
crossref pmid pmc pdf
72. Bhensdadia NM, Hunt KJ, Lopes-Virella MF, et al. Urine haptoglobin levels predict early renal functional decline in patients with type 2 diabetes. Kidney Int 2013;83:1136–1143.
crossref pmid pmc
73. Gerszten RE, Wang TJ. The search for new cardiovascular biomarkers. Nature 2008;451:949–952.
crossref pmid pdf
74. Hocher B, Adamski J. Metabolomics for clinical use and research in chronic kidney disease. Nat Rev Nephrol 2017;13:269–284.
crossref pmid pdf
75. Han LD, Xia JF, Liang QL, et al. Plasma esterified and non-esterified fatty acids metabolic profiling using gas chromatography-mass spectrometry and its application in the study of diabetic mellitus and diabetic nephropathy. Anal Chim Acta 2011;689:85–91.
crossref pmid
76. Hirayama A, Nakashima E, Sugimoto M, et al. Metabolic profiling reveals new serum biomarkers for differentiating diabetic nephropathy. Anal Bioanal Chem 2012;404:3101–3109.
crossref pmid pdf
77. Zhang J, Yan L, Chen W, et al. Metabonomics research of diabetic nephropathy and type 2 diabetes mellitus based on UPLC-oaTOF-MS system. Anal Chim Acta 2009;650:16–22.
crossref pmid
78. Colombo M, Looker HC, Farran B, et al. Serum kidney injury molecule 1 and β2-microglobulin perform as well as larger biomarker panels for prediction of rapid decline in renal function in type 2 diabetes. Diabetologia 2019;62:156–168.
crossref pmid pmc pdf
79. Ng DP, Salim A, Liu Y, et al. A metabolomic study of low estimated GFR in non-proteinuric type 2 diabetes mellitus. Diabetologia 2012;55:499–508.
crossref pmid pdf
80. Pena MJ, Lambers Heerspink HJ, Hellemons ME, et al. Urine and plasma metabolites predict the development of diabetic nephropathy in individuals with type 2 diabetes mellitus. Diabet Med 2014;31:1138–1147.
crossref pmid
81. Kwan B, Fuhrer T, Zhang J, et al. Metabolomic markers of kidney function decline in patients with diabetes: evidence from the Chronic Renal Insufficiency Cohort (CRIC) Study. Am J Kidney Dis 2020;76:511–520.
crossref pmid pmc
82. Mutter S, Valo E, Aittomäki V, et al. Urinary metabolite profiling and risk of progression of diabetic nephropathy in 2670 individuals with type 1 diabetes. Diabetologia 2022;65:140–149.
crossref pmid pmc pdf
83. Pezzolesi MG, Satake E, McDonnell KP, Major M, Smiles AM, Krolewski AS. Circulating TGF-β1-regulated miRNAs and the risk of rapid progression to ESRD in type 1 diabetes. Diabetes 2015;64:3285–3293.
crossref pmid pmc pdf
84. Barutta F, Bruno G, Matullo G, et al. MicroRNA-126 and micro-/macrovascular complications of type 1 diabetes in the EURODIAB Prospective Complications Study. Acta Diabetol 2017;54:133–139.
crossref pmid pdf
85. Peng H, Zhong M, Zhao W, et al. Urinary miR-29 correlates with albuminuria and carotid intima-media thickness in type 2 diabetes patients. PLoS One 2013;8:e82607.
crossref pmid pmc
86. Barutta F, Tricarico M, Corbelli A, et al. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One 2013;8:e73798.
crossref pmid pmc
87. An Y, Zhang C, Xu F, et al. Increased urinary miR-196a level predicts the progression of renal injury in patients with diabetic nephropathy. Nephrol Dial Transplant 2020;35:1009–1016.
crossref pmid pdf
88. Kato M. Noncoding RNAs as therapeutic targets in early stage diabetic kidney disease. Kidney Res Clin Pract 2018;37:197–209.
crossref pmid pmc
89. Lindhardt M, Persson F, Currie G, et al. Proteomic prediction and renin angiotensin aldosterone system inhibition prevention of early diabetic nephropathy in type 2 diabetic patients with normoalbuminuria (PRIORITY): essential study design and rationale of a randomised clinical multicentre trial. BMJ Open 2016;6:e010310.
crossref pmid pmc
90. Steno Diabetes Center Copenhagen. Effects of dapagliflozin treatment on urinary proteomic patterns in patients with type 2 diabetes (DapKid) [Internet]. Herlev (Denmark): Steno Diabetes Center Copenhagen, c2018 [cited 2022 Mar 12]. Available from: https://clinicaltrials.gov/ct2/show/NCT02914691.
91. VU University Medical Center. Renoprotective effects of dapagliflozin in type 2 diabetes (RED) [Internet]. Amsterdam (Netherlands): VU University Medical Center, c2020 [cited 2022 Mar 12]. Available from: https://clinicaltrials.gov/ct2/show/NCT02682563.
92. Nanjing First Hospital. Ivabradine to treat microalbuminuria in patients with type 2 diabetes and coronary heart disease (BENCH) [Internet]. Nanjing (China): Nanjing First Hospital, c2020 [cited Mar 12]. Available from: https://clinicaltrials.gov/ct2/show/NCT03105219.


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
#301, (Miseung Bldg.) 23, Apgujenog-ro 30-gil, Gangnam-gu, Seoul 06022, Korea
Tel: +82-2-3486-8736    Fax: +82-2-3486-8737    E-mail: registry@ksn.or.kr                

Copyright © 2024 by The Korean Society of Nephrology.

Developed in M2PI

Close layer