Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Jul 25, 2016; 7(14): 290-301
Published online Jul 25, 2016. doi: 10.4239/wjd.v7.i14.290
Early detection of diabetic kidney disease: Present limitations and future perspectives
Chih-Hung Lin, Yi-Cheng Chang, Lee-Ming Chuang, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
Chih-Hung Lin, Lee-Ming Chuang, Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
Yi-Cheng Chang, Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100, Taiwan
Yi-Cheng Chang, Institute of Biomedical Science, Academia Sinica, Taipei 115, Taiwan
Author contributions: Lin CH and Chang YC drafted the manuscript and tables; Chuang LM conceived the outlines and approved the final manuscript.
Conflict-of-interest statement: The authors declare no conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Lee-Ming Chuang, MD, PhD, Department of Internal Medicine, National Taiwan University Hospital, 7, Chung Shan S. Rd, Taipei 100, Taiwan. leeming@ntu.edu.tw
Telephone: +886-2-23123456 Fax: +886-2-23938859
Received: March 28, 2016
Peer-review started: March 28, 2016
First decision: May 16, 2016
Revised: May 29, 2016
Accepted: June 27, 2016
Article in press: June 29, 2016
Published online: July 25, 2016

Abstract

Diabetic kidney disease (DKD) is one of the most common diabetic complications, as well as the leading cause of chronic kidney disease and end-stage renal disease around the world. To prevent the dreadful consequence, development of new assays for diagnostic of DKD has always been the priority in the research field of diabetic complications. At present, urinary albumin-to-creatinine ratio and estimated glomerular filtration rate (eGFR) are the standard methods for assessing glomerular damage and renal function changes in clinical practice. However, due to diverse tissue involvement in different individuals, the so-called “non-albuminuric renal impairment” is not uncommon, especially in patients with type 2 diabetes. On the other hand, the precision of creatinine-based GFR estimates is limited in hyperfiltration status. These facts make albuminuria and eGFR less reliable indicators for early-stage DKD. In recent years, considerable progress has been made in the understanding of the pathogenesis of DKD, along with the elucidation of its genetic profiles and phenotypic expression of different molecules. With the help of ever-evolving technologies, it has gradually become plausible to apply the thriving information in clinical practice. The strength and weakness of several novel biomarkers, genomic, proteomic and metabolomic signatures in assisting the early diagnosis of DKD will be discussed in this article.

Key Words: Diabetic kidney disease, Early diagnosis, Genomics, Biomarkers

Core tip: Estimated glomerular filtration rate (eGFR) and albuminuria are currently the standard method for detecting diabetic kidney disease (DKD). Creatinine-based GFR estimates are affected by muscle mass and diet pattern, as well as the formula chosen. Albuminuria majorly reflects glomerular dysfunction, and is less sensitive to tubulointerstitial and vascular damages. These facts limit the application of eGFR and albuminuria in the early diagnosis of DKD, especially in heterogeneous type 2 diabetic patients. Through the assistance of genetic information for screening of susceptible patients, together with novel biomarkers to reflect diverse renal tissue damage, early diagnosis of DKD could be facilitated.



INTRODUCTION

Diabetes mellitus is currently one of the most rapidly-growing “epidemics” around the world. According to the International Diabetes Federation, 415 million people are currently affected by this disease worldwide[1]. By the year 2040, the patient number is expected to rise up to 642 million, reaching a global prevalence of 10%[1]. This increasing number of patients, mostly with type 2 diabetes mellitus (T2DM), has influenced the rate of diabetic complications, including diabetic kidney disease (DKD). In developed countries, DKD is one of the most common complications of both type 1 diabetes mellitus (T1DM) and T2DM[2], and is also the leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD)[3-5]. The costs of care for patients with DKD are extremely high, especially after they enter ESRD. In the United States, for the patients covered by Medicare, the average cost per person per year was USD 20000, whereas it was USD 40000 in the younger group (below 65 years of age)[2]. This leads to an increasing burden on the finance and health care systems. Therefore, different methods for identification and management of patients with DKD, especially in the early stages, have always been the priority in the research field of diabetic complications. At present, diagnosis of DKD in clinical settings relies upon the assessment of kidney function, usually by calculating estimated glomerular filtration rate (eGFR), and the assessment of kidney damage, usually by checking urinary albumin-to-creatinine ratio [UACR, urine albumin (mg/L)/urine creatinine (mmol/L)] in random spot urine samples[6]. Although these tests can be performed easily, they have certain limitations. Therefore, understanding these limitations is important to both clinical applications and the future quest for better diagnostic methods.

NATURAL HISTORY OF DKD

The first clinical sign suggestive of DKD is glomerular hyperfiltration, which is observed in about 70% and 50% of the patients with T1DM and T2DM, respectively[7]. Due to the increased intraglomerular pressure, the elevation in GFR may exceed 120 mL/min per 1.73 m2[8]. In some patients, hyperfiltration is followed by the development of albuminuria. Most patients with T1DM have a normal UACR (< 3.4 mg/mmol) during the first 5 years after the disease onset. In the subsequent 10-15 years, albuminuria develops in some patients, and progresses gradually if no intervention is taken. Once UACR is over 34 mg/mmol, the GFR decreases progressively at a variable rate. Approximately 50% of the patients with UACR > 34 mg/mmol progress to ESRD over a period of 10 years and approximately 75% of the patients over a period of 20 years[6]. In patients with T2DM, however, the natural course of DKD is less understood, as the diagnosis is usually delayed by many years. Some patients already display various degrees of albuminuria at the time of diagnosis; however, only 20% of the patients with UACR > 34 mg/mmol progress to ESRD over a period of 20 years[9,10].

LIMITATIONS OF EGFR

In terms of renal excretory functions, GFR is considered the best overall index. However, due to its time-consuming nature, the measurement of 24-h creatinine clearance to assess GFR is not always easily performed in clinical settings. Instead, to assess renal function, calculating eGFR using serum creatinine level and formulae such as the modification of diet in renal disease [MDRD, eGFR = 175 × standardized Scr-1.154× age-0.203× 1.212 (if black) × 0.742 (if female), where Scr is serum creatinine][11] or the chronic kidney disease epidemiology collaboration [CKD-EPI, eGFR = 141 × min (Scr/k, 1)α× max (Scr/k, 1)-1.209× 0.993Age× 1.018 (if female) × 1.159 (if black), where k is 0.7 for females and 0.9 for males, α is -0.329 for females and -0.411 for males, min indicates the minimum of Scr/k or 1, and max indicates the maximum of Scr/k or 1][12] equations has become a routine practice. The National Kidney Foundation uses eGFR to classify stages of CKD[13]. Nonetheless, there are some potential flaws in using eGFR as a marker for the early diagnosis of DKD. First, serum creatinine levels are affected by the muscle mass and diet pattern (especially meat intake)[14,15], and therefore may interfere with the eGFR calculation. Second, the formula used may also cause imprecision in certain conditions. The MDRD equations become less reliable in patients with GFR > 60 mL/min per 1.73 m2[16,17]. This would cause a considerable problem in the early diagnosis of DKD, as glomerular hyperfiltration appears early in the course of the disease. The CKD-EPI equation, on the other hand, is more accurate in patients whose GFR is > 90 mL/min per 1.73 m2[18] and is, therefore, preferred when applying it in patients with diabetes[6]. However, Camargo et al[19] reported a marked underestimation of GFR calculated with the CKD-EPI equation in diabetic patients compared to healthy individuals. Moreover, the MDRD and CKD-EPI equations have a P30 value between 80% and 90%, which means that the eGFR generated from these equations has, at best, a 90% chance of being within ± 30% of the measured GFR[2]. To sum up, caution should be exercised when using eGFR as the sole marker for diagnosis of DKD.

LIMITATIONS OF ALBUMINURIA

Albuminuria is considered a marker of kidney damage, especially with glomerular dysfunction. An assay for detecting low concentration of urinary albumin was first described in the 1960s[20]. When compared with semi-quantitative method, it is more sensitive and specific for disease survey and monitoring. Similar to GFR, measurement of 24-h urine albumin is time-consuming, and adds little to prediction or accuracy[13,21]. Therefore, calculating UACR by checking albumin and creatinine levels in random spot urine samples is currently the standard of clinical practice. However, urinary albumin excretion may also increase for reasons other than DKD, such as physical activity, diet pattern, infection, fever, congestive heart failure, marked hyperglycemia, menstruation, and marked hypertension[22]. Therefore, the diagnosis of persistent albuminuria is based on abnormal UACR in two out of three specimens collected within a period of 3-6 mo[6].

A crucial point of clinical significance is the discordance between the presence of albuminuria and the decline in renal function. Perkins et al[23] reported the development of advanced CKD (GFR < 60 mL/min per 1.73 m2) without concomitant progression of albuminuria in patients with T1DM enrolled in the Joslin Kidney Study. In the Third National Health and Nutrition Examination Survey (NHANES III), a normal urinary albumin level was identified in 36% of the 1197 patients with T2DM who had advanced CKD[24,25]. In the United Kingdom Prospective Diabetes Study 74, only 49% of the patients with renal impairment had preceding albuminuria[26]. In the Developing Education on Microalbuminuria for Awareness of Renal and Cardiovascular Risk in Diabetes study, advanced CKD was noticed in 17% of those with normal UACR[27]. This discordance might be caused by the heterogeneous nature of renal injury, especially in T2DM. As mentioned above, albuminuria is a marker of glomerular dysfunction, which is characteristic of DKD in T1DM[28,29]. However, glomerulopathy is a less common pathogenesis in DKD of T2DM. In fact, tubulointerstitial and/or vascular lesions are sometimes the major histological changes[30-32]. Penno et al[33] described a strong association between prevalence of cardiovascular diseases and “non-albuminuric renal impairment”, suggesting a predominance of macroangiopathy as the underlying renal pathology. Further studies are required to clarify this assumption.

ALTERNATIVE BIOMARKERS

Due to the limitations of eGFR and albuminuria in the early diagnosis of DKD, enormous efforts have been made to investigate and validate alternative biomarkers in recent decades. A tremendous amount of biomarkers have been evaluated for the diagnosis of DKD, and many studies have shown promising preliminary results (Table 1). However, large-scale studies are still required to validate the value of these biomarkers over and above that of eGFR and UACR.

Table 1 Advantages of novel biomarkers in the early diagnosis of diabetic kidney disease.
BiomarkerValidation study designSample sizeType of diabetesSpecimenAdvantagesRef.
CysCCO52[38]2SerumNot affected by lean body mass[35-39]
30[39]Estimates more accurate than creatinine-based ones when GFR > 60 mL/min per 1.73 m2
NGALCC1122UrineIndicator of glomerular hyperfiltration[44]
KIM1CC1122UrineIndicator of glomerular hyperfiltration[44]
NAGCC4341UrineBaseline level predicts development of DKD[51]
CC9462[52]
8-oxodGPC3962UrineBaseline level predicts development of DKD[59]
PentosidineCC4341UrineBaseline level predicts progression of albuminuria[51]
TNFR1/2RC6281SerumBaseline level predicts development of advanced CKD[65]
RC4102[66]

Cystatin C (CysC) is a 13.3 kDa plasma protein freely filtered through the glomerulus. It does not re-enter the bloodstream in an intact form after being re-absorbed and catabolized by tubular cells[34]. Validation studies have showed that serum CysC levels are not affected by muscle mass, which is a major defect of creatinine, and are well-correlated with GFR[35-37]. In addition, CysC-based GFR estimation is more accurate than creatinine-based estimation when GFR remains > 60 mL/min per 1.73 m2[38,39], suggesting that CysC might serve as a better marker of glomerular function in the early stages of DKD. However, a greater intra-individual variability compared to serum creatinine[37], together with a higher cost, should be considered before its clinical application.

Neutrophil gelatinase-associated lipocalin (NGAL) is a 25 kDa molecule which belongs to the lipocalin superfamily. It serves as a binder and transporter of small hydrophobic molecules, and a factor of innate antibacterial responses[40]. Urinary NGAL is closely related to the severity of renal impairment in various kidney disease. It is considered to play a protective role in such harmful conditions, as it is capable of promoting the proliferation and differentiation of renal cells[41]. Yang et al[42] reported that urinary NGAL correlated positively with serum CysC and creatinine levels, and inversely with GFR, whereas serum NGAL correlated negatively with serum CysC, in patients with T2DM. Furthermore, urinary NGAL has been shown to correlate positively with the severity of albuminuria in both T1DM[43] and T2DM[42] patients. In patients with short duration (less than 5 years) of T2DM, Fu et al[44] described a positive correlation between urinary NGAL and glomerular hyperfiltration. Such compelling evidences suggest the potential of NGAL as a novel biomarker for the early detection of DKD.

Kidney injury molecule 1 (KIM1) is a transmembrane protein with immunoglobulin-like and mucin domains in its ectodomain. Upregulated expression of KIM1 in renal tubules has been observed in ischemic, toxic, and proteinuric kidney diseases, suggesting its potential role as a marker of renal damage[45]. Similar to NGAL, elevated urinary KIM1 concentrations were identified in T2DM patients with glomerular hyperfiltration[44]. Nielsen et al[43] reported higher urinary KIM1 excretion in patients with T1DM than in healthy controls. Vaidya et al[46] showed that lower baseline concentration of urinary KIM1 was predictive of subsequent regression of albuminuria. These results indicate that the role of KIM1 in the early diagnosis of DKD is worth further investigation.

N-acetyl-β-(D)-glucosaminidase (NAG) is a 130 kDa lysosomal enzyme located in the brush border of proximal renal tubular cells. Under normal conditions, NAG is excreted in low amounts in urine during the process of exocytosis. Elevated urinary NAG has been observed in various kidney diseases, suggesting a reflection of renal damage[47,48]. In patients with diabetes, increased excretion of NAG in urine has been identified to associate with the severity of albuminuria[49-51]. Despite inconsistency has been observed in the correlation between urinary NAG and glomerular hyperfiltration[44], results from the studies of Kern et al[51] and Hong et al[52] have indicated that higher baseline concentrations of urinary NAG were predictive of future development of DKD. On the other hand, lower baseline urinary concentration of urinary NAG was associated with the subsequent regression of albuminuria[46]. In addition to DKD, increased excretion of NAG in urine has also been reported to predict macrovascular complications in patients with T2DM[52-54].

Oxidative stress has been considered to play an important part in the pathogenesis of diabetic complications[55]. 8-oxo-7,8-dihydro-2’-deoxyguanosine (8-oxodG) is an oxidized nucleoside - one of the major product of oxidative damage in nuclear and mitochondrial DNA[56]. Upon DNA repair, 8-oxodG is directly excreted into urine without further metabolization, so its urine concentration may serve as a generalized index of oxidative stress[57]. The study conducted by Hinokio et al[58] demonstrated a close correlation between urinary 8-oxodG excretion and the severity of microsvascular diabetic complications. In a 5-year cohort study of 532 Japanese patients with T2DM, baseline concentration of urinary 8-oxodG predicted subsequent development of DKD[59], indicating its potential as a predictive marker.

Hyperglycemia irreversibly modifies long-lived macromolecules by forming advanced glycation end products (AGEs), which cause qualitative and quantitative changes of the components of extracellular matrix. By affecting cell adhesion, growth, and matrix accumulation, AGE-induced changes are associated with the pathogenesis of diabetes complications[60]. One of the best chemically characterized AGEs found in human is pentosidine, which has been considered as a marker of formation and accumulation of AGEs[61]. Elevated urinary and plasma pentosidine levels were identified in T2DM patients with DKD[62]. Both urinary[51] and plasma[63] pentosidine levels have been demonstrated to correlate positively with the severity of albuminuria in patients with diabetes. In the study conducted by Kern et al[51], baseline urinary pentosidine excretion in patients with T1DM predicted the progression of albuminuria, with a seven-fold increase in risk for every 50% increase in urinary pentosidine.

Tumor necrosis factor (TNF)-α is a key mediator of inflammation and apoptosis. The signal transduction of TNF-α is commenced via two distinct receptors, TNF receptor (TNFR) 1 and TNFR2, which are presented in both membrane-bound form and soluble form in serum[64]. Serum levels of TNFR1 and TNFR2 were shown to correlate with GFR in patients with diabetes, and was independent of the status of albuminuria[64]. Recent studies in both T1DM[65] and T2DM[66] patients have indicated that plasma TNFR levels were capable of predicting the development of advanced CKD independently over 12 years of follow-up. These evidences suggest that serum concentrations of TNFR1 and TNFR2 may be utilized as predictors of DKD progression.

GENETIC SUSCEPTIBILITY

Genetic studies provide a powerful tool in the understanding of disease mechanisms. Emerging evidences have suggested that DKD is heritable[67-69]. Prior to the deployment of modern high-throughput technologies such as single nucleotide polymorphism microarray analysis and next-generation sequencing, linkage analysis had revealed variants on different chromosomal regions associated with DKD. For instance, variants on chromosome 18q have been identified to be associated with albuminuria and decreased renal function in different ethnic groups[70,71]. With the application of genome-wide association studies (GWASs) over the past decade, considerable progress has been made in the understanding of genetic background of DKD. Genes such as engulfment and cell motility 1[72-77], FERM domain containing 3[78-81], cysteinyl-tRNA synthase[78,79,81], apolipoprotein L3-non-muscle myosin heavy chain 9[82,83] have been identified to be associated with the phenotypic presentations of DKD. Other risk loci have also been reported, yet data from different GWASs are not consistent[84]. Several fundamental problems remain to be solved before applying these results in clinical practice. First, genetic heterogeneity is always a major consideration when assessing the genetic background of any disease. Replication studies are essential for patients with DKD in different populations. Second, in most GWASs, DKD was defined as the co-existence of hyperglycemia and proteinuria; therefore, it is likely that these results are confounded by patients with renal damage due to causes other than diabetes. Last but not least, the actual functions of many genes which contain loci of risk are still unknown. Further studies are required to elucidate their roles in the pathogenesis of DKD.

EPIGENETIC MODIFICATIONS

Epigenetic modifications refer to DNA methylation, histone methylation, and histone acetylation, which alter the expression of a gene by changing its accessibility rather than nucleotide sequence[85]. In patients with diabetes, multiple factors, such as hyperglycemia, reactive oxygen species, and inflammation, can trigger epigenetic modifications[86]. Knowledge about the role of epigenetic modifications in the pathogenesis of DKD is currently very limited; however, since epigenetics is very sensitive to environmental factors, it is plausible that epigenetic imprints are responsible for the “metabolic memory” linked to diabetic complications[87]. Hasegawa et al[88] demonstrated that differentially methylated genes correlated with fibrogenesis in microdissected tubules obtained from patients with DKD. In a case-control study of 192 Irish patients with T1DM, Bell et al[89] reported that methylation at 19 CpG cites in several genes, including UNC13B, was associated with the time to development of DKD. Sapienza et al[90] identified 187 genes that were differentially methylated on at least two CpG sites among African American and Hispanic diabetic patients with ESRD. Intriguingly, many of these genes have been recognized previously through genome association or transcription profiling studies, and are associated with inflammation, oxidative stress, ubiquitination, fibrosis, drug metabolism, and development of DKD. These results suggest a very close connection between epigenetic modifications and genetic dysregulartions in the pathogenesis of DKD.

MICRORNA PROFILES

MicroRNAs (miRNAs) are small non-coding RNAs composed of 21-25 nucleotides that are produced by genes. By binding to target mRNAs, miRNAs induce degradation of RNAs or, more frequently, repression of protein translation[91]. Being packed within exosomes, miRNAs are stable in serum, plasma, and urine[92]. The stability makes miRNAs as potential candidate biomarkers for the non-invasive diagnosis of many diseases[93].

In vitro and in vivo studies have revealed the potential roles of miRNAs in the pathogenesis of DKD, especially in the early mesangial expansion stage. Changes in the expression of many miRNAs, such as miR-192[94-97], miR-216a[98], miR-377[99], miR-29c[100], miR-200b/c[101], miR-21[102], miR-1207-5p[103], miR-200a[104], and miR-23b[105], have been identified to be involved in the process of extracellular matrix expansion and fibrosis, interaction with transforming growth factor β and other pro-fibrotic genes. Long et al[106] identified miR-93 as a novel regulator of vascular endothelial growth factor in in vitro and in vivo experimental models under hyperglycemic conditions. Fu et al[107] described a significant reduction of endogenous miR-25 in rat mesangial cells treated with high glucose concentrations and in the kidneys of diabetic rats associated with increased nicotinamide adenine dinucleotide phosphate hydrogen oxidase (NOX) activity characterized by high NOX4 expression levels. Zhang et al[108] reported that over-expression of miR-451, which targets tyrosine3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta and p38 mitogen-activated protein kinase signaling pathways, resulted in reduced glomerular mesangial cell proliferation in vitro and in vivo. These experimental findings are summarized in Table 2.

Table 2 In vitro and in vivo renal cell models demonstrating the potential involvement of miRNAs in development of diabetic kidney disease.
miRNASpeciesSpecimenmiRNA expressionMechanism of actionRef.
miR-192Mice/RatM, Te, KTInconsistent resultsInteraction with TGFβ-associated and other pro-fibrotic genes[94-96]
HumanTe, KTReduced[97]
miR-216aMiceM, KTElevated[98]
miR-377MiceM, KTElevated[99]
HumanM
miR-29cMiceP, KTElevated[100]
miR-200b/cMiceM, KTElevated[101]
miR-21MiceKTElevated[102]
HumanTe
miR-1207-5pHumanP, M, TeElevated[103]
miR-200aRatTeReduced[104]
MiceKT
miR-23bMiceKTReduced[105]
HumanTe, HEK-293A
miR-93MiceP, En, KTReducedRegulation of VEGF expression[106]
miR-25RatM, KTReducedRegulation of NOX4 expression[107]
miR-451MiceM, KTReducedTargeting YwhaZ and p38 MAPK signaling pathways[108]

The urinary and serum miRNA in patients with DKD have also been profiled. In T1DM patients with albuminuria, Argyropoulos et al[109] showed underexpression of urinary miR-323b-5p, miR-221-3p, miR-524-5p, and miR-188-3p, whereas miR-214-3p, miR-92b-5p, hsa-miR-765, hsa-miR-429, miR-373-5p, miR-1913, and miR-638 were overexpressed. On the other hand, an elevation in urinary miR-130a and miR-145 levels, with a reduction in miR-155 and miR-424, were reported by Barutta et al[110] in a similar setting. In patients with T2DM, Peng et al[111] described a positive correlation between urinary miR-29 levels and the severity of albuminuria.

Expression of miRNAs was also measured in venous blood from Chinese T2DM patients with and without DKD. Using a microarray-based approach, Zhou et al[112] confirmed the downregulation of miR-let-7a in the patients with DKD. Intriguingly, the authors also observed that the distribution of a specific variant within let-7a (rs1143770) was significantly higher in patients with diabetes than in healthy controls. These results are summarized in Table 3.

Table 3 Urinary and serum miRNA profiles in patients with diabetic kidney disease.
Type of diabetesSpecimenmiRNA expressionRef.
1UrineDecreasedIncreased[109]
miR-323b-5p, miR-221-3p, miR-524-5p, miR-188-3pmiR-214-3p, miR-92b-5p, hsa-miR-765, hsa-miR-429, miR-373-5p, miR-1913, miR-638
1UrineDecreasedIncreased[110]
miR-155, miR-424miR-130a, miR-145
2UrinemiR-29 expression positively correlated to the severity of albuminuria[111]
2BloodReduced expression of miR-let-7a[112]
PROTEOMIC SIGNATURES

Proteomics is defined as “the knowledge of the structure, function, and expression of all proteins in the biochemical or biological context of organisms”[113]. The most attractive feature of proteomics is that it allows the monitoring of patterns of multiple urine and plasma proteins simultaneously. Considering the sophisticated nature of DKD, especially in patients with T2DM, it is plausible that early diagnosis of this disease, which relies only on a single biomarker, might eventually fail to reach optimal sensitivity and specificity[114]. The role of proteomics in the early diagnosis of DKD, therefore, is worthy of further evaluation.

DN65 is a panel , type I collagen. In the study conducted by Rossing et al[115], DN65 was capable of distinguishing between diabetic patients without albuminuria from those with DKD. It was also proved to be sensitive and specific in distinguishing DKD from CKD of other etiologies, as well as predicting the progression toward overt DKD in patients with diabetes who had albuminuria over 3 years. First described by Good et al[116] in 2010, CKD273 is another panel of 273 urinary peptides and proteins capable of identifying CKD of any cause with excellent sensitivity and specificity. In a cohort of 35 patients with diabetes, Zürbig et al[117] showed that the CKD273 classifier was capable of detectingthose who were at risk of DKD progression up to 5 years prior to development of overt albuminuria. In the Prevention of Renal and Vascular End-stage Disease (PREVEND) cohort, Roscioni et al[118] showed that the baseline CKD273 classifier score was independently associated with the progression of albuminuria. In urine samples obtained from 165 patients with T2DM at 9 different centers, Siwy et al[119] demonstrated that the classifier could identify DKD patients with high consistency.

METABOLOMIC SIGNATURES

Metabolomics refers to the identification of low molecular weight intermediate and end-products of cellular functions in a biological sample with nuclear magnetic resonance and mass spectrometry-based profiling techniques[120,121]. As metabolome represents the complete collection of metabolites in an organism, understanding the perturbations in human metabolome might help with early unveiling of the pathological changes in disease processes.

Several studies have assessed the potential of metabolomics in diagnosis of DKD (Table 4). Han et al[122] described the diverse profiles of plasma fatty acids in different stages of DKD. In 82 patients with T2DM, Zhu et al[123] demonstrated that a panel of six plasma phospholipids was capable of distinguishing between patients with and without DKD. In 78 patients with diabetes, Hirayama et al[124] identified a panel of 19 serum metabolites correlated significantly with UACR. A multiple logistic regression model composed of the five best performing markers (including γ-butyrobetaine, symmetric dimethylarginine, azelaic acid, and two unknowns) yielded remarkable sensitivity and specificity for the diagnosis of DKD. Sharma et al[125] quantified 94 metabolites in urine obtained from healthy control, diabetic patients with and without DKD. A decrease in the urine levels of 13 metabolites, many potentially related to mitochondrial function, was found to be associated with DKD. Pena et al[126] described the different metabolomic profiles in the urine and plasma samples from the T2DM cohort of the PREVEND study. Differences were observed in the levels of plasma histidine, butenoylcarnitine, as well as urine hexose, glutamine, and tyrosine, between those who with and without albuminuria. Adding these metabolites to a predictive model composed of baseline urinary albumin excretion and eGFR improve risk estimation for the progression of albuminuria. In the T2DM cohort of the Joslin Kidney Study, Niewczas et al[127] identified a panel of 5 plasma metabolites capable of predicting progression toward ESRD, which was independent of UACR, eGFR, and hemoglobin A1c. Although these results seems promising, the complexity of the analysis techniques and the incomplete coverage of the human metabolome at present are problems than may need to be addressed before the application of metabolomics in everyday practice.

Table 4 Applications of metabolomics in the diagnosis of diabetic kidney disease.
SpecimenPanelApplicationRef.
PlasmaFatty acids C10:0, C12:0, C14:0, C16:1n-9, C16:0, C18:2, C18:1n-9, C18:1n-11, C18:0, C20:4, C20:5, C20:3, C20:2, C20:0, C22:6Diverse profiles in different stages of DKD[122]
PlasmaPhospholipids C18:2-LPC, C16:0/18:1-PE, pC18:0/20:4-PE, C18:0/22:6-PI, C18:0/18:0-PS, dC18:0/20:2-SMDiagnosis of DKD[123]
Serumγ-butyrobetaine, SDMA, azelaic acid, MID 114, MID 127Diagnosis of DKD[124]
Urine3-hydroxy isovalerate, aconitic acid, citric acid, 2-ethyl 3-OH propionate, glycolic acid, homovanillic acid, 3-hydroxy isobutyrate, 2-methyl acetoacetate, 3-methyl adipic acid, 3-methyl crotonyl glycine, 3-hydroxy propionate, tiglylglycine, uracilReduced expression in DKD patients[125]
Plasma and urinePlasma: Histidine, butenoylcarnitine Urine: Hexose, glutamine, tyrosineAddition to the original predictive model improved risk estimation for albuminuria progression[126]
PlasmaP-cresol sulfate, phenylacetylglutamine, myoinositol, pseudouridine, uratePredicting progression toward ESRD[127]
CONCLUSION

The development of DKD involves the dysfunction and damage of different renal tissues in multiple stages. Due to the complex nature of this disease, whether there is a “universal” biomarker is questionable. With extensive validations, albuminuria and eGFR are currently the standard diagnostic criteria for DKD. Nonetheless, the abilities of these markers to detect tissue damage and functional change in the early stage are limited. With the increasing understanding of pathogenesis and promising preliminary data, applying the information generated from the studies of novel biomarkers, genomic, and proteomic profiles to assist in the early diagnosis of DKD has gradually become plausible. An integration of the “traditional” and “next-generation” markers might be more practical in everyday settings, considering the financial and technical requirements of these novel assays. To sum up, large longitudinal cohort studies are still required to validate the abilities of the aforementioned novel early diagnosis and prediction techniques.

Footnotes

Manuscript Source: Invited manuscript

Specialty Type: Endocrinology and Metabolism

Country of Origin: Taiwan

Peer-Review Report Classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Haidara MAA, Kaya C, Markic D, Sun CK S- Editor: Ji FF L- Editor: A E- Editor: Li D

References
1.  International Diabetes Federation. IDF Diabetes Atlas, 7th ed. Brussels, Belgium: International Diabetes Federation, 2015. .  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, Hirsch IB, Kalantar-Zadeh K, Narva AS, Navaneethan SD. Diabetic kidney disease: a report from an ADA Consensus Conference. Diabetes Care. 2014;37:2864-2883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 726]  [Cited by in F6Publishing: 674]  [Article Influence: 67.4]  [Reference Citation Analysis (0)]
3.  United States Renal Data System. 2015 USRDS annual data report: Epidemiology of kidney disease in the United States. National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 2015. .  [PubMed]  [DOI]  [Cited in This Article: ]
4.  ERA-EDTA Registry. ERA-EDTA Registry Annual Report 2013. Academic Medical Center, Department of Medical Informatics, Amsterdam, The Netherlands, 2015. .  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Wu MS, Wu IW, Shih CP, Hsu KH. Establishing a Platform for Battling End-stage Renal Disease and Continuing Quality Improvement in Dialysis Therapy in Taiwan- Taiwan Renal Registry Data System (TWRDS). Acta Nephrologica. 2011;25:148-153 Available from: http://www.tsn.org.tw/tsnFile/journal/catalog/D8CE4CABE4354AB6/148-153.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  American Diabetes Association. 9. Microvascular Complications and Foot Care. Diabetes Care. 2016;39 Suppl 1:S72-S80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 90]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
7.  Helal I, Fick-Brosnahan GM, Reed-Gitomer B, Schrier RW. Glomerular hyperfiltration: definitions, mechanisms and clinical implications. Nat Rev Nephrol. 2012;8:293-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 388]  [Cited by in F6Publishing: 447]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
8.  Espinel E, Agraz I, Ibernon M, Ramos N, Fort J, Serón D. Renal Biopsy in Type 2 Diabetic Patients. J Clin Med. 2015;4:998-1009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 42]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
9.  Ritz E, Orth SR. Nephropathy in patients with type 2 diabetes mellitus. N Engl J Med. 1999;341:1127-1133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 487]  [Cited by in F6Publishing: 539]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
10.  Remuzzi G, Schieppati A, Ruggenenti P. Clinical practice. Nephropathy in patients with type 2 diabetes. N Engl J Med. 2002;346:1145-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 380]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
11.  Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of Diet in Renal Disease Study Group. Ann Intern Med. 1999;130:461-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11183]  [Cited by in F6Publishing: 11520]  [Article Influence: 460.8]  [Reference Citation Analysis (0)]
12.  Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF, Feldman HI, Kusek JW, Eggers P, Van Lente F, Greene T. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150:604-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15626]  [Cited by in F6Publishing: 17702]  [Article Influence: 1180.1]  [Reference Citation Analysis (0)]
13.  Levey AS, Coresh J, Balk E, Kausz AT, Levin A, Steffes MW, Hogg RJ, Perrone RD, Lau J, Eknoyan G. National Kidney Foundation practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Ann Intern Med. 2003;139:137-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3156]  [Cited by in F6Publishing: 3112]  [Article Influence: 148.2]  [Reference Citation Analysis (0)]
14.  Perrone RD, Madias NE, Levey AS. Serum creatinine as an index of renal function: new insights into old concepts. Clin Chem. 1992;38:1933-1953.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Baxmann AC, Ahmed MS, Marques NC, Menon VB, Pereira AB, Kirsztajn GM, Heilberg IP. Influence of muscle mass and physical activity on serum and urinary creatinine and serum cystatin C. Clin J Am Soc Nephrol. 2008;3:348-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 503]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
16.  Rule AD, Larson TS, Bergstralh EJ, Slezak JM, Jacobsen SJ, Cosio FG. Using serum creatinine to estimate glomerular filtration rate: accuracy in good health and in chronic kidney disease. Ann Intern Med. 2004;141:929-937.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 736]  [Cited by in F6Publishing: 753]  [Article Influence: 37.7]  [Reference Citation Analysis (0)]
17.  Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, Kusek JW, Van Lente F. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145:247-254.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Michels WM, Grootendorst DC, Verduijn M, Elliott EG, Dekker FW, Krediet RT. Performance of the Cockcroft-Gault, MDRD, and new CKD-EPI formulas in relation to GFR, age, and body size. Clin J Am Soc Nephrol. 2010;5:1003-1009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 338]  [Cited by in F6Publishing: 404]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
19.  Camargo EG, Soares AA, Detanico AB, Weinert LS, Veronese FV, Gomes EC, Silveiro SP. The Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation is less accurate in patients with Type 2 diabetes when compared with healthy individuals. Diabet Med. 2011;28:90-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
20.  Keen H, Chlouverakis C. An immunoassay method for urinary albumin at low concentrations. Lancet. 1963;2:913-914.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Eknoyan G, Hostetter T, Bakris GL, Hebert L, Levey AS, Parving HH, Steffes MW, Toto R. Proteinuria and other markers of chronic kidney disease: a position statement of the national kidney foundation (NKF) and the national institute of diabetes and digestive and kidney diseases (NIDDK). Am J Kidney Dis. 2003;42:617-622.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  National Kidney Foundation. KDOQI Clinical Practice Guidelines and Clinical Practice Recommendations for Diabetes and Chronic Kidney Disease. Am J Kidney Dis. 2007;49:S12-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 603]  [Cited by in F6Publishing: 814]  [Article Influence: 47.9]  [Reference Citation Analysis (0)]
23.  Perkins BA, Ficociello LH, Roshan B, Warram JH, Krolewski AS. In patients with type 1 diabetes and new-onset microalbuminuria the development of advanced chronic kidney disease may not require progression to proteinuria. Kidney Int. 2010;77:57-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 202]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
24.  Garg AX, Kiberd BA, Clark WF, Haynes RB, Clase CM. Albuminuria and renal insufficiency prevalence guides population screening: results from the NHANES III. Kidney Int. 2002;61:2165-2175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 216]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
25.  Kramer HJ, Nguyen QD, Curhan G, Hsu CY. Renal insufficiency in the absence of albuminuria and retinopathy among adults with type 2 diabetes mellitus. JAMA. 2003;289:3273-3277.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 425]  [Cited by in F6Publishing: 419]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
26.  Retnakaran R, Cull CA, Thorne KI, Adler AI, Holman RR. Risk factors for renal dysfunction in type 2 diabetes: U.K. Prospective Diabetes Study 74. Diabetes. 2006;55:1832-1839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 646]  [Cited by in F6Publishing: 649]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
27.  Dwyer JP, Parving HH, Hunsicker LG, Ravid M, Remuzzi G, Lewis JB. Renal Dysfunction in the Presence of Normoalbuminuria in Type 2 Diabetes: Results from the DEMAND Study. Cardiorenal Med. 2012;2:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 81]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
28.  Mauer SM, Steffes MW, Ellis EN, Sutherland DE, Brown DM, Goetz FC. Structural-functional relationships in diabetic nephropathy. J Clin Invest. 1984;74:1143-1155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 862]  [Cited by in F6Publishing: 837]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
29.  Caramori ML, Kim Y, Huang C, Fish AJ, Rich SS, Miller ME, Russell G, Mauer M. Cellular basis of diabetic nephropathy: 1. Study design and renal structural-functional relationships in patients with long-standing type 1 diabetes. Diabetes. 2002;51:506-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 151]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
30.  Fioretto P, Mauer M, Brocco E, Velussi M, Frigato F, Muollo B, Sambataro M, Abaterusso C, Baggio B, Crepaldi G. Patterns of renal injury in NIDDM patients with microalbuminuria. Diabetologia. 1996;39:1569-1576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 311]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
31.  Brocco E, Fioretto P, Mauer M, Saller A, Carraro A, Frigato F, Chiesura-Corona M, Bianchi L, Baggio B, Maioli M. Renal structure and function in non-insulin dependent diabetic patients with microalbuminuria. Kidney Int Suppl. 1997;63:S40-S44.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Dalla Vestra M, Saller A, Bortoloso E, Mauer M, Fioretto P. Structural involvement in type 1 and type 2 diabetic nephropathy. Diabetes Metab. 2000;26 Suppl 4:8-14.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Penno G, Solini A, Bonora E, Fondelli C, Orsi E, Zerbini G, Trevisan R, Vedovato M, Gruden G, Cavalot F. Clinical significance of nonalbuminuric renal impairment in type 2 diabetes. J Hypertens. 2011;29:1802-1809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 164]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
34.  Ogawa Y, Goto T, Tamasawa N, Matsui J, Tando Y, Sugimoto K, Tomotsune K, Kimura M, Yasujima M, Suda T. Serum cystatin C in diabetic patients. Not only an indicator for renal dysfunction in patients with overt nephropathy but also a predictor for cardiovascular events in patients without nephropathy. Diabetes Res Clin Pract. 2008;79:357-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
35.  Kyhse-Andersen J, Schmidt C, Nordin G, Andersson B, Nilsson-Ehle P, Lindström V, Grubb A. Serum cystatin C, determined by a rapid, automated particle-enhanced turbidimetric method, is a better marker than serum creatinine for glomerular filtration rate. Clin Chem. 1994;40:1921-1926.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Finney H, Newman DJ, Price CP. Adult reference ranges for serum cystatin C, creatinine and predicted creatinine clearance. Ann Clin Biochem. 2000;37:49-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 172]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
37.  Laterza OF, Price CP, Scott MG. Cystatin C: an improved estimator of glomerular filtration rate? Clin Chem. 2002;48:699-707.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Mussap M, Dalla Vestra M, Fioretto P, Saller A, Varagnolo M, Nosadini R, Plebani M. Cystatin C is a more sensitive marker than creatinine for the estimation of GFR in type 2 diabetic patients. Kidney Int. 2002;61:1453-1461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 168]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
39.  Perkins BA, Nelson RG, Ostrander BE, Blouch KL, Krolewski AS, Myers BD, Warram JH. Detection of renal function decline in patients with diabetes and normal or elevated GFR by serial measurements of serum cystatin C concentration: results of a 4-year follow-up study. J Am Soc Nephrol. 2005;16:1404-1412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 262]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
40.  Goetz DH, Willie ST, Armen RS, Bratt T, Borregaard N, Strong RK. Ligand preference inferred from the structure of neutrophil gelatinase associated lipocalin. Biochemistry. 2000;39:1935-1941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 137]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
41.  Bolignano D, Donato V, Coppolino G, Campo S, Buemi A, Lacquaniti A, Buemi M. Neutrophil gelatinase-associated lipocalin (NGAL) as a marker of kidney damage. Am J Kidney Dis. 2008;52:595-605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 386]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
42.  Yang YH, He XJ, Chen SR, Wang L, Li EM, Xu LY. Changes of serum and urine neutrophil gelatinase-associated lipocalin in type-2 diabetic patients with nephropathy: one year observational follow-up study. Endocrine. 2009;36:45-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
43.  Nielsen SE, Schjoedt KJ, Astrup AS, Tarnow L, Lajer M, Hansen PR, Parving HH, Rossing P. Neutrophil Gelatinase-Associated Lipocalin (NGAL) and Kidney Injury Molecule 1 (KIM1) in patients with diabetic nephropathy: a cross-sectional study and the effects of lisinopril. Diabet Med. 2010;27:1144-1150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
44.  Fu WJ, Li BL, Wang SB, Chen ML, Deng RT, Ye CQ, Liu L, Fang AJ, Xiong SL, Wen S. Changes of the tubular markers in type 2 diabetes mellitus with glomerular hyperfiltration. Diabetes Res Clin Pract. 2012;95:105-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
45.  van Timmeren MM, van den Heuvel MC, Bailly V, Bakker SJ, van Goor H, Stegeman CA. Tubular kidney injury molecule-1 (KIM-1) in human renal disease. J Pathol. 2007;212:209-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 335]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
46.  Vaidya VS, Niewczas MA, Ficociello LH, Johnson AC, Collings FB, Warram JH, Krolewski AS, Bonventre JV. Regression of microalbuminuria in type 1 diabetes is associated with lower levels of urinary tubular injury biomarkers, kidney injury molecule-1, and N-acetyl-β-D-glucosaminidase. Kidney Int. 2011;79:464-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 167]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
47.  Bazzi C, Petrini C, Rizza V, Arrigo G, Napodano P, Paparella M, D’Amico G. Urinary N-acetyl-beta-glucosaminidase excretion is a marker of tubular cell dysfunction and a predictor of outcome in primary glomerulonephritis. Nephrol Dial Transplant. 2002;17:1890-1896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 169]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
48.  Liangos O, Perianayagam MC, Vaidya VS, Han WK, Wald R, Tighiouart H, MacKinnon RW, Li L, Balakrishnan VS, Pereira BJ. Urinary N-acetyl-beta-(D)-glucosaminidase activity and kidney injury molecule-1 level are associated with adverse outcomes in acute renal failure. J Am Soc Nephrol. 2007;18:904-912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 344]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
49.  Widstam-Attorps U, Berg U. Urinary protein excretion and renal function in young people with diabetes mellitus. Nephrol Dial Transplant. 1992;7:487-492.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Uslu S, Efe B, Alataş O, Kebapçi N, Colak O, Demirüstü C, Yörük A. Serum cystatin C and urinary enzymes as screening markers of renal dysfunction in diabetic patients. J Nephrol. 2005;18:559-567.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Kern EF, Erhard P, Sun W, Genuth S, Weiss MF. Early urinary markers of diabetic kidney disease: a nested case-control study from the Diabetes Control and Complications Trial (DCCT). Am J Kidney Dis. 2010;55:824-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 74]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
52.  Hong CY, Chia KS, Ling SL. Urinary protein excretion in Type 2 diabetes with complications. J Diabetes Complications. 2000;14:259-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
53.  Weitgasser R, Schnoell F, Gappmayer B, Kartnig I. Prospective evaluation of urinary N-acetyl-beta-D-glucosaminidase with respect to macrovascular disease in elderly type 2 diabetic patients. Diabetes Care. 1999;22:1882-1886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
54.  Yoshikawa R, Wada J, Seiki K, Matsuoka T, Miyamoto S, Takahashi K, Ota S, Taniai K, Hida K, Yamakado M. Urinary PGDS levels are associated with vascular injury in type 2 diabetes patients. Diabetes Res Clin Pract. 2007;76:358-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
55.  Macisaac RJ, Ekinci EI, Jerums G. Markers of and risk factors for the development and progression of diabetic kidney disease. Am J Kidney Dis. 2014;63:S39-S62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 206]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
56.  Cooke MS, Evans MD, Herbert KE, Lunec J. Urinary 8-oxo-2’-deoxyguanosine--source, significance and supplements. Free Radic Res. 2000;32:381-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 163]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
57.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Hinokio Y, Suzuki S, Hirai M, Chiba M, Hirai A, Toyota T. Oxidative DNA damage in diabetes mellitus: its association with diabetic complications. Diabetologia. 1999;42:995-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 156]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
59.  Hinokio Y, Suzuki S, Hirai M, Suzuki C, Suzuki M, Toyota T. Urinary excretion of 8-oxo-7, 8-dihydro-2’-deoxyguanosine as a predictor of the development of diabetic nephropathy. Diabetologia. 2002;45:877-882.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 114]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
60.  Brownlee M. Glycation products and the pathogenesis of diabetic complications. Diabetes Care. 1992;15:1835-1843.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Price DL, Rhett PM, Thorpe SR, Baynes JW. Chelating activity of advanced glycation end-product inhibitors. J Biol Chem. 2001;276:48967-48972.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 246]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
62.  Calabrese V, Mancuso C, Sapienza M, Puleo E, Calafato S, Cornelius C, Finocchiaro M, Mangiameli A, Di Mauro M, Stella AM. Oxidative stress and cellular stress response in diabetic nephropathy. Cell Stress Chaperones. 2007;12:299-306.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Piarulli F, Sartore G, Ceriello A, Ragazzi E, Reitano R, Nollino L, Cosma C, Fedele D, Lapolla A. Relationship between glyco-oxidation, antioxidant status and microalbuminuria in type 2 diabetic patients. Diabetologia. 2009;52:1419-1425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
64.  Niewczas MA, Ficociello LH, Johnson AC, Walker W, Rosolowsky ET, Roshan B, Warram JH, Krolewski AS. Serum concentrations of markers of TNFalpha and Fas-mediated pathways and renal function in nonproteinuric patients with type 1 diabetes. Clin J Am Soc Nephrol. 2009;4:62-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 111]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
65.  Gohda T, Niewczas MA, Ficociello LH, Walker WH, Skupien J, Rosetti F, Cullere X, Johnson AC, Crabtree G, Smiles AM. Circulating TNF receptors 1 and 2 predict stage 3 CKD in type 1 diabetes. J Am Soc Nephrol. 2012;23:516-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 256]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
66.  Niewczas MA, Gohda T, Skupien J, Smiles AM, Walker WH, Rosetti F, Cullere X, Eckfeldt JH, Doria A, Mayadas TN. Circulating TNF receptors 1 and 2 predict ESRD in type 2 diabetes. J Am Soc Nephrol. 2012;23:507-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 327]  [Cited by in F6Publishing: 336]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
67.  Forsblom CM, Kanninen T, Lehtovirta M, Saloranta C, Groop LC. Heritability of albumin excretion rate in families of patients with Type II diabetes. Diabetologia. 1999;42:1359-1366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 73]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
68.  Fogarty DG, Rich SS, Hanna L, Warram JH, Krolewski AS. Urinary albumin excretion in families with type 2 diabetes is heritable and genetically correlated to blood pressure. Kidney Int. 2000;57:250-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
69.  Langefeld CD, Beck SR, Bowden DW, Rich SS, Wagenknecht LE, Freedman BI. Heritability of GFR and albuminuria in Caucasians with type 2 diabetes mellitus. Am J Kidney Dis. 2004;43:796-800.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Vardarli I, Baier LJ, Hanson RL, Akkoyun I, Fischer C, Rohmeiss P, Basci A, Bartram CR, Van Der Woude FJ, Janssen B. Gene for susceptibility to diabetic nephropathy in type 2 diabetes maps to 18q22.3-23. Kidney Int. 2002;62:2176-2183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 126]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
71.  Bowden DW, Colicigno CJ, Langefeld CD, Sale MM, Williams A, Anderson PJ, Rich SS, Freedman BI. A genome scan for diabetic nephropathy in African Americans. Kidney Int. 2004;66:1517-1526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 140]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
72.  Shimazaki A, Kawamura Y, Kanazawa A, Sekine A, Saito S, Tsunoda T, Koya D, Babazono T, Tanaka Y, Matsuda M. Genetic variations in the gene encoding ELMO1 are associated with susceptibility to diabetic nephropathy. Diabetes. 2005;54:1171-1178.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Pezzolesi MG, Katavetin P, Kure M, Poznik GD, Skupien J, Mychaleckyj JC, Rich SS, Warram JH, Krolewski AS. Confirmation of genetic associations at ELMO1 in the GoKinD collection supports its role as a susceptibility gene in diabetic nephropathy. Diabetes. 2009;58:2698-2702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 94]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
74.  Leak TS, Perlegas PS, Smith SG, Keene KL, Hicks PJ, Langefeld CD, Mychaleckyj JC, Rich SS, Kirk JK, Freedman BI. Variants in intron 13 of the ELMO1 gene are associated with diabetic nephropathy in African Americans. Ann Hum Genet. 2009;73:152-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 82]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
75.  Craig DW, Millis MP, DiStefano JK. Genome-wide SNP genotyping study using pooled DNA to identify candidate markers mediating susceptibility to end-stage renal disease attributed to Type 1 diabetes. Diabet Med. 2009;26:1090-1098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
76.  Williams WW, Salem RM, McKnight AJ, Sandholm N, Forsblom C, Taylor A, Guiducci C, McAteer JB, McKay GJ, Isakova T. Association testing of previously reported variants in a large case-control meta-analysis of diabetic nephropathy. Diabetes. 2012;61:2187-2194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 69]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
77.  Wu HY, Wang Y, Chen M, Zhang X, Wang D, Pan Y, Li L, Liu D, Dai XM. Association of ELMO1 gene polymorphisms with diabetic nephropathy in Chinese population. J Endocrinol Invest. 2013;36:298-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 19]  [Reference Citation Analysis (0)]
78.  Pezzolesi MG, Poznik GD, Mychaleckyj JC, Paterson AD, Barati MT, Klein JB, Ng DP, Placha G, Canani LH, Bochenski J. Genome-wide association scan for diabetic nephropathy susceptibility genes in type 1 diabetes. Diabetes. 2009;58:1403-1410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 234]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
79.  Maeda S, Araki S, Babazono T, Toyoda M, Umezono T, Kawai K, Imanishi M, Uzu T, Watada H, Suzuki D. Replication study for the association between four Loci identified by a genome-wide association study on European American subjects with type 1 diabetes and susceptibility to diabetic nephropathy in Japanese subjects with type 2 diabetes. Diabetes. 2010;59:2075-2079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
80.  Park H, Kim HJ, Lee S, Yoo YJ, Ju YS, Lee JE, Cho SI, Sung J, Kim JI, Seo JS. A family-based association study after genome-wide linkage analysis identified two genetic loci for renal function in a Mongolian population. Kidney Int. 2013;83:285-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
81.  Pezzolesi MG, Jeong J, Smiles AM, Skupien J, Mychaleckyj JC, Rich SS, Warram JH, Krolewski AS. Family-based association analysis confirms the role of the chromosome 9q21.32 locus in the susceptibility of diabetic nephropathy. PLoS One. 2013;8:e60301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
82.  McDonough CW, Palmer ND, Hicks PJ, Roh BH, An SS, Cooke JN, Hester JM, Wing MR, Bostrom MA, Rudock ME. A genome-wide association study for diabetic nephropathy genes in African Americans. Kidney Int. 2011;79:563-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 125]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
83.  Cooke JN, Bostrom MA, Hicks PJ, Ng MC, Hellwege JN, Comeau ME, Divers J, Langefeld CD, Freedman BI, Bowden DW. Polymorphisms in MYH9 are associated with diabetic nephropathy in European Americans. Nephrol Dial Transplant. 2012;27:1505-1511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 67]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
84.  Chang YC, Chang EY, Chuang LM. Recent progress in the genetics of diabetic microvascular complications. World J Diabetes. 2015;6:715-725.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
85.  Conserva F, Gesualdo L, Papale M. A Systems Biology Overview on Human Diabetic Nephropathy: From Genetic Susceptibility to Post-Transcriptional and Post-Translational Modifications. J Diabetes Res. 2016;2016:7934504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
86.  Richter K, Konzack A, Pihlajaniemi T, Heljasvaara R, Kietzmann T. Redox-fibrosis: Impact of TGFβ1 on ROS generators, mediators and functional consequences. Redox Biol. 2015;6:344-352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 161]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
87.  Pirola L, Balcerczyk A, Okabe J, El-Osta A. Epigenetic phenomena linked to diabetic complications. Nat Rev Endocrinol. 2010;6:665-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 179]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
88.  Hasegawa K, Wakino S, Simic P, Sakamaki Y, Minakuchi H, Fujimura K, Hosoya K, Komatsu M, Kaneko Y, Kanda T. Renal tubular Sirt1 attenuates diabetic albuminuria by epigenetically suppressing Claudin-1 overexpression in podocytes. Nat Med. 2013;19:1496-1504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 339]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
89.  Bell CG, Teschendorff AE, Rakyan VK, Maxwell AP, Beck S, Savage DA. Genome-wide DNA methylation analysis for diabetic nephropathy in type 1 diabetes mellitus. BMC Med Genomics. 2010;3:33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 215]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
90.  Sapienza C, Lee J, Powell J, Erinle O, Yafai F, Reichert J, Siraj ES, Madaio M. DNA methylation profiling identifies epigenetic differences between diabetes patients with ESRD and diabetes patients without nephropathy. Epigenetics. 2011;6:20-28.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Chua JH, Armugam A, Jeyaseelan K. MicroRNAs: biogenesis, function and applications. Curr Opin Mol Ther. 2009;11:189-199.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105:10513-10518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5636]  [Cited by in F6Publishing: 6041]  [Article Influence: 377.6]  [Reference Citation Analysis (0)]
93.  Schena FP, Sallustio F, Serino G. microRNAs in glomerular diseases from pathophysiology to potential treatment target. Clin Sci (Lond). 2015;128:775-788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
94.  Kato M, Zhang J, Wang M, Lanting L, Yuan H, Rossi JJ, Natarajan R. MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via inhibition of E-box repressors. Proc Natl Acad Sci USA. 2007;104:3432-3437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 545]  [Cited by in F6Publishing: 560]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
95.  Wang B, Herman-Edelstein M, Koh P, Burns W, Jandeleit-Dahm K, Watson A, Saleem M, Goodall GJ, Twigg SM, Cooper ME. E-cadherin expression is regulated by miR-192/215 by a mechanism that is independent of the profibrotic effects of transforming growth factor-beta. Diabetes. 2010;59:1794-1802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 209]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
96.  Putta S, Lanting L, Sun G, Lawson G, Kato M, Natarajan R. Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J Am Soc Nephrol. 2012;23:458-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 275]  [Article Influence: 22.9]  [Reference Citation Analysis (0)]
97.  Krupa A, Jenkins R, Luo DD, Lewis A, Phillips A, Fraser D. Loss of MicroRNA-192 promotes fibrogenesis in diabetic nephropathy. J Am Soc Nephrol. 2010;21:438-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 284]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
98.  Kato M, Wang L, Putta S, Wang M, Yuan H, Sun G, Lanting L, Todorov I, Rossi JJ, Natarajan R. Post-transcriptional up-regulation of Tsc-22 by Ybx1, a target of miR-216a, mediates TGF-{beta}-induced collagen expression in kidney cells. J Biol Chem. 2010;285:34004-34015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 141]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
99.  Wang Q, Wang Y, Minto AW, Wang J, Shi Q, Li X, Quigg RJ. MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy. FASEB J. 2008;22:4126-4135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 302]  [Cited by in F6Publishing: 321]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
100.  Long J, Wang Y, Wang W, Chang BH, Danesh FR. MicroRNA-29c is a signature microRNA under high glucose conditions that targets Sprouty homolog 1, and its in vivo knockdown prevents progression of diabetic nephropathy. J Biol Chem. 2011;286:11837-11848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 217]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
101.  Kato M, Arce L, Wang M, Putta S, Lanting L, Natarajan R. A microRNA circuit mediates transforming growth factor-β1 autoregulation in renal glomerular mesangial cells. Kidney Int. 2011;80:358-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 199]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
102.  Wang JY, Gao YB, Zhang N, Zou DW, Wang P, Zhu ZY, Li JY, Zhou SN, Wang SC, Wang YY. miR-21 overexpression enhances TGF-β1-induced epithelial-to-mesenchymal transition by target smad7 and aggravates renal damage in diabetic nephropathy. Mol Cell Endocrinol. 2014;392:163-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 119]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
103.  Alvarez ML, Khosroheidari M, Eddy E, Kiefer J. Role of microRNA 1207-5P and its host gene, the long non-coding RNA Pvt1, as mediators of extracellular matrix accumulation in the kidney: implications for diabetic nephropathy. PLoS One. 2013;8:e77468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 121]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
104.  Wang B, Koh P, Winbanks C, Coughlan MT, McClelland A, Watson A, Jandeleit-Dahm K, Burns WC, Thomas MC, Cooper ME. miR-200a Prevents renal fibrogenesis through repression of TGF-β2 expression. Diabetes. 2011;60:280-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 274]  [Article Influence: 21.1]  [Reference Citation Analysis (0)]
105.  Zhao B, Li H, Liu J, Han P, Zhang C, Bai H, Yuan X, Wang X, Li L, Ma H. MicroRNA-23b Targets Ras GTPase-Activating Protein SH3 Domain-Binding Protein 2 to Alleviate Fibrosis and Albuminuria in Diabetic Nephropathy. J Am Soc Nephrol. 2016; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 76]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
106.  Long J, Wang Y, Wang W, Chang BH, Danesh FR. Identification of microRNA-93 as a novel regulator of vascular endothelial growth factor in hyperglycemic conditions. J Biol Chem. 2010;285:23457-23465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 218]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
107.  Fu Y, Zhang Y, Wang Z, Wang L, Wei X, Zhang B, Wen Z, Fang H, Pang Q, Yi F. Regulation of NADPH oxidase activity is associated with miRNA-25-mediated NOX4 expression in experimental diabetic nephropathy. Am J Nephrol. 2010;32:581-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 133]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
108.  Zhang Z, Luo X, Ding S, Chen J, Chen T, Chen X, Zha H, Yao L, He X, Peng H. MicroRNA-451 regulates p38 MAPK signaling by targeting of Ywhaz and suppresses the mesangial hypertrophy in early diabetic nephropathy. FEBS Lett. 2012;586:20-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 82]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
109.  Argyropoulos C, Wang K, McClarty S, Huang D, Bernardo J, Ellis D, Orchard T, Galas D, Johnson J. Urinary microRNA profiling in the nephropathy of type 1 diabetes. PLoS One. 2013;8:e54662.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 121]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
110.  Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, Bruno G, Cimino D, Taverna D, Deregibus MC. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One. 2013;8:e73798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 226]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
111.  Peng H, Zhong M, Zhao W, Wang C, Zhang J, Liu X, Li Y, Paudel SD, Wang Q, Lou T. Urinary miR-29 correlates with albuminuria and carotid intima-media thickness in type 2 diabetes patients. PLoS One. 2013;8:e82607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
112.  Zhou J, Peng R, Li T, Luo X, Peng H, Zha H, Yin P, Wen L, Zhang Z. A potentially functional polymorphism in the regulatory region of let-7a-2 is associated with an increased risk for diabetic nephropathy. Gene. 2013;527:456-461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
113.  Kenyon GL, DeMarini DM, Fuchs E, Galas DJ, Kirsch JF, Leyh TS, Moos WH, Petsko GA, Ringe D, Rubin GM. Defining the mandate of proteomics in the post-genomics era: workshop report. Mol Cell Proteomics. 2002;1:763-780.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Currie G, McKay G, Delles C. Biomarkers in diabetic nephropathy: Present and future. World J Diabetes. 2014;5:763-776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 76]  [Cited by in F6Publishing: 73]  [Article Influence: 7.3]  [Reference Citation Analysis (1)]
115.  Rossing K, Mischak H, Dakna M, Zürbig P, Novak J, Julian BA, Good DM, Coon JJ, Tarnow L, Rossing P. Urinary proteomics in diabetes and CKD. J Am Soc Nephrol. 2008;19:1283-1290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 223]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
116.  Good DM, Zürbig P, Argilés A, Bauer HW, Behrens G, Coon JJ, Dakna M, Decramer S, Delles C, Dominiczak AF. Naturally occurring human urinary peptides for use in diagnosis of chronic kidney disease. Mol Cell Proteomics. 2010;9:2424-2437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 377]  [Article Influence: 26.9]  [Reference Citation Analysis (0)]
117.  Zürbig P, Jerums G, Hovind P, Macisaac RJ, Mischak H, Nielsen SE, Panagiotopoulos S, Persson F, Rossing P. Urinary proteomics for early diagnosis in diabetic nephropathy. Diabetes. 2012;61:3304-3313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 191]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
118.  Roscioni SS, de Zeeuw D, Hellemons ME, Mischak H, Zürbig P, Bakker SJ, Gansevoort RT, Reinhard H, Persson F, Lajer M. A urinary peptide biomarker set predicts worsening of albuminuria in type 2 diabetes mellitus. Diabetologia. 2013;56:259-267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 106]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
119.  Siwy J, Schanstra JP, Argiles A, Bakker SJ, Beige J, Boucek P, Brand K, Delles C, Duranton F, Fernandez-Fernandez B. Multicentre prospective validation of a urinary peptidome-based classifier for the diagnosis of type 2 diabetic nephropathy. Nephrol Dial Transplant. 2014;29:1563-1570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 92]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
120.  Lindon JC, Holmes E, Bollard ME, Stanley EG, Nicholson JK. Metabonomics technologies and their applications in physiological monitoring, drug safety assessment and disease diagnosis. Biomarkers. 2004;9:1-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 372]  [Cited by in F6Publishing: 316]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
121.  Wishart DS, Knox C, Guo AC, Eisner R, Young N, Gautam B, Hau DD, Psychogios N, Dong E, Bouatra S. HMDB: a knowledgebase for the human metabolome. Nucleic Acids Res. 2009;37:D603-D610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1397]  [Cited by in F6Publishing: 1387]  [Article Influence: 86.7]  [Reference Citation Analysis (0)]
122.  Han LD, Xia JF, Liang QL, Wang Y, Wang YM, Hu P, Li P, Luo GA. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 104]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
123.  Zhu C, Liang QL, Hu P, Wang YM, Luo GA. Phospholipidomic identification of potential plasma biomarkers associated with type 2 diabetes mellitus and diabetic nephropathy. Talanta. 2011;85:1711-1720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
124.  Hirayama A, Nakashima E, Sugimoto M, Akiyama S, Sato W, Maruyama S, Matsuo S, Tomita M, Yuzawa Y, Soga T. Metabolic profiling reveals new serum biomarkers for differentiating diabetic nephropathy. Anal Bioanal Chem. 2012;404:3101-3109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 117]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
125.  Sharma K, Karl B, Mathew AV, Gangoiti JA, Wassel CL, Saito R, Pu M, Sharma S, You YH, Wang L. Metabolomics reveals signature of mitochondrial dysfunction in diabetic kidney disease. J Am Soc Nephrol. 2013;24:1901-1912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 406]  [Article Influence: 36.9]  [Reference Citation Analysis (0)]
126.  Pena MJ, Lambers Heerspink HJ, Hellemons ME, Friedrich T, Dallmann G, Lajer M, Bakker SJ, Gansevoort RT, Rossing P, de Zeeuw D. Urine and plasma metabolites predict the development of diabetic nephropathy in individuals with Type 2 diabetes mellitus. Diabet Med. 2014;31:1138-1147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 101]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
127.  Niewczas MA, Sirich TL, Mathew AV, Skupien J, Mohney RP, Warram JH, Smiles A, Huang X, Walker W, Byun J. Uremic solutes and risk of end-stage renal disease in type 2 diabetes: metabolomic study. Kidney Int. 2014;85:1214-1224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 163]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]