Kidney Res Clin Pract > Epub ahead of print
Li and Szeto: Urinary podocyte markers in diabetic kidney disease

Abstract

Podocytes are involved in maintaining kidney function and are a major focus of research on diabetic kidney disease (DKD). Urinary biomarkers derived from podocyte fragments and molecules have been proposed for the diagnosis and monitoring of DKD. Various methods have been used to detect intact podocytes and podocyte-derived microvesicles in urine, including centrifugation, visualization, and molecular quantification. Quantification of podocyte-specific protein targets and messenger RNA levels can be performed by Western blotting or enzyme-linked immunosorbent assay and quantitative polymerase chain reaction, respectively. At present, many of these techniques are expensive and labor-intensive, all limiting their widespread use in routine clinical tests. While the potential of urinary podocyte markers for monitoring and risk stratification of DKD has been explored, systematic studies and external validation are lacking in the current literature. Standardization and automation of laboratory methods should be a priority for future research, and the added value of these methods to routine clinical tests should be defined.

Introduction

Epidemiology of chronic kidney disease

Chronic kidney disease (CKD) affects over 10% of the world’s population worldwide, amounting to over 800 million individuals [1]. With the advancement in the pharmaceutical industry and the increasing availability of dialysis, the healthcare expenditure used for the treatment of CKD has risen drastically in the past decades [2]. In 2022, between 4.90 and 7.08 million patients worldwide had end-stage kidney disease and need kidney replacement therapy [3]. Of note, CKD represents an especially large burden in low- and middle-income countries, which are least equipped to deal with its consequences [4].
CKD is more common among older individuals, women, racial minorities, and people with diabetes mellitus and hypertension. Over the past two decades, CKD has become one of the leading causes of death worldwide. This disease is one of the few non-communicable diseases that has seen an increase in associated deaths during this time period [1,5]. Effective means for the diagnosis, risk stratification, and monitoring of kidney disease are much needed.

Podocyte as the focus of kidney disease

The primary function of the kidney is the excretion of metabolic wastes and excessive body water [6]. This objective is achieved by three processes: 1) filtration of plasma through the glomerulus; 2) reabsorption of useful substances via renal tubule; and 3) secretion of other metabolic wastes from peritubular capillary to the tubular fluid [6]. Although the kidney has a delicate three-dimensional (3D) structure with multiple cell types, the podocyte plays a central role in the maintenance of glomerular architecture and is the primary focus of many kidney diseases. Podocytes are terminally differentiated cells with a voluminous cell body that gives rise to primary processes, each of which has an extensive array of foot processes that affix to the glomerular capillary basement membrane (GCBM) and, together with the endothelial cells, from the glomerular filtration barrier [7]. The cell body of podocytes could be divided into three distinct compartments: the base, the top, and the slid diaphragm (SD) [8]. Specific cell membrane proteins are present in each compartment, and their interaction with the cytoplasmic cytoskeleton is responsible for the stability of podocyte structure and function [9].
Podocyte dysfunction plays a pivotal role in the pathogenesis of many kidney diseases. Germline mutations of podocyte-related genes result in kidney diseases, which typically present as steroid-resistant nephrotic syndrome and focal glomerulosclerosis (FGS). On the other hand, acquired podocyte dysfunctions, including a reduction in podocyte number or the density and fusion of podocyte foot processes, lead to the damage of glomerular filtration barrier and proteinuria [10]. Notably, many types of podocyte injury lead to a weakened interaction with GCBM, resulting in the detachment of podocyte and its cellular fragments to the urinary space, which may serve as biomarkers of kidney diseases (Fig. 1).

Podocyte damage and metabolic stress

A hot topic in podocyte research in recent years is the metabolic stress of podocyte during disease processes. Glycolysis and oxidative phosphorylation (OXPHOS) are the two major cellular pathways for energy production. Most cell types may switch between these two pathways in order to cope with any changes in energy demand, and podocyte is not an exception [11]. Podocytes are rich in mitochondria and heavily dependent on them for energy to maintain normal functions. A previous study found that mitochondrial OXPHOS contributes to over 50% of adenosine triphosphate production in mature podocytes [11]. Impairment of mitochondrial function can result in an energy crisis, oxidative stress, inflammation, and cell death. In diabetic kidney disease (DKD), mitochondrial pathways involved in podocyte injury include mitochondrial dynamics and mitophagy, mitochondrial biogenesis, mitochondrial OXPHOS and oxidative stress, and mitochondrial protein quality control. Mitochondria are dynamic organelles that respond to pathophysiologic cues by altering mitochondrial content, fusion, fission, mitophagy, and the unfolded protein response. Fission and fusion work together to maintain mitochondrial morphology, while mitophagy selectively removes damaged mitochondria from the network [12]. In DKD, excessive mitochondrial fission combined with decreased mitochondrial fusion is a typical feature of podocytes [13]. Additionally, the lack of proper mitochondrial turnover in the diabetic kidney is due to the inhibition of mitophagy [14,15]. Another key feature of mitochondrial dysfunction in podocytes is the reduced efficiency of mitochondrial biogenesis [15]. Under high glucose conditions, intracellular reactive oxygen species (ROS) production, mitochondrial DNA damage, and protein and lipid peroxidation are enhanced [16]. Furthermore, maintaining mitochondrial protein homeostasis is challenging due to the continuous exposure of mitochondrial proteins to mitochondrial ROS. Mitochondria cannot exist in isolation within a cell. They interact with the endoplasmic reticulum by forming mitochondrial-associated membranes (MAMs). The disruption of MAMs leads to abnormal intracellular Ca2+ influx, mitochondrial damage, and apoptosis [17]. The combination of the above factors results in podocyte injury and the progression of DKD.
However, mitochondria are mainly located around the podocyte nucleus and are scarce in the foot processes, probably because mitochondria are larger than the foot processes [18]. As a result, the energy expenditure of foot processes is largely supported by glycolysis, which probably explains the predominant expression of phosphofructokinase, the rate-limiting enzyme of glycolysis, in podocyte foot processes [11,18], and there were relatively low levels of tricarboxylic acid cycle intermediates in the glomerulus [18]. Recent evidence suggests that anaerobic glycolysis and fermentation of glucose to lactate are the primary sources of energy for podocytes. Additionally, anaerobic glycolysis maintains the glomerular filtration barrier independently of mitochondrial metabolism [18]. In diabetic mice, compromised glycolysis results in increased amino acid catabolism in podocytes [19]. In DKD, ornithine catabolism contributes to activating mammalian target of rapamycin (mTOR) signaling and cytoskeletal remodeling in podocytes. Therefore, inhibiting ornithine catabolism or mTOR signaling may help mitigate podocyte damage in diabetic mice [19]. Further studies showed that the blockade of glycolysis reduces the migration ability of podocytes [11], while cell-specific knockout of mitochondrial genes in podocytes does not lead to any major changes in the structure and function of podocytes [18].
Available evidence showed that the metabolic pathway of podocytes alters in response to cell damage. Under high glucose conditions, the cellular levels of glycolytic enzymes significantly increased, while that of tricarboxylic acid cycle and mitochondrial respiratory chain proteins significantly decreased [20], and similar changes were observed in kidney tissue samples of diabetes [21]. In streptozotocin-induced diabetic mice, podocyte-specific knockout of pyruvate kinase M2 (PKM2), another key enzyme of the glycolysis pathway, resulted in significantly aggravated kidney injury [21], indicating that increased glycolysis is part of the protective mechanism in response to podocyte injury.
It follows that the impairment of podocyte glycolysis promotes the development of DKD. Notably, activation of the renin-angiotensin system leads to PKM2 inhibition [22]. When podocytes are treated with angiotensin II, PKM2 expression rapidly decreases [22]. Specific deletion of PKM2 in mouse podocytes further exacerbates angiotensin II-induced podocyte damage, which was followed by foot process detachment and proteinuria [22].
Increased glycolysis leads to the formation of pyruvate and lactate, resulting in a certain degree of intracellular acidosis. Podocytes express the G protein-coupled receptor 81 (GPR81), the lactate receptor that is known to modulate the vascular endothelial growth factor A and N-methyl-D-aspartate receptor signaling cascades in podocytes, and regulate glomerular permeability and podocyte remodeling, respectively [23]. Lactate accumulation may therefore contribute to podocyte dysfunction in a diabetic milieu [24,25]. Since glomerular mesangial cells also express lactate transporters [26,27], it is possible that lactate accumulation in diabetic podocytes may also affect mesangial cell function.
Other metabolic disturbances in podocytes probably come secondary to the impaired glycolysis. Hyperglycemia leads to the accumulation of lipids in podocytes, which may accelerate the development of DKD, and this process may be aggravated in an environment with high angiotensin II level [28]. Adipose triglyceride lipase (ATGL), which is the first step in the hydrolysis of triglycerides, is important for maintaining the dynamic balance between lipid droplet storage and metabolism [29]. High-fat diet reduces ATGL expression and increases the accumulation of lipid droplets in podocytes, and the process can be reverted by promoting ATGL expression [30]. The depletion of ATGL in podocytes increases the intracellular level of ROS, promotes cytoskeleton redistribution, induces foot process fusion, increases the permeability of the glomerular filtration barrier, and promotes cell apoptosis [31]. The hyperglycemia-induced reduction in ATGL expression in podocytes is found to be the result of lactate accumulation, which activates the GPR81 receptor, with the downstream suppression of the cyclic adenosine monophosphate/protein kinase A pathway [31]. On the other hand, the facilitation of lipid beta-oxidation in human podocytes by the inhibition of acetyl-CoA carboxylase 2 alleviates glucose-induced insulin resistance via the sirtuin 1/peroxisome proliferator-activated receptor gamma coactivator-1 alpha pathway [32]. In addition, Luo et al. [19] showed that in a diabetic environment, impaired glycolysis of podocytes enhanced ornithine catabolism. Putrescine, the metabolite of ornithine catabolism, activated Rheb, thereby promoting the activation of mTOR signaling, which leads to cytoskeleton remodeling and fusion of podocyte foot processes [19].

Podocyte-specific targets as biomarkers

The major podocyte-derived molecules that are potential targets for biomarker development are summarized in Table 1.

Nuclear targets

Wilms’ tumor suppressor gene-1 (WT1) is a zinc finger-like transcription factor in the podocyte nucleus and is the most commonly used podocyte marker for histological study [33].

Slit-diaphragm protein complex

Components of the podocyte SD have been extensively studied as biomarkers of kidney diseases. The major targets in this group include nephrin, podocin, CD2-associated protein (CD2AP), and podoplanin [34]. Nephrin is a transmembrane protein with extracellular domains that form the core of the SD. It acts as a physical sieve barrier as well as a signaling scaffold [35]. Podocin connects nephrin to the actin cytoskeleton. CD2AP is a scaffolding molecule that interacts with nephrin and actin and is implicated in actin remodeling and membrane trafficking of podocytes [36]. Podoplanin is a small transmembrane glycoprotein in the SD compartment and has been implicated in the non-canonical signaling of mineralocorticoids [37].

Glomerular capillary basement membrane targets

The major protein components of the podocyte basal region are the α3β1 integrin and α-dystroglycan (α-DG). These proteins anchor podocytes to the GCBM and are critical for maintaining the orientation of podocyte as well as the integrity of the filtration barrier [38]. The α3β1 integrin is a transmembrane protein that connects GCBM with the cytoskeleton via α-actinin-4. The α-DG is a glycosylated protein that is connected to the cytoskeleton via utropin, and to the podocyte and GCBM via its interaction with laminin.

Targets at the top membrane

Podocalyxin is the major transmembrane protein on the apical side of podocyte. It is a negatively charged sialic acid protein and contributes to the charge barrier of the glomerulus [39]. It is connected to the actin cytoskeleton by ezrin and sodium-proton exchanger regulatory factor 2 (NHERF2).

Cytoskeleton targets

Several podocyte-specific proteins are linked to the actin cytoskeleton and maintain the 3D structure of the podocyte. The best-studied ones are synaptopodin and α-actinin-4. Both of them bind to the actin skeleton via interaction with the membrane-associated guanylate kinase with inverted orientation-1 (MAGI-1) [40]. Synaptopodin is only expressed in differentiating podocytes after they develop the foot processes, and it is often considered as the marker of mature podocytes [41].

Other cytoplasmic targets

Cytoplasmic proteins of podocytes are less well studied as biomarkers. The aarF domain containing kinase 4 (ADCK4) is specifically present in the mitochondria within the foot processes of rat podocytes [42]. Changes in ADCK4 level may represent metabolic disturbance of the podocyte [43].

Methods of study

Urinary podocytes

Intact podocytes are detectable in the urine of healthy people and patients with kidney diseases [44]. In fact, urinary loss is probably the major cause of podocyte depletion in CKD [45]. The traditional method for the identification of podocytes in urine involves cytospin and immunofluorescence study with specific antibodies (most commonly anti-podocalyxin antibody) [46]. The technique can be partly automated but has limited sensitivity and specificity because of cell debris contamination. With this technique, healthy people excrete less than 0.5 podocytes/mg creatinine, whereas patients with glomerular disease excrete up to 400 podocytes/mg creatinine [44].
In addition to the cytospin technique, podocytes could also be identified by the detection of podocyte-specific tryptic peptides with liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Although the equipment cost is high, this technique is not operator-dependent and therefore highly reproducible [47].
The majority of urinary podocytes are viable when tested with propidium iodide exclusion and TUNEL staining [44]. In theory, a culture of viable podocytes ex vivo would improve the specificity of podocyte identification. However, podocytes do not normally proliferate in vivo, and special experimental conditions are required for the ex vivo culture [48]. This method has great value in translational study and therapeutic target identification but is too complicated for routine clinical use.

Urinary podocyte-derived fragments

Extracellular vesicles (EVs) are spherical membranous bodies released by various cell types [49]. In urine, EVs containing apical membrane and intracellular fluid are shed from all nephron segments, including podocytes, renal tubular epithelial cells, and uroepithelial cells. Urinary EVs may contain protein markers for kidney damage. In addition, EVs may contribute to intercellular communication within the nephron [50]. Previous studies showed that apical cell membrane fragments are shed from injured podocytes into the urine and could be identified as podocalyxin-positive granular structures by electron microscopy [51]. Urinary exosomes from podocytes could also be isolated by serial centrifugation, and microparticles quantitated by flow cytometry with annexin V antibody (which detects all microparticles), followed by antibody to podocalyxin or podoplanin (which specifically identifies podocyte-derived microparticles) [52,53]. Additional light-scattering analyses have been applied to determine the size of the microparticles [54]. With these sophisticated techniques, it has been shown that podocyte-derived microparticles are increased in mice upon exposure to high glucose [53], as well as in diabetic mice before the onset of albuminuria [54]. However, the application of these techniques to human kidney disease has not been explored.
In addition to measuring the number and size of podocyte-derived EVs and microparticles in the urine, there is a growing interest in measuring podocyte-specific molecules in urinary exosomes. For example, podocyte-derived signal transduction factors in urinary EVs have been found to be a promising candidate for the assessment of podocyte injuries [54]. However, their clinical applications remain under development.

Urinary podocyte-specific protein targets

The urinary levels of several podocyte-specific protein targets can be measured with ease, and their role as biomarkers of kidney disease has been studied extensively. In healthy adults, urinary podocalyxin and nephrin levels are generally detectable by enzyme-linked immunosorbent assay but not by traditional Western blotting [55]. Urinary nephrin level correlates with the severity of proteinuria [56] and kidney function [57]. Urinary podocalyxin level can also be measured by indirect immunofluorescence or flow cytometry [58], and it has been proposed as a marker for urinary podocyte count. Notably, urinary podocalyxin originated from the apical membrane of injured podocytes, and its level is increased in early kidney injury [51]. The major problem of using a urinary level of podocyte-specific protein targets as a biomarker is the confounding effect of urine concentration dilution. Urinary creatinine concentration is commonly used for adjustment, but this is only appropriate for free proteins in urinary supernatant but not for the levels in the cellular sediment.

Urinary podocyte-specific messenger RNAs

Measurement of urinary podocyte-specific messenger RNA (mRNA) level has been explored as the marker of intrarenal podocyte injury. Podocyte-specific mRNA level in urine is readily measured by real-time quantitative polymerase chain reaction (RT-QPCR). Two previous studies showed that urinary nephrin mRNA levels had a close correlation with urinary podocyte count [56,59]. In an animal study, urinary podocyte-specific mRNA levels also correlated with the rate of glomerular podocyte loss as determined by kidney biopsy [54].
Recent studies, however, have shifted the focus to derived urinary mRNA indices. In a rat model, the urinary podocin-to-nephrin mRNA ratio had a better correlation with the severity of histological progression than the absolute mRNA levels [60]. In healthy adults, urinary podocin mRNA-to-creatinine ratio (a marker of podocyte detachment), podocin-to-nephrin mRNA ratio (a marker of podocyte stress), and urinary podocin-to-aquaporin-2 mRNA ratio (a marker of relative podocyte injury versus tubular injury) correlated with the arterial blood pressure [61]. Glomerular injury is specifically associated with increased urinary podocin-to-aquaporin-2 and nephrin-to-aquaporin-2 mRNA ratios [62].

Urinary podocyte-specific micro-RNAs

Micro-RNAs (miRNAs) are short noncoding RNAs that regulate many biological pathways by targeting specific mRNAs [63]. Similar to mRNA, urinary miRNA level is easily measured by RT-QPCR [64]. Unlikely mRNA, miRNAs are resistant to degradation, which makes them promising biomarkers for clinical use [64]. A number of specific miRNA changes have been reported in kidney diseases [65]. For example, podocyte-specific loss of miR-23b, miR-24, and miR-26a was associated with rapidly progressive glomerular injury [66], and urinary miR-21, miR-124, and miR-192 levels correlated urinary nephrin, synaptopodin, and podocalyxin levels in diabetic patients [67]. In addition, there are studies that delineated the pathophysiological mechanism of miRNA-mediated podocyte dysfunction. Notably, miR-193a suppresses the expression of WT-1 and affects podocyte differentiation [68]. Similarly, miR-26a was the most abundantly expressed miRNA in the glomerulus of normal C57BL/6 mouse, and miR-26a was responsible for the regulation of podocyte differentiation and cytoskeletal integrity [69]. In human studies, urinary exosomal miR-26a levels were significantly higher in lupus nephritis than in healthy controls [69].
However, although many miRNA targets may be significantly altered in podocyte dysfunction, their urinary levels would unlikely to serve as podocyte markers. This is because podocyte contributes to only a small proportion of urinary miRNA. Any specific miRNA alterations observed in the podocytes may not be readily discernable in the urine, while any alteration in the urinary miRNA levels may reflect the pathological change in other renal cell types [70,71]. For example, miR-21 inhibits the expression of tissue inhibitor of metalloproteinase 3 (TIMP3) [72], miR-26a inhibits the expression of transforming growth factor beta 1 [73], miR-23b targets Ras GTPase-activating protein SH3 domain-binding protein 2 (G3BP2) [74], and miR-29c targets Sprouty homolog 1 (Spry1) [75]. Although these miRNA targets and their corresponding pathways are relevant for the pathogenesis of podocyte dysfunction, they may also be affected in other cell types in the kidney. Similarly, although urinary levels of miR-155, miR-663, and miR-1915 are significantly different between patients with FGS or minimal change nephropathy and healthy controls [76], it has not been shown that miR-155, miR-663, or miR-1915 specifically originates from podocytes.

Adjusting for urinary concentration

The key advantages and disadvantages of the above-mentioned methodologies are summarized in Table 2. An important methodological consideration for studying any urinary biomarker is how to adjust for the concentration-dilution effect of the urine. For the study of urinary electrolytes, the traditional methods are via the calculation of fractional excretion or the trans-tubular gradient [77]. Both of these methods require the concomitant measurement of plasma electrolyte level. For urinary biomarkers, a common method is to adjust for urinary creatinine concentration, which originates from the practice of meuring urinary protein- or albumin-creatinine ratio in chronic kidney disease [78]. However, this method is actually only suitable for molecules that are freely soluble in the urine, and therefore the concentration of which is truly affected by urinary concentration-dilution and could be adjusted appropriately by the urinary creatinine concentration. For molecular targets present in urinary sediment, the quantity in a centrifuged sample is not affected by any concentration-dilution effect of the urine, but by the duration of urine collection (i.e., if the rate of excretion is constant, an 8-hour urine sample should contain twice the amount of the molecule as compared to a 4-hour urine sample). A timed urine sample or expressing the result as a rate of excretion per hour would seem appropriate for this kind of molecular target—akin to the reporting of albuminuria [79]. For molecular targets in urinary microvesicles, the optimal method of adjustment is less well-defined, and it probably depends on the centrifugation protocol.

Urinary podocyte markers in diabetic kidney disease

There is a wealth of literature on the use of various urinary podocyte markers for the diagnosis and monitoring of DKD. Amongst all podocyte-specific protein targets, the most promising ones are podocalyxin and glycogen synthase kinase 3β (GSK3β). Podocalyxin level in urine was increased in diabetic patients before the onset of microalbuminuria, and therefore likely valuable for the early detection of DKD [80]. In patients with overt DKD, urine podocalyxin levels correlated with albumin-to-creatinine ratio [81,82]. On the other hand, GSK3β levels in urinary exfoliated cells were more accurate than albuminuria in discriminating diabetic patients with and without progressive renal impairment [83], this assay is cumbersome and may not be applicable to routine clinical practice. Our recent study showed that urinary phosphorylated GSK3β (p-GSK3β) level correlated with kidney function, but urinary total GSK3β, p-GSK3β level, its mRNA level, or the p-GSK3β/GSK3β ratio did not predict dialysis-free survival or the rate of kidney function decline [84].
As to other podocyte-specific mRNA targets, urinary mRNA levels of nephrin, podocin, synaptopodin, WT-1, and α-actin-4 were elevated in DKD [85], and they also correlated with urinary podocyte count, urinary nephrin level, albuminuria, and the severity of renal impairment [86]. More importantly, urinary podocyte-specific mRNA levels preceded the onset of microalbuminuria in diabetic patients [86], and the urinary podocin mRNA-to-creatinine ratio (a marker of podocyte detachment) predicted the subsequent rate of kidney function decline [87]. However, all of these studies have small sample sizes and the inclusion criteria are prone to selection bias. Further large-scale studies are necessary to validate their results before routine clinical application can be considered.
In addition to biomarkers specific for DKD, the urinary level of several podocyte-specific targets has been explored as biomarkers for CKD in general. For example, urine synaptopodin level correlated with kidney function in both diabetic and non-diabetic CKD, regardless of the degree of albuminuria [88]. However, not all kidney diseases have podocyte injury [89], and the association between podocyturia and proteinuria varied markedly in different diseases, indicating that urinary podocyte markers should be explored as disease-specific biomarkers [89].

Challenges and future studies

Although podocytes play a crucial role in the development and progression of DKD, the use of podocyte-associated molecules in urine as biomarkers of DKD is still in its early stages of development [90,91]. Before this technology can be applied in clinical practice, much work is needed to define the molecular target (or combination of targets) to be tested. Since the amount of mRNA and miRNA in urine is small, there is no reliable method to determine the gene expression library in urine [92,93], and identifying suitable targets is often a fishing exercise. Moreover, a urine sample contains separate compartments (i.e., conventional sediment, microvesicles, and truly cell-free supernatant), and podocyte-associated molecules (proteins, mRNA, and miRNA) are present in them at different concentrations [94,95]. Since the compartment to be tested determines the centrifugation protocol [50,96], it is a necessary but arduous task to define the suitable molecular target in each specific urinary compartment for clinical use.
In developing biomarkers for DKD, there is another theoretical aspect that is often overlooked. Specifically, two types of biomarkers can be identified: those that are specific to DKD, and others that are generic for any type of kidney damage. The type of biomarker identified depends on the study protocol and the selection of the control group [97]. It can be difficult to identify markers that are specific to podocyte injury or dysfunction, as opposed to markers of kidney inflammation or other pathological processes. However, if a reliable generic marker of kidney disease (a predictor of kidney function loss regardless of the underlying cause of kidney damage) were identified, it would have great clinical potential. Since podocyte loss is a central factor in the progression of many kidney diseases, we believe it would be beneficial to focus on the search for urinary biomarkers related to podocytes.
Taken together, although available studies provide preliminary ideas on the potential of podocyte-specific targets as biomarkers, there is much to do before they can be applied to clinical practice. While the published data focused on the potential of these biomarkers, they did not clearly define their clinical relevance or practical implications. It would be crucial to delineate the prospect of translation of these biomarkers into clinical practice, including the potential challenges and opportunities that may arise. Additionally, the validation of these biomarkers in large-scale studies is necessary to ensure their accuracy and reliability. Finally, it is important to consider the prospective impact of these biomarkers on patient management, including potential changes in treatment plans. By addressing these important considerations, we can gain a deeper understanding of the potential of podocyte-specific targets as biomarkers and their impact on clinical practice.

Conclusion

Podocyte injury plays an important role in the pathogenesis and progression of many kidney diseases. Urinary levels of podocyte-derived cellular fragments and podocyte-specific molecules may serve as biomarkers for the diagnosis and monitoring of kidney diseases. With the advance in our understanding of podocyte biology, the development of new technologies, and the increasing availability of existing ones, urinary podocyte markers are expected to have an expanding application. On the other hand, the identification of specific urinary podocyte markers may shed light on the pathophysiology of kidney diseases. Further research should focus on the standardization and automation of laboratory methods, as well as defining their added value to the available tests.

Notes

Conflicts of interest

All authors have no conflicts of interest to declare.

Funding

This work was supported by the Richard Yu Chinese University of Hong Kong (CUHK) PD Research Fund, and CUHK research accounts 6905134 and 8601286. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Data sharing statement

The data presented in this study are available upon reasonable request from the corresponding author.

Authors’ contributions

Conceptualization: CCS

Methodology: CL

Writing–original draft: CL

Writing–review & editing: CCS

All authors read and approved the final manuscript.

Figure 1.

Podocyte injury and potential podocyte-derived markers in urine.

Injury to podocytes results in three inter-related pathological processes: morphological change, apoptosis, and detachment. Podocytes may remain viable but manifest as morphological changes that have functional downstream consequences, such as proteinuria and tubulointerstitial fibrosis. Podocyte apoptosis may develop as a result of cumulated morphological changes or directly from specific insults. Loss of podocyte in the glomeruli (i.e., podocytopenia) leads to architectural changes of the glomerular capillary loop and results in glomerulosclerosis. Podocytes, either apoptotic or viable ones due to loss of α3β1 integrin, may detach to the urinary space, be identified in the urine, and serve as markers of kidney disease. Vesicles, microparticles, or podocyte-specific molecules derived from injured or apoptotic podocyte may also be detected in the urine and serve as biomarkers.
j-krcp-23-109f1.jpg
Table 1.
Major podocyte-related molecules that are potential targets for biomarker development
Molecules Gene Associated genetic disease
Nucleus
 Wilms’ tumor suppressor gene-1 WT1 Deny-Drash syndrome, Frasier syndrome
Cytoskeleton
 Synaptopodin SYNPO Congenital nephrotic syndrome
 Alpha-actinin-4 ACTN4 Early or late onset SRNS
Slit diaphragm
 Nephrin NPHS1 Early onset SRNS
 Podocin NPHS2 Early or late onset SRNS
 CD2-associated protein CD2AP Early onset SRNS
 Podoplanin PDPN (None reported in human)
Foot process
 α3β1 Integrin ITGA3, ITGB1 Kidney development defect (α3 integrin)
 α-Dystroglycan DAG1 Muscular dystrophy
Top membrane
 Podocalyxin PODXL Congenital nephrotic syndrome
Other cytoplasmic/mitochondrial targets
 AarF domain containing kinase-4 ADCK4 SRNS

SRNS, steroid-resistant nephrotic syndrome.

Table 2.
Methods of detecting urinary podocyte markers
Methods Advantage Disadvantage and limitations
Podocyte quantification
 Cytospin and immunofluorescence Possibility of automation Limited sensitivity and specificity; long turn-around time
Ex vivo culture Specific Technically demanding, time consuming; unlikely feasible for routine clinical practice
Podocyte-derived extracellular vesicles
 Electron microscopy Standardized method Result difficult to interpret and bd quantified; long turn-around time
 Serial centrifugation/flow cytometry Able to identify sublethal podocyte injury Protocol not standardized; expensive and labor intensive
Podocyte-specific proteins ELISA-based method is simple and often commercially available Uncertain pathophysiological relevance and clinical utility; variable degradation of the protein target after sample collection; method to adjust for urinary concentration effect controversial; choice of target (or combination of targets) to be defined
Podocyte-specific mRNA PCR-based method is standardized, new targets readily accommodated Handling of urine sample is tedious and often not feasible for archive materials; method to adjust for urinary concentration effect controversial; choice of target (or combination of targets) to be defined
MiRNA markers PCR-based method is method standardized, new targets readily accommodated, handling of archive sample is simple Targets unlikely to be podocyte-specific, uncertain pathophysiological relevance; method to adjust for urinary concentration effect controversial; choice of target (or combination of targets) to be defined

ELISA, enzyme-linked immunosorbent assay; mRNA, messenger RNA; miRNA, microRNA; PCR, polymerase chain reaction.

References

1. Kovesdy CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int Suppl (2011) 2022;12:7–11.
crossref pmid pmc
2. Tummalapalli SL, Anderson GF. Comparative health care spending for dialysis: an example of public cost containment? J Am Soc Nephrol 2021;32:2103–2104.
crossref pmid pmc
3. Bello AK, Okpechi IG, Osman MA, et al. Epidemiology of haemodialysis outcomes. Nat Rev Nephrol 2022;18:378–395.
crossref pmid pmc pdf
4. Ulasi II, Awobusuyi O, Nayak S, et al. Chronic kidney disease burden in low-resource settings: regional perspectives. Semin Nephrol 2022;42:151336.
crossref pmid
5. Sundström J, Bodegard J, Bollmann A, et al. Prevalence, outcomes, and cost of chronic kidney disease in a contemporary population of 2•4 million patients from 11 countries: the CaReMe CKD study. Lancet Reg Health Eur 2022;20:100438.
crossref pmid pmc
6. Denic A, Rule AD, Glassock RJ. Healthy and unhealthy aging on kidney structure and function: human studies. Curr Opin Nephrol Hypertens 2022;31:228–234.
crossref pmid pmc
7. Jiang A, Song A, Zhang C. Modes of podocyte death in diabetic kidney disease: an update. J Nephrol 2022;35:1571–1584.
crossref pmid pdf
8. Ichimura K, Miyazaki N, Sadayama S, et al. Three-dimensional architecture of podocytes revealed by block-face scanning electron microscopy. Sci Rep 2015;5:8993.
crossref pmid pmc pdf
9. Schell C, Huber TB. The evolving complexity of the podocyte cytoskeleton. J Am Soc Nephrol 2017;28:3166–3174.
crossref pmid pmc
10. Reynolds PA. The mechanobiology of kidney podocytes in health and disease. Clin Sci (Lond) 2020;134:1245–1253.
crossref pmid pdf
11. Ozawa S, Ueda S, Imamura H, et al. Glycolysis, but not mitochondria, responsible for intracellular ATP distribution in cortical area of podocytes. Sci Rep 2015;5:18575.
crossref pmid pmc pdf
12. Nisoli E, Falcone S, Tonello C, et al. Mitochondrial biogenesis by NO yields functionally active mitochondria in mammals. Proc Natl Acad Sci U S A 2004;101:16507–16512.
crossref pmid pmc
13. Qin X, Zhao Y, Gong J, et al. Berberine protects glomerular podocytes via inhibiting Drp1-mediated mitochondrial fission and dysfunction. Theranostics 2019;9:1698–1713.
crossref pmid pmc
14. Li W, Du M, Wang Q, et al. FoxO1 promotes mitophagy in the podocytes of diabetic male mice via the PINK1/Parkin pathway. Endocrinology 2017;158:2155–2167.
crossref pmid
15. Li SY, Susztak K. The role of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in kidney disease. Semin Nephrol 2018;38:121–126.
crossref pmid pmc
16. Coughlan MT, Higgins GC, Nguyen TV, et al. Deficiency in apoptosis-inducing factor recapitulates chronic kidney disease via aberrant mitochondrial homeostasis. Diabetes 2016;65:1085–1098.
crossref pmid pdf
17. Inoue T, Maekawa H, Inagi R. Organelle crosstalk in the kidney. Kidney Int 2019;95:1318–1325.
crossref pmid
18. Brinkkoetter PT, Bork T, Salou S, et al. Anaerobic glycolysis maintains the glomerular filtration barrier independent of mitochondrial metabolism and dynamics. Cell Rep 2019;27:1551–1566.
crossref pmid pmc
19. Luo Q, Liang W, Zhang Z, et al. Compromised glycolysis contributes to foot process fusion of podocytes in diabetic kidney disease: role of ornithine catabolism. Metabolism 2022;134:155245.
crossref pmid
20. Imasawa T, Obre E, Bellance N, et al. High glucose repatterns human podocyte energy metabolism during differentiation and diabetic nephropathy. FASEB J 2017;31:294–307.
crossref pmid pmc pdf
21. Qi W, Keenan HA, Li Q, et al. Pyruvate kinase M2 activation may protect against the progression of diabetic glomerular pathology and mitochondrial dysfunction. Nat Med 2017;23:753–762.
pmid pmc
22. Chen Z, Zhu Z, Liang W, et al. Reduction of anaerobic glycolysis contributes to angiotensin II-induced podocyte injury with foot process effacement. Kidney Int 2023;103:735–748.
crossref pmid
23. Liu C, Wu J, Zhu J, et al. Lactate inhibits lipolysis in fat cells through activation of an orphan G-protein-coupled receptor, GPR81. J Biol Chem 2009;284:2811–2822.
crossref pmid
24. Roshanravan H, Kim EY, Dryer SE. NMDA receptors as potential therapeutic targets in diabetic nephropathy: increased renal NMDA receptor subunit expression in akita mice and reduced nephropathy following sustained treatment with memantine or MK-801. Diabetes 2016;65:3139–3150.
crossref pmid pmc pdf
25. Shen J, Wang R, He Z, et al. NMDA receptors participate in the progression of diabetic kidney disease by decreasing Cdc42-GTP activation in podocytes. J Pathol 2016;240:149–160.
crossref pmid pdf
26. Thangaraju M, Ananth S, Martin PM, et al. c/ebpdelta Null mouse as a model for the double knock-out of slc5a8 and slc5a12 in kidney. J Biol Chem 2006;281:26769–26773.
pmid
27. Nagasawa K, Nagai K, Sumitani Y, et al. Monocarboxylate transporter mediates uptake of lovastatin acid in rat cultured mesangial cells. J Pharm Sci 2002;91:2605–2613.
crossref pmid
28. Yang Q, Hu J, Yang Y, et al. Sirt6 deficiency aggravates angiotensin II-induced cholesterol accumulation and injury in podocytes. Theranostics 2020;10:7465–7479.
crossref pmid pmc
29. Ahmed K, Tunaru S, Tang C, et al. An autocrine lactate loop mediates insulin-dependent inhibition of lipolysis through GPR81. Cell Metab 2010;11:311–319.
crossref pmid
30. Lubojemska A, Stefana MI, Sorge S, et al. Adipose triglyceride lipase protects renal cell endocytosis in a Drosophila dietary model of chronic kidney disease. PLoS Biol 2021;19:e3001230.
crossref pmid pmc
31. Chen W, Jiang Y, Han J, et al. Atgl deficiency induces podocyte apoptosis and leads to glomerular filtration barrier damage. FEBS J 2017;284:1070–1081.
crossref pmid pdf
32. Wang Q, Zhao B, Zhang J, et al. Faster lipid β-oxidation rate by acetyl-CoA carboxylase 2 inhibition alleviates high-glucose-induced insulin resistance via SIRT1/PGC-1α in human podocytes. J Biochem Mol Toxicol 2021;35:e22797.
crossref pmid pdf
33. Zhou L, Liu Y. Wnt/β-catenin signalling and podocyte dysfunction in proteinuric kidney disease. Nat Rev Nephrol 2015;11:535–545.
crossref pmid pmc pdf
34. Vogtländer NP, Dijkman H, Bakker MA, Campbell KP, van der Vlag J, Berden JH. Localization of alpha-dystroglycan on the podocyte: from top to toe. J Histochem Cytochem 2005;53:1345–1353.
crossref pmid pdf
35. Verma R, Venkatareddy M, Kalinowski A, et al. Nephrin is necessary for podocyte recovery following injury in an adult mature glomerulus. PLoS One 2018;13:e0198013.
crossref pmid pmc
36. Sever S, Reiser J. CD2AP, dendrin, and cathepsin L in the kidney. Am J Pathol 2015;185:3129–3130.
crossref pmid pmc
37. Koop K, Eikmans M, Wehland M, et al. Selective loss of podoplanin protein expression accompanies proteinuria and precedes alterations in podocyte morphology in a spontaneous proteinuric rat model. Am J Pathol 2008;173:315–326.
crossref pmid pmc
38. Torban E, Braun F, Wanner N, et al. From podocyte biology to novel cures for glomerular disease. Kidney Int 2019;96:850–861.
crossref pmid
39. Nielsen JS, McNagny KM. The role of podocalyxin in health and disease. J Am Soc Nephrol 2009;20:1669–1676.
crossref pmid
40. Kannan N, Tang VW. Synaptopodin couples epithelial contractility to α-actinin-4-dependent junction maturation. J Cell Biol 2015;211:407–434.
crossref pmid pmc pdf
41. Ning L, Suleiman HY, Miner JH. Synaptopodin is dispensable for normal podocyte homeostasis but is protective in the context of acute podocyte injury. J Am Soc Nephrol 2020;31:2815–2832.
crossref pmid pmc
42. Ashraf S, Gee HY, Woerner S, et al. ADCK4 mutations promote steroid-resistant nephrotic syndrome through CoQ10 biosynthesis disruption. J Clin Invest 2013;123:5179–5189.
crossref pmid pmc
43. Widmeier E, Yu S, Nag A, et al. ADCK4 deficiency destabilizes the coenzyme Q Complex, which is rescued by 2,4-dihydroxybenzoic acid treatment. J Am Soc Nephrol 2020;31:1191–1211.
crossref pmid pmc
44. Vogelmann SU, Nelson WJ, Myers BD, Lemley KV. Urinary excretion of viable podocytes in health and renal disease. Am J Physiol Renal Physiol 2003;285:F40–F48.
crossref pmid pmc
45. Kriz W, Lemley KV. A potential role for mechanical forces in the detachment of podocytes and the progression of CKD. J Am Soc Nephrol 2015;26:258–269.
crossref pmid pmc
46. Hara M, Yamamoto T, Yanagihara T, et al. Urinary excretion of podocalyxin indicates glomerular epithelial cell injuries in glomerulonephritis. Nephron 1995;69:397–403.
crossref pmid pdf
47. Garovic VD, Craici IM, Wagner SJ, et al. Mass spectrometry as a novel method for detection of podocyturia in pre-eclampsia. Nephrol Dial Transplant 2013;28:1555–1561.
crossref pmid pmc
48. Shankland SJ, Pippin JW, Reiser J, Mundel P. Podocytes in culture: past, present, and future. Kidney Int 2007;72:26–36.
crossref pmid
49. van der Pol E, Böing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev 2012;64:676–705.
crossref pmid
50. Zhou H, Yuen PS, Pisitkun T, et al. Collection, storage, preservation, and normalization of human urinary exosomes for biomarker discovery. Kidney Int 2006;69:1471–1476.
crossref pmid pmc
51. Hara M, Yanagihara T, Kihara I, Higashi K, Fujimoto K, Kajita T. Apical cell membranes are shed into urine from injured podocytes: a novel phenomenon of podocyte injury. J Am Soc Nephrol 2005;16:408–416.
pmid
52. Sharma K. Tell-tale signs of perturbed podocytes. J Am Soc Nephrol 2014;25:1367–1369.
crossref pmid pmc
53. Burger D, Thibodeau JF, Holterman CE, Burns KD, Touyz RM, Kennedy CR. Urinary podocyte microparticles identify prealbuminuric diabetic glomerular injury. J Am Soc Nephrol 2014;25:1401–1407.
crossref pmid pmc
54. Fukuda A, Minakawa A, Sato Y, et al. Urinary podocyte and TGF-β1 mRNA as markers for disease activity and progression in anti-glomerular basement membrane nephritis. Nephrol Dial Transplant 2017;32:1818–1830.
crossref pmid pmc
55. Garg P. A review of podocyte biology. Am J Nephrol 2018;47 Suppl 1:3–13.
crossref pmid pdf
56. Wang P, Li M, Liu Q, Chen B, Ji Z. Detection of urinary podocytes and nephrin as markers for children with glomerular diseases. Exp Biol Med (Maywood) 2015;240:169–174.
crossref pmid pmc pdf
57. Son GH, Kwon JY, Lee S, et al. Comparison of serum and urinary nephrin levels between normal pregnancies and severe preeclampsia. Eur J Obstet Gynecol Reprod Biol 2013;166:139–144.
crossref pmid
58. Palacios de Franco Y, Velazquez K, Segovia N, et al. Urinary podocalyxin as a marker of preeclampsia in a Hispanic population. Int J Physiol Pathophysiol Pharmacol 2014;6:115–124.
pmid pmc
59. Kelder TP, Penning ME, Uh HW, et al. Quantitative polymerase chain reaction-based analysis of podocyturia is a feasible diagnostic tool in preeclampsia. Hypertension 2012;60:1538–1544.
crossref pmid
60. Fukuda A, Wickman LT, Venkatareddy MP, et al. Urine podocin: nephrin mRNA ratio (PNR) as a podocyte stress biomarker. Nephrol Dial Transplant 2012;27:4079–4087.
crossref pmid pmc
61. Naik AS, Le D, Aqeel J, et al. Podocyte stress and detachment measured in urine are related to mean arterial pressure in healthy humans. Kidney Int 2020;98:699–707.
crossref pmid pmc
62. Sato Y, Wharram BL, Lee SK, et al. Urine podocyte mRNAs mark progression of renal disease. J Am Soc Nephrol 2009;20:1041–1052.
crossref pmid pmc
63. Trionfini P, Benigni A. MicroRNAs as master regulators of glomerular function in health and disease. J Am Soc Nephrol 2017;28:1686–1696.
crossref pmid pmc
64. Wang G, Szeto CC. Methods of microRNA quantification in urinary sediment. Methods Mol Biol 2013;1024:211–220.
crossref pmid
65. Ishii H, Kaneko S, Yanai K, et al. MicroRNAs in podocyte injury in diabetic nephropathy. Front Genet 2020;11:993.
crossref pmid pmc
66. Ho J, Ng KH, Rosen S, Dostal A, Gregory RI, Kreidberg JA. Podocyte-specific loss of functional microRNAs leads to rapid glomerular and tubular injury. J Am Soc Nephrol 2008;19:2069–2075.
crossref pmid pmc
67. Milas O, Gadalean F, Vlad A, et al. Deregulated profiles of urinary microRNAs may explain podocyte injury and proximal tubule dysfunction in normoalbuminuric patients with type 2 diabetes mellitus. J Investig Med 2018;66:747–754.
crossref pmid pdf
68. Li TS, Chen L, Wang SC, et al. Magnesium isoglycyrrhizinate ameliorates fructose-induced podocyte apoptosis through downregulation of miR-193a to increase WT1. Biochem Pharmacol 2019;166:139–152.
crossref pmid
69. Ichii O, Otsuka-Kanazawa S, Horino T, et al. Decreased miR-26a expression correlates with the progression of podocyte injury in autoimmune glomerulonephritis. PLoS One 2014;9:e110383.
crossref pmid pmc
70. Liu Y, Gao G, Yang C, et al. Stability of miR-126 in urine and its potential as a biomarker for renal endothelial injury with diabetic nephropathy. Int J Endocrinol 2014;2014:393109.
crossref pmid pmc pdf
71. Szeto CC, Ching-Ha KB, Ka-Bik L, et al. Micro-RNA expression in the urinary sediment of patients with chronic kidney diseases. Dis Markers 2012;33:137–144.
crossref pmid pmc pdf
72. Wang F, Gao X, Zhang R, Zhao P, Sun Y, Li C. LncRNA TUG1 ameliorates diabetic nephropathy by inhibiting miR-21 to promote TIMP3-expression. Int J Clin Exp Pathol 2019;12:717–729.
pmid pmc
73. Koga K, Yokoi H, Mori K, et al. MicroRNA-26a inhibits TGF-β-induced extracellular matrix protein expression in podocytes by targeting CTGF and is downregulated in diabetic nephropathy. Diabetologia 2015;58:2169–2180.
crossref pmid pdf
74. Zhao B, Li H, Liu J, et al. 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;27:2597–2608.
crossref pmid pmc
75. 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.
crossref pmid pmc
76. Ramezani A, Devaney JM, Cohen S, et al. Circulating and urinary microRNA profile in focal segmental glomerulosclerosis: a pilot study. Eur J Clin Invest 2015;45:394–404.
pmid pmc
77. Kamel KS, Halperin ML. Use of urine electrolytes and urine osmolality in the clinical diagnosis of fluid, electrolytes, and acid-base disorders. Kidney Int Rep 2021;6:1211–1224.
crossref pmid pmc
78. Rossing P, Caramori ML, Chan JC, et al. Executive summary of the KDIGO 2022 clinical practice guideline for diabetes management in chronic kidney disease: an update based on rapidly emerging new evidence. Kidney Int 2022;102:990–999.
crossref pmid pdf
79. Fagerstrom P, Sallsten G, Akerstrom M, Haraldsson B, Barregard L. Urinary albumin excretion in healthy adults: a cross sectional study of 24-hour versus timed overnight samples and impact of GFR and other personal characteristics. BMC Nephrol 2015;16:8.
crossref pmid pmc pdf
80. Kostovska I, Tosheska-Trajkovska K, Topuzovska S, et al. Urinary nephrin is earlier, more sensitive and specific marker of diabetic nephropathy than microalbuminuria. J Med Biochem 2020;39:83–90.
crossref pmid pmc
81. Hara M, Yamagata K, Tomino Y, et al. Urinary podocalyxin is an early marker for podocyte injury in patients with diabetes: establishment of a highly sensitive ELISA to detect urinary podocalyxin. Diabetologia 2012;55:2913–2919.
crossref pmid pmc
82. Shoji M, Kobayashi K, Takemoto M, Sato Y, Yokote K. Urinary podocalyxin levels were associated with urinary albumin levels among patients with diabetes. Biomarkers 2016;21:164–167.
crossref pmid
83. Liang X, Wang P, Chen B, et al. Glycogen synthase kinase 3β hyperactivity in urinary exfoliated cells predicts progression of diabetic kidney disease. Kidney Int 2020;97:175–192.
crossref pmid
84. Zeng L, Ng JK, Fung WW, Chan GC, Chow KM, Szeto CC. Intra-renal and urinary glycogen synthase kinase 3 beta levels in diabetic and non-diabetic chronic kidney disease. Kidney Blood Press Res 2023;48:241–248.
crossref pmid pmc pdf
85. Wang G, Lai FM, Lai KB, Chow KM, Li KT, Szeto CC. Messenger RNA expression of podocyte-associated molecules in the urinary sediment of patients with diabetic nephropathy. Nephron Clin Pract 2007;106:c169–c179.
crossref pmid pdf
86. Petrica L, Ursoniu S, Gadalean F, et al. Urinary podocyte-associated mRNA levels correlate with proximal tubule dysfunction in early diabetic nephropathy of type 2 diabetes mellitus. Diabetol Metab Syndr 2017;9:31.
crossref pmid pmc pdf
87. Fukuda A, Minakawa A, Kikuchi M, et al. Urinary podocyte mRNAs precede microalbuminuria as a progression risk marker in human type 2 diabetic nephropathy. Sci Rep 2020;10:18209.
crossref pmid pmc pdf
88. Kwon SK, Kim SJ, Kim HY. Urine synaptopodin excretion is an important marker of glomerular disease progression. Korean J Intern Med 2016;31:938–943.
crossref pmid pmc pdf
89. Wickman L, Afshinnia F, Wang SQ, et al. Urine podocyte mRNAs, proteinuria, and progression in human glomerular diseases. J Am Soc Nephrol 2013;24:2081–2095.
crossref pmid pmc
90. Zeng L, Fung WW, Chan GC, Ng JK, Chow KM, Szeto CC. Urinary and kidney podocalyxin and podocin levels in diabetic kidney disease: a kidney biopsy study. Kidney Med 2023;5:100569.
crossref pmid
91. Kravets I, Mallipattu SK. The role of podocytes and podocyte-associated biomarkers in diagnosis and treatment of diabetic kidney disease. J Endocr Soc 2020;4:bvaa029.
crossref pmid pmc pdf
92. Mamdouh S, Sherif H, Romeih M, Elesaily K. Urine micro-RNA signature as a potential non-invasive diagnostic biomarker in bladder cancer. Asian Pac J Cancer Prev 2023;24:121–131.
crossref pmid pmc
93. Baumgart S, Meschkat P, Edelmann P, et al. MicroRNAs in tumor samples and urinary extracellular vesicles as a putative diagnostic tool for muscle-invasive bladder cancer. J Cancer Res Clin Oncol 2019;145:2725–2736.
crossref pmid pdf
94. Tepus M, Tonoli E, Verderio EAM. Molecular profiling of urinary extracellular vesicles in chronic kidney disease and renal fibrosis. Front Pharmacol 2022;13:1041327.
crossref pmid
95. Mighty J, Rubio-Navarro A, Shi C, et al. Extracellular vesicles of human diabetic retinopathy retinal tissue and urine of diabetic retinopathy patients are enriched for the junction plakoglo bin protein. Front Endocrinol (Lausanne) 2022;13:1077644.
crossref pmid
96. Barreiro K, Dwivedi OP, Rannikko A, et al. Capturing the kidney transcriptome by urinary extracellular vesicles-from pre-analytical obstacles to biomarker research. Genes (Basel) 2023;14:1415.
crossref pmid pmc
97. Colhoun HM, Marcovecchio ML. Biomarkers of diabetic kidney disease. Diabetologia 2018;61:996–1011.
crossref pmid pmc pdf


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