1,633
Views
0
CrossRef citations to date
0
Altmetric
Invited Reviews

Progress in understanding primary glomerular disease: insights from urinary proteomics and in-depth analyses of potential biomarkers based on bioinformatics

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 346-365 | Received 17 Oct 2022, Accepted 06 Feb 2023, Published online: 23 Feb 2023

Abstract

Chronic kidney disease (CKD) has become a global public health challenge. While primary glomerular disease (PGD) is one of the leading causes of CKD, the specific pathogenesis of PGD is still unclear. Accurate diagnosis relies largely on invasive renal biopsy, which carries risks of bleeding, pain, infection and kidney vein thrombosis. Problems with the biopsy procedure include lack of glomeruli in the tissue obtained, and the sampling site not being reflective of the overall lesion in the kidney. Repeated renal biopsies to monitor disease progression cannot be performed because of the significant risks of bleeding and kidney vein thrombosis. On the other hand, urine collection, a noninvasive method, can be performed repeatedly, and urinary proteins can reflect pathological changes in the urinary system. Advancements in proteomics technologies, especially mass spectrometry, have facilitated the identification of candidate biomarkers in different pathological types of PGD. Such biomarkers not only provide insights into the pathogenesis of PGD but also are important for diagnosis, monitoring treatment, and prognosis. In this review, we summarize the findings from studies that have used urinary proteomics, among other omics screens, to identify potential biomarkers for different types of PGD. Moreover, we performed an in-depth bioinformatic analysis to gain a deeper understanding of the biological processes and protein–protein interaction networks in which these candidate biomarkers may participate. This review, including a description of an integrated analysis method, is intended to provide insights into the pathogenesis, noninvasive diagnosis, and personalized treatment efforts of PGD and other associated diseases.

Introduction

In the past few decades, chronic kidney disease (CKD) has become a global public health challenge [Citation1]. The prevalence of CKD is high, between 11 and 13%, and glomerular disease (GD), especially primary glomerular disease (PGD), is one of the most common causes of CKD [Citation2]. Patients with PGD may develop end-stage renal disease (ESRD) [Citation3]. Determining the pathological type of PGD together with understanding the pathogenesis of the disease is therefore pertinent and conducive to treatment. However, the invasive method of renal biopsy remains the gold standard for determining the pathological type of PGD, and there is a lack of painless diagnostic methods for PGD. Renal biopsy is challenging for patients with severely impaired renal function. Risks associated with renal biopsy include bleeding, pain, infection and kidney vein thrombosis. Problems with biopsy include lack of glomeruli in the tissue obtained, and the sampling site not being reflective of the overall lesion in the kidney. Repeated renal biopsies to monitor disease progression cannot be performed because of the significant risks of bleeding and kidney vein thrombosis. Thus there is great interest in identifying biomarkers for the diagnosis of PGD.

PGD is an immune-mediated inflammatory disease, and the immune complex can damage the glomeruli, leading to clinical symptoms [Citation4]. The application of proteomics technology using mass spectrometry (MS) has helped to identify a number of potential biomarkers in urine and has recently played an important role in studying the pathogenesis of several diseases as well as achieving diagnosis and prognosis without the use of biopsies [Citation5].

In this review, we summarize articles on urinary proteomics involving PGD and provided an overview of the developments in this area. Additionally, differentially expressed proteins (DEPs) were selected for in-depth analysis using a bioinformatic approach.

Advantages of urinary proteomics in the study of glomerular disease

Urine is an easily accessible biological fluid with the advantage of noninvasive collection [Citation6]. When there is an abnormality in the urinary system or a kidney-related disease, the levels of several proteins in the urine change. Thus, urine is an ideal source for identifying biomarkers related to kidney disease [Citation7].

Proteomics is the study of all proteins expressed by specific cells, tissues, organs, or organisms, and it can reveal changes in protein levels at a given time. Various functional proteins are not only important active substances with distinct physiological roles but also can be directly related to pathological states. Urinary proteomics describes all the proteins in the urine, the levels of which may change in disorders of the kidney and following kidney damage. Increasing attention has been given to urinary proteomics studies, especially in the following fields: identifying biomarkers for noninvasive diagnosis, studying the function of DEPs, and investigating interactions between target proteins to understand the mechanisms of pathogenesis.

Many emerging technologies have been developed based on core proteomics methods, and proteomics technologies based on MS have played crucial roles in identifying biomarkers in urine for the diagnosis of GD, and in expanding our knowledge regarding its origin.

The roadmap for the study of biomarkers in the urinary proteome

When investigating biomarkers in urine, the first step is to identify individuals with the disease of interest along with appropriate controls. Then, their urine is collected. The participants should be divided into discovery and validation groups. Urine samples should be processed by a suitable method [Citation8]. The urine collected from individuals in the discovery group may be analyzed using proteomics technologies to identify DEPs. The most commonly-used proteomic technique for screening biomarkers is MS. Chu’s team identified three DEPs using MS between patients with type 2 diabetes and HCs [Citation9]. Fang et al. collected urine samples from four patients with IgAN, four with Henoch-Schönlein purpura nephritis (HSPN), and four HCs [Citation10], and applied LC-MS/MS to identify DEPs between the disease groups and HCs. While 276 DEPs between IgA nephropathy (IgAN) patients and HCs, and 125 DEPs between HSPN patients and HCs were found, they observed that urinary α-1B-glycoprotein and afamin were considerably increased in children with IgAN and HSPN compared with HCs [Citation10]. The expression of these candidate biomarkers was assessed in a validation set of 15 patients with IgAN, 15 patients with HSPN, and 10 HCs. Then they performed bioinformatic analysis using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and protein–protein interaction (PPI) analyses of the DEPs [Citation10]. Through GO annotation enriched analysis, they found that most of the DEPs in the two diseases related to cell adhesion and biological regulation and that the DEPs were distributed mainly in the extracellular matrix and intrinsic component of the plasma membrane. KEGG pathway annotation analysis showed that the DEPs of IgAN and HSPN were involved in cell adhesion and the complement and coagulation cascades. The PPI analysis showed the complex interactions between these DEPs.

The protein concentration in urine is very low under physiological conditions (urine protein excretion <150 mg/24 h), approximately 1/1000th the plasma protein content. Thus, the presence of serum protein in urine could be indicative of glomeruli damage or another pathological state of the urinary system; consequently, changes in the levels of several target proteins in the urine may help to characterize the underlying molecular pathways of certain diseases.

Despite the importance of MS-based urine proteomics in the screening of biomarkers and the study of the pathological mechanisms of kidney diseases, urine proteomics studies also have a number of drawbacks. Proteins of low abundance are not easily detected, and urine contains high concentrations of urea and salts, which can interfere with MS results. Therefore, the removal of high-concentration proteins such as albumin, as well as salts, from urine is necessary for the identification of low-abundance proteins in MS studies. It is important to note that 70–75% of urine protein is produced by the urinary system, and 25–30% comes from plasma filtration, thereby making urine a suitable source for the study for biomarkers of urinary diseases. In this regard, urinary proteomics has a key role in further delineating the pathogenesis of PGD.

The common procedures for studies of urinary proteomics are shown in .

Figure 1. Common research steps on urinary proteomics. The first stage is to identify candidate biomarkers via proteomics analysis. When differentially expressed proteins are screened, candidate biomarkers in the validation group can be targeted. After identifying biomarkers, functional analysis of specific proteins can be carried out.

Figure 1. Common research steps on urinary proteomics. The first stage is to identify candidate biomarkers via proteomics analysis. When differentially expressed proteins are screened, candidate biomarkers in the validation group can be targeted. After identifying biomarkers, functional analysis of specific proteins can be carried out.

Urinary proteomics in healthy people

In 1996, Wilkins first put forward the concept of proteomics as the study of all the protein components expressed by the genome in specific cells or tissues [Citation11]. In the same year, Marshall et al. for the first time separated proteins from human urine using two-dimensional electrophoresis, which helped in creating a two-dimensional gel map of the human urine proteome [Citation12]. The following year, Heine et al. identified 34 highly expressed proteins via coupled microbore high-performance liquid chromatography and electrospray ionization coupled with tandem MS [Citation13]. Since then, proteomics research has developed rapidly.

The combination of two different separation techniques, acetone precipitation and ultracentrifugation, contributed to the identification of more proteins, and a relatively simple urinary proteome map of healthy people was constructed [Citation14]. Adachi et al. analyzed urinary proteins using liquid chromatography combined with linear ion trap-Fourier transform and a linear ion trap-orbitrap MS and drew up the first comprehensive protein expression map of healthy human urine [Citation15]. Zhang et al. observed the differential expression of urinary proteins between healthy Li and Han ethnic groups in a study using LC-MS/MS [Citation16]. A total of 25 urinary proteins were found to differ significantly, 16 of which had been previously reported to be biomarkers of many diseases, e.g. Ig kappa chain V-III region (which is increased in the urine of type 2 diabetic patients [Citation17]) and neutrophil gelatinase-associated lipocalin (reported to be a urinary biomarker of patients with acute kidney injury and urinary tract infection [Citation18,Citation19]). The authors confirmed that protein variability was based on ethnic rather than random differences [Citation16]; thus, ethnic group differences may be an influencing factor in the study of urinary proteomics and should be considered when urinary samples are used for biomarker discovery. Using a new enrichment method (lectin affinity chromatography), Marimuthu et al. identified 671 new proteins [Citation20]. Moreover, several proteins that were previously thought to be present only in plasma were detected in urine. Thus, there are numerous connections between blood and urinary proteins among healthy people.

Because different techniques were used in these studies, the direct comparison between results is difficult, which has in turn limited the development of proteomics to a certain extent [Citation21]. Nevertheless, progress made in research on urinary proteomics of healthy people has concomitantly allowed research on kidney diseases to flourish.

Progress of urinary proteomics in studying primary glomerular disease

Urinary proteomics and IgA nephropathy

IgA nephropathy (IgAN) is the most common chronic proliferative glomerulonephritis worldwide. Approximately one-third of patients progress to ESRD, and the associated mortality is steadily increasing [Citation22,Citation23]. In Europe, IgAN accounts for 19–51% of the total renal biopsy patients with GD [Citation24,Citation25]. The disease is related to galactose-deficient IgA1 (Gd-IgA1) [Citation26,Citation27]. While the pathogenesis of IgAN remains unclear, the theory of “four blows” is current the common consensus [Citation28–30]. This theory suggests that the development of the disease involves four main steps: hit 1. production of galactose-deficient lgA1; hit 2. production of anti-galactose-deficient lgA1 autoantibodies; hit 3. formation and deposition of immune complexes; and hit 4. deposition of immune complexes in the mesangial area and activation of the complement system, leading to glomerular damage (). The main pathological feature of IgAN is the granular or massive deposition of immune complexes, mainly IgA, in the glomerular mesangial region [Citation31–34] (). Interestingly, glycosylation of autoantibodies also occurs in other autoimmune diseases such as rheumatoid arthritis [Citation35]. Therefore, there may be a relationship between IgAN and rheumatoid arthritis. Although potentially promising, this hypothesis warrants further examination.

Figure 2. The current widely accepted pathogenesis and pathological characteristics of several glomerular diseases. (a) The popular theory of “four blows” on the pathogenesis of IgA: hit 1, production of galactose-deficient lgA1; hit 2, production of anti-galactose-deficient lgA1 autoantibodies; hit 3, immune complex formation and deposition; hit 4, deposition of immune complexes in the mesangial area, activating the complement system and causing glomerular damage. (b) Depiction of immune complexes deposited under the epithelial cells of GBM, resulting in its thickening and podocyte injury, which could damage the barrier of glomerular filtration and cause further proteinuria. (c) Depiction of glomerular changes in patients with MCD and FSGS. The glomerular lesion of the former is mild, while that of the latter shows local sclerosis. GBM: glomerular basement membrane.

Figure 2. The current widely accepted pathogenesis and pathological characteristics of several glomerular diseases. (a) The popular theory of “four blows” on the pathogenesis of IgA: hit 1, production of galactose-deficient lgA1; hit 2, production of anti-galactose-deficient lgA1 autoantibodies; hit 3, immune complex formation and deposition; hit 4, deposition of immune complexes in the mesangial area, activating the complement system and causing glomerular damage. (b) Depiction of immune complexes deposited under the epithelial cells of GBM, resulting in its thickening and podocyte injury, which could damage the barrier of glomerular filtration and cause further proteinuria. (c) Depiction of glomerular changes in patients with MCD and FSGS. The glomerular lesion of the former is mild, while that of the latter shows local sclerosis. GBM: glomerular basement membrane.

Figure 3. Pathological features of different glomerular diseases. (a) Immunofluorescence microscopy in a patient with IgA shows IgA deposition along the mesangial area (original magnification, 200×). (b) Glomerulus from a patient with idiopathic membranous nephropathy shows the thickened GBM stained with PASM (arrow) under a light microscope. (c) HE staining shows mild glomerular lesions and mild mesangial hyperplasia (arrow) from a patient with MCD. (d) The glomeruli with segmental sclerosis (arrow) are shown via PAS staining in a patient with FSGS. These images were provided by the Department of Nephrology of Shengjing Hospital. HE: hematoxylin-eosin; PAS: Schiff periodic acid; PASM: periodic acid-silver metheramine.

Figure 3. Pathological features of different glomerular diseases. (a) Immunofluorescence microscopy in a patient with IgA shows IgA deposition along the mesangial area (original magnification, 200×). (b) Glomerulus from a patient with idiopathic membranous nephropathy shows the thickened GBM stained with PASM (arrow) under a light microscope. (c) HE staining shows mild glomerular lesions and mild mesangial hyperplasia (arrow) from a patient with MCD. (d) The glomeruli with segmental sclerosis (arrow) are shown via PAS staining in a patient with FSGS. These images were provided by the Department of Nephrology of Shengjing Hospital. HE: hematoxylin-eosin; PAS: Schiff periodic acid; PASM: periodic acid-silver metheramine.

Preliminary studies on the urinary proteome of patients with IgAN were performed to detect DEPs that would differentiate IgAN from other nephropathies. A previous study established the urinary polypeptide maps of patients with IgAN using two-dimensional electrophoresis, which distinguished these individuals from HCs and patients with membranous nephropathy (MN), minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS), as well as diabetic nephropathy [Citation36]. Another team also used DEPs to distinguish IgAN from HCs and other kidney diseases. [Citation37]. It is worth mentioning, the overall specificity of this panel was as high as 82.3%. Subsequently, with more data being obtained, together with advances in recent technology, several researcher groups screened for markers related to the severity of IgAN and established relevant models [Citation38,Citation39]. The well-known IgAN 237 classifier, which contains 237 different urinary peptides of progressive and nonprogressive IgAN and was developed through CE-MS, was found to indicate the severity of IgAN accurately [Citation40].

One team examined the urinary proteome of Uygur-ethnic patients with IgAN in Xinjiang, and screened out four candidate biomarkers (adiponectin, antithrombin-III, intercellular adhesion molecule 1, and metalloproteinase inhibitor 1) [Citation41]. However, another study that included urine samples from children revealed different biomarkers through CE-MS/MS, while also reporting that the contents of α-1B-glycoprotein and afamin in the urine were higher in children with IgAN than in healthy children [Citation10]. The variability of the results might be caused by variances in the chosen sample sources and techniques used as per the particular purpose of each study.

We have summarized urinary proteomic studies on IgAN in .

Table 1. Summary of research progress on urinary proteomics of glomerular disease.

As these collective findings highlight, potential IgAN biomarkers are related mainly to podocytes, immune complexes, and complement components. Podocytes, together with basement membrane and capillary endothelial cells, constitute the glomerular hemofiltration barrier and are one of the main cell types that maintain normal structure and function of the glomerular hemofiltration membrane. When podocytes are damaged, symptoms such as proteinuria occur. IgAN is an autoimmune disease. Galactose-deficient IgA1 exposes its hinge region epitopes, resulting in the production of IgA antibodies against abnormal IgA1, along with the formation and deposition of immune complexes in the mesangial region. IgA deposited in the mesangial region can activate the complement system through the bypass pathway and mannose-binding lectin pathway, which promotes inflammation and further pathological injury. Therefore, the level of complement components in urine may be closely related to the disease.

Urinary proteomics and membranous nephropathy

Membranous nephropathy (MN) is the main pathological type of adult nephrotic syndrome. It can occur in all age groups, although children are less likely than adults to develop the disease [Citation58–62]. Approximately, 30% of MN patients progress to ESRD [Citation63]. The main pathological feature of MN is the appearance of autoantibodies and activation of the complement system, which together form a membrane attack complex that targets the glomerular podocytes and causes podocyte damage and proteinuria. Immune complexes are deposited under epithelial cells of the glomerular basement membrane, leading to its thickening [Citation64,Citation65] ( and Citation3(B)). The M-type phospholipase A2 receptor (PLA2R) is the main antigen of primary MN [Citation66,Citation67]. The detection of serum anti-PLA2R antibody levels has been widely used as a noninvasive diagnostic method in clinical settings [Citation68,Citation69].

Several studies have been published on the urinary proteomics of MN. In a study using MALDI-TOF-MS/MS to compare the urinary proteome of passive Heymann nephritis in mice with the proteome prior to modeling, the content of α-1-antitrypsin (A1AT) increased in the urine of the nephritic mice [Citation70], Several proteomic studies have also found A1AT to be abnormally expressed in the urine of patients with kidney diseases [Citation40,Citation42,Citation45,Citation47,Citation48,Citation71]. A1AT is the most abundant serum serine protease inhibitor, whose main physiological role is to inhibit the activity of specific serine proteases [Citation48,Citation72]. A1AT overexpression can lead to inhibition of neutrophil elastase, accelerating the accumulation of mesangial matrix [Citation72]. As MN is a disease related to glomerular podocyte injury and A1AT is found in the cytoplasm of sclerotic glomeruli podocytes, further studies that examine the relationship between A1AT and podocyte dysfunction may help in deepening our understanding of this disease [Citation73]. Lin et al. analyzed polypeptides in the urine of patients with MN using LC-MS/MS and screened for DEPs [Citation50]. They predicted the proteases that were related to the DEPs through relevant database investigations and provided the impetus for identifying biomarkers of other kidney diseases using a similar method. Another study found that levels of C3a and C5a in the urine of patients with MN increased significantly compared to controls, indicating that the complement system was strongly activated [Citation49]. Renal biopsy has demonstrated the deposition of complement complexes in patients with MN. The products of complement activation are expected to serve as sensitive biomarkers for disease diagnosis, and the mechanism related to complement activation may also provide additional insight for the treatment of idiopathic membranous nephropathy.

Choi et al. performed LC-MS/MS on urine samples from 16 patients with MCD, MN, and FSGS and HC (4 individuals each) to identify candidate biomarkers for the differential diagnosis of these diseases [Citation47]. After validating several proteins by enzyme-linked immunosorbent assay in a validation set of 51 patients, they reported that SERPINA7 and CD44 were specific to MN. The urinary proteomic studies performed by different teams related to MN along with the relevant contents are summarized in .

Urinary proteomics and minimal change disease and focal segmental glomerulosclerosis

Minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS) are diseases related to podocyte injury and are characterized by excessive proteinuria [Citation74–77]. Of the two, MCD often occurs in children and adolescents [Citation78,Citation79]. Glomeruli do not change drastically in patients with MCD, but extensive fusion of the podocyte foot process can be observed ( and Citation3(C)). Patients with FSGS often present with hematuria and hypertension, while podocytes are also damaged. Segmental hyaline sclerosis can be detected in a number of glomeruli during light microscopy ().

Podocyte injuries in MCD and FSGS are similar, a finding that has prompted several researchers to regard MCD and FSGS as the same disease [Citation80]. Under this assumption, MCD was thought to represent early podocyte injury and FSGS, a late form [Citation81]. Despite their similar characteristics, there are many differences between the two pathologies. Patients with MCD often have better prognosis, whereas patients with FSGS are more likely to develop renal insufficiency. Glucocorticoid therapy is more effective against MCD than against FSGS [Citation1,Citation75,Citation82]. In addition, urinary CD80 levels are increased in patients with MCD, but not in those with FSGS [Citation83]. C1q level is high in FSGS patients, but mannose-binding lectin is rarely observed [Citation84]. The foot process width in patients with FSGS is higher than that seen in patients with MCD, which may suggest more severe podocyte pathological changes [Citation85,Citation86].

Several classic omics studies have identified biomarkers of MCD and FSGS. Williams et al. were the first to undertake molecular analysis at the genomic level in glomeruli and tubulointerstitial regions with histological characteristics and to compare the differences in small RNA maps between patients with FSGS and HCs [Citation87]. They observed a large number of small RNAs that were differentially expressed between the two groups [Citation87]. Nafar et al. analyzed urine samples of six patients with IgAN, eleven individuals with FSGS and eight healthy people using nano-scale liquid chromatography tandem mass spectrometry and reported significant changes in the differential expression of CD59, CD44, IBP7, Robo4 and DPEP1 in people with FSGS [Citation54]. Another study utilizing this technique found differences in the urinary protein landscape of patients with steroid-resistant and steroid-sensitive FSGS [Citation88]; in particular, they observed that apolipoprotein A-1 (APOA1) expression was markedly elevated in the urine samples of the latter patients compared to the former [Citation88].

Pérez et al. compared the urinary proteome between patients with MCD and FSGS using two-dimensional differential gel electrophoresis coupled with MS, and reported that levels of A1AT, transferrin, histatin-3 and 39S ribosomal protein L17 increased, while levels of calretinin decreased, in MCD patients relative to those in FSGS patients [Citation55]. Urinary fibrinogen could also aid in distinguishing between the two diseases [Citation89]. Choi’s study found that CD14, C9, and SERPINA1 were specific to MCD. [Citation47]. Certain candidate biomarkers specific to FSGS include calmodulin-like mucin 26, RNase A family 1, DIS3-like nucleic acid exonuclease 1, and Golgi-associated olfactory signaling regulators [Citation47,Citation56].

These DEPs may be involved in the development of MCD and FSGS and could potentially be used for the noninvasive differential diagnosis and monitoring of treatment of the two pathologies.

The abnormal expression of these proteins in urine may reflect mass cell death and release of cell contents during the separation of podocytes from glomeruli. These results may also suggest specific roles for immunity, inflammation, and apoptosis in the development of FSGS. Cell proliferation, differentiation, and death may be related to the development of MCD [Citation90]. Other potential biomarkers include CD44, MXRA8, and APOA1. The CD44 glycoprotein reflects the activation of parietal epithelial cells that trigger glomerulosclerosis, while MXRA8 is involved in fibrosis and disease progression. Finally, APOA1 indicates that oxidative stress is related to hyperlipidemia, which is one of the pathogenic factors in the development and progression of FSGS. Thus, it has become evident that uroproteomics could be instrumental for the development of noninvasive differential diagnostic methods for MCD and FSGS as well as for the expansion of our understanding of each disease’s pathogenesis.

The collective results of urinary proteomics studies for MCD and FSGS are presented in .

Bioinformatic analysis

In the context of gaining additional insight related to candidate biomarkers of PGD, their predicted protein interactions and their molecular functions, we performed an in-depth bioinformatic analysis of 42 DEPs obtained from the data of key urinary proteomics studies for biomarker screening by means of comparing the urine protein profiles between PGD and HC via MS discussed in this review. The analyzed DEPs were: AFAM, A2GL, A1M, HEMO, APOA1, CO3, APOA4, B2MG, RET4, ZA2G, A2MG, CAH1, CERU, CO4A, FINC, THRB, VTDB, HPT, IBP-7, A1AT, AACT, ANT3, CD44, CLM-9, PCDH1, UTER, DPP4, NHLC3, CO9, IGKC, K1C10, K2C1, K2C5, ADIPO, ICAM1, TIMP1, A1BG, UROM, SCRB2, THBG, CD59, and SH3L3 ().

Table 2. Summary of research on urinary protein biomarkers of glomerular diseases.

To reveal the possible physiological roles of DEPs, GO and KEGG enrichment analyses were carried out using Metascape (https://metascape.org). The result of the GO enrichment analysis is shown in . The molecular functions, biological processes, cellular components, and pathways in which the identified DEPs are predicted to be involved are presented in . Interactions between DEPs were analyzed via the STRING database (https://cn.string-db.org), while Cytoscape (version 3.9.1, https://cytoscape.org) was used to construct and visualize the resulting PPI network (). By utilizing these approaches, we found that DEPs were related to complement activation, immune response, cell adhesion, and cytoskeleton dynamics.

Figure 4. GO and KEGG enrichment analysis. Bar graph of enriched terms across input gene lists (colored according to p-values). (a) Enrichment analysis results of GO molecular functions. (b) Enrichment analysis results of GO biological process. (c) Enrichment analysis results of GO cellular components. (d) Enrichment analysis results of KEGG pathways. Adjusted p-value <0.05 was considered significant.

Figure 4. GO and KEGG enrichment analysis. Bar graph of enriched terms across input gene lists (colored according to p-values). (a) Enrichment analysis results of GO molecular functions. (b) Enrichment analysis results of GO biological process. (c) Enrichment analysis results of GO cellular components. (d) Enrichment analysis results of KEGG pathways. Adjusted p-value <0.05 was considered significant.

Figure 5. PPI network diagram of candidate biomarkers. Excluding four proteins without segment connections, the PPI network diagram includes 37 DEPs. At the outermost part of the concentric circle, the BC value that represents the centrality of a protein increases counterclockwise from SERPINA7, while decreasing clockwise from HP for the middle circle. The PPI enrichment p-value was lower than 1.0e−16 in STRING. The number of edges on a protein is proportional to its the connectivity with other proteins in the network. The BC was selected to represent the importance of differentially expressed proteins. The higher BC values correlate with stronger centrality of these proteins and consequently with more protein cross interactions through the relevant mediations. The size of the circle and the font size reflect the BC value, with bigger circles and fonts corresponding to higher betweenness values. BC: betweenness centrality

Figure 5. PPI network diagram of candidate biomarkers. Excluding four proteins without segment connections, the PPI network diagram includes 37 DEPs. At the outermost part of the concentric circle, the BC value that represents the centrality of a protein increases counterclockwise from SERPINA7, while decreasing clockwise from HP for the middle circle. The PPI enrichment p-value was lower than 1.0e−16 in STRING. The number of edges on a protein is proportional to its the connectivity with other proteins in the network. The BC was selected to represent the importance of differentially expressed proteins. The higher BC values correlate with stronger centrality of these proteins and consequently with more protein cross interactions through the relevant mediations. The size of the circle and the font size reflect the BC value, with bigger circles and fonts corresponding to higher betweenness values. BC: betweenness centrality

The complement system plays a vital role in many diseases and can be activated in three ways. Its activation can also trigger an attack against the body’s own cells, consequently causing autoimmune diseases [Citation91,Citation92]. It is also associated with kidney pathologies, especially MN. Cell adhesion is a dynamic process that plays an essential role in cell proliferation, differentiation, and migration, while perturbations in cell adhesion often lead to severe pathological alterations [Citation93]. Abnormal cell adhesion may cause kidney disease such as IgAN, which is associated with mesangial cell and cell adhesion matrix proliferation [Citation94,Citation95]. Further delineating the relationship between IgAN and cell adhesion may help toward promoting noninvasive methods for diagnosing this disease. Cytoskeleton. the protein fiber grid system in eukaryotic cells, is a dynamic scaffolding network that is essential in maintaining basic cell morphology. Cytoskeleton proteins have been found to be abnormally expressed in patients with MCN and FSGS [Citation96], while podocyte cytoskeleton injury can cause proteinuria [Citation97]. Another study reported that rearrangements of the cytoskeleton could lead to functional abnormality of podocyte foot processes and symptoms of albuminuria, which trigger podocyte-related diseases [Citation98]. Additional studies on cytoskeleton dynamics may help to characterize the pathogenesis of MCD and FSGS in greater detail. Further research in these three areas may lead to new discoveries about PGD.

We believe that the occurrence of PGD may be due to complement attacks on glomerular cells, which disrupt cytoskeleton integrity and further lead to abnormal expression of certain proteins and other macromolecules through a complex intracellular protein interaction network, culminating in the collapse of the glomerular filtration barrier and the development of albuminuria, among other pathological symptoms. However, the specific pathogenesis of PGD warrants more investigation.

Performing in-depth studies of these proteins may in turn lead to advancements toward the pain-free diagnosis of kidney diseases and facilitate research in the design of drugs for the personalized treatment and assessment of prognosis of patients, as well as improve our current knowledge regarding the pathogenesis of PGD.

Conclusions and prospects

In this review, we have discussed studies on urinary proteomics of primary glomerular diseases and we have selected potential biomarkers of PGD to analyze via bioinformatics to obtain a more informed overview of their molecular functions, biological processes, and protein–protein crosstalk interactions, to gain a deeper insight into the pathogenesis of PGD. Based on the results gleaned from our analysis, PGD appears to be related to complement activation, immune response, cell adhesion, and cytoskeleton dynamics.

PGD is an autoimmune disease with unknown pathogenesis, and there is no effective pain-free diagnosis method that can replace renal biopsy in clinical practice. Compared with blood and tissue fluid, urine as the fluid for analysis offers the advantages of noninvasiveness, sustainable collection, and repeated detection. As urine is generated and excreted by the urinary system, it may also be used for detecting urinary system-related abnormalities. In recent years, research in the fields of noninvasive diagnosis, treatment, prognostic evaluation and in-depth understanding of the pathogenesis of PGD has progressed significantly, owing to advances in urinary proteomics. Nevertheless, as results are largely dependent on the choice of proteomic techniques, these methods should be carefully selected according to the purpose of the study. The combination of urine, blood, and kidney tissues, together with high-throughput omics studies, may reveal key findings relevant to the study of PGD. In this regard, urine proteomics and bioinformatics may provide a sound basis for the development of novel biomarkers to diagnose disease and assess prognosis, and to delineate the molecular mechanisms of the disease.

Although there is a number of reviews on urine proteomics of PGD, few articles have integrated the results of different studies to assess their biological impact on the mechanism of PGD through bioinformatic analysis. In this context, protein network analysis may be more informative than protein analysis alone owing to the complexity of the biological processes in which proteins participate. Our present review discusses candidate biomarkers from several studies while also analyzing these data in a descriptive bioinformatic process, thereby proposing an integrated method for future proteomics research.

However, our analysis has some limitations. We assumed that all the proteins analyzed are involved in at least some relevant biological processes during the occurrence and development of PGD and are related to its pathogenesis. Owing to the complex network of protein interactions in an organism, the level of some proteins might be influenced by other proteins, and the change in their levels may not have an impact on PGD.

In conclusion, numerous studies discuss urine proteomics in PGD and several potential biomarkers have been identified. Here, our bioinformatic analysis of candidate biomarkers showed that they tend to be associated with processes such as complement response, cell adhesion, and cytoskeleton dynamics. The network of protein interactions is complex, and our bioinformatic analysis of these potential biomarkers may facilitate research efforts for the further elucidation of the pathogenesis of PGD. Our summary of urinary proteomics studies of PGD is helpful in promoting diagnostic alternatives, in the form of biomarkers, for PGD. We believe that urinary proteomics combined with bioinformatic approaches will not only help in the noninvasive diagnosis of PGD but also provide a framework for precision therapy and a deeper understanding of the pathogenesis for this disease.

Abbreviations
CKD=

chronic kidney disease

DEPs=

differentially expressed proteins

ESRD=

end-stage renal disease

FSGS=

focal segmental glomerulosclerosis

GD=

glomerular disease

GO=

Gene Ontology

HCs=

healthy controls

IgAN=

IgA nephropathy

KEGG=

Kyoto Encyclopedia of Genes and Genomes

MCD=

minimal change disease

MN=

membranous nephropathy

MS=

mass spectrometry

PGD=

primary glomerular disease

PPI=

protein–protein interaction

Acknowledgments

We thank Figdraw (https://www.figdraw.com/) and Untitled (https://app.biorender.com/) for providing us with a drawing platform.

Disclosure statement

The research was conducted in the absence of any commercial or financial relationships that might be construed as potential conflicts of interest. No potential conflict of interest was reported by the author(s).

Additional information

Funding

Our study was supported by “345 Talent Project” of Shengjing Hospital of China Medical University [2022146], Liaoning Province Central Government’s special project to guide local scientific and technological development [2019JH6/1], and National Key Research and Development Program of China [2018YFE0207300].

References

  • Chebotareva N, Vinogradov A, McDonnell V, et al. Urinary protein and peptide markers in chronic kidney disease. Int J Mol Sci. 2021;22(22):12123.
  • Hill NR, Fatoba ST, Oke JL, et al. Global prevalence of chronic kidney disease – a systematic review and meta-analysis. PLOS One. 2016;11(7):e0158765.
  • Levey AS, Eckardt KU, Tsukamoto Y, et al. Definition and classification of chronic kidney disease: a position statement from kidney disease: improving global outcomes (KDIGO). Kidney Int. 2005;67(6):2089–2100.
  • Pani A. Standard immunosuppressive therapy of immune-mediated glomerular diseases. Autoimmun Rev. 2013;12(8):848–853.
  • Navas-Carrillo D, Rodriguez JM, Montoro-García S, et al. High-resolution proteomics and metabolomics in thyroid cancer: deciphering novel biomarkers. Crit Rev Clin Lab Sci. 2017;54(7–8):446–457.
  • Paul P, Antonydhason V, Gopal J, et al. Bioinformatics for renal and urinary proteomics: call for aggrandization. Int J Mol Sci. 2020;21(3):961.
  • Wen Y, Parikh CR. Current concepts and advances in biomarkers of acute kidney injury. Crit Rev Clin Lab Sci. 2021;58(5):354–368.
  • Wu J, Chen YD, Gu W. Urinary proteomics as a novel tool for biomarker discovery in kidney diseases. J Zhejiang Univ Sci B. 2010;11(4):227–237.
  • Chu L, Fu G, Meng Q, et al. Identification of urinary biomarkers for type 2 diabetes using bead-based proteomic approach. Diabetes Res Clin Pract. 2013;101(2):187–193.
  • Fang X, Lu M, Xia Z, et al. Use of liquid chromatography-tandem mass spectrometry to perform urinary proteomic analysis of children with IgA nephropathy and Henoch-Schönlein purpura nephritis. J Proteomics. 2021;230:103979.
  • Wilkins MR, Sanchez JC, Gooley AA, et al. Progress with proteome projects: why all proteins expressed by a genome should be identified and how to do it. Biotechnol Genet Eng Rev. 1996;13:19–50.
  • Marshall T, Williams K. Two-dimensional electrophoresis of human urinary proteins following concentration by dye precipitation. Electrophoresis. 1996;17(7):1265–1272.
  • Heine G, Raida M, Forssmann WG. Mapping of peptides and protein fragments in human urine using liquid chromatography-mass spectrometry. J Chromatogr A. 1997;776(1):117–124.
  • Thongboonkerd V, McLeish KR, Arthur JM, et al. Proteomic analysis of normal human urinary proteins isolated by acetone precipitation or ultracentrifugation. Kidney Int. 2002;62(4):1461–1469.
  • Adachi J, Kumar C, Zhang Y, et al. The human urinary proteome contains more than 1500 proteins, including a large proportion of membrane proteins. Genome Biol. 2006;7(9):R80.
  • Zhang F, Li X, Ni Y, et al. Preliminary study of the urinary proteome in Li and Han ethnic individuals from Hainan. Sci China Life Sci. 2020;63(1):125–137.
  • Bellei E, Rossi E, Lucchi L, et al. Proteomic analysis of early urinary biomarkers of renal changes in type 2 diabetic patients. Proteomics Clin Appl. 2008;2(4):478–491.
  • Albert C, Albert A, Kube J, et al. Urinary biomarkers may provide prognostic information for subclinical acute kidney injury after cardiac surgery. J Thorac Cardiovasc Surg. 2018;155(6):2441–2452.e13.
  • Chu Y, Lai YH, Lee MC, et al. Calsyntenin-1, clusterin and neutrophil gelatinase-associated lipocalin are candidate serological biomarkers for lung adenocarcinoma. Oncotarget. 2017;8(64):107964–107976.
  • Marimuthu A, O'Meally RN, Chaerkady R, et al. A comprehensive map of the human urinary proteome. J Proteome Res. 2011;10(6):2734–2743.
  • Papadopoulou-Marketou N, Kanaka-Gantenbein C, Marketos N, et al. Biomarkers of diabetic nephropathy: a 2017 update. Crit Rev Clin Lab Sci. 2017;54(5):326–342.
  • Jarrick S, Lundberg S, Welander A, et al. Mortality in IgA nephropathy: a nationwide population-based cohort study. J Am Soc Nephrol. 2019;30(5):866–876.
  • Reid S, Cawthon PM, Craig JC, et al. Non-immunosuppressive treatment for IgA nephropathy. Cochrane Database Syst Rev. 2011;16(3):Cd003962.
  • McGrogan A, Franssen CF, de Vries CS. The incidence of primary glomerulonephritis worldwide: a systematic review of the literature. Nephrol Dial Transplant. 2011;26(2):414–430.
  • Zaza G, Bernich P, Lupo A, ‘Triveneto’ Register of Renal Biopsies (TVRRB). Incidence of primary glomerulonephritis in a large North-Eastern Italian area: a 13-year renal biopsy study. Nephrol Dial Transplant. 2013;28(2):367–372.
  • Cambier A, Gleeson PJ, Abbad L, et al. Soluble CD89 is a critical factor for mesangial proliferation in childhood IgA nephropathy. Kidney Int. 2022;101(2):274–287.
  • Dotz V, Visconti A, Lomax-Browne HJ, et al. O- and N-glycosylation of serum immunoglobulin A is associated with IgA nephropathy and glomerular function. J Am Soc Nephrol. 2021;32(10):2455–2465.
  • Tortajada A, Gutierrez E, Pickering MC, et al. The role of complement in IgA nephropathy. Mol Immunol. 2019;114:123–132.
  • Lafayette RA, Kelepouris E. Immunoglobulin a nephropathy: advances in understanding of pathogenesis and treatment. Am J Nephrol. 2018;47(Suppl 1):43–52.
  • Pattrapornpisut P, Avila-Casado C, Reich HN. IgA nephropathy: core curriculum 2021. Am J Kidney Dis. 2021;78(3):429–441.
  • Zambrano S, He L, Kano T, et al. Molecular insights into the early stage of glomerular injury in IgA nephropathy using single-cell RNA sequencing. Kidney Int. 2022;101(4):752–765.
  • Li H, Chen Z, Chen W, et al. MicroRNA-23b-3p deletion induces an IgA nephropathy-like disease associated with dysregulated mucosal IgA synthesis. J Am Soc Nephrol. 2021;32(10):2561–2578.
  • Barratt J, Feehally J. IgA nephropathy. J Am Soc Nephrol. 2005;16(7):2088–2097.
  • Rops A, Jansen E, van der Schaaf A, et al. Interleukin-6 is essential for glomerular immunoglobulin a deposition and the development of renal pathology in Cd37-deficient mice. Kidney Int. 2018;93(6):1356–1366.
  • van Delft MAM, Huizinga TWJ. An overview of autoantibodies in rheumatoid arthritis. J Autoimmun. 2020;110:102392.
  • Haubitz M, Wittke S, Weissinger EM, et al. Urine protein patterns can serve as diagnostic tools in patients with IgA nephropathy. Kidney Int. 2005;67(6):2313–2320.
  • Julian BA, Wittke S, Novak J, et al. Electrophoretic methods for analysis of urinary polypeptides in IgA-associated renal diseases. Electrophoresis. 2007;28(23):4469–4483.
  • He Q, Shao L, Yu J, et al. Urinary proteome analysis by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry with magnetic beads for identifying the pathologic presentation of clinical early IgA nephropathy. J Biomed Nanotechnol. 2012;8(1):133–139.
  • Kalantari S, Rutishauser D, Samavat S, et al. Urinary prognostic biomarkers and classification of IgA nephropathy by high resolution mass spectrometry coupled with liquid chromatography. PLOS One. 2013;8(12):e80830.
  • Rudnicki M, Siwy J, Wendt R, PERSTIGAN Working Group, et al. Urine proteomics for prediction of disease progression in patients with IgA nephropathy. Nephrol Dial Transplant. 2021;37(1):42–52.
  • Guo Z, Wang Z, Lu C, et al. Analysis of the differential urinary protein profile in IgA nephropathy patients of uygur ethnicity. BMC Nephrol. 2018;19(1):358.
  • Mucha K, Bakun M, Jaźwiec R, et al. Complement components, proteolysis‑related, and cell communication‑related proteins detected in urine proteomics are associated with IgA nephropathy. Pol Arch Med Wewn. 2014;124(7-8):380–386.
  • Samavat S, Kalantari S, Nafar M, et al. Diagnostic urinary proteome profile for immunoglobulin a nephropathy. Iran J Kidney Dis. 2015;9(3):239–248.
  • Ning X, Yin Z, Li Z, et al. Comparative proteomic analysis of urine and laser microdissected glomeruli in IgA nephropathy. Clin Exp Pharmacol Physiol. 2017;44(5):576–585.
  • Navarro-Muñoz M, Ibernon M, Bonet J, et al. Uromodulin and α(1)-antitrypsin urinary peptide analysis to differentiate glomerular kidney diseases. Kidney Blood Press Res. 2012;35(5):314–325.
  • Rood IM, Merchant ML, Wilkey DW, et al. Increased expression of lysosome membrane protein 2 in glomeruli of patients with idiopathic membranous nephropathy. Proteomics. 2015;15(21):3722–3730.
  • Choi YW, Kim YG, Song MY, et al. Potential urine proteomics biomarkers for primary nephrotic syndrome. Clin Proteomics. 2017;14:18.
  • Pang L, Li Q, Li Y, et al. Urine proteomics of primary membranous nephropathy using nanoscale liquid chromatography tandem mass spectrometry analysis. Clin Proteomics. 2018;15:5.
  • Zhang MF, Huang J, Zhang YM, et al. Complement activation products in the circulation and urine of primary membranous nephropathy. BMC Nephrol. 2019;20(1):313.
  • Lin B, Liu J, Zhang Y, et al. Urinary peptidomics reveals proteases involved in idiopathic membranous nephropathy. BMC Genomics. 2021;22(1):852.
  • Araumi A, Osaki T, Ichikawa K, et al. Urinary and plasma proteomics to discover biomarkers for diagnosing between diabetic nephropathy and minimal change nephrotic syndrome or membranous nephropathy. Biochem Biophys Rep. 2021;27:101102.
  • Pérez V, Ibernón M, López D, et al. Urinary peptide profiling to differentiate between minimal change disease and focal segmental glomerulosclerosis. PLOS One. 2014;9(1):e87731.
  • Kalantari S, Nafar M, Samavat S, et al. Urinary prognostic biomarkers in patients with focal segmental glomerulosclerosis. Nephrourol Mon. 2014;6(2):e16806.
  • Nafar M, Kalantari S, Samavat S, et al. The novel diagnostic biomarkers for focal segmental glomerulosclerosis. Int J Nephrol. 2014;2014:574261.
  • Pérez V, López D, Boixadera E, et al. Comparative differential proteomic analysis of minimal change disease and focal segmental glomerulosclerosis. BMC Nephrol. 2017;18(1):49.
  • Siwy J, Zürbig P, Argiles A, et al. Noninvasive diagnosis of chronic kidney diseases using urinary proteome analysis. Nephrol Dial Transplant. 2017;32(12):2079–2089.
  • Wang Y, Zheng C, Wang X, et al. Proteomic profile‑based screening of potential protein biomarkers in the urine of patients with nephrotic syndrome. Mol Med Rep. 2017;16(5):6276–6284.
  • Couser WG. Primary membranous nephropathy. Clin J Am Soc Nephrol. 2017;12(6):983–997.
  • Ronco P, Debiec H. Pathophysiological advances in membranous nephropathy: time for a shift in patient’s care. Lancet. 2015;385(9981):1983–1992.
  • Safar-Boueri L, Piya A, Beck LH, Jr., et al. Membranous nephropathy: diagnosis, treatment, and monitoring in the post-PLA2R era. Pediatr Nephrol. 2021;36(1):19–30.
  • Liu W, Gao C, Liu Z, et al. Idiopathic membranous nephropathy: glomerular pathological pattern caused by extrarenal immunity activity. Front Immunol. 2020;11:1846.
  • Liu W, Gao C, Dai H, et al. Immunological pathogenesis of membranous nephropathy: focus on PLA2R1 and its role. Front Immunol. 2019;10:1809.
  • Gu Y, Xu H, Tang D. Mechanisms of primary membranous nephropathy. Biomolecules. 2021;11(4):513.
  • Sethi S. New 'antigens’ in membranous nephropathy. J Am Soc Nephrol. 2021;32(2):268–278.
  • Lv M, Li W, Tao R, et al. Spatial-spectral density peaks-based discriminant analysis for membranous nephropathy classification using microscopic hyperspectral images. IEEE J Biomed Health Inform. 2021;25(8):3041–3051.
  • Beck LH, Jr., Bonegio RG, Lambeau G, et al. M-type phospholipase A2 receptor as target antigen in idiopathic membranous nephropathy. N Engl J Med. 2009;361(1):11–21.
  • Coenen MJ, Hofstra JM, Debiec H, et al. Phospholipase A2 receptor (PLA2R1) sequence variants in idiopathic membranous nephropathy. J Am Soc Nephrol. 2013;24(4):677–683.
  • Bobart SA, De Vriese AS, Pawar AS, et al. Noninvasive diagnosis of primary membranous nephropathy using phospholipase A2 receptor antibodies. Kidney Int. 2019;95(2):429–438.
  • Qin W, Beck LH, Jr., Zeng C, et al. Anti-phospholipase A2 receptor antibody in membranous nephropathy. J Am Soc Nephrol. 2011;22(6):1137–1143.
  • Ngai HH, Sit WH, Jiang PP, et al. Markedly increased urinary preprohaptoglobin and haptoglobin in passive Heymann nephritis: a differential proteomics approach. J Proteome Res. 2007;6(8):3313–3320.
  • Aregger F, Uehlinger DE, Witowski J, et al. Identification of IGFBP-7 by urinary proteomics as a novel prognostic marker in early acute kidney injury. Kidney Int. 2014;85(4):909–919.
  • Yang Y, Wei J, Huang X, et al. iTRAQ-based proteomics of chronic renal failure rats after FuShengong decoction treatment reveals haptoglobin and alpha-1-antitrypsin as potential biomarkers. Evid Based Complement Alternat Med. 2017;2017:1480514.
  • Smith A, L'Imperio V, De Sio G, et al. α-1-Antitrypsin detected by MALDI imaging in the study of glomerulonephritis: its relevance in chronic kidney disease progression. Proteomics. 2016;16(11–12):1759–1766.
  • Vivarelli M, Massella L, Ruggiero B, et al. Minimal change disease. Clin J Am Soc Nephrol. 2017;12(2):332–345.
  • Hogan J, Radhakrishnan J. The treatment of minimal change disease in adults. J Am Soc Nephrol. 2013;24(5):702–711.
  • Meyrier AY. Treatment of focal segmental glomerulosclerosis with immunophilin modulation: when did we stop thinking about pathogenesis? Kidney Int. 2009;76(5):487–491.
  • De Vriese AS, Wetzels JF, Glassock RJ, et al. Therapeutic trials in adult FSGS: lessons learned and the road forward. Nat Rev Nephrol. 2021;17(9):619–630.
  • Meyrier A, Niaudet P. Acute kidney injury complicating nephrotic syndrome of minimal change disease. Kidney Int. 2018;94(5):861–869.
  • Chugh SS, Clement LC, Macé C. New insights into human minimal change disease: lessons from animal models. Am J Kidney Dis. 2012;59(2):284–292.
  • Maas RJ, Deegens JK, Smeets B, et al. Minimal change disease and idiopathic FSGS: manifestations of the same disease. Nat Rev Nephrol. 2016;12(12):768–776.
  • Ahn W, Bomback AS. Approach to diagnosis and management of primary glomerular diseases due to podocytopathies in adults: core curriculum 2020. Am J Kidney Dis. 2020;75(6):955–964.
  • Moura LR, Franco MF, Kirsztajn GM. Minimal change disease and focal segmental glomerulosclerosis in adults: response to steroids and risk of renal failure. J Bras Nefrol. 2015;37(4):475–480.
  • Garin EH, Mu W, Arthur JM, et al. Urinary CD80 is elevated in minimal change disease but not in focal segmental glomerulosclerosis. Kidney Int. 2010;78(3):296–302.
  • van de Lest NA, Zandbergen M, Wolterbeek R, et al. Glomerular C4d deposition can precede the development of focal segmental glomerulosclerosis. Kidney Int. 2019;96(3):738–749.
  • Deegens JK, Dijkman HB, Borm GF, et al. Podocyte foot process effacement as a diagnostic tool in focal segmental glomerulosclerosis. Kidney Int. 2008;74(12):1568–1576.
  • Taneda S, Honda K, Ohno M, et al. Podocyte and endothelial injury in focal segmental glomerulosclerosis: an ultrastructural analysis. Virchows Arch. 2015;467(4):449–458.
  • Williams AM, Jensen DM, Pan X, et al. Histologically resolved small RNA maps in primary focal segmental glomerulosclerosis indicate progressive changes within glomerular and tubulointerstitial regions. Kidney Int. 2022;101(4):766–778.
  • Kalantari S, Nafar M, Rutishauser D, et al. Predictive urinary biomarkers for steroid-resistant and steroid-sensitive focal segmental glomerulosclerosis using high resolution mass spectrometry and multivariate statistical analysis. BMC Nephrol. 2014;15:141.
  • Wang Y, Zheng C, Xu F, et al. Urinary fibrinogen and renal tubulointerstitial fibrinogen deposition: discriminating between primary FSGS and minimal change disease. Biochem Biophys Res Commun. 2016;478(3):1147–1152.
  • Schwaller B. Calretinin: from a "simple" Ca(2+) buffer to a multifunctional protein implicated in many biological processes. Front Neuroanat. 2014;8:3.
  • Biryukov S, Stoute JA. Complement activation in malaria: friend or foe? Trends Mol Med. 2014;20(5):293–301.
  • Niculescu F, Rus H. The role of complement activation in atherosclerosis. Immunol Res. 2004;30(1):73–80.
  • Bachmann M, Kukkurainen S, Hytönen VP, et al. Cell adhesion by integrins. Physiol Rev. 2019;99(4):1655–1699.
  • Tada H, Nishioka T, Takase A, et al. Porphyromonas gingivalis induces the production of interleukin-31 by human mast cells, resulting in dysfunction of the gingival epithelial barrier. Cell Microbiol. 2019;21(3):e12972.
  • Kochi S, Yamashiro K, Hongo S, et al. Aggregatibacter actinomycetemcomitans regulates the expression of integrins and reduces cell adhesion via integrin α5 in human gingival epithelial cells. Mol Cell Biochem. 2017;436(1–2):39–48.
  • Shi SF, Wang SX, Zhang YK, et al. Ultrastructural features and expression of cytoskeleton proteins of podocyte from patients with minimal change disease and focal segmental glomerulosclerosis. Ren Fail. 2008;30(5):477–483.
  • Ye Q, Lan B, Liu H, et al. A critical role of the podocyte cytoskeleton in the pathogenesis of glomerular proteinuria and autoimmune podocytopathies. Acta Physiol (Oxf). 2022;235(4):e13850.
  • Gao W, Liu Y, Fan L, et al. Role of γ-adducin in actin cytoskeleton rearrangements in podocyte pathophysiology. Am J Physiol Renal Physiol. 2021;320(1):F97–F113.