[go: up one dir, main page]

Skip to main content

Future embracing: exosomes driving a revolutionary approach to the diagnosis and treatment of idiopathic membranous nephropathy

Abstract

Membranous nephropathy (MN) is a leading cause of nephrotic syndrome in adults and is associated with high rates of end-stage renal disease. Early detection and precise interventions are crucial for improving patient prognosis and quality of life. However, the current diagnosis primarily relies on renal biopsies and traditional biomarkers, which have limitations. Additionally, targeted therapeutic strategies are lacking. Exosomes, small vesicles that facilitate intercellular communication, have emerged as potential noninvasive diagnostic markers due to their stability, diverse cargo, and rapid detectability. They also hold promise as carriers for gene and drug delivery, presenting innovative opportunities in renal disease prognosis and treatment. However, research on exosomes in the context of idiopathic membranous nephropathy (IMN) remains limited, with a focus on exploring urinary exosomes as IMN markers. In this review, we summarize the current status of MN diagnosis and treatment, highlight the fundamental characteristics of exosomes, and discuss recent advancements in their application to IMN diagnosis and therapy. We provide insights into the clinical prospects of exosomes in IMN and acknowledge potential challenges. This article aims to offer forward-looking insights into the future of exosome-mediated IMN diagnosis and treatment, indicating a revolutionary transformation in this field.

Graphical Abstract

Introduction

Idiopathic membranous nephropathy (IMN) is an autoimmune glomerular podocytosis syndrome [1]. Its clinical features mainly include proteinuria or nephrotic syndrome with insidious onset, which is a common cause of adult nephrotic syndrome in China. In recent years, both domestic and international studies have demonstrated a progressively increasing incidence of IMN [2,3,4,5,6,7]. In Caucasians, IMN accounts for approximately 30–40% of primary nephrotic syndrome cases [8]. In China, the prevalence of IMN has increased significantly in recent renal biopsy cases. According to data from a retrospective study covering 10 years and 6049 cases of renal pathology from Peking University First Hospital in 2015 [9], the incidence of IMN increased from 16.8% in 2003–2007 to 29.35% in 2008–2012. Moreover, another study led by academician Hou Fanfan in 2016, which included data from nearly 70,000 patient renal biopsies across 938 hospitals in 282 cities throughout China, revealed that while the incidences of other glomerular diseases remained stable, the incidence of MN doubled from 12.2% in 2004 to 24.9% in 2014, positioning it as the second most common cause of primary glomerulopathy [5]. In the same year, after calibrating data on kidney disease types across 11 years in the Chinese population, Xu and colleagues reported an annual increase of 13% in MN patients, demonstrating a trend toward surpassing IgA nephropathy [5]. Research has suggested that in the northeastern region of China, the incidence of IMN has exceeded that of IgA nephropathy, suggesting that IMN is the leading cause of primary glomerulopathies [10, 11] and poses a serious threat to human health.

The natural course of MN exhibits significant variability. Approximately 30% of patients experience spontaneous remission, while approximately 70% manifest persistent proteinuria. Within 5–20 years, 40–60% of patients progress to end-stage renal disease (ESRD) [3], making it a significant contributor to ESRD. Due to the high incidence and recurrence rates of this disease, early detection and preventive treatment are essential for improving patient prognosis and quality of life. Currently, the diagnosis of IMN still relies on invasive renal biopsies, which carry potential risks of complications such as bleeding and infections and are not suitable for repeated evaluations of renal changes. However, traditional IMN biomarkers, such as the serum creatinine concentration, estimated glomerular filtration rate (eGFR), serum albumin (ALB) concentration, and urine protein concentration, all have limitations in terms of sensitivity and specificity [12]. Moreover, novel biomarkers, including autoantibodies against intrinsic podocyte antigens such as anti-PLA2R and anti-THSD7A, exhibit increased sensitivity and specificity. However, they still do not encompass all IMN patients. For instance, there is a possibility of underdiagnosing anti-PLA2R- and/or anti-THSD7A-negative IMN patients. Therefore, in-depth research into the pathogenesis of IMN and the exploration of new biomarkers are pressing challenges in the current field of kidney disease research. Currently, specific drugs capable of halting or reversing the progression of MN are lacking. Although the International Kidney Disease Guidelines (KDIGO) have issued guidelines for the treatment of MN, recommending medications that provide varying degrees of therapeutic effects for MN, corresponding biologics and low-dose steroids combined with immunosuppressive regimens exhibit unstable efficacy. These approaches can cause potential immune-related side effects, increased rates of relapse, and increased economic burdens [13,14,15], preventing patients from fully meeting the treatment needs of MN patients. As a result, there is an urgent need to identify noninvasive diagnostic markers and specific therapeutic targets for IMN.

Moreover, related studies have reported that almost all intrinsic renal cells, such as endothelial cells, podocytes, and tubular epithelial cells, can secrete exosomes and mediate crosstalk between different types of cells in the kidney [16, 17]. There is a characteristic change in exosome content in renal diseases such as acute kidney injury [18], IgA nephropathy [19], diabetic nephropathy [20], renal tubular acidosis [21], and polycystic kidney disease [22]. In addition, exosomal miRNAs are more stable than circulating miRNAs, and they are protected from degradation by rRNA enzymes [23]. Based on these features, exosomes have great potential as biomarkers and therapeutic agents for the early diagnosis of IMN. Currently, exosomes have been widely studied as biomarkers for the diagnosis of renal diseases and as therapeutic means for renal diseases, but there is a relative lack of application of exosomes in the diagnosis and treatment of idiopathic membranous nephropathy (IMN). In this paper, we hope to systematically review the progress in the use of exosomes in the diagnosis and treatment of IMN and provide a reference for the future diagnosis and treatment of IMN. This review first describes the generation and origin of exosomes, their composition and contents, and their biological properties and functions and then explores the application of exosomes in IMN diagnosis, pathogenesis and therapy. Finally, we look ahead to current limitations and challenges, as well as potential directions for future research and clinical translation of exosomes.

Overview of exosomes

Generation and origins of exosomes

The biogenesis of exosomes begins with the maturation of early endosomes to late endosomes or multivesicular bodies (MVBs) containing intraluminal vesicles (ILVs). Endosomes are the focal point of the endocytosis pathway and determine whether internalized proteins and lipids are degraded or recycled [24]. Endosomes are categorized as early endosomes, late endosomes, or recycling endosomes. The biogenesis of exosomes involves double invagination of the plasma membrane, with the initial inward bending of the plasma membrane forming a cup-shaped structure that includes cell surface proteins and extracellular components such as soluble proteins, lipids, metabolites, small molecules, and ions. These components can be internalized through endocytosis and membrane invagination along with cell surface proteins. The de novo formation of early-sorting endosomes (ESEs) is initiated. Sometimes, it may be directly fused with pre-ESE, which is preformed from components of the endoplasmic reticulum (ER), the trans-Golgi network (TGN), and the mitochondrion. Early-sorting endosomes (marked by Rab5) mature through acidification and substance exchange to become late-sorting endosomes (LSEs) (marked by Rab7). Ultimately, the second internalization of LSEs results in the formation of MVBs. These MVBs contain ILVs with diameters ranging from 40 to 150 nm, which are formed by inward budding of the MVB membrane. ILVs can be directly fused with lysosomes or autophagic lysosomes to undergo degradation, and the degradation products can be recycled by the cell. The other pathway is plasma membrane fusion, in which the contents are released into the extracellular space in the form of exosomes [25, 26]. This process is referred to as exosome biogenesis and distinguishes it from other forms of vesicle release, such as budding from the plasma membrane, apoptotic body formation, or membrane rupture (Fig. 1). There are two distinct mechanisms involved in the formation of ILVs: ESCRT-independent and ESCRT-dependent mechanisms required for cargo sorting into endosomes. ESCRT consists of four complexes and auxiliary proteins: ESCRT-0, ESCRT-I, ESCRT-II, and ESCRT-III. These complexes collaborate in an orderly manner, recognizing ubiquitinated proteins on the endosomal membrane and inducing inward budding to form ILVs. Another mechanism, which is ESCRT-independent, relies on lipid raft microdomains enriched in sphingomyelinase and microdomains enriched in tetraspanins [27,28,29].

Fig. 1
figure 1

Exosome production and contents. A The process of exosome generation. B The contents of exosomes

Composition and contents of exosomes

Exosomes are lipid bilayer cup-shaped vesicles with sizes ranging from 30 to 200 nm [30]. Embedded within their phospholipid bilayer membrane are numerous proteins and lipids believed to have evolved from parent cells [31]. The lipid composition of these exosome bilayers includes phosphatidylcholines, phosphatidylethanolamine, phosphatidylinositol, phosphatidylserine, and sphingomyelin [32]. These components are more balanced in the exosome when their proportions are 26:26:19:19:20 and 43:23:12:12:9, respectively [33]. Elevated levels of sphingomyelin and phosphatidylinositol ensure their stability in biological fluids with varying pH levels, guarding against lipid or protein hydrolysis that might occur during systemic circulation [34]. In addition, exosome membranes are enriched with lipid rafts of various proteins, such as tyrosine kinase Src and glycosylphosphatidylinositol-owned proteins [35]. The proteins contained in exosomes can be categorized into two groups: nonspecific and specific. Among them, nonspecific proteins are widely present in all types of exosomes, regardless of their cell of origin. These proteins include tetraspanins (such as CD9, CD63, CD81, and CD82) serving as exosomal surface markers, proteins involved in exosome trafficking and binding to target cells (such as GTPases, annexins, and flotillin) [36], proteins participating in the biogenesis of MVBs like TSG101, Alix, ESCRT complexes, heat shock proteins (Hsc70, Hsp90), GTPases, and membrane-associated proteins [37], as well as cytoskeletal proteins (such as heterotrimeric G proteins, 14-3-3, syntenin). On the other hand, specific proteins within exosomes encompass tissue-specific proteins, such as the major histocompatibility complex class II (MHC-II), present on the surface of nearly all dendritic cells (DC) and B lymphocytes [38], and proteins unique to specific cell types. These specific proteins do not exist independently from nonspecific proteins. For instance, the shell of tetraspanin proteins is composed of various cell-specific transmembrane proteins, including α and β integrin chains (such as αM found on T cells and dendritic cells, α4β1 present on reticular cells, and β2 located on the apex of dendritic cells), cholesterol, and flotillin as lipid raft components. Additionally, certain members of the immunoglobulin family (such as A33 antigen on intestinal cells, intercellular adhesion molecule 1 (ICAM-1)/CD54 on B cells, and P-selectin on platelets) as well as cell surface peptidases (such as aminopeptidase N/CD13 on M cells and dipeptidyl peptidase IV/CD26 on intestinal cells) are also included [31]. Exosomes also encompass molecules involved in signaling pathways, such as β-catenin, ADP-ribosylation factor 1 (ARF1), epidermal growth factor receptor (EGFR), mucin 1 (MUC1), phosphoinositide 3-kinase (PI3K), G-proteins, cytoskeletal proteins, and cell division control protein 42 (CDC42) [35]. Simultaneously, exosomes harbor a diverse array of RNA types, including mRNA and noncoding RNAs such as miRNA, lncRNA, and tRNA. These RNAs exhibit functional roles, capable of influencing the transcriptome of recipient cells [39,40,41,42]. Among them, miRNA represents the most abundant RNA species in exosomes [43, 44].

Biological characteristics and functions of exosomes

The functions of exosomes are contingent upon their originating cells [45]. They participate in immune responses, inflammation, angiogenesis, coagulation, intercellular communication, as well as the dissemination of pathogens such as prions and viruses [17]. The attributes of exosomes underscore their significant role in disease diagnosis, noncellular therapies, and the delivery of proteins, genes, and chemical substances [46,47,48]. First, the composition of exosomes varies due to the cell type, stimuli, stress, transformation, and differentiation functions of the source cells, rendering their detection and characterization in bodily fluids promising as diagnostic markers and prognostic indicators of diseases [49,50,51,52,53,54,55]; Second, exosomes are of natural origin and are inherently highly biocompatible and less immunogenic, and can be used as endogenous carriers [44]. At the same time, because of their nanoscale [33], exosomes are capable of crossing biological barriers, evading the mononuclear phagocytic system (MPS), and other advantages, and are easy to deliver drugs to the target organs [44, 56]. In addition, exosomes can be isolated from a variety of body fluids and can be stored at −80 °C for long periods of time and have a relatively long lifespan in vivo [17, 45, 57]. Finally, exosomes contain bioactive substances and proteins, and their lipid bilayer structure protects them from enzymatic degradation [58, 59]. Based on the above characteristics such as endogenous, biocompatible and multifunctional properties, exosomes are expected to be a new means of drug delivery system, immunotherapy, and precision therapy.

The application of exosomes in the diagnosis and treatment of IMN

Role of exosomes in IMN diagnosis

The optimization of treatment for kidney diseases relies on the availability of diagnostic and prognostic biomarkers. Early diagnosis and treatment of IMN present significant challenges in the field of kidney disease [60]. Currently, diagnosing IMN requires the exclusion of secondary factors such as hepatitis B infection, systemic lupus erythematosus, cancer, or drug-related effects that can cause IMN [1]. Although renal biopsy remains the gold standard for diagnosing IMN [61], it poses the risk of potential severe postoperative complications such as bleeding and infection, and it is inconvenient for repetitive procedures to assess and evaluate kidney damage. Moreover, inappropriate sampling or lack of representativeness can affect the credibility of renal biopsy results [62]. The anti-PLA2R antibody is currently the best noninvasive biomarker, yet its positivity rate ranges from 50 to 72% in different ethnicities within the IMN population [62, 63], leaving some patients unsatisfied. Additionally, traditional biomarkers such as the serum creatinine concentration, eGFR, and urinary protein concentration exhibit low sensitivity, particularly in the early stages of kidney damage [12, 64]. Therefore, the search for novel noninvasive diagnostic biomarkers capable of identifying IMN has emerged as a vibrant area of research within the current landscape of glomerular disease studies [65, 66].

Exosomes, as potential biomarkers, were recognized by the Massachusetts Institute of Technology Technology Review as one of the "Top 10 Breakthroughs of 2015" [60, 64]. Unlike renal biopsies, exosomal biomarkers are not only exempt from the limitations of potentially nonrepresentative sampling but also sidestep the traumatic nature and potential complications associated with tissue biopsies [64]. The encapsulation of exosomes shields their cargo from RNA enzymes and repeated freeze‒thaw cycles in both intracellular and extracellular environments, ensuring the integrity and stability of the biomolecular information they carry [21, 67]. Furthermore, exosomes express origin-specific markers, allowing for the monitoring of changes in specific cellular compartments within tissues, thereby enabling the tracking of lesion locations [64]. For instance, the presence of podocyte proteins like podocin [68], nephrin, and podocalyxin [69, 70] determines the increased in podocyte or endothelial-origin exosomes, potentially implying podocyte damage. Analysis of urinary exosomes may be useful in the diagnostic classification of other disease processes involving the renal tubules, such as polycystic kidneys [71], lysosomal storage diseases like Niemann-Pick disease and cystinosis, and transporter mutations like Gitelman and Bartter syndromes. Similarly, elevated levels of endothelial proteins in urinary exosomes, such as PL-VAP, CD31, and CD144 [72], indicate endothelial damage. Of particular interest, urinary exosomal miR-200b is associated with renal fibrosis in chronic kidney disease (CKD) only when measured in CD13 + exosomes (those not derived from proximal tubules) [73]. This suggests that exosomal biomarkers associated with this cellular subset might possess unique advantages. Previous research has also shown characteristic changes in exosome content in various kidney diseases, such as acute kidney injury [18], IgA nephropathy [19], diabetic nephropathy (DN) [20], renal tubular acidosis [21], and polycystic kidney disease [22]. This indicates the potential and substantial promise of exosomes as biomarkers in the field of kidney diseases.

Urinary exosomes in diagnosing IMN

Normal urine contains exosomes from each type of epithelial cell in the urinary space, including podocytes, endothelial cells, mesangial cells of the glomerulus, tubular cells of the nephron, and transitional epithelial cells of the urinary excretion system, and isolation of urinary exosomes allows identification of their sources [74, 75]. Thus, through urine collection and analysis, changes in the function of the entire renal, prostate, and bladder urinary systems can be monitored [67, 76,77,78]. This finding aligns with the findings of Miranda et al., who reported that exosomes isolated from human urine exhibited a comprehensive RNA profile similar to that of the kidneys [21]. Additionally, urinary exosomes offer advantages, such as large volume, rich content, and noninvasive collection [79,80,81]. Compared to the original urine metabolic pattern, the exosome metabolic pattern holds greater potential for MN diagnostics [79] and demonstrates increased stability [82,83,84]. Currently, urinary exosomes have been established as biomarkers for numerous kidney disorders, including CKD [85], DN [86, 87], autosomal dominant polycystic kidney disease [88], renal cell carcinoma [89], and renal fibrosis [73, 90]. These exosomal markers in urine can be detected in quantities as low as 0.5 mL, suggesting high sensitivity [91]. With further research, urinary exosomes have also been found to be useful for assessing the severity of kidney diseases.

  1. (1)

    Urinary exosomal proteins as IMN biomarkers

Under normal circumstances, approximately 3% of the total protein content in urine originates from urinary exosomes, with 70% originating from the urinary system and 30% from the circulatory system [76]. During the formation of urinary exosomes, various components undergo selective enrichment, and changes in their protein composition may reflect pathological processes in the urinary system or systemic diseases [21, 92]. Moreover, proteins in urinary exosomes more accurately reflect changes in kidney tissue compared to urinary proteins [93], underscoring the significant potential of urinary exosomes proteins as biomarkers for both the urinary system and systemic conditions [76, 94]. For instance, urinary exosomal ceruloplasmin (CP) is significantly elevated by 10–20 times in CKD patients compared to healthy controls and increases significantly before the onset of proteinuria [95]. Urinary exosomal transcription factor Elf3 protein is exclusively detected in DN and can reflect irreversible podocyte damage, serving as an early noninvasive biomarker for DN podocyte injury [96]. Urinary exosomal fibroblast-specific protein 1 (FSP1) correlates with the diagnosed glomerular crescent formation rate and total crescent formation rate in kidney biopsies, reflecting ongoing glomerular injury activity (crescent formation) [97]. Polycystin-1 (PC-1), the protein product of the autosomal dominant polycystic kidney disease gene, is readily detectable in urinary exosomes, despite its lower abundance in renal tissue [75]. Other proteins in urinary exosomes, such as Fetuin-A [98], activating transcription factor 3 (ATF3) [99, 100] and aquaporin-1 [101], show significant changes in the early stages of AKI and may be potential markers for early detection of AKI. In certain hereditary kidney diseases, the production of pathological proteins regulated by defective genes in exosomes may be reduced (PKD1 in polycystic kidney disease) [102] or completely absent (SLC12A1 in Bartter syndrome type 1) [67] (see Table 1). These studies collectively indicate the widespread utility of urinary exosomal proteins as biomarkers in the field of kidney diseases, revealing a certain feasibility of urinary exocytosis in the diagnosis of IMN, although it has not been directly elucidated.

Table 1 Potential biomarkers of IMN from urinary exosomal proteins

In patients with IMN, the urinary exosomal marker proteins (Alix, CD63, and TSG101) were significantly greater than those in the control group, exhibiting a positive correlation with proteinuria [103]. This can reflect the active pathological changes in renal tissue associated with IMN and holds the potential to become a noninvasive biomarker for IMN diagnosis, disease assessment, and prognosis prediction [103]. Urinary exosomal ceruloplasmin is notably elevated in patients with CKDs, including MN [92, 95], and further investigation using the rat Heymann nephritis model revealed that this elevation occurred prior to the onset of proteinuria. Additionally, studies have indicated a positive correlation between the urinary exosomal proteins Nrf2 and NLRP3 and serum anti-PLA2R antibodies. Lower levels of Nrf2 or NLRP3 are suggestive of better treatment outcomes, suggesting their potential as prospective biomarkers for prognosis assessment [104].

  1. (2)

    Urinary exosomal mRNA as biomarkers for IMN.

Urine presents a potential source of nucleic acids, although these may arise from apoptotic cells and potentially not accurately reflect the functional state of viable cells [21]. Furthermore, urine's intricate composition, coupled with a lack of specificity in its components, may introduce interference into component detection [21]. However, urinary exosomes selectively encapsulate mRNA and miRNA, overcoming these drawbacks by enriching for relatively specific components [21]. Moreover, the bilayer membrane structure of exosomes shields against degradation by both intracellular and extracellular RNases, rendering exosomal RNA more stable than total urine RNA [21]. Recent research, as indicated in Table 2, underscores the practicality of urinary exosomal mRNA as biomarkers for various kidney diseases, including IMN, DN, FSGS, IgA nephropathy, renal fibrosis, and CKD. For example, in IMN patients, CCL2 mRNA expression was significantly elevated compared to healthy controls [118]. Similarly, in patients with renal fibrosis, urinary exosomal CD2AP mRNA downregulation correlated negatively with renal function, proteinuria levels, severity of fibrosis, and glomerular sclerosis [119]. Among DN, those with proteinuria displayed notably elevated levels of urinary exosomal WT1 mRNA expression compared to nonproteinuric patients. WT1 levels were indicative of the extent of diabetic glomerular injury [68, 106]. These findings collectively underscore the critical role of urinary exosomal vesicle mRNA as essential diagnostic and prognostic tools for various kidney diseases, including IMN. This methodology capitalizes on the enrichment of specific mRNA types and the inherent stability of exosomal RNA, ultimately amplifying the potential for early detection and management of renal pathologies [68, 118, 120].

  1. (3)

    Urinary Exosomal Non-Coding RNAs as Biomarkers for IMN.

Table 2 Potential biomarkers of IMN from urinary exosomal mRNAs

MicroRNAs (miRNAs) are a class of noncoding RNAs that play a crucial role in the regulation of gene expression. Typically, they interact with the 3' UTR of target mRNAs to suppress gene expression [123], influencing various biological processes [124]. Aberrant expression of miRNAs has been linked to numerous human diseases [125, 126], suggesting that they could be potential biomarkers for a variety of kidney disorders [127,128,129] (Table 3). Moreover, urinary exosomes contain abundant miRNAs, rendering them potential biomarkers for diverse diseases [21, 130, 131]. They can also reflect kidney dysfunction and structural damage [21, 127,128,129, 131]. For instance, in CKD, the overexpression of the urinary exosomes miR-181a-5p [85] and miR-451 [132] individually contributes to CKD pathogenesis through lipid metabolism modulation, renal fibrosis, and mesangial hypertrophy [132]. Renal fibrosis serves as an indicator of permanent CKD-related damage, and correlations between elevated miR-200b [73] and decreased miR-29c [90, 119] levels and CKD-related fibrosis have been established. In DN patients, urinary exosomal miR-21-5p [133], miR-15b, miR-34a, miR-636 [134], and miR-30b-5p [133] hold promise as potential biomarkers. In lupus nephritis (LN) patients, urinary exosomal miR-21, miR-29c, and miR-150 are potential predictive biomarkers for disease progression [135]. Notably, reduced levels of urinary exosomal miR-29a and miR-29c are associated with disease severity, tubulointerstitial fibrosis, and glomerulosclerosis in DN, focal segmental glomerulosclerosis, IgA nephropathy, MN, and membranoproliferative glomerulonephritis [90, 119]. These findings underscore the invaluable diagnostic advantage of urinary extracellular vesicle miRNAs in early-stage kidney diseases. In patients with IMN, Ma et al. [16] identified MUC3A in blood and urinary exosomes as a potential diagnostic biomarker for IMN. The implication is that the MUC3A gene encodes amino acids pertinent to IMN pathogenesis, possibly involving the lectin pathway via mannose binding.

Table 3 Potential biomarkers of IMN from urinary exosomal noncoding RNA

Role of exosomes in the pathogenesis of IMN

Elucidating the pathogenic mechanisms underlying IMN through the use of exosomes is imperative for improving the diagnosis and treatment of this disease. Exosomes are not only cellular entities but also pivotal players within the framework of disease mechanisms [159]. Previously, it was widely believed that the primary physiological role of urinary exosomes was the disposal of senescent proteins from cells, possibly through a more effective protein elimination method than proteasomal and lysosomal degradation [76]. This process is akin to the shedding of outdated membrane proteins and subsequent membrane remodeling by mature reticulocytes via the exosomal route [160]. However, an increasing body of evidence suggests that the role of urinary exosomes extends beyond the elimination of extracellular cellular waste [161, 162]. Another potential role of miRNAs is their ability to impact recipient cell mRNAs and miRNAs by secreting and reabsorbing their contents, thus regulating collaborative functions among various parts of the kidney [74]. Songjia Guo, Jinshi Zhang, and their colleagues employed high-throughput sequencing to analyze urinary exosomal miRNA expression profiles in healthy controls and IMN patients. These authors revealed significant downregulation of miRNAs, including miR-532-3p, miR-9-5p, miR-30b-5p, miR-129-5p, miR-125b, and miR-338-5p, in IMN patients [163, 164]. These findings suggest the potential involvement of these miRNAs in the pathogenesis of IMN.

  1. (1)

    Associated with PLA2R1 and HLA-DQA1.

PLA2R1 and HLA-DQA1 have been confirmed to be risk factors for IMN [165]. Currently, anti-PLA2R antibodies serve as crucial diagnostic markers for IMN, with approximately 70% of IMN patients exhibiting their presence via kidney biopsies. A search of the TargetScanHuman8.0 database (https://www.targetscan.org/vert_80] revealed that differentially expressed genes, such as miR-30b-5p and miR-9-5p, in the urinary exosomes of IMN patients potentially regulate PLA2R1. Additionally, other members of the miR-30 family (miR-30 s) are associated with HLA-DQA1. Further Spearman correlation analysis indicated a significant negative correlation between miR-30b-5p and anti-PLA2R antibodies [164]. Hence, we postulate that urinary exosomes may participate in the pathogenesis of IMN by potentially modulating anti-PLA2R antibodies and/or HLA-DQA1 (Fig. 2).

  1. (2)

    Regulating extracellular matrix and combating renal fibrosis.

Fig. 2
figure 2

The role of exosomes in the pathogenesis of IMN. The role of exosomes in the pathogenesis of IMN. The five miRNAs in the figure are the differential miRNAs identified by miRNA flux sequencing in IMN patients compared with healthy controls, and they are involved in the pathogenesis of IMN through four main aspects, namely, the regulation of anti-PLA2R antibody and/or HLA-DQA1, renal fibrosis, podocyte injury, and immune homeostasis of Tregs

It is well known that both MNs and DNs are associated with varying degrees of excessive accumulation of extracellular matrix, leading to gradual glomerular sclerosis and renal fibrosis. Renal fibrosis is the ultimate outcome of CKD development and a major contributor to ESRD. Research has indicated that miR-30b-5p and miR-9-5p may be involved in the process of renal fibrosis [166, 167]. In DN mouse models and human kidney tissues, miR-30b-5p is significantly downregulated, thereby promoting epithelial–mesenchymal transition (EMT) in DN. Moreover, overexpression of miR-30b-5p can mitigate high glucose-induced EMT [166]. This effect is likely achieved by targeting the key EMT activator SNAI1. In unilateral ureteral obstruction (UUO) mice, miR-9-5p protects against renal fibrosis by inhibiting the downregulation of genes associated with key metabolic pathways, including mitochondrial function, oxidative phosphorylation, fatty acid oxidation (FAO), and glycolysis [167]. In IMN patients, there are differences in the expression of urinary exosomal miR-30b-5p and miR-9-5p. The downregulation of urinary exosomal miR-9-5p in IMN patients may reflect the active metabolism of pathways related to kidney fibrosis. Based on the above findings, it can be inferred that miR-30b-5p and miR-9-5p might also play a role in the renal fibrosis process in IMN [164] (Fig. 2).

  1. (3)

    Associated with podocyte injury.

Podocytes are terminally differentiated visceral epithelial cells of the glomerulus in the kidney; together with the basement membrane and endothelial cells, these cells form the glomerular filtration barrier [168]. Podocyte injury leads to proteinuria, and reduced podocyte numbers are considered a relative risk factor for progressive kidney damage [169]. The primary pathological change in IMN is kidney glomerular podocyte injury caused by immune complex deposition. MiRNAs are essential for maintaining podocyte homeostasis. Studies have shown that differentially expressed miR-9-5p and miR-30 s in IMN urinary exosomes may be involved in maintaining podocyte stability [164]. Wu et al. reported that downregulation of miR-30 induces proteinuria and podocyte injury [145]. Further confirmation in a rat model demonstrated that miR-30 exerts a protective effect by directly inhibiting Notch1 and p53, which mediate podocyte injury [137]. Moreover, recent research has suggested that miR-30 may enhance mouse podocyte injury and proteinuria improvement by potentially regulating calcium/calcineurin signaling and disrupting urokinase-type plasminogen activator receptor-integrin β3 (uPAR-ITGB3) signal transduction [170]. In addition, miR-9-5p, regulated by tumor susceptibility candidate gene 2 (CASC2), targets PPARγ and can alleviate podocyte injury [171]. Furthermore, relevant literature indicates that differentially expressed genes associated with urinary exosomal miRNAs, such as miR-532-3p [172], miR-429 [173], miR-129-5p [174], miR-107 [172], miR-25-3p [175], and miR-206 [176, 177], are associated with glomerular podocyte injury, and miR-532 and miR-107 have been confirmed to participate in podocyte injury in MN [178] (Fig. 2).

  1. (4)

    Tregs are involved in the regulation and modulation of the immune response.

Currently, antigen‒antibody reactions are considered the primary immunopathogenic mechanism of MN [179], and CD4 + T cells are recognized as key cellular participants in immune responses [180]. CD4 + T cells consist of helper T cells (Th) and regulatory T cells (Tregs), with the former playing a pivotal role in the immune response by secreting cytokines that mediate inflammatory reactions and pathogen clearance [180]. Treg cells primarily regulate the intensity of Th cell responses to prevent excessive immune reactions, causing self-repair damage [180]. Clinically, MN is also characterized by evident Th cell subset imbalances. Multiple studies have indicated that Th17 cell expression is enhanced in MN patients, along with upregulated IL-17 and other cytokines [181,182,183], while the proportion of Treg cells is reduced [183]. In the urine of IMN patients, differentially expressed miRNAs, including miR-532-3p [184], miR-9-5p [185], miR-30b-5p [186], miR-129-5p [186], miR-125b [187], and miR-338-5p [188], have been found to participate in the regulation of Tregs across various diseases. Therefore, it is inferred that in IMN, the differential expression of miRNAs, including miR-532-3p, miR-9-5p, miR-30b-5p, miR-129-5p, miR-125b, and miR-338-5p, in urine exosomes might also be involved in Treg regulation to prevent kidney damage potentially caused by excessive immune reactions [184]. However, these assumptions await further experimental validation [164] (Fig. 2).

The role of exosomes in the treatment of IMN

Treatment for IMN primarily involves the use of steroids in combination with alkylating agents in modern medicine. The latest 2021 guidelines from KDIGO [189] included rituximab as a first-line treatment for IMN. However, challenges persist, such as inconsistent efficacy, substantial side effects, and a high relapse rate, which fail to fully meet the therapeutic needs of MN patients [190]. Consequently, exploring safer and more effective treatment approaches is imperative. Exosomes have demonstrated potential as cellular therapy alternatives in preclinical and clinical studies, with data indicating the feasibility and safety of exosome-based treatments. For instance, exosomes derived from dendritic cells (DCs), which contain major histocompatibility complex/peptide complexes and promote T-cell immune responses, have been tested in clinical trials as vaccines against metastatic melanoma and non-small cell lung cancer [191,192,193]. Furthermore, exosomes sourced from stem cells have been developed for applications in cardiovascular disease, diabetes, graft-versus-host disease, and neurological and orthopedic disorders [194,195,196]. Clinical trials have also explored the use of plant-derived exosomes for curcumin delivery [197, 198]. In the field of kidney diseases, multiple preclinical, clinical, and in vitro models have been used to investigate the potential therapeutic applications of exosomes in conditions such as DN [199], hypertension-related cardiorenal syndrome [200], acute kidney injury [201, 202], IgA nephropathy [203], cadmium nephropathy [204], obstructive kidney diseases [205], and ischemia/reperfusion injury [206]. Exosomes, which can act as therapeutic agents or drug delivery vehicles, exhibit significant potential to mitigate systemic consequences in patients with CKD [207], suggesting that they are promising candidates for treatment [208,209,210]. Moreover, the discovery of mRNAs and miRNAs in exosomes and their role in cell-to-cell communication signify a novel direction for utilizing exosomes as delivery vehicles for therapeutic drugs [76].

  1. (1)

    Therapeutic agents: exosomes with inherent healing activity.

Exosomes carrying RNA can selectively deliver their contents to specific target cells, temporarily correcting dysfunctional processes [76]. This endows exosomes with immense potential as therapeutic delivery vehicles. Exosomes have found widespread application in kidney diseases, such as modulating kidney transplant rejection, rectifying metabolic defects, and fostering renal regeneration. These therapeutic extracellular vesicles (EVs) seem to primarily derive from various sources of mesenchymal stem cells [211]. Mesenchymal stem cells (MSCs), recognized as among the most effective stem cell types for inducing kidney regeneration and having diverse differentiation potential [212], predominantly treat kidney ailments through the paracrine release of EVs [213, 214]. For instance, they have demonstrated the ability to reverse acute and chronic kidney injuries in various experimental models [215]. These effects are partly driven by paracrine enhancement of recovery [215,216,217] and are strongly mediated by the cargo of RNA within exosomes and/or microvesicles [218, 219]. Injection of exosomes derived from bone marrow mesenchymal stem cells (BMSCs) into DN rats significantly improves renal tissue oxidative stress damage, reduces urinary protein excretion, and safeguards renal function [220]. Injection of exosomes isolated from urine-derived stem cells (SCs) into DN rats decreases cellular apoptosis and urinary ALB while enhancing glomerular endothelial cell growth [221]. Moreover, urinary stem cells have been shown to repair podocyte injury through exosome-mediated mechanisms [222] (see Fig. 3 for details). Additionally, exosomes from cultured epithelial cells also exhibit some effects in vitro [223].

Fig. 3
figure 3

Exosomes as therapeutic agents for IMN. Exosomes as therapeutic agents for IMN. Exosomes used as therapeutic agents for IMN are mainly derived from mesenchymal stem cells (MSCs), which can be subdivided into bone marrow mesenchymal stem cells (BMSCs) and urothelial stem cells (SCs). Each of the three pathways shown in the figure treats IMN through different pathways

For many kidney-related diseases, the primary targets for potential exosome-based therapies are endothelial cells, which play important roles in regulating blood pressure, locally regulating blood flow, modulating blood coagulation, and removing plasma lipids and are readily accessible to exosomes from the circulation [94]. Dysregulation of these processes constitutes a significant factor contributing to common CKDs. Endothelial cells face the bloodstream, positioning them as "low-hanging fruits" for exosome-based therapies and largely circumventing targeting issues [76]. Although current research lacks the application of exosomes as therapeutic agents for IMN, the future may involve utilizing mesenchymal stem cells or epithelial cells as sources, with endothelial cells as targets, potentially ushering in a new paradigm for treating IMN.

  1. (2)

    Drug delivery vehicles: exosomes as therapeutic carriers.

Currently, various drug delivery vehicles, such as liposomes, micelles, nanoparticles, and hydrogels, are being extensively investigated. However, many of these materials face significant challenges, such as low bioavailability and high systemic toxicity [33]. Recently, exosomes and microvesicles have garnered substantial attention as novel drug delivery vehicles due to the following attributes: (a) Safety: Exosomes, which are endogenous carriers, exhibit excellent biocompatibility, low immunogenicity, and good tolerability, thereby establishing safer and more effective drug delivery systems (DDSs) [224,225,226,227,228,229]. (b) Barrier penetration: Exosomes and microvesicles, owing to their small size and flexibility, can traverse major biological barriers, including the blood‒brain barrier (BBB) [230,231,232,233]. Zhuang et al. discovered that exosomes effectively transport curcumin to the brain to treat neuroinflammation-related diseases without side effects [227]. (c) Specificity: Analysis of the proteins on the surface of exosome membranes aids in developing drug delivery systems for targeted cell-specific delivery [234]. (d) Stability: The bilayer structure of exosomes shields their cargo from RNases and proteases, enhancing drug stability and efficacy [94]; see Fig. 4 for details. Additionally, research suggests that the bioavailability of exosome delivery systems surpasses that of other systems. For instance, doxorubicin loaded into exosomes has been shown to be more effective than other delivery systems and to cause fewer adverse effects on major organ systems, especially the heart [235]. In the future, exosomes hold promise for delivering drugs or traditional Chinese medicine monomers for treating IMN to target organs, enhancing treatment precision and effectiveness.

Fig. 4
figure 4

Exosomes as drug delivery carriers. Exosomes, as drug delivery carriers, consist of three main components: harvesting, loading engineering and targeted delivery

Future outlook and challenges

As novel biological signaling molecules and therapeutic carriers, exosomes have unique advantages in the field of kidney disease diagnosis and treatment. Compared with other renal diseases, there are relatively few studies on the use of exosomes in the treatment of IMN, which also means that exosome development in IMN will help revolutionize the diagnosis and treatment of IMN. Inevitably, there are some challenges to overcome.

  1. (1)

    Extension of clinical applications: exosomes face the challenges of standardization and standardized methods in the clinical treatment of IMN. Ensuring consistency and accuracy in the exosome collection, purification and assay process is critical to ensure the efficacy and reproducibility of the results. Large-scale multicenter clinical trials are necessary to extensively validate the efficacy and safety of exosomes in patients with IMN and to develop relevant guidelines and standards.

  2. (2)

    In-depth mechanistic exploration of exosomes: although exosomes play an important role in the pathogenesis of IMN, their specific regulatory mechanisms and targets of action are not yet fully understood. Future studies should further explore the relationship between exosomes and IMN and reveal their regulatory networks and signaling pathways to better understand the occurrence and progression of IMN.

  3. (3)

    The development of individualized therapeutic strategies: IMN is a heterogeneous disease with significant clinical variability. As potential biomarkers and therapeutic targets, exosomes hold promise for individualized diagnosis and treatment. Future research should focus on the transition from discovery to application. Exosomes can be used for early diagnosis, patient staging and severity prediction, as well as for more accurate identification of underlying etiologies, improved patient categorization, and stratification of patients with IMN. Appropriate exosome-targeted therapies should be selected based on individual patient characteristics. After specific therapeutic strategies have been defined, continuous monitoring of therapeutic efficacy allows for close individualization of diagnosis and treatment.

  4. (4)

    Optimization of drug delivery vehicles and associated techniques: the foremost challenges of using exosomes as drug delivery vehicles include imperfect extraction and separation techniques, which can lead to low yields and low encapsulation and loading efficiencies. Functionalizing exosomes is needed for encapsulating hydrophilic macromolecules. Concurrently, advancing and optimizing exosome delivery systems and technologies is crucial for enhancing exosome stability and targeting within the body. Additionally, additional pharmacological studies are needed to validate the safety and efficacy of exosome-targeted therapies for eventual clinical translation.

  5. (5)

    Interdisciplinary Collaboration and Data Sharing: exosome research demands interdisciplinary collaboration involving experts from fields such as nephrology, molecular biology, and bioinformatics. Future efforts should strengthen collaboration and communication between different domains, facilitating data and resource sharing to expedite research progress and promote exosome applications in IMNs.

In conclusion, exosomes hold immense potential for the diagnosis and treatment of IMN. Future research and clinical practices should further refine the techniques and methods, explore their mechanisms comprehensively, develop personalized treatment strategies, intensify drug development, and foster interdisciplinary collaboration to realize the widespread application of exosomes in IMN. These findings could lead to more precise and effective diagnostic and therapeutic tools for IMN patients, significantly improving disease management and prognosis.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

ARF1:

ADP-ribosylation factor 1

ATF3:

Activating transcription factor 3

CDC42:

Cell division control protein 42

CP:

Cerulo plasmin

DCs:

Dendritic cells

eGFR:

Estimated glomerular filtration rate

EMT:

Epithelial–mesenchymal transition

ER:

Endoplasmic reticulum

ESE:

Early-sorting endosome

Evs:

Extracellular vesicles

FSP1:

Fibroblast-specific protein1

MSCs:

Mesenchymal stem cells

NGAL:

Neutrophil gelatinase-associated lipocalin

PI3K:

Phosphoinos itide 3-kinase

SCs:

Stem cells

Th:

Helper T cells

Treg:

Regulatory Tcells

UUO:

Unilateral ureteral obstruction

AKI:

Acute kidney injury

BMSCs:

Bone marrowmesenchymal stem cells

CKD:

Chronic kidney disease

DN:

Diabetic nephropathy

ESRD:

End-stage renal disease

FSGS:

Focal segmental glomerulo sclerosis

IgAN:

IgA nephropathy

ILVs:

Intra luminal vesicles

IMN:

Idiopathic membranous nephropathy

KDIGO:

International Kidney Disease Guidelines

LSEs:

Late-sorting endosomes

MCN:

Minimal change nephrosis

MCNS:

Minimal change nephrotic syndrome

MHC-II:

Major histocompatibility complex class II

MN:

Membranous nephropathy

MPS:

Mononuclear phagocyte system

MUC1:

Mucin1

MVBs:

Multi vesicula rbodies

NGAL:

Neutrophil gelatinase-associated lipocalin

SSNS:

Steroid-sensitive nephrotic syndrome

TBMN:

Thin basement membrane nephropathy

TGN:

Trans-Golgi network

UE:

Urinary exosomes

References

  1. Couser WG. Primary membranous nephropathy. Clin J Am Soc Nephrol. 2017;12(6):983.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Murugapandian S, Mansour I, Hudeeb M, Hamed K, Hammode E, Bijin B, Daheshpour S, Thajudeen B, Kadambi P. Epidemiology of glomerular disease in Southern Arizona: review of 10-year renal biopsy data. Medicine. 2016;95(18):e3633.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Tang L, Yao J, Kong X, Sun Q, Wang Z, Zhang Y, Wang P, Liu Y, Li W, Cui M, Zhen J, Xu D. Increasing prevalence of membranous nephropathy in patients with primary glomerular diseases: a cross-sectional study in China. Nephrology. 2017;22(2):168–73.

    Article  PubMed  Google Scholar 

  4. Ronco P, Beck L, Debiec H, Fervenza FC, Hou FF, Jha V, Sethi S, Tong A, Vivarelli M, Wetzels J. Membranous nephropathy. Nat Rev Dis Primers. 2021;7(1):69.

    Article  PubMed  Google Scholar 

  5. Xu X, Wang G, Chen N, Lu T, Nie S, Xu G, Zhang P, Luo Y, Wang Y, Wang X, Schwartz J, Geng J, Hou FF. Long-term exposure to air pollution and increased risk of membranous nephropathy in China. J Am Soc Nephrol. 2016;27(12):3739–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hogan SL, Muller KE, Jennette JC, Falk RJ. A review of therapeutic studies of idiopathic membranous glomerulopathy. Am J Kidney Dis. 1995;25(6):862–75.

    Article  CAS  PubMed  Google Scholar 

  7. Hamilton P, Blaikie K, Roberts SA, Gittins M, Downie ML, Gupta S, Voinescu C, Kanigicherla D, Stanescu H, Kleta R, Brenchley P. Membranous nephropathy in the UK Biobank. PLoS ONE. 2023;18(4):e0281795.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Keri KC, Blumenthal S, Kulkarni V, Beck L, Chongkrairatanakul T. Primary membranous nephropathy: comprehensive review and historical perspective. Postgrad Med J. 2019;95(1119):23–31.

    Article  CAS  PubMed  Google Scholar 

  9. Zhu P, Zhou FD, Wang SX, Zhao MH, Wang HY. Increasing frequency of idiopathic membranous nephropathy in primary glomerular disease: a 10-year renal biopsy study from a single Chinese nephrology centre. Nephrology. 2015;20(8):560–6.

    Article  PubMed  Google Scholar 

  10. Wang K. Clinical and pathological characterization of 3899 renal biopsy patients in a single center. Jilin University, 2020.

  11. Shang Z, Sun Y, Wu R, Qiao Y, Yang H. To explore the current status of research on membranous nephropathy in China from 2011 to 2020 based on bibliometrics. Chin J Integr Tradit West Nephrol. 2022;23(06):527–9.

    Google Scholar 

  12. Campion CG, Sanchez-Ferras O, Batchu SN. Potential role of serum and urinary biomarkers in diagnosis and prognosis of diabetic nephropathy. Can J Kidney Health Dis. 2017;4:2054358117705371.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Coban M, Eke RN, Kizilates F, Ucar S, Dede F. Effect of steroid and cyclosporine in membranous nephropathy that is resistant to steroid and/or cytotoxic treatment. Int J Clin Exp Med. 2014;7(1):255–61.

    PubMed  PubMed Central  Google Scholar 

  14. Troyanov S, Wall CA, Miller JA, Scholey JW, Cattran DC. Idiopathic membranous nephropathy: definition and relevance of a partial remission. Kidney Int. 2004;66(3):1199–205.

    Article  PubMed  Google Scholar 

  15. van den Brand J, Ruggenenti P, Chianca A, Hofstra JM, Perna A, Ruggiero B, Wetzels JFM, Remuzzi G. Safety of rituximab compared with steroids and cyclophosphamide for idiopathic membranous nephropathy. J Am Soc Nephrol. 2017;28(9):2729–37.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhang W, Zhou X, Zhang H, Yao Q, Liu Y, Dong Z. Extracellular vesicles in diagnosis and therapy of kidney diseases. Am J Physiol Renal Physiol. 2016;311(5):F844–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Karpman D, Ståhl AL, Arvidsson I. Extracellular vesicles in renal disease. Nat Rev Nephrol. 2017;13(9):545–62.

    Article  CAS  PubMed  Google Scholar 

  18. Chen HH, Lai PF, Lan YF, Cheng CF, Zhong WB, Lin YF, Chen TW, Lin H. Exosomal ATF3 RNA attenuates pro-inflammatory gene MCP-1 transcription in renal ischemia-reperfusion. J Cell Physiol. 2014;229(9):1202–11.

    Article  CAS  PubMed  Google Scholar 

  19. Asao R, Asanuma K, Kodama F, Akiba-Takagi M, Nagai-Hosoe Y, Seki T, Takeda Y, Ohsawa I, Mano S, Matsuoka K, Kurosawa H, Ogasawara S, Hirayama Y, Sekine S, Horikoshi S, Hara M, Tomino Y. Relationships between levels of urinary podocalyxin, number of urinary podocytes, and histologic injury in adult patients with IgA nephropathy. Clin J Am Soc Nephrol. 2012;7(9):1385–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hara M, Yamagata K, Tomino Y, Saito A, Hirayama Y, Ogasawara S, Kurosawa H, Sekine S, Yan K. 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(11):2913–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Miranda KC, Bond DT, McKee M, Skog J, Păunescu TG, Da Silva N, Brown D, Russo LM. Nucleic acids within urinary exosomes/microvesicles are potential biomarkers for renal disease. Kidney Int. 2010;78(2):191–9.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Hogan MC, Bakeberg JL, Gainullin VG, Irazabal MV, Harmon AJ, Lieske JC, Charlesworth MC, Johnson KL, Madden BJ, Zenka RM, McCormick DJ, Sundsbak JL, Heyer CM, Torres VE, Harris PC, Ward CJ. Identification of biomarkers for PKD1 using urinary exosomes. J Am Soc Nephrol. 2015;26(7):1661–70.

    Article  CAS  PubMed  Google Scholar 

  23. Gallo A, Tandon M, Alevizos I, Illei GG. The majority of microRNAs detectable in serum and saliva is concentrated in exosomes. PLoS ONE. 2012;7(3):e30679.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Jovic M, Sharma M, Rahajeng J, Caplan S. The early endosome: a busy sorting station for proteins at the crossroads. Histol Histopathol. 2010;25(1):99–112.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol. 1983;97(2):329–39.

    Article  CAS  PubMed  Google Scholar 

  26. Pan BT, Teng K, Wu C, Adam M, Johnstone RM. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol. 1985;101(3):942–8.

    Article  CAS  PubMed  Google Scholar 

  27. Tschuschke M, Kocherova I, Bryja A, Mozdziak P, Angelova Volponi A, Janowicz K, Sibiak R, Piotrowska-Kempisty H, Iżycki D, Bukowska D, Antosik P, Shibli JA, Dyszkiewicz-Konwińska M, Kempisty B. Inclusion biogenesis, methods of isolation and clinical application of human cellular exosomes. J Clin Med. 2020;9(2):436.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kosaka N, Iguchi H, Hagiwara K, Yoshioka Y, Takeshita F, Ochiya T. Neutral sphingomyelinase 2 (nSMase2)-dependent exosomal transfer of angiogenic microRNAs regulate cancer cell metastasis. J Biol Chem. 2013;288(15):10849–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hanson PI, Cashikar A. Multivesicular body morphogenesis. Annu Rev Cell Dev Biol. 2012;28:337–62.

    Article  CAS  PubMed  Google Scholar 

  30. Keerthikumar S, Gangoda L, Gho YS, Mathivanan S. Bioinformatics tools for extracellular vesicles research. Methods Mol Biol. 2017;1545:189–96. https://doi.org/10.1007/978-1-4939-6728-5_13.

  31. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569–79.

    Article  PubMed  Google Scholar 

  32. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Ann Rev Cell Dev Biol. 2014;30:255–89.

    Article  CAS  Google Scholar 

  33. Deb A, Gupta S, Mazumder PB. Exosomes: a new horizon in modern medicine. Life Sci. 2021;264:118623.

    Article  CAS  PubMed  Google Scholar 

  34. Laulagnier K, Motta C, Hamdi S, Roy S, Fauvelle F, Pageaux JF, Kobayashi T, Salles JP, Perret B, Bonnerot C, Record M. Mast cell- and dendritic cell-derived exosomes display a specific lipid composition and an unusual membrane organization. Biochem J. 2004;380(Pt 1):161–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Staubach S, Razawi H, Hanisch FG. Proteomics of MUC1-containing lipid rafts from plasma membranes and exosomes of human breast carcinoma cells MCF-7. Proteomics. 2009;9(10):2820–35.

    Article  CAS  PubMed  Google Scholar 

  36. Amiri A, Bagherifar R, Ansari Dezfouli E, Kiaie SH, Jafari R, Ramezani R. Exosomes as bio-inspired nanocarriers for RNA delivery: preparation and applications. J Transl Med. 2022;20(1):125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. De Toro J, Herschlik L, Waldner C, Mongini C. Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications. Front Immunol. 2015;6:203.

    PubMed  PubMed Central  Google Scholar 

  38. Tamkovich S, Tutanov O, Laktionov P. Exosomes: generation, structure, transport, biological activity, and diagnostic application. Biochem Moscow Suppl Ser A Membr Cell Biol. 2016;10:163–73.

    Article  Google Scholar 

  39. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9(6):654–9.

    Article  CAS  PubMed  Google Scholar 

  40. Melo SA, Sugimoto H, O’Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA, Lucci A, Ivan C, Calin GA, Kalluri R. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26(5):707–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Deregibus MC, Cantaluppi V, Calogero R, Lo Iacono M, Tetta C, Biancone L, Bruno S, Bussolati B, Camussi G. Endothelial progenitor cell derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA. Blood. 2007;110(7):2440–8.

    Article  CAS  PubMed  Google Scholar 

  42. Chen WX, Liu XM, Lv MM, Chen L, Zhao JH, Zhong SL, Ji MH, Hu Q, Luo Z, Wu JZ, Tang JH. Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs. PLoS ONE. 2014;9(4):e95240.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Zhang J, Li S, Li L, Li M, Guo C, Yao J, Mi S. Exosome and exosomal microRNA: trafficking, sorting, and function. Genomics Proteomics Bioinform. 2015;13(1):17–24.

    Article  CAS  Google Scholar 

  44. Momen LT, Abdolmaleki A, Asadi A, Akram M. Regeneration and diagnosis of kidney disease using exosomes. J Cell Mol Biol. 2021. https://doi.org/10.5812/jjcmb.120113.

    Article  Google Scholar 

  45. Miao C, Wang X, Zhou W, Huang J. The emerging roles of exosomes in autoimmune diseases, with special emphasis on microRNAs in exosomes. Pharmacol Res. 2021;169:105680.

    Article  CAS  PubMed  Google Scholar 

  46. Gurunathan S, Kang MH, Kim JH. A comprehensive review on factors influences biogenesis, functions, therapeutic and clinical implications of exosomes. Int J Nanomed. 2021;16:1281–312.

    Article  Google Scholar 

  47. Gurunathan S, Kang MH, Jeyaraj M, Qasim M, Kim JH. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes. Cells. 2019;8(4):307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Choi JY, Kim S, Kwak HB, Park DH, Park JH, Ryu JS, Park CS, Kang JH. extracellular vesicles as a source of urological biomarkers: lessons learned from advances and challenges in clinical applications to major diseases. Int Neurourol J. 2017;21(2):83–96.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Camussi G, Deregibus MC, Bruno S, Cantaluppi V, Biancone L. Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 2010;78(9):838–48.

    Article  CAS  PubMed  Google Scholar 

  50. Dear JW, Street JM, Bailey MA. Urinary exosomes: a reservoir for biomarker discovery and potential mediators of intrarenal signaling. Proteomics. 2013;13(10–11):1572–80.

    Article  CAS  PubMed  Google Scholar 

  51. Lässer C, Alikhani VS, Ekström K, Eldh M, Paredes PT, Bossios A, Sjöstrand M, Gabrielsson S, Lötvall J, Valadi H. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med. 2011;9:9.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Palanisamy V, Sharma S, Deshpande A, Zhou H, Gimzewski J, Wong DT. Nanostructural and transcriptomic analyses of human saliva derived exosomes. PLoS ONE. 2010;5(1):e8577.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, Zheng Y, Hoshino A, Brazier H, Xiang J, Williams C, Rodriguez-Barrueco R, Silva JM, Zhang W, Hearn S, Elemento O, Paknejad N, Manova-Todorova K, Welte K, Bromberg J, Peinado H, Lyden D. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. 2014;24(6):766–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Tu M, Wei F, Yang J, Wong D. Detection of exosomal biomarker by electric field-induced release and measurement (EFIRM). Biosens Bioelectron. 2015;95:e52439.

    Google Scholar 

  55. Zhang Y, Liu Y, Liu H, Tang WH. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 2019;9:19.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Chen B-Y, Sung CW-H, Chen C, Cheng C-M, Lin DP-C, Huang C-T, Hsu M-Y. Advances in exosomes technology. Clin Chimica Acta. 2019;493:14–9.

    Article  CAS  Google Scholar 

  57. Ge Q, Zhou Y, Lu J, Bai Y, Xie X, Lu Z. miRNA in plasma exosome is stable under different storage conditions. Molecules. 2014;19(2):1568–75.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Myers LW. The use of extracellular vesicles (EVs) in regenerative medicine: a move toward cell-derived EV-based therapeutics and their use as novel biomarkers, College of Medicine-Mayo Clinic, 2022.

  59. Zhang L-Y, Yang X, Wang S-B, Chen H, Pan H-Y, Hu Z-M. Membrane derived vesicles as biomimetic carriers for targeted drug delivery system. Curr Top Med Chem. 2020;20(27):2472–92.

    Article  CAS  PubMed  Google Scholar 

  60. Chen P. The disruptive effects of renal disease on the peripheral biological clock system. Peking Union Medical College, 2018.

  61. Liu Q, Liu J, Lin B, Zhang Y, Ma M, Yang M, Qin X. Novel biomarkers in membranous nephropathy. Front Immunol. 2022;13:845767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Radice A, Pieruzzi F, Trezzi B, Ghiggeri G, Napodano P, D’Amico M, Stellato T, Brugnano R, Ravera F, Rolla D, Pesce G, Giovenzana ME, Londrino F, Cantaluppi V, Pregnolato F, Volpi A, Rombolà G, Moroni G, Ortisi G, Sinico RA. Diagnostic specificity of autoantibodies to M-type phospholipase A2 receptor (PLA2R) in differentiating idiopathic membranous nephropathy (IMN) from secondary forms and other glomerular diseases. J Nephrol. 2018;31(2):271–8.

    Article  CAS  PubMed  Google Scholar 

  63. van de Logt AE, Fresquet M, Wetzels JF, Brenchley P. The anti-PLA2R antibody in membranous nephropathy: what we know and what remains a decade after its discovery. Kidney Int. 2019;96(6):1292–302.

    Article  PubMed  Google Scholar 

  64. Masaoutis C, Al Besher S, Koutroulis I, Theocharis S. Exosomes in nephropathies: a rich source of novel biomarkers. Dis Markers. 2020. https://doi.org/10.1155/2020/8897833.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Fraser SD, Roderick PJ, McIntyre NJ, Harris S, McIntyre C, Fluck R, Taal MW. Assessment of proteinuria in patients with chronic kidney disease stage 3: albuminuria and nonalbumin proteinuria. PLoS ONE. 2014;9(5):e98261.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Bazzi C, Petrini C, Rizza V, Arrigo G, Beltrame A, Pisano L, D’Amico G. Urinary excretion of IgG and alpha(1)-microglobulin predicts clinical course better than extent of proteinuria in membranous nephropathy. Am J Kidney Dis. 2001;38(2):240–8.

    Article  CAS  PubMed  Google Scholar 

  67. Gonzales PA, Pisitkun T, Hoffert JD, Tchapyjnikov D, Star RA, Kleta R, Wang NS, Knepper MA. Large-scale proteomics and phosphoproteomics of urinary exosomes. J Am Soc Nephrol. 2009;20(2):363. https://doi.org/10.1681/ASN.2008040406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Abe H, Sakurai A, Ono H, Hayashi S, Yoshimoto S, Ochi A, Ueda S, Nishimura K, Shibata E, Tamaki M. Urinary exosomal mRNA of WT1 as diagnostic and prognostic biomarker for diabetic nephropathy. J Med Investig. 2018;65(3.4):208–15.

    Article  Google Scholar 

  69. Zhang L-H, Zhu X-Y, Eirin A, Nargesi AA, Woollard JR, Santelli A, Sun IO, Textor SC, Lerman LO. Early podocyte injury and elevated levels of urinary podocyte-derived extracellular vesicles in swine with metabolic syndrome: role of podocyte mitochondria. Am J Physiol Renal Physiol. 2019;317(7):F12–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kwon SH, Woollard JR, Saad A, Garovic VD, Zand L, Jordan KL, Textor SC, Lerman LO. Elevated urinary podocyte-derived extracellular microvesicles in renovascular hypertensive patients. Nephrol Dial Transplant. 2017;32(5):800–7.

    CAS  PubMed  Google Scholar 

  71. Hogan MC, Manganelli L, Woollard JR, Masyuk AI, Masyuk TV, Tammachote R, Huang BQ, Leontovich AA, Beito TG, Madden BJ, Charlesworth MC, Torres VE, LaRusso NF, Harris PC, Ward CJ. Characterization of PKD protein-positive exosome-like vesicles. J Am Soc Nephrol. 2009;20(2):278–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Sun IO, Santelli A, Abumoawad A, Eirin A, Ferguson CM, Woollard JR, Lerman A, Textor SC, Puranik AS, Lerman LO. Loss of renal peritubular capillaries in hypertensive patients is detectable by urinary endothelial microparticle levels. Hypertension. 2018;72(5):1180–8.

    Article  CAS  PubMed  Google Scholar 

  73. Yu Y, Bai F, Qin N, Liu W, Sun Q, Zhou Y, Yang J. Non-proximal renal tubule-derived urinary exosomal miR-200b as a biomarker of renal fibrosis. Nephron. 2018;139(3):269–82.

    Article  CAS  PubMed  Google Scholar 

  74. Dimov I, Jankovic Velickovic L, Stefanovic V. Urinary exosomes. Sci World J. 2009;9:1107–18.

    Article  CAS  Google Scholar 

  75. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci USA. 2004;101(36):13368–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. van Balkom BW, Pisitkun T, Verhaar MC, Knepper MA. Exosomes and the kidney: prospects for diagnosis and therapy of renal diseases. Kidney Int. 2011;80(11):1138–45.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Wang YT, Shi T, Srivastava S, Kagan J, Liu T, Rodland KD. Proteomic analysis of exosomes for discovery of protein biomarkers for prostate and bladder cancer. Cancers. 2020;12(9):2335.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Salih M, Zietse R, Hoorn EJ. Urinary extracellular vesicles and the kidney: biomarkers and beyond. Am J Physiol Renal Physiol. 2014;306(11):F1251–9.

    Article  CAS  PubMed  Google Scholar 

  79. Chen H, Zhang N, Wu Y, Yang C, Xie Q, Deng C, Sun N. Investigation of urinary exosome metabolic patterns in membranous nephropathy by Titania-assisted intact exosome mass spectrometry. Small Sci. 2022;2(5):2100118.

    Article  CAS  Google Scholar 

  80. Vitorino R, Ferreira R, Guedes S, Amado F, Thongboonkerd V. What can urinary exosomes tell us? Cell Mol Life Sci. 2021;78(7):3265–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Gonzales PA, Zhou H, Pisitkun T, Wang NS, Star RA, Knepper MA, Yuen PS. Isolation and purification of exosomes in urine. Methods Mol Biol. 2010;641:89–99.

    Article  CAS  PubMed  Google Scholar 

  82. Jin C, Wu P, Li L, Xu W, Qian H. Exosomes: emerging therapy delivery tools and biomarkers for kidney diseases. Stem Cells Int. 2021;2021:7844455.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Zhou H, Yuen PS, Pisitkun T, Gonzales PA, Yasuda H, Dear JW, Gross P, Knepper MA, Star RA. Collection, storage, preservation, and normalization of human urinary exosomes for biomarker discovery. Kidney Int. 2006;69(8):1471–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Yu W, Hurley J, Roberts D, Chakrabortty SK, Enderle D, Noerholm M, Breakefield XO, Skog JK. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32(4):466–77.

    Article  CAS  PubMed  Google Scholar 

  85. Khurana R, Ranches G, Schafferer S, Lukasser M, Rudnicki M, Mayer G, Hüttenhofer A. Identification of urinary exosomal noncoding RNAs as novel biomarkers in chronic kidney disease. RNA. 2017;23(2):142–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Zhao Y, Shen A, Guo F, Song Y, Jing N, Ding X, Pan M, Zhang H, Wang J, Wu L, Ma X, Feng L, Qin G. Urinary exosomal MiRNA-4534 as a novel diagnostic biomarker for diabetic kidney disease. Front Endocrinol. 2020;11:590.

    Article  CAS  Google Scholar 

  87. Eissa S, Matboli M, Bekhet MM. Clinical verification of a novel urinary microRNA panal: 133b, -342 and -30 as biomarkers for diabetic nephropathy identified by bioinformatics analysis. Biomed Pharmacother. 2016;83:92–9.

    Article  CAS  PubMed  Google Scholar 

  88. Magayr TA, Song X, Streets AJ, Vergoz L, Chang L, Valluru MK, Yap HL, Lannoy M, Haghighi A, Simms RJ, Tam FWK, Pei Y, Ong ACM. Global microRNA profiling in human urinary exosomes reveals novel disease biomarkers and cellular pathways for autosomal dominant polycystic kidney disease. Kidney Int. 2020;98(2):420–35.

    Article  CAS  PubMed  Google Scholar 

  89. Song S, Long M, Yu G, Cheng Y, Yang Q, Liu J, Wang Y, Sheng J, Wang L, Wang Z, Xu B. Urinary exosome miR-30c-5p as a biomarker of clear cell renal cell carcinoma that inhibits progression by targeting HSPA5. J Cell Mol Med. 2019;23(10):6755–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Lv LL, Cao YH, Ni HF, Xu M, Liu D, Liu H, Chen PS, Liu BC. MicroRNA-29c in urinary exosome/microvesicle as a biomarker of renal fibrosis. Am J Physiol Renal Physiol. 2013;305(8):F1220–7.

    Article  CAS  PubMed  Google Scholar 

  91. Cheruvanky A, Zhou H, Pisitkun T, Kopp JB, Knepper MA, Yuen PS, Star RA. Rapid isolation of urinary exosomal biomarkers using a nanomembrane ultrafiltration concentrator. Am J Physiol Renal Physiol. 2007;292(5):F1657–61.

    Article  CAS  PubMed  Google Scholar 

  92. Gudehithlu KP, Garcia-Gomez I, Vernik J, Brecklin C, Kraus M, Cimbaluk DJ, Hart P, Dunea G, Arruda JA, Singh AK. In diabetic kidney disease urinary exosomes better represent kidney specific protein alterations than whole urine. Am J Nephrol. 2015;42(6):418–24.

    Article  CAS  PubMed  Google Scholar 

  93. Zubiri I, Posada-Ayala M, Sanz-Maroto A, Calvo E, Martin-Lorenzo M, Gonzalez-Calero L, de la Cuesta F, Lopez JA, Fernandez-Fernandez B, Ortiz A, Vivanco F, Alvarez-Llamas G. Diabetic nephropathy induces changes in the proteome of human urinary exosomes as revealed by label-free comparative analysis. J Proteomics. 2014;96:92–102.

    Article  CAS  PubMed  Google Scholar 

  94. Fang DY, King HW, Li JY, Gleadle JM. Exosomes and the kidney: blaming the messenger. Nephrology. 2013;18(1):1–10.

    Article  CAS  PubMed  Google Scholar 

  95. Gudehithlu KP, Hart P, Joshi A, Garcia-Gomez I, Cimbaluk DJ, Dunea G, Arruda JAL, Singh AK. Urine exosomal ceruloplasmin: a potential early biomarker of underlying kidney disease. Clin Exp Nephrol. 2019;23(8):1013–21.

    Article  CAS  PubMed  Google Scholar 

  96. Sakurai A, Ono H, Ochi A, Matsuura M, Yoshimoto S, Kishi S, Murakami T, Tominaga T, Nagai K, Abe H, Doi T. Involvement of Elf3 on Smad3 activation-dependent injuries in podocytes and excretion of urinary exosome in diabetic nephropathy. PLoS ONE. 2019;14(5):e0216788.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Morikawa Y, Takahashi N, Kamiyama K, Nishimori K, Nishikawa Y, Morita S, Kobayashi M, Fukushima S, Yokoi S, Mikami D, Kimura H, Kasuno K, Yashiki T, Naiki H, Hara M, Iwano M. Elevated levels of urinary extracellular vesicle fibroblast-specific protein 1 in patients with active crescentic glomerulonephritis. Nephron. 2019;141(3):177–87.

    Article  CAS  PubMed  Google Scholar 

  98. Zhou H, Pisitkun T, Aponte A, Yuen PS, Hoffert JD, Yasuda H, Hu X, Chawla L, Shen RF, Knepper MA, Star RA. Exosomal Fetuin-A identified by proteomics: a novel urinary biomarker for detecting acute kidney injury. Kidney Int. 2006;70(10):1847–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Panich T, Chancharoenthana W, Somparn P, Issara-Amphorn J, Hirankarn N, Leelahavanichkul A. Urinary exosomal activating transcription factor 3 as the early diagnostic biomarker for sepsis-induced acute kidney injury. BMC Nephrol. 2017;18(1):10.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Zhou H, Cheruvanky A, Hu X, Matsumoto T, Hiramatsu N, Cho ME, Berger A, Leelahavanichkul A, Doi K, Chawla LS, Illei GG, Kopp JB, Balow JE, Austin HA 3rd, Yuen PS, Star RA. Urinary exosomal transcription factors, a new class of biomarkers for renal disease. Kidney Int. 2008;74(5):613–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sonoda H, Yokota-Ikeda N, Oshikawa S, Kanno Y, Yoshinaga K, Uchida K, Ueda Y, Kimiya K, Uezono S, Ueda A, Ito K, Ikeda M. Decreased abundance of urinary exosomal aquaporin-1 in renal ischemia-reperfusion injury. Am J Physiol Renal Physiol. 2009;297(4):F1006–16.

    Article  CAS  PubMed  Google Scholar 

  102. Hogan MC, Bakeberg JL, Gainullin VG, Irazabal MV, Harmon AJ, Lieske JC, Charlesworth MC, Johnson KL, Madden BJ, Zenka RM. Identification of biomarkers for PKD1 using urinary exosomes. J Am Soc Nephrol. 2015;26(7):1661.

    Article  CAS  PubMed  Google Scholar 

  103. Li R, Li C, Geng Le, Tian Xu, Lily W, Zhou H, Zhang B, Sun G, Xuesong Su, Wang Y. Expression of urinary exosomes in patients with idiopathic membranous nephropathy and its clinical significance. Chin J Pract Intern Med. 2020;40(06):487–92.

    CAS  Google Scholar 

  104. Li R-M. Expression of urinary exosomes as well as exosomal proteins Nrf2 and NLRP3 in patients with idiopathic membranous nephropathy and its clinical significance, China Medical University, 2020.

  105. Benito-Martin A, Ucero AC, Zubiri I, Posada-Ayala M, Fernandez-Fernandez B, Cannata-Ortiz P, Sanchez-Nino MD, Ruiz-Ortega M, Egido J, Alvarez-Llamas G, Ortiz A. Osteoprotegerin in exosome-like vesicles from human cultured tubular cells and urine. PLoS ONE. 2013;8(8):e72387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Kalani A, Mohan A, Godbole MM, Bhatia E, Gupta A, Sharma RK, Tiwari S. Wilm’s tumor-1 protein levels in urinary exosomes from diabetic patients with or without proteinuria. PLoS ONE. 2013;8(3):e60177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Raimondo F, Corbetta S, Morosi L, Chinello C, Gianazza E, Castoldi G, Di Gioia C, Bombardi C, Stella A, Battaglia C, Bianchi C, Magni F, Pitto M. Urinary exosomes and diabetic nephropathy: a proteomic approach. Mol BioSyst. 2013;9(6):1139–46.

    Article  CAS  PubMed  Google Scholar 

  108. Rossi L, Nicoletti MC, Carmosino M, Mastrofrancesco L, Di Franco A, Indrio F, Lella R, Laviola L, Giorgino F, Svelto M, Gesualdo L, Procino G. Urinary excretion of kidney aquaporins as possible diagnostic biomarker of diabetic nephropathy. J Diabetes Res. 2017;2017:4360357.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Sun H, Yao W, Tang Y, Zhuang W, Wu D, Huang S, Sheng H. Urinary exosomes as a novel biomarker for evaluation of α-lipoic acid’s protective effect in early diabetic nephropathy. J Clin Lab Anal. 2017;31(6):e22129.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Zubiri I, Posada-Ayala M, Benito-Martin A, Maroto AS, Martin-Lorenzo M, Cannata-Ortiz P, de la Cuesta F, Gonzalez-Calero L, Barderas MG, Fernandez-Fernandez B, Ortiz A, Vivanco F, Alvarez-Llamas G. Kidney tissue proteomics reveals regucalcin downregulation in response to diabetic nephropathy with reflection in urinary exosomes. Transl Res. 2015;166(5):474-484.e4.

    Article  CAS  PubMed  Google Scholar 

  111. Moon PG, Lee JE, You S, Kim TK, Cho JH, Kim IS, Kwon TH, Kim CD, Park SH, Hwang D, Kim YL, Baek MC. Proteomic analysis of urinary exosomes from patients of early IgA nephropathy and thin basement membrane nephropathy. Proteomics. 2011;11(12):2459–75.

    Article  CAS  PubMed  Google Scholar 

  112. Abdeen A, Sonoda H, El-Shawarby R, Takahashi S, Ikeda M. Urinary excretion pattern of exosomal aquaporin-2 in rats that received gentamicin. Am J Physiol Renal Physiol. 2014;307(11):F1227–37.

    Article  CAS  PubMed  Google Scholar 

  113. Alvarez S, Suazo C, Boltansky A, Ursu M, Carvajal D, Innocenti G, Vukusich A, Hurtado M, Villanueva S, Carreño JE, Rogelio A, Irarrazabal CE. Urinary exosomes as a source of kidney dysfunction biomarker in renal transplantation. Transpl Proc. 2013;45(10):3719–23.

    Article  CAS  Google Scholar 

  114. Du J, Li Y, Sun Q, Wang Z, Wang F, Chen F, Wang H, Liu Y, Zhou H, Shang G, Chen X, Ding S, Li C, Wu D, Zhang W, Zhong M. Urinary exosomal CD26 is associated with recovery from acute kidney injury in intensive care units: a prospective cohort study. Clin Chem Lab Med. 2021;59(9):1535–46.

    Article  CAS  PubMed  Google Scholar 

  115. Trnka P, Ivanova L, Hiatt MJ, Matsell DG. Urinary biomarkers in obstructive nephropathy. Clin J Am Soc Nephrol. 2012;7(10):1567–75.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Zhou H, Kajiyama H, Tsuji T, Hu X, Leelahavanichkul A, Vento S, Frank R, Kopp JB, Trachtman H, Star RA, Yuen PS. Urinary exosomal Wilms’ tumor-1 as a potential biomarker for podocyte injury. Am J Physiol Renal Physiol. 2013;305(4):F553–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Lee H, Han KH, Lee SE, Kim SH, Kang HG, Cheong HI. Urinary exosomal WT1 in childhood nephrotic syndrome. Pediatr Nephrol. 2012;27(2):317–20.

    Article  CAS  PubMed  Google Scholar 

  118. Feng Y, Lv LL, Wu WJ, Li ZL, Chen J, Ni HF, Zhou LT, Tang TT, Wang FM, Wang B, Chen PS, Crowley SD, Liu BC. Urinary exosomes and exosomal CCL2 mRNA as biomarkers of active histologic injury in IgA nephropathy. Am J Pathol. 2018;188(11):2542–52.

    Article  CAS  PubMed  Google Scholar 

  119. Lv LL, Cao YH, Pan MM, Liu H, Tang RN, Ma KL, Chen PS, Liu BC. CD2AP mRNA in urinary exosome as biomarker of kidney disease. Clin Chimica Acta. 2014;428:26–31.

    Article  CAS  Google Scholar 

  120. Spanu S, van Roeyen CR, Denecke B, Floege J, Mühlfeld AS. Urinary exosomes: a novel means to noninvasively assess changes in renal gene and protein expression. PLoS ONE. 2014;9(10):e109631.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Koyner JL, Garg AX, Shlipak MG, Patel UD, Sint K, Hong K, Devarajan P, Edelstein CL, Zappitelli M, Thiessen-Philbrook H, Parikh CR. Urinary cystatin C and acute kidney injury after cardiac surgery. Am J Kidney Dis. 2013;61(5):730–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Dieterle F, Perentes E, Cordier A, Roth DR, Verdes P, Grenet O, Pantano S, Moulin P, Wahl D, Mahl A, End P, Staedtler F, Legay F, Carl K, Laurie D, Chibout SD, Vonderscher J, Maurer G. Urinary clusterin, cystatin C, beta2-microglobulin and total protein as markers to detect drug-induced kidney injury. Nat Biotechnol. 2010;28(5):463–9.

    Article  CAS  PubMed  Google Scholar 

  123. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 2014;15(8):509–24.

    Article  CAS  PubMed  Google Scholar 

  124. Fu G, Brkić J, Hayder H, Peng C. MicroRNAs in human placental development and pregnancy complications. Int J Mol Sci. 2013;14(3):5519–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Tüfekci KU, Oner MG, Meuwissen RL, Genç S. The role of microRNAs in human diseases. Methods Mol Biol. 2014;1107:33–50.

    Article  PubMed  Google Scholar 

  126. Paul P, Chakraborty A, Sarkar D, Langthasa M, Rahman M, Bari M, Singha RS, Malakar AK, Chakraborty S. Interplay between miRNAs and human diseases. J Cell Physiol. 2018;233(3):2007–18.

    Article  CAS  PubMed  Google Scholar 

  127. Hayes J, Peruzzi PP, Lawler S. MicroRNAs in cancer: biomarkers, functions and therapy. Trends Mol Med. 2014;20(8):460–9.

    Article  CAS  PubMed  Google Scholar 

  128. Wang J, Chen J, Sen S. MicroRNA as biomarkers and diagnostics. J Cell Physiol. 2016;231(1):25–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Huang W. MicroRNAs: biomarkers, diagnostics, and therapeutics. Methods Mol Biol. 2017;1617:57–67.

    Article  CAS  PubMed  Google Scholar 

  130. Cheng L, Sun X, Scicluna BJ, Coleman BM, Hill AF. Characterization and deep sequencing analysis of exosomal and nonexosomal miRNA in human urine. Kidney Int. 2014;86(2):433–44.

    Article  CAS  PubMed  Google Scholar 

  131. Min QH, Chen XM, Zou YQ, Zhang J, Li J, Wang Y, Li SQ, Gao QF, Sun F, Liu J, Xu YM, Lin J, Huang LF, Huang B, Wang XZ. Differential expression of urinary exosomal microRNAs in IgA nephropathy. J Clin Lab Anal. 2018;32(2):e22226.

    Article  PubMed  Google Scholar 

  132. Kumari M, Mohan A, Ecelbarger CM, Gupta A, Prasad N, Tiwari S. miR-451 loaded exosomes are released by the renal cells in response to injury and associated with reduced kidney function in human. Front Physiol. 2020;11:234.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Zang J, Maxwell AP, Simpson DA, McKay GJ. Differential expression of urinary exosomal MicroRNAs miR-21-5p and miR-30b-5p in individuals with diabetic kidney disease. Sci Rep. 2019;9(1):10900.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Eissa S, Matboli M, Aboushahba R, Bekhet MM, Soliman Y. Urinary exosomal microRNA panel unravels novel biomarkers for diagnosis of type 2 diabetic kidney disease. J Diabetes Complicat. 2016;30(8):1585–92.

    Article  Google Scholar 

  135. Solé C, Moliné T, Vidal M, Ordi-Ros J, Cortés-Hernández J. An exosomal urinary miRNA signature for early diagnosis of renal fibrosis in lupus nephritis. Cells. 2019;8(8):773.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Ma H, Xu Y, Zhang R, Guo B, Zhang S, Zhang X. Differential expression study of circular RNAs in exosomes from serum and urine in patients with idiopathic membranous nephropathy. Arch Med Sci. 2019;15(3):738–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Sonoda H, Lee BR, Park KH, Nihalani D, Yoon JH, Ikeda M, Kwon SH. miRNA profiling of urinary exosomes to assess the progression of acute kidney injury. Sci Rep. 2019;9(1):4692.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Lange T, Artelt N, Kindt F, Stracke S, Rettig R, Lendeckel U, Chadjichristos CE, Kavvadas P, Chatziantoniou C, Endlich K, Endlich N. MiR-21 is upregulated in urinary exosomes of chronic kidney disease patients and after glomerular injury. J Cell Mol Med. 2019;23(7):4839–43.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Xie Y, Jia Y, Cuihua X, Hu F, Xue M, Xue Y. Urinary exosomal MicroRNA profiling in incipient type 2 diabetic kidney disease. J Diabetes Res. 2017;2017:6978984.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Prabu P, Rome S, Sathishkumar C, Gastebois C, Meugnier E, Mohan V, Balasubramanyam M. MicroRNAs from urinary extracellular vesicles are noninvasive early biomarkers of diabetic nephropathy in type 2 diabetes patients with the “Asian Indian phenotype.” Diabetes Metab. 2019;45(3):276–85.

    Article  CAS  PubMed  Google Scholar 

  141. Li W, Yang S, Qiao R, Zhang J. Potential value of urinary exosome-derived let-7c-5p in the diagnosis and progression of type II diabetic nephropathy. Clin Lab. 2018;64(5):709–18.

    CAS  PubMed  Google Scholar 

  142. Mohan A, Singh RS, Kumari M, Garg D, Upadhyay A, Ecelbarger CM, Tripathy S, Tiwari S. Urinary exosomal microRNA-451-5p is a potential early biomarker of diabetic nephropathy in rats. PLoS ONE. 2016;11(4):e0154055.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Lv LL, Feng Y, Wu M, Wang B, Li ZL, Zhong X, Wu WJ, Chen J, Ni HF, Tang TT, Tang RN, Lan HY, Liu BC. Exosomal miRNA-19b-3p of tubular epithelial cells promotes M1 macrophage activation in kidney injury. Cell Death Differ. 2020;27(1):210–26.

    Article  CAS  PubMed  Google Scholar 

  144. Lee WC, Li LC, Ng HY, Lin PT, Chiou TT, Kuo WH, Lee CT. Urinary exosomal MicroRNA signatures in nephrotic, biopsy-proven diabetic nephropathy. J Clin Med. 2020;9(4):1220.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Wu J, Zheng C, Fan Y, Zeng C, Chen Z, Qin W, Zhang C, Zhang W, Wang X, Zhu X, Zhang M, Zen K, Liu Z. Downregulation of microRNA-30 facilitates podocyte injury and is prevented by glucocorticoids. J Am Soc Nephrol. 2014;25(1):92–104.

    Article  CAS  PubMed  Google Scholar 

  146. Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, Bruno G, Cimino D, Taverna D, Deregibus MC, Rastaldi MP, Perin PC, Gruden G. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS ONE. 2013;8(11):e73798.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Delić D, Eisele C, Schmid R, Baum P, Wiech F, Gerl M, Zimdahl H, Pullen SS, Urquhart R. Urinary exosomal miRNA signature in type II diabetic nephropathy patients. PLoS ONE. 2016;11(3):e0150154.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Güllülü M, Tuncel E, Peynirci H, Alemdar A, Tunca B, Egeli Ü, Çeçener G, Bayindir M, Cosgun G. Biomarker potential of urine miR-451 at different stages of diabetic nephropathy. J Diabetes Metab. 2016. https://doi.org/10.4172/2155-6156.1000650.

    Article  Google Scholar 

  149. Gong D, Chen X, Middleditch M, Huang L, Vazhoor Amarsingh G, Reddy S, Lu J, Zhang S, Ruggiero K, Phillips AR, Cooper GJ. Quantitative proteomic profiling identifies new renal targets of copper(II)-selective chelation in the reversal of diabetic nephropathy in rats. Proteomics. 2009;9(18):4309–20.

    Article  CAS  PubMed  Google Scholar 

  150. Tangtanatakul P, Klinchanhom S, Sodsai P, Sutichet T, Promjeen C, Avihingsanon Y, Hirankarn N. Downregulation of let-7a and miR-21 in urine exosomes from lupus nephritis patients during disease flare. Asian Pac J Allergy Immunol. 2019;37(4):189–97.

    CAS  PubMed  Google Scholar 

  151. Solé C, Cortés-Hernández J, Felip ML, Vidal M, Ordi-Ros J. miR-29c in urinary exosomes as predictor of early renal fibrosis in lupus nephritis. Nephrol Dial Transpl. 2015;30(9):1488–96.

    Article  Google Scholar 

  152. Garcia-Vives E, Solé C, Moliné T, Vidal M, Agraz I, Ordi-Ros J, Cortés-Hernández J. The urinary exosomal miRNA expression profile is predictive of clinical response in lupus nephritis. Int J Mol Sci. 2020;21(4):1372.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Perez-Hernandez J, Forner MJ, Pinto C, Chaves FJ, Cortes R, Redon J. Increased urinary exosomal MicroRNAs in patients with systemic lupus erythematosus. PLoS ONE. 2015;10(9):e0138618.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Li Y, Xu X, Tang X, Bian X, Shen B, Zhao H, Luo S, Chen Z, Zhang K. MicroRNA expression profile of urinary exosomes in Type IV lupus nephritis complicated by cellular crescent. J Biol Res. 2018;25:16.

    Google Scholar 

  155. Huang Z, Zhang Y, Zhou J, Zhang Y. Urinary exosomal miR-193a can be a potential biomarker for the diagnosis of primary focal segmental glomerulosclerosis in children. Biomed Res Int. 2017;2017:7298160. https://doi.org/10.1155/2017/7298160.

    PubMed  PubMed Central  Google Scholar 

  156. Awdishu L, Le A, Amato J, Jani V, Bal S, Mills RH, Carrillo-Terrazas M, Gonzalez DJ, Tolwani A, Acharya A, Cerda J, Joy MS, Nicoletti P, Macedo E, Vaingankar S, Mehta R, RamachandraRao SP, I. On Behalf Of The Direct. Urinary exosomes identify inflammatory pathways in vancomycin associated acute kidney injury. Int J Mol Sci. 2021;22(6):2784.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Chun-Yan L, Zi-Yi Z, Tian-Lin Y, Yi-Li W, Bao L, Jiao L, Wei-Jun D. Liquid biopsy biomarkers of renal interstitial fibrosis based on urinary exosome. Exp Mol Pathol. 2018;105(2):223–8.

    Article  CAS  PubMed  Google Scholar 

  158. Yun CY, Lim JH, Oh JH, Cho AY, Lee KY, Sun IO. Urinary exosomal microRNA-21 as a marker for scrub typhus-associated acute kidney injury. Genet Test Mol Biomarkers. 2021;25(2):140–4.

    Article  CAS  PubMed  Google Scholar 

  159. Shimasaki T, Yamamoto S, Arisawa T. Exosome research and coculture study. Biol Pharm Bull. 2018;41(9):1311–21.

    Article  CAS  PubMed  Google Scholar 

  160. Johnstone RM, Mathew A, Mason AB, Teng K. Exosome formation during maturation of mammalian and avian reticulocytes: evidence that exosome release is a major route for externalization of obsolete membrane proteins. J Cell Physiol. 1991;147(1):27–36.

    Article  CAS  PubMed  Google Scholar 

  161. Keller S, Rupp C, Stoeck A, Runz S, Fogel M, Lugert S, Hager HD, Abdel-Bakky MS, Gutwein P, Altevogt P. CD24 is a marker of exosomes secreted into urine and amniotic fluid. Kidney Int. 2007;72(9):1095–102.

    Article  CAS  PubMed  Google Scholar 

  162. Pisitkun T, Johnstone R, Knepper MA. Discovery of urinary biomarkers. Mol Cell Proteomics. 2006;5(10):1760–71.

    Article  CAS  PubMed  Google Scholar 

  163. Zhang J, Zhu Y, Cai R, Jin J, He Q. Differential expression of urinary exosomal small RNAs in idiopathic membranous nephropathy. Biomed Res Int. 2020;2020:3170927.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Guo S, Hao H, Li S, Zhang L, Li R. Differential expression of urinary exosomal miRNA in idiopathic membranous nephropathy and evaluation of its diagnostic value. Tohoku J Exp Med. 2022;256(4):327–36.

    Article  CAS  PubMed  Google Scholar 

  165. Fernando MM, Vyse TJ. Risk alleles in idiopathic membranous nephropathy. N Engl J Med. 2011;364(21):2072.

    Article  CAS  PubMed  Google Scholar 

  166. Wang Y, Liu Y, Zhang L, Bai L, Chen S, Wu H, Sun L, Wang X. miR-30b-5p modulate renal epithelial–mesenchymal transition in diabetic nephropathy by directly targeting SNAI1. Biochem Biophys Res Commun. 2021;535:12–8.

    Article  CAS  PubMed  Google Scholar 

  167. Fierro-Fernández M, Miguel V, Márquez-Expósito L, Nuevo-Tapioles C, Herrero JI, Blanco-Ruiz E, Tituaña J, Castillo C, Cannata P, Monsalve M, Ruiz-Ortega M, Ramos R, Lamas S. MiR-9-5p protects from kidney fibrosis by metabolic reprogramming. FASEB J. 2020;34(1):410–31.

    Article  PubMed  Google Scholar 

  168. Li J, Zheng S, Ma C, Chen X, Li X, Li S, Wang P, Chen P, Wang Z, Li W, Liu Y. Research progress on exosomes in podocyte injury associated with diabetic kidney disease. Front Endocrinol. 2023;14:1129884.

    Article  Google Scholar 

  169. He P, Liu D, Zhang B, Zhou G, Su X, Wang Y, Li D, Yang X. Hepatitis B virus X protein reduces podocyte adhesion via downregulation of α3β1 integrin. Cell Physiol Biochem. 2017;41(2):689–700.

    Article  CAS  PubMed  Google Scholar 

  170. Lang Y, Zhao Y, Zheng C, Lu Y, Wu J, Zhu X, Zhang M, Yang F, Xu X, Shi S, Liu Z. MiR-30 family prevents uPAR-ITGB3 signaling activation through calcineurin-NFATC pathway to protect podocytes. Cell Death Dis. 2019;10(6):401.

    Article  PubMed  PubMed Central  Google Scholar 

  171. Li F, Dai B, Ni X. Long noncoding RNA cancer susceptibility candidate 2 (CASC2) alleviates the high glucose-induced injury of CIHP-1 cells by regulating miR-9-5p/PPARγ axis in diabetes nephropathy. Diabetol Metab Syndr. 2020;12:68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Barbagallo C, Passanisi R, Mirabella F, Cirnigliaro M, Costanzo A, Lauretta G, Barbagallo D, Bianchi C, Pagni F, Castorina S, Granata A, Di Pietro C, Ragusa M, Malatino LS, Purrello M. Upregulated microRNAs in membranous glomerulonephropathy are associated with significant downregulation of IL6 and MYC mRNAs. J Cell Physiol. 2019;234(8):12625–36.

    Article  CAS  PubMed  Google Scholar 

  173. Li Z, Yin H, Hao S, Wang L, Gao J, Tan X, Yang Z. miR-200 family promotes podocyte differentiation through repression of RSAD2. Sci Rep. 2016;6:27105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Huang X, Hou X, Chuan L, Wei S, Wang J, Yang X, Ru J. miR-129-5p alleviates LPS-induced acute kidney injury by targeting HMGB1/TLRs/NF-kappaB pathway. Int Immunopharmacol. 2020;89(Pt A):107016.

    Article  CAS  PubMed  Google Scholar 

  175. Huang H, Liu H, Tang J, Xu W, Gan H, Fan Q, Zhang W. M2 macrophage-derived exosomal miR-25-3p improves high glucose-induced podocytes injury through activation autophagy by inhibiting DUSP1 expression. IUBMB Life. 2020;72(12):2651–62.

    Article  CAS  PubMed  Google Scholar 

  176. Ding XQ, Gu TT, Wang W, Song L, Chen TY, Xue QC, Zhou F, Li JM, Kong LD. Curcumin protects against fructose-induced podocyte insulin signaling impairment through upregulation of miR-206. Mol Nutr Food Res. 2015;59(12):2355–70.

    Article  CAS  PubMed  Google Scholar 

  177. Guo N, Guo J, Su D. MicroRNA-206 and its downregulation of Wilms’Tumor-1 dictate podocyte health in adriamycin-induced nephropathy. Ren Fail. 2016;38(6):989–95.

    Article  CAS  PubMed  Google Scholar 

  178. Ardalan M, Hosseiniyan Khatibi SM, Rahbar Saadat Y, Bastami M, Nariman-Saleh-Fam Z, Abediazar S, Khalilov R, Zununi Vahed S. Migrasomes and exosomes; different types of messaging vesicles in podocytes. Cell Biol Int. 2022;46(1):52–62.

    Article  CAS  PubMed  Google Scholar 

  179. Debiec H, Guigonis V, Mougenot B, Decobert F, Haymann JP, Bensman A, Deschênes G, Ronco PM. Antenatal membranous glomerulonephritis due to anti-neutral endopeptidase antibodies. N Engl J Med. 2002;346(26):2053–60.

    Article  PubMed  Google Scholar 

  180. Geginat J, Paroni M, Maglie S, Alfen JS, Kastirr I, Gruarin P, De Simone M, Pagani M, Abrignani S. Plasticity of human CD4 T-cell subsets. Front Immunol. 2014;5:630.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Li H, Wu H, Guo Q, Yu H, Xu Y, Yu J, Wang Z, Yi H. Myeloid-derived suppressor cells promote the progression of primary membranous nephropathy by enhancing Th17 response. Front Immunol. 2020;11:1777.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Cremoni M, Brglez V, Perez S, Decoupigny F, Zorzi K, Andreani M, Gérard A, Boyer-Suavet S, Ruetsch C, Benzaken S, Esnault V, Seitz-Polski B. Th17-immune response in patients with membranous nephropathy is associated with thrombosis and relapses. Front Immunol. 2020;11:574997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Motavalli R, Etemadi J, Soltani-Zangbar MS, Ardalan MR, Kahroba H, Roshangar L, Nouri M, Aghebati-Maleki L, Khiavi FM, Abediazar S, Mehdizadeh A, Hojjat-Farsangi M, Mahmoodpoor A, Kafil HS, Zolfaghari M, Ahmadian Heris J, Yousefi M. Altered Th17/Treg ratio as a possible mechanism in pathogenesis of idiopathic membranous nephropathy. Cytokine. 2021;141:155452.

    Article  CAS  PubMed  Google Scholar 

  184. Jiang W, Zheng L, Yan Q, Chen L, Wang X. MiR-532–3p inhibits metastasis and proliferation of non-small cell lung cancer by targeting FOXP3. J BUON. 2019;24(6):2287–93.

    PubMed  Google Scholar 

  185. Majd M, Hosseini A, Ghaedi K, Kiani-Esfahani A, Tanhaei S, Shiralian-Esfahani H, Rahnamaee SY, Mowla SJ, Nasr-Esfahani MH. MiR-9-5p and miR-106a-5p dysregulated in CD4(+) T cells of multiple sclerosis patients and targeted essential factors of T helper17/regulatory T cells differentiation. Iran J Basic Med Sci. 2018;21(3):277–83.

    PubMed  PubMed Central  Google Scholar 

  186. Chen L, Ma H, Hu H, Gao L, Wang X, Ma J, Gao Q, Liu B, Zhou G, Liang C. Special role of Foxp3 for the specifically altered microRNAs in regulatory T cells of HCC patients. BMC Cancer. 2014;14:489.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Zheng J, Zeng M, Nian JB, Zeng LY, Fu Z, Huang QJ, Wei X. The CXCR4/miR-125b/FoxP3 axis regulates the function of the epithelial barrier via autophagy in allergic rhinitis. Am J Transl Res. 2020;12(6):2570–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Holla S, Stephen-Victor E, Prakhar P, Sharma M, Saha C, Udupa V, Kaveri SV, Bayry J, Balaji KN. Mycobacteria-responsive sonic hedgehog signaling mediates programmed death-ligand 1- and prostaglandin E2-induced regulatory T-cell expansion. Sci Rep. 2016;6:24193.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. KDIGO. Clinical practice guideline for the management of glomerular diseases. Kidney Int. 2021;100(4s):S1-s276.

    Google Scholar 

  190. Fervenza FC, Appel GB, Barbour SJ, Rovin BH, Lafayette RA, Aslam N, Jefferson JA, Gipson PE, Rizk DV, Sedor JR, Simon JF, McCarthy ET, Brenchley P, Sethi S, Avila-Casado C, Beanlands H, Lieske JC, Philibert D, Li T, Thomas LF, Green DF, Juncos LA, Beara-Lasic L, Blumenthal SS, Sussman AN, Erickson SB, Hladunewich M, Canetta PA, Hebert LA, Leung N, Radhakrishnan J, Reich HN, Parikh SV, Gipson DS, Lee DK, da Costa BR, Jüni P, Cattran DC. Rituximab or cyclosporine in the treatment of membranous nephropathy. N Engl J Med. 2019;381(1):36–46.

    Article  CAS  PubMed  Google Scholar 

  191. Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med. 1998;4(5):594–600.

    Article  CAS  PubMed  Google Scholar 

  192. Escudier B, Dorval T, Chaput N, André F, Caby MP, Novault S, Flament C, Leboulaire C, Borg C, Amigorena S, Boccaccio C, Bonnerot C, Dhellin O, Movassagh M, Piperno S, Robert C, Serra V, Valente N, Le Pecq JB, Spatz A, Lantz O, Tursz T, Angevin E, Zitvogel L. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. J Transl Med. 2005;3(1):10.

    Article  PubMed  PubMed Central  Google Scholar 

  193. Morse MA, Garst J, Osada T, Khan S, Hobeika A, Clay TM, Valente N, Shreeniwas R, Sutton MA, Delcayre A, Hsu DH, Le Pecq JB, Lyerly HK. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. J Transl Med. 2005;3(1):9.

    Article  PubMed  PubMed Central  Google Scholar 

  194. Newton WC, Kim JW, Luo JZQ, Luo L. Stem cell-derived exosomes: a novel vector for tissue repair and diabetic therapy. J Mol Endocrinol. 2017;59(4):R155-r165.

    Article  CAS  PubMed  Google Scholar 

  195. Heldring N, Mäger I, Wood MJ, Le Blanc K, Andaloussi SE. Therapeutic potential of multipotent mesenchymal stromal cells and their extracellular vesicles. Hum Gene Ther. 2015;26(8):506–17.

    Article  CAS  PubMed  Google Scholar 

  196. Le Blanc K, Frassoni F, Ball L, Locatelli F, Roelofs H, Lewis I, Lanino E, Sundberg B, Bernardo ME, Remberger M, Dini G, Egeler RM, Bacigalupo A, Fibbe W, Ringdén O. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet. 2008;371(9624):1579–86.

    Article  PubMed  Google Scholar 

  197. Wang Q, Zhuang X, Mu J, Deng ZB, Jiang H, Zhang L, Xiang X, Wang B, Yan J, Miller D, Zhang HG. Delivery of therapeutic agents by nanoparticles made of grapefruit-derived lipids. Nat Commun. 2013;4:1867.

    Article  PubMed  Google Scholar 

  198. Sun D, Zhuang X, Xiang X, Liu Y, Zhang S, Liu C, Barnes S, Grizzle W, Miller D, Zhang HG. A novel nanoparticle drug delivery system: the anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther. 2010;18(9):1606–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Grange C, Tritta S, Tapparo M, Cedrino M, Tetta C, Camussi G, Brizzi MF. Stem cell-derived extracellular vesicles inhibit and revert fibrosis progression in a mouse model of diabetic nephropathy. Sci Rep. 2019;9(1):4468.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Lindoso RS, Lopes JA, Binato R, Abdelhay E, Takiya CM, Miranda KR, Lara LS, Viola A, Bussolati B, Vieyra A, Collino F. Adipose mesenchymal cells-derived EVs alleviate DOCA-salt-induced hypertension by promoting cardio-renal protection. Mol Ther Methods Clin Dev. 2020;16:63–77.

    Article  CAS  PubMed  Google Scholar 

  201. Collino F, Lopes JA, Tapparo M, Tortelote GG, Kasai-Brunswick TH, Lopes GMC, Almeida DB, Skovronova R, Wendt CHC, Miranda KR, Bussolati B, Vieyra A, Lindoso RS. Extracellular vesicles derived from induced pluripotent stem cells promote renoprotection in acute kidney injury model. Cells. 2020;9(2):453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Liu C, Wang J, Hu J, Fu B, Mao Z, Zhang H, Cai G, Chen X, Sun X. Extracellular vesicles for acute kidney injury in preclinical rodent models: a meta-analysis. Stem Cell Res Ther. 2020;11(1):11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Bai L, Li J, Li H, Song J, Zhou Y, Lu R, Liu B, Pang Y, Zhang P, Chen J, Liu X, Wu J, Liang C, Zhou J. Renoprotective effects of artemisinin and hydroxychloroquine combination therapy on IgA nephropathy by suppressing NF-κB signaling and NLRP3 inflammasome activation by exosomes in rats. Biochem Pharmacol. 2019;169:113619.

    Article  CAS  PubMed  Google Scholar 

  204. Matsukura T, Inaba C, Weygant EA, Kitamura D, Janknecht R, Matsumoto H, Hyink DP, Kashiwada S, Obara T. Extracellular vesicles from human bone marrow mesenchymal stem cells repair organ damage caused by cadmium poisoning in a medaka model. Physiol Rep. 2019;7(14):e14172.

    Article  PubMed  PubMed Central  Google Scholar 

  205. Zhang A, Wang H, Wang B, Yuan Y, Klein JD, Wang XH. Exogenous miR-26a suppresses muscle wasting and renal fibrosis in obstructive kidney disease. FASEB J. 2019;33(12):13590–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Zhu G, Pei L, Lin F, Yin H, Li X, He W, Liu N, Gou X. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury by transferring miR-199a-3p. J Cell Physiol. 2019;234(12):23736–49.

    Article  CAS  PubMed  Google Scholar 

  207. Nassar W, El-Ansary M, Sabry D, Mostafa MA, Fayad T, Kotb E, Temraz M, Saad AN, Essa W, Adel H. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomater Res. 2016;20:21.

    Article  PubMed  PubMed Central  Google Scholar 

  208. Wang B, Wang J, He W, Zhao Y, Zhang A, Liu Y, Hassounah F, Ma F, Klein JD, Wang XH, Wang H. Exogenous miR-29a attenuates muscle atrophy and kidney fibrosis in unilateral ureteral obstruction mice. Hum Gene Ther. 2020;31(5–6):367–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Wang B, Zhang A, Wang H, Klein JD, Tan L, Wang ZM, Du J, Naqvi N, Liu BC, Wang XH. miR-26a limits muscle wasting and cardiac fibrosis through exosome-mediated microRNA transfer in chronic kidney disease. Theranostics. 2019;9(7):1864–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Février B, Raposo G. Exosomes: endosomal-derived vesicles shipping extracellular messages. Curr Opin Cell Biol. 2004;16(4):415–21.

    Article  PubMed  Google Scholar 

  211. Ranghino A, Dimuccio V, Papadimitriou E, Bussolati B. Extracellular vesicles in the urine: markers and mediators of tissue damage and regeneration. Clin Kidney J. 2015;8(1):23–30.

    Article  CAS  PubMed  Google Scholar 

  212. Nikfarjam S, Rezaie J, Zolbanin NM, Jafari R. Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine. J Transl Med. 2020;18(1):449.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Bochon B, Kozubska M, Surygała G, Witkowska A, Kuźniewicz R, Grzeszczak W, Wystrychowski G. Mesenchymal stem cells-potential applications in kidney diseases. Int J Mol Sci. 2019;20(10):2462.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Wu L, Tian X, Zuo H, Zheng W, Li X, Yuan M, Tian X, Song H. miR-124-3p delivered by exosomes from heme oxygenase-1 modified bone marrow mesenchymal stem cells inhibits ferroptosis to attenuate ischemia-reperfusion injury in steatotic grafts. J Nanobiotechnol. 2022;20(1):196.

    Article  CAS  Google Scholar 

  215. Bruno S, Grange C, Collino F, Deregibus MC, Cantaluppi V, Biancone L, Tetta C, Camussi G. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS ONE. 2012;7(3):e33115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, Morando L, Busca A, Falda M, Bussolati B, Tetta C, Camussi G. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J Am Soc Nephrol. 2009;20(5):1053–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Kunter U, Rong S, Djuric Z, Boor P, Müller-Newen G, Yu D, Floege J. Transplanted mesenchymal stem cells accelerate glomerular healing in experimental glomerulonephritis. J Am Soc Nephrol. 2006;17(8):2202–12.

    Article  CAS  PubMed  Google Scholar 

  218. Gatti S, Bruno S, Deregibus MC, Sordi A, Cantaluppi V, Tetta C, Camussi G. Microvesicles derived from human adult mesenchymal stem cells protect against ischemia-reperfusion-induced acute and chronic kidney injury. Nephrol Dial Transpl. 2011;26(5):1474–83.

    Article  CAS  Google Scholar 

  219. Biancone L, Bruno S, Deregibus MC, Tetta C, Camussi G. Therapeutic potential of mesenchymal stem cell-derived microvesicles. Nephrol Dial Transpl. 2012;27(8):3037–42.

    Article  CAS  Google Scholar 

  220. Mao R, Shen J, Hu X. BMSCs-derived exosomal microRNA-let-7a plays a protective role in diabetic nephropathy via inhibition of USP22 expression. Life Sci. 2021;268:118937.

    Article  CAS  PubMed  Google Scholar 

  221. Jiang ZZ, Liu YM, Niu X, Yin JY, Hu B, Guo SC, Fan Y, Wang Y, Wang NS. Exosomes secreted by human urine-derived stem cells could prevent kidney complications from type I diabetes in rats. Stem Cell Res Ther. 2016;7:24.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Lang R, Liu G, Shi Y, Bharadwaj S, Leng X, Zhou X, Liu H, Atala A, Zhang Y. Self-renewal and differentiation capacity of urine-derived stem cells after urine preservation for 24 hours. PLoS ONE. 2013;8(1):e53980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Borges FT, Melo SA, Özdemir BC, Kato N, Revuelta I, Miller CA, Gattone VH 2nd, LeBleu VS, Kalluri R. TGF-β1-containing exosomes from injured epithelial cells activate fibroblasts to initiate tissue regenerative responses and fibrosis. J Am Soc Nephrol. 2013;24(3):385–92.

    Article  CAS  PubMed  Google Scholar 

  224. Batrakova EV, Kim MS. Using exosomes, naturally equipped nanocarriers, for drug delivery. J Control Release. 2015;219:396–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Duan L, Xu L, Xu X, Qin Z, Zhou X, Xiao Y, Liang Y, Xia J. Exosome-mediated delivery of gene vectors for gene therapy. Nanoscale. 2021;13(3):1387–97.

    Article  CAS  PubMed  Google Scholar 

  226. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341–5.

    Article  CAS  PubMed  Google Scholar 

  227. Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, Ju S, Mu J, Zhang L, Steinman L, Miller D, Zhang HG. Treatment of brain inflammatory diseases by delivering exosome encapsulated anti-inflammatory drugs from the nasal region to the brain. Mol Ther. 2011;19(10):1769–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Lee JY, Kim HS. Extracellular vesicles in neurodegenerative diseases: a double-edged sword. Tissue Eng Regener Med. 2017;14(6):667–78.

    Article  CAS  Google Scholar 

  229. Ma YS, Yang XL, Xin R, Liu JB, Fu D. Power and promise of exosomes as clinical biomarkers and therapeutic vectors for liquid biopsy and cancer control. Biochim Biophys Acta. 2021;1875(1):188497.

    CAS  Google Scholar 

  230. Zamay TN, Zamay GS, Shnayder NA, Dmitrenko DV, Zamay SS, Yushchenko V, Kolovskaya OS, Susevski V, Berezovski MV, Kichkailo AS. Nucleic acid aptamers for molecular therapy of epilepsy and blood-brain barrier damages. Mol Ther Nucleic Acids. 2020;19:157–67.

    Article  CAS  PubMed  Google Scholar 

  231. May J-N, Golombek SK, Baues M, Dasgupta A, Drude N, Rix A, Rommel D, Von Stillfried S, Appold L, Pola R. Multimodal and multiscale optical imaging of nanomedicine delivery across the blood-brain barrier upon sonopermeation. Theranostics. 2020;10(4):1948.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Li X, Corbett AL, Taatizadeh E, Tasnim N, Little JP, Garnis C, Daugaard M, Guns E, Hoorfar M, Li ITS. Challenges and opportunities in exosome research-Perspectives from biology, engineering, and cancer therapy. APL Bioeng. 2019;3(1):011503.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Yang T, Martin P, Fogarty B, Brown A, Schurman K, Phipps R, Yin VP, Lockman P, Bai S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm Res. 2015;32(6):2003–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Bai C, Gao S, Hu S, Liu X, Li H, Dong J, Huang A, Zhu L, Zhou P, Li S, Shao N. Self-assembled multivalent aptamer nanoparticles with potential CAR-like characteristics could activate T cells and inhibit melanoma growth. Mol Ther Oncolytics. 2020;17:9–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Qiao L, Hu S, Liu S, Zhang H, Ma H, Huang K, Li Z, Su T, Vandergriff A, Tang J, Allen T, Dinh PU, Cores J, Yin Q, Li Y, Cheng K. microRNA-21-5p dysregulation in exosomes derived from heart failure patients impairs regenerative potential. J Clin Investig. 2019;129(6):2237–50.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (No. 2019YFC1709401, 82372552), the Tianjin Municipal Health Commission (No. 2021012), the Guangdong Basic and Applied Basic Research Foundation (No. 2023A1515111044), the Shenzhen Science and Technology Program (grant no. RCBS20231211090733052; RCBS20231211090701008), the Excellent Youth of Natural Science Research Projects in Anhui Province Universities (2023AH030060), the Research Fund of Anhui Institute of Translational Medicine (2022zhyx-C01), the Basic and Clinical Cooperative Research and Promotion Program of Anhui Medical University (2021xkT028), and the Postdoctoral Fellowship Program of CPSF (No. GZC20233233). All the authors contributed to the article and approved the submitted version.

Author information

Authors and Affiliations

Authors

Contributions

LW, JW, KY: writing, review and editing and designed the manuscript. LW, TS, MP, AX and XX: modified the tables and figures and revised the manuscript. LF: writing, review and editing. PY: writing, review, editing, and supervision. HY and XW: writing-review & editing, supervision, resources, conceptualization.

Corresponding authors

Correspondence to Lingyan Fei, Yihang Pan, Hongtao Yang or Xianwen Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare that there are no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, L., Wang, J., Xu, A. et al. Future embracing: exosomes driving a revolutionary approach to the diagnosis and treatment of idiopathic membranous nephropathy. J Nanobiotechnol 22, 472 (2024). https://doi.org/10.1186/s12951-024-02633-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-024-02633-y

Keywords