Open access peer-reviewed chapter

Extracellular Vesicles in Kidney Disease

Written By

Chunyan Lv

Submitted: 18 July 2023 Reviewed: 13 September 2023 Published: 03 November 2023

DOI: 10.5772/intechopen.113200

From the Edited Volume

Extracellular Vesicles - Applications and Therapeutic Potential

Edited by Manash K. Paul

Chapter metrics overview

59 Chapter Downloads

View Full Metrics

Abstract

The kidney is the mainly apparatus in the human body, with a complex organizational structure and diverse pathological changes closely related to other organs. Extracellular vesicles are vesicles with diameters ranging from tens of nanometers to several micrometers, originating from multiple intracellular vesicles or local cell membranes. They carry various information from the source cells and operate between various cells in the kidney and extrarenal organs, conveying substances between cells. They play a large part in signal transmission within the kidney and between the kidney and other organs. Detecting changes in extracellular vesicles and their cargo can monitor both renal neoplastic and nonneoplastic diseases. Extracellular vesicles derived from various stem cells, loaded with bioactive substances, can be applied to some extent to treat kidney diseases. Bioengineering drugs using extracellular vesicles as carriers are also playing an increasingly big role in treating kidney diseases. Research on extracellular vesicles has achieved certain results and has some preclinical applications, but there is still a process for large-scale and widespread application.

Keywords

  • kidney disease
  • extracellular vesicles
  • cell-to-cell communication
  • biomarker
  • therapeutic potential

1. Introduction

Extracellular vesicles (EVs) are a heterogeneous population of bilayered lipid vesicles [1, 2, 3] regarded as a vital interactive courier between cells and secreted by most types of cells. EVs can be differentiated into three main categories according to their origin and size: exosomes, local microdomains assembled in endocytic membranes with the size of 30–100 nm; microvesicles, released from the plasma membrane with a size of 100 nm-1 μm; apoptotic bodies, released by cells undergoing apoptosis with a size of 1 μm–5 μm [4, 5]. The giant tumor-derived vesicles (oncosomes) and mitochondrial-originating vesicles (mitovesicles) are also considered as EVs [6, 7].

Despite the heterogeneous and diverse nature of EVs, they are released by normal cells and move in endless cycles in humor, with a major role in numerous physiological and pathological conditions. EVs are abundant in body fluids and are easy to separate and enrich, with complicated cargo reflecting the physiological and pathological condition of the source cells. EVs can be efficient transport through the cell membrane of target cells, thus enabling communication between cells and modulating the gene expression and function of target cells [8, 9].

The kidney is one of the important organs to remain steady in the internal milieu. EVs feature in renal physiology and are widely involved in the occurrence and progression of various kidney diseases. It can be used for the diagnosis of kidney diseases and be instrumental in the treatment of renal diseases [10, 11].

Advertisement

2. Role of extracellular vesicles in cell-cell communication in kidney

As the characteristics mentioned above, EVs act major affection in nephron intercellular communication [12]. Proteomics results show that the exosomes in nephron mainly come from glomerular podocyte and tubular cells located in the proximal tubules, the thick segments of ascending medullary branches, the distal tubules, and collecting ducts. Some studies have confirmed that EVs transmit information in the nephron between the glomerulus and renal tubules. The upper tubule cells can release EVs, which are internalized by the cells in the lower segment of the tubule, transmitting activated cell molecules. The earliest research found that EVs could be expelled and internalized by murine renal collecting duct cells in cultivation and transfer the functional aquaporin 2 [13]. Further research has found that the EVs from proximal renal tubules can be absorbed by the distal tubules and collecting ducts [14]. A study on podocyte RNA labeling found that this RNA was subsequently detected in renal tubular cells, thus confirming the material transfer between glomerulus and renal tubular cells [15].

Not only that, the maintenance of electrolyte and acid-base equipoise is one of the crux functions of the kidney, which is fulfilled by tubular transport. A variety of proteins and transporters rich in urinary exosomes come from cells in the nephron, for example, Na+-Cl symporter (distal tubule), aquaporin (AQP)-2 protein (concentrated pipe), Na+-K+-2Cl symporter (thick ascending limb), AQP-1 (proximal tubule), podoglycocalyx protein (podocyte). The urinary extracellular vesicles (uEVs) produced at the top membrane of kidney tubular cells, cargo water, electrolyte, and acid-base transporters. When the proximal tubular cell line is affected by inflammation, they excrete more EVs from apical and basal membrane, both of which have different molecular and functional characteristics [16]. It has proved that miRNAs acted as a major role in EV-mediated intercellular communication. Jella et al. recorded by unipath patch clamp that the probability of ENaC opening Xenopus cells and isolated splitopen tubules could be reduced by proximal EVs, and this could be weakened by EVs transfected with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) inhibitors [17]. Another research confirmed that ENaC activity could be regulated by EVs through purinergic signaling [18].

The distant tissues can release EVs into circulation and mediate inter-organ crosstalk. EVs released from the placenta of preeclampsia can carry anti-angiogenic factors and lead to maternal glomerular endothelial dysfunction and proteinuria [19]. EVs released from autoimmune diseases, such as antiphospholipid syndrome, thrombotic microangiopathy, systemic lupus erythematosus, and ANCA-vasculitis, can promote coagulation, thrombosis, and immune-mediated renal pathology [20].

Increasing evidence shows that circulating cells (monocytes, neutrophils, and red blood cells) and platelets attacked by toxins release the EVs, and the latter are the major elements of pathogenesis in hemolytic uremia syndrome [21]. The ability of EVs derived from mesenchymal stem cells (MSC) to alleviate myocardial injury in experimental metabolic renovascular hypertension is partly mediated by IL-10-containing EVs [22]. The bioactive cargo of EVs in kidney transplantation (KT) includes implant antigens, cytokines, growth factors, costimulation/inhibitory molecules, and functional microRNAs (miRNAs) that may regulate gene expression in receptor cells, which act as the immune modulators and play a crucial role in maintaining complex crosstalk between graft tissue and innate/adaptive immune system. EVs are of great importance in allogeneic recognition, ischemia–reperfusion injury (IRI), autoimmunity, and allogeneic immunity and are expected to become biomarkers and therapeutic tools for KT [23].

The kidneys can also release EVs, affecting the function of other tissues and organs. A study on the proteomics of EVs in murine hearts found that some proteins come from other organs, including the kidneys [24]. Cardiovascular dysfunction is caused by a high level of circulating endothelial cell-derived particles in chronic kidney disease (CKD).

These particles may mediate inflammation, vascular wall damage, and remodeling and act as an incremental risk of Vascular calcification (VC) [25, 26, 27, 28, 29, 30]. VC is a pathological manifestation with high mortality, mainly manifested as abnormal calcium deposition in the vascular wall. During the process of VC, vascular smooth muscle cells (VSMCs) suffer osteogenic transformation and secrete EVs with various sources and compositions. The secreted EVs may obtain calcium-promoting properties, thus acting as the nucleation focus of hydroxyapatite crystallization and calcium transmission [31]. The serum calpain particles (CPPs) and EVs in uremia are meaningful participants in the extensive calcification mechanism of CKD, and cGRP (a protein rich in Gla) plays an inhibitory role in preventing calcification at the system and tissue levels [32].

The deterioration of renal damage during liver or heart disease may be caused by EVs, while the advancement of hepatorenal or cardiorenal syndrome may involve EVs [33, 34]. Nonvalvular atrial fibrillation is associated with kidney disease because the increase in thromboembolism is mediated by a higher level of EVs from prethrombotic endothelial platelets rather than by thrombotic status and other markers of cell activation, even in anticoagulant patients [35]. In patients with hypertension, the presence of EVs indicating podocyte injury and peritubular capillary injury is detected [36]. The endothelial cells derived EVs released from peritubular capillaries were checked in the primary and renovascular hypertension patients’ urine, with the density of EVs directly associated with clinical parameters and the scarcity of capillaries, but was inversely proportional to renal perfusion [37]. Therefore, the change of urinary EVs in hypertensive patients can be regarded as an early marker of renal injury caused by peritubular capillary injury, and it will change with the improvement of renal function after drug treatment in patients with primary and renovascular hypertension (Figure 1).

Figure 1.

Extracellular vesicles as delivery vans in cell-cell communication between renal cells and extrarenal organs (cells).

Advertisement

3. Extracellular vesicles as biomarkers of renal diseases

EVs are a type of disc-shape vesicles with single concave, secreted by active cell and enveloped by phospholipid bilayers [38], with sizes ranging from nanometers to micrometers [39, 40, 41, 42]. EVs grow out from the straight sprout of small vesicles wrapped in membranes or through blend on the superficial multivesicular bodies, engage in the intercellular commutation of materials and message, with cargo proteins, lipids, and nucleic acids (microRNA, messenger RNA, circ RNA, and lncRNA, etc.) [43, 44, 45, 46, 47]. With the pivotal roles in an exchange between renal cells and target cells, EVs are expected to serve as new molecular markers for the detection of kidney diseases, carrying specific molecular substances in source renal cells with the lipid structure to protect the contents from being degraded [48, 49]. At present, the study of EVs, especially exosomes, has been involved in the field of liquid biopsy [50, 51].

3.1 Extracellular vesicles as liquid biopsy markers for renal cell carcinoma

Renal cell carcinoma (RCC) is a kind of malignant tumor in the urinary system originating from the renal tubular epithelium, accounting for about 3% of adult malignant tumors, ranking second in the urinary system. The partial or total nephrectomy are the current therapeutic strategy, following or not following with immune-checkpoint-inhibitors-based targeted therapies to which patients are often drug-resistant. It lacks specific symptoms at the early stage [52]. Diagnosis mostly relys on ultrasound and other examinations. At the initial diagnosis, about 17% of RCCs have distant metastasis, with a 5-year survival rate close to 12% [53]. Therefore, early diagnosis and prediction of metastasis risk are crucial to the treatment and prognosis of patients. Data has shown that EVs are the proficiency biomarkers for tumor liquid biopsy due to their specific content, species conservation, stability, and high abundance under different sources and physiological and pathological conditions [54].

3.1.1 EVs as biomarkers for the diagnosis and staging of RCC

Some researchers conducted multi-omics analysis on tumor-related proteins and mRNA in EVs from different RCC cell lines, identified multiple candidate molecular markers, and confirmed that EVs could not only distinguish between RCC and benign lesions, but also assisted in determining the subtypes of RCC, with great potential for clinical transformation [55]. Research has found that compared to the control HK-2 cell line, the expression level of miR-150 in EVs from the 786-O RCC cell line was upregulated by 5.2 times, while miR-205 was downregulated by 10,000 times, indicating that miR-205 and miR-150 in EVs were new prospective biomarkers for the diagnosis of RCC [56].

However, in vitro cell models cannot fully and in real-time reflect tumor conditions, and it is still essential to screen and validate in clinical specimens. There are reports that compared to healthy control, the expression of miR-149-3p and miR-424-3p in plasma EVs of RCC patients are upregulated, while miR-92a-1-5p is obviously downregulated, which is helpful for the diagnosis of RCC [57]. Zhang et al. observed that the levels of miR-210 and miR-1233 in serum EVs were markedly upregulated in patients with clear cell renal cell carcinoma (ccRCC) than those in the control group, and their expression were dramatically reduced after tumor resection, they has worthwhile value for RCC diagnosis and surgical effect monitoring [58]. Additionally, the expression of miR-210 is also related to their clinical prognosis in serum EVs of RCC patients, and the diagnostic performance for early patients is better than that of serum miR-155 [59]. Similarly, serum EVs miR-4525 can serve as a potential diagnostic marker for advanced RCC patients [60]. EVs from other sources of bodily fluids, besides blood also have the diagnostic potential for RCC. Urinary exo-miR-30c-5p may regulate the expression of a protein related to the progression of ccRCC (heat shock 70 kDa protein 5) and is regarded as a latent diagnostic biomarker for early ccRCC [61]. Some researchers have found that a combination detection of urine EVs miR-126-3p and miR-449a or miR-34b-5p can prominently differentiate between ccRCC and healthy individuals [62].

Recently, although most research on EVs as molecular markers of RCC has focused on miRNAs, it is also important to recognize that other specific molecules (proteins, mRNA, DNA, etc.) in EVs can also be used for auxiliary diagnosis of RCC [63]. Palma et al. found the obvious diverse in urinary exosomal shuttle RNA (esRNA) type of ccRCC patients compared to healthy population and non-ccRCC, and observed that this specific pattern increased to reach the normal level one month after partial or radical nephrectomy. They identified that the urinary esRNA levels of CEBPA, GSTA1, and PCBD1 were downregulated in patients than the healthy subjects, then suggested the RNA content in urinary EVs could be the prospective diagnostic applications for the ccRCC [64]. Some scholars describe CA9, CD147, and CD70 in EVs as tumor markers that are specific to ccRCC [65].

Some scholars characterized CA9, CD147, and CD70 as tumor-specific markers on EVs in CCRCC [65]. Vergori observed that CA9 in circulating large EVs plays a part in the diagnosis and prognosis of ccRCC [66]. Researchers regarded Polymerase I and transcript release factor (PTRF)/CAVIN1 in urinary exosomes as prospective biomarkers of ccRCC [67]. Li et al. believed that exosomal circRNAs may be the potential cancer diagnosis marker [68]. The above studies have confirmed that EVs and their contents are expected to become diagnostic and staging markers for RCC.

3.1.2 EVs as markers for RCC metastasis monitoring

The messages in EVs actively loading by cancer cells can support tumor spread; the double layer of EV protects the peculiarity. Thus EVs play a major role in metastasis by improving the content’s half-life and stability, and then EVs act as markers for metastasis monitoring. Many scholars have confirmed this by animal and cell experiments.

EVs released from RCC cell line (786-O) can transfer long-chain noncoding RNA (LncRNA)_human lung adenocarcinoma of the metastasis related transcription factor 1 (MALAT1) to adjacent nonmetastatic RCC cells and enhance their growth, invasion, and metastasis abilities [69]. Meanwhile, CD103+ EVs separated from cancer stem cell (CSC) of ccRCC patients targeted regulate the protein level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which is tightly relevant to cell migration, by transporting miR-19b-3p to ccRCC cells; Cancer stem cell EVs also can be guided by CD103 to target cancer cells and organs, making ccRCC have higher lung metastasis capacity [70].

In addition, the expression of miR-1293 decreased, and the expression of miR-301a-3p increased in plasma EVs of RCC patients with metastasis, which are regarded as presumable biomarkers for the metastasis of RCC [71]. The downregulated miR-483-5p was observed in ccRCC, and Wang et al. believed that it contributed to cell proliferation, metastasis, and inflammation [72]. Evs-derived proteins also can be the marker of RCC metastasis monitoring. Some investigators observed that the serum EVs of prostate-specific membrane antigen (PMSA) levels in metastatic patients of RCC were sharply increased compared with nonmetastatic patients by establishing a sandwich ELISA method, and also believed that PMSA can reflect the angiogenesis of the primary tumor and metastasis, then monitoring the metastatic RCC in real time [73]. Furthermore, the surface or content of EVs can provide targeted recognition sites for tumor metastasis, providing a theoretical basis for developing the predicting RCC metastasis markers and targeted therapy strategies.

3.1.3 EVs as biomarkers for drug resistance and prognosis in RCC

The prognosis of RCC patients is closely related to tumor staging, grading, metastasis, and patient sensitivity to therapeutic drugs. It is deemed that partial nephrectomy is the preferred treatment strategy because some researchers observed a better ending and better postoperation renal function in the advanced stage RCC patients treated by partial nephrectomy than those that undergo radical nephrectomy [74]. The survival rate of early RCC is 90%, while that of locally advanced or metastatic cases is only 13%. Therefore, early diagnosis and treatment of RCC is extremely prominence for improving the 5-year survival rate of this disease. Liquid biopsy techniques provided for early RCC detection may bring about a superior outcome. EVs detection can guide clinical rationalization, individualization, and precision of medication by assessing the sensitivity of patients to drugs. Ras-related protein Rab-27B is one of the chief proteins involved in the secretion of EVs. Previous studies have confirmed that the high expression of this protein is related to the poor prognosis of hepatocellular carcinoma, ovarian cancer, and colorectal cancer. Tsuruda et al. identified that Rab-27B was significantly overexpressed in sunitinib-resistant renal cell carcinoma cell lines [75]. Dias et al. found that compared with patients with nonmetastatic in situ ccRCC, patients with metastatic disease had higher levels of matrix metalloproteinase inhibitor of metalloproteinase-1 (TIMP-1) mRNA in plasma EVs and lower overall survival rate. This means TIMP-1 mRNA derived from EVs is a potential prognostic marker for ccRCC [76].

Additionally, researchers also found that after 1 day of cryoablation treatment, the miR-17-5p, miR-126-3p, and miR-21-3p in mouse serum-derived EVs rapidly decreased, reflecting the number of active tumors, which can be used to evaluate tumor clearance efficacy and dynamically monitor tumor burden [77]. Du et al. applied Cox regression analysis and Kaplan Meier analysis to confirm the significant overall survival (OS) association of three EVs source miRNAs (miR-let-7i-5p, miR-615-3p, and miR-26a-1-3p. Then, these miRNAs in plasma exosomes were used as prognostic markers for metastatic kidney cancer [78]. Nakanori et al. found that intracellular miR-224 increases in ccRCC and is obviously relevant to cancer metastasis and invasion. Then, they explored the relevance between the level of exo-miR-224 and the prognosis of 108 ccRCC patients, and they observed that progression-free survival and overall survival are shorter in the group with high exo-miR-224 expression level. Thus, they deemed that extracellular miR-224 in ccRCC patient’s exosomes is a prospective prognostic marker [79]. Sunitinib is a novel multi-targeted oral drug for treating tumors and is also used for treating advanced renal cell carcinoma. The drug resistance is the main challenge of current treatment. Sun et al. detected lncRNA Activated in RCC with sunitinib resistance (lncARSR), a lncRNA related to the clinically poor response to sunitinib. LncARSR may improve the level of AXL and c-MET in RCC cells by competitively binding to miR-34/miR-449 and recovery the drug resistance of sunitinib. In addition, this lncRNA also transmits its drug resistance mechanism to sensitive cells through exosomes. Therefore, it is suggested that lncARSR may predict sunitinib resistance and a potential therapeutic target (Table 1) [80].

MarkerExpression disorderOriginApplicationReferences
miR-150UpregulatedRCC cell line (786-O)Diagnosis[56]
miR-205Downregulated
miR-92a-1-5pDownregulatedPlasmaDiagnosis[57]
miR-149-3p & miR-424-3pUpregulated
miR-210 & miR-1233UpregulatedSerumDiagnosis[58]
miR-4525UpregulatedSerumDiagnosis[59]
combination of miR-34b-5p or miR-449a & miR-126-3pUrine and cell line modeDiagnosis[61]
GSTA1, CEBPA, and PCBD1DownregulatedUrineDiagnosis[63]
CD147, CA9, and CD70UpregulatedCell lines and tissue samplesDiagnosis[64]
CA9IncreasedPlasmaDiagnosis and prognosis[65]
PTRF/CAVIN1IncreasedUrineDiagnosis[66]
MALAT1Cell lineMetastasis monitoring[68]
CD103Cancer stem cellMetastasis Monitoring[69]
miR-1293IncreasedPlasmaMetastasis monitoring[70]
miR-301a-3pDecreased
miR-483-5pDownregulatedPlasma and tissuesMetastasis monitoring[71]
hsa-miR-301a-3pIncreasedPlasmaMetastasis monitoring[72]
hsa-miR-1293Decreased
PMSAIncreasedSerumMetastasis monitoring[73]
RAB27BIncreasedCell linesDrug resistance[75]
TIMP-1 mRNAIncreasedPlasmaPrognosis[76]
miR-126-3pDecreasedSerumPrognosis[77]
miR-17-5pDecreased
miR-21-3pDecreased
miR-let-7i-5pIncreasedPlasmaPrognosis[78]
miR-26a-1-3pIncreased
miR-615-3pIncreased
miR-224IncreasedBlood and cell linesPrognosis[79]
lncARSRIncreasedCell line, plasma, and tissueDrug resistance[80]

Table 1.

A list of candidate dysregulated EV contents as molecular markers for RCC.

3.2 Extracellular vesicles as diagnostic markers for renal tubulointerstitial injury

Some biomolecules carried in urine EVs are closely related to the degree of renal tubulointerstitial inflammation and fibrosis; the detection of these markers in EVs can reflect the degree of renal inflammatory fibrosis progression. After genetic screening of EVs in urine of chronic kidney disease (CKD) patients, it was found that miR-29c and CD2AP mRNA were positively correlated with the degree of renal fibrosis and renal function, which may have meaningful diagnostic value for the progression of renal fibrosis [81, 82]. Based on previous findings that EVs CCL2 mRNA released by renal tubular epithelial cells (TECs) can directly transfer to macrophages and promote tubulointerstitial inflammation, some researchers found that the expression of CCL2 mRNA in urine EVs of IgA nephropathy (IgAN) patients is related to the level of eGFR and can predict the progress of renal function, which may become a new marker for IgAN prognosis monitoring [83]. Hypertension is a noteworthy cause of CKD, and the loss of peritubular capillaries (PTC) is one of the characteristics of hypertension; the level of endothelial microparticles (EMP) in the loop can report the endothelial injury systematically. Urinary EMPs levels were examined as possible markers of PTC and decreased fibrotic density. Ptc-emps were peculiar proteins that were identified as plasmalemmal vesicle-associated protein (PL-VAP) positive urinary exosomes, which were showed in PTC and Vas deferens endothelium but not in glomeruli and arteries and are supposed to be a novel biomarker of intrarenal capillary loss [84]. In obstructive kidney disease, uEVs may help assess the risk of developing renal dysfunction, and some studies have observed that the profibrotic factor TGF-β1 level in uEVs is related to glomerular filtration rate [85]. It was the upregulated of the urinary exosomal miR-181a (200-fold) in CKD patients [86] and the downregulated of the exosomal level of secreting transglutaminase-2 (a fibrosis-activating enzyme) in UUO mice [87]. CKD patients have a decrease in miR-200b of urinary exosomes, with the greatest reduction in urinary exosomes originating from cells outside the proximal renal tubules [88]. The upregulation of the urinary exosomal ceruloplasmin in CKD patients and animals with passive Heyman nephritis was observed [89]. Moreover, osteoprotegerin, an inflammatory marker, was shown to be increased in the uEVs of patients with CKD [90].

The kidney is a major organ that regulates the body’s water and salt metabolism. Ions and aquaporin are distributed in different segments of TECs. During the injury of TECs, these ions and water transporters in urine EVs also changed correspondingly. For example, the decrease of aquaporin (AQP) expression in urine EVs was observed in the rat model of Acute kidney injury (AKI) induced by cisplatin and ischemia/reperfusion, suggesting its potential impact on the change of renal concentration function [91]. The level of EVs Na + transporters in urine may also indicate certain pathological conditions in the kidneys. The activation of renin angiotensin aldosterone system (RAAS) after sodium-restricted diet acute aldosterone infusion in patients with hypertension increases the content of epithelial sodium channel (ENaC) in urine EVs by nearly 20 times, suggesting that it may be a novel means to monitor RAAS activation [92]. Moreover, the significant reduction of furosemide-sensitive sodium potassium chloride cotransporter (NKCC2) and sodium chloride cotransporter (NCC) in urine EVs has been used to distinguish different phenotypes of hereditary desalinization renal tubular disease [93, 94]. These studies suggest that changes in ion and water transport proteins in urine EVs may reflect renal tubular damage.

3.3 Extracellular vesicles as diagnostic markers for glomerular injury

EVs derived from glomeruli are continuously released into urine under physiological conditions, so changes in urine EVs can reflect the degree of glomerular disease, including podocyte damage. In early studies, Wilms Tumor Protein 1 (WT1) has been identified as a key regulator of podocyte expression, and the WT1 target gene is crucial to maintain the glomerular filtration barrier [95]. WT1 in uEVs was confirmed to be detectable before obvious glomerular sclerosis and urinary EVs WT1 appeared prior to proteinuria and glomerular histological damage in focal segmental glomerulosclerosis (FSGS) animal models [96]; moreover, urinary exosomal WT-1 was significantly decreased in patients in remission for either FSGS or steroid-sensitive nephrotic syndrome (SSNS) or following steroid treatment of patients with SSNS [97]. Compared with patients with minimal change nephropathy (MCN), the WT1 mRNA expression was increased significantly in urine EVs of patients with diabetes nephropathy (DN) and was associated with decreased eGFR; thus the high expression of WT1 mRNA in urine EVs distinguishes patients with DN from patients with MCN [98]. In addition, hyperglycemia can stimulate the release of WT1 from podocyte to urine EVs, so the detection of WT1 in urine EVs can indicate early damage of podocyte in diabetes patients. WT-1 may be a biomarker for early diagnosis of podocyte injury, as suggested by these data. Recently, it has been found that the expression of the upper pitt-specific transcription factor ELF3 protein in urine EVs is in patients with DN, but not in patients with MCN. Urine EVs ELF3 can predict the decline of eGFR in patients with DN in the next few years [99]. It is a practical diagnostic tool that uEVs is applied to distinguish early IgA nephropathy (IgAN) and thin basement membrane nephropathy with microscopic hematuria in children and adults. Moon et al. discovered four various biomarkers that have different expressions in the uEV of these patients and the high levels of aminopeptidase N and vasoactive precursors in the thin basement membrane nephropathy group, while α-1-Elevated levels of antitrypsin and ceruloplasmin were higher in the IgAN group [100].

3.4 Extracellular vesicles as diagnostic markers for other kidney disease

The biological processes involved in cytoskeleton-regulating and Ca(2+)-binding proteins are closely related to the pathogenic state of renal tubular epithelial cells in autosomal dominant polycystic kidney disease (ADPKD). Some study found by iTRAQ-based quantitative proteomics that this differential expression of proteins in urine EV of ADPKD demonstrates the possibility of using urine EV to monitor patient status [101, 102]. Fabry disease, also known as Anderson-Fabry disease, is the most common lysosome accumulation disease [103]. It is an X-linked congenital defect in the pathway of glycosphingolipid metabolism, which causes the accumulation of globotriaosylceramide (Gb3) in a variety of lysosome, leading to a series of clinical manifestations. Fabry nephropathy is kidney impairment, mainly manifested as hypertension, hematuria, mild proteinuria and fatty urine, and various renal tubular dysfunction, such as concentration and dilution function [104]. Some scholars observed the increased expression of miR-29a-3p and miR-200a-3p in uEVs of Fabry nephropathy patients may reveal an attempt by this organism to inhibit the progression of renal lesions leading to end-stage renal disease (ESRD).

Meanwhile, the expression of miR-30b-5p increased within 10 years in uEVs of patients without renal dysfunction, which may play a defensive part in podocyte trauma and may play a vital role in Fabry nephropathy [105].

In summary, the “liquid biopsy” based on EVs currently shows good application prospects in the diagnosis of kidney disease. In future research, it is necessary to further explore the standardization of urine sample separation, storage, and processing [106], strengthen the large-scale cohort study of the diagnostic, prognostic and predictive value of EVs biomarkers in different kidney diseases, and promote the technical research on the traceability of urine EVs kidney cells (Table 2).

PathemaLiquid biopsy markerLevel related to healthyType of regeneration or lesionDetection way
Autosomal dominant polycystic kidney disease (ADPKD)S100-A9, S100-A8, annexin A1, and annexin A2increasedurinary EVsiTRAQ-based quantitative proteomics [101, 102]
Fabry nephropathymiR-200a-3p, miR-29a-3pincreasedUrine EVsqPCR [105]
Acute kidney injuryAQP1DecreasedTubular cell injuryWestern blot [107]
ATF3IncreasedqRT-PCR [96]
Fetuin AIncreasedLC-MS/MS, MALDI-TOF, western blot [108]
miR-130a and miR-145IncreasedGlomerularqRT-PCR [109]
miR-155 and miR-424Decreasedmesangial damage
ELF3increasedPodocyte damageWestern blotting [99]
WT1IncreasedPodocyte damageWestern blot [110]
AEBP1 mRNAincreasedNot knownBioinformatics analysis [111]
hsa-miR-503 and hsa-miR-451aincreaseduEVsnext-generation small RNA sequencing & qRT-PCR [112]
Hypertensive kidney injuryPL-VAPincreasedUrineflow cytometry [84]
Focal segmental glomerulosclerosisWT1IncreasedPodocyte damageWestern blot [97]
Chronic kidney diseaseCCL2 mRNAIncreasedtubular epithelial cellsqRT-PCR [83]
CD2AP mRNADecreasedPodocyte damageqRT-PCR [82]
miR-29DecreasedFibrosisqRT-PCR [81]
miR-200bIncreased
secreting transglutaminase-2IncreasedUUOProteomic [87]
ceruloplasminincreasedUrineELISA [89]
miR-181a-5p and miR-451IncreasedNot knownqRT-PCR [86, 113]
IgA nephropathymiR-378, miR-215-5pIncreasedNot knownqRT-PCR [114]
miR-205-5p, miR-29cDecreased
α-1-antitrypsin & ceruloplasminincreasedUrinesemi-quantitative immunoblot analysis [100]
Kidney transplantationNGALIncreasedTubular cell injuryWestern blot [115]
CD133DecreasedTubular cell regenerationFACS, western blot [116]
Medullary Sponge Kidney DiseaseFicolin 1, Mannan-binding lectin serine protease 2, and Complement component 4-binding protein βincreasedUrine EVsELISA [117]
Lupus nephritismiR-146a, miR-150, miR-21IncreasedFibrosisqRT-PCR [118]

Table 2.

Deregulated EVs and their contents as the molecular marker of nonneoplastic kidney disease.

Advertisement

4. Extracellular vesicles in the therapeutic role

Renal pathophysiology is a multivariate procedure concerned with different kidney structures. Acute kidney injury (AKI) is a group of clinical syndromes, the manifestations include oliguria, anuria, edema, loss of appetite, etc., with the character of tubular necrosis and glomerular hyperfiltration. The maladaptive repair after AKI can easily lead to chronic kidney disease, also known as CKD. CKD is a growing and irretrievable damage of kidney function, the athological manifestation being fibrosis that could lead to ESRD. Currently, kidney disease therapies based on EVs can be divided into two categories: firstly, certain specific cells, such as stem cell-derived EVs, can be directly applied as therapeutic drugs due to their carrying bioactive molecules; Secondly, EVs can serve as delivery carriers for various types of drugs for the treatment of kidney diseases [119, 120].

4.1 Application of stem cell-derived EVs in the treatment of kidney disease

Timmers et al. first confirmed that the conditioned medium of human mesenchymal stem cells had a protective effect on myocardium in 2007, they also confirmed that the main active substances involved this were at 100–220 nm by size analysis [121]. The team further isolated and identified the substance in 2010, identifying it as exosomes. Since then, more and more studies have confirmed that the EVs derived from mesenchymal stem cell (MSC-EVs) play a major part in tissue damage repair and immune regulation, and have developed their therapeutic potential in the matter of regenerative medicine. In recent years, stem cell-derived EVs have been studied in different animal kidney disease models in vivo, including AKI, diabetes nephropathy, hypertensive nephropathy, unilateral ureteral obstruction, and subtotal nephrectomy.

4.1.1 Acute kidney injury (AKI)

AKI is a general clinical condition, and there is no clear and effective treatment method. In recent years, stem cell EVs have been proven to play therapeutic roles in AKI, including anti-apoptotic, anti-inflammatory, and antioxidant stress. BRUNO et al. proved that in the glycerol-induced AKI mouse model, the bone marrow mesenchymal stem cells derived EVs accelerate the repair of damaged renal tubular cells, promote renal tubular cell proliferation, and protect cells from apoptosis [122]. Extracellular vesicles derived from bone marrow mesenchymal stem cells were also verified in the toxic AKI model induced by cisplatin and gentamicin [123, 124], which can improve renal function, reduce histological damage, and alleviate renal fibrosis. The bone marrow mesenchymal stem cells derived EVs have also achieved the same effect in renal ischemia–reperfusion (I/R) injury models [125, 126].

There are many mechanisms by which stem cell-derived EVs can improve AKI. Studies have found that the bone marrow mesenchymal stem cells derived EVs may carry specific mRNAs to activate the proliferation process of surviving renal tubular cells after injury, so that the impaired cells can re-enter the cell cycle.

Recently, CAO and his team sequenced human umbilical cord mesenchymal stem cells derived EVs and found that they are rich in miR-125b-5p and can inhibit G2/M cell cycle arrest and apoptosis of TECs by targeting p53, thereby promoting renal tubular repair and improving ischemic AKI [127]. Li et al. found that human urine-derived stem cells EVs can defend renal function of ischemia–reperfusion rats by carrying miR-146a-5p, which can target the 3′-untranslated coding region of IL-1 receptor-associated kinase (IRAK), thereby inhibiting NF-κ activation of the B signaling pathway and infiltration of inflammatory cells [128]. Lately, someone conducted EVs derived from human bone marrow combined with pulse-focused ultrasound therapy on a cisplatin-induced mouse model and found that the decreased expression of NLRP3 inflammasome and its downstream pro-inflammatory cytokine IL-1βand IL-18 decreased promoted renal repair after AKI [129]. These studies suggest that MSC-EVs exert the therapeutic effect of AKI through anti-inflammatory effects. The study also found that MSC-EVs can also inhibit mitochondrial damage in the IRI model through various pathways, thereby alleviating AKI. MSC-EVs derived from human placenta activate the Keap1-Nrf2 signaling pathway, stimulate mitochondrial antioxidant defense mechanisms to maintain the stability of TEC mitochondrial structure and regulate mitochondrial function, participate in TEC damage repair, and promote renal function recovery [130]. In addition, there are reports that MSCs-EVs can reduce mitochondrial damage and inflammation caused by AKI through the mitochondrial transcription factor A (TFAM) pathway [131]. EVs originating in umbilical stalk blood mesenchymal stem cells promote dedifferentiation and proliferation of renal tubular cells; extracellular vesicles derived from umbilical cord Wharton glue mesenchymal stem cells stimulate cell proliferation, reduce inflammation, and apoptosis through mitochondrial protection. 3D cultured placental mesenchymal stem cells derived EVs more effectively inhibit cell apoptosis, inflammation and improve renal function [132, 133, 134]; Human urinary mesenchymal stem cells derived EVa accelerate renal recovery, stimulate renal tubular cell proliferation, reduce the expression of inflammatory and injury markers, restore endogenous Klotho loss, and thus protect renal function [135, 136].

4.1.2 Chronic kidney disease (CKD)

It is a significant pathological manifestation in CKD progression of persistent kidney tubulointerstitial fibration. Scholars have found that hBMSC-EVs can inhibit TGF through their rich miR-294/miR-133-β1 mediates epithelial-mesenchymal transformation in CKD rats, thereby alleviating renal interstitial fibrosis (RIF) [137]. Moreover, particles derived from renal-derived MSCs can improve peritubular capillary sparsity in the kidney and delay the progression of renal injury by inhibiting tubulointerstitial fibrosis in mice UUO [138]. EVs produced by bone marrow mesenchymal stem cells derived EVs alleviate UUO renal fibrosis, partially by inhibiting the RhoA/ROCK pathway [139]. In a mouse CKD model induced by chronic cyclosporin, bone marrow derived EVs can improve the kidney function in the inflammatory microenvironment [140]. Extracellular vesicles rich in miR-196b-5p mediate crosstalk between proximal tubular epithelial cells and fibroblasts, which may be related to the STAT3/SOCO2 signaling pathway and mediate aldosterone-induced renal fibrosis with diabetes [141]. However, miR-221 in the EVs derived from podocyte reversed DN by inhibiting Wnt/β-Catenin signaling mediated proximal tubular cell damage [142]. EVs from human liver stem cell through miR29b reduce renal fibrosis by disturbing the β-Catenin pathway [143]. In a recent study, researchers synthesized a biological scaffold, which integrates MSC-EVs, extracellular matrix, poly (lactic acid Glycolic acid) copolymer, poly (Deoxyribonucleotide), etc. This biological scaffold achieved renal tissue remodeling in a partial nephrectomy mouse model by promoting cell proliferation, angiogenesis, and inhibiting fibrosis and inflammation [144]. Eirin et al. found that intrarenal injection of adipose-derived mesenchymal stem cells EVs improved pigs with metabolic syndrome and renal artery stenosis disease by ameliorating renal inflammation and fibrosis. Further research confirmed that these renal protective effects were mediated by the carrier of anti-inflammatory cytokine IL-10 carried by MSC-EVs [145]. The secretion of EVs from mesenchymal stem cells stimulated by melatonin inhibits fibrosis in renal tissue by regulating cell apoptosis and proliferation of fibrosis related cells [146]. Stem cell-derived EVs also improve CKD related lesions. CHOI et al. found that renal mesenchymal stem cells expressing erythropoietin EVs improve renal anemia in mice with chronic kidney disease [147].

4.1.3 Diabetic nephropathy (DKD)

Diabetes nephropathy (DKD) is one of the elemental etiology of terminal-stage kidney disease [148]. For a long time, there is no specific drug for DKD, and the current treatment is limited to blood glucose control, blocking of renin-angiotensin-aldosterone system, and changes in lifestyle [149]. MSC-EVs can protect cells from high glucose-induced damage by promoting regeneration through anti-apoptosis, anti-fibrosis, and autophagy. Jin et al. confirmed that miR-486 carried by EVs derived from fat MSCs can be transferred to podocyte, which leads to increased autophagy flux and decreased apoptosis by inhibiting the Smad1/mTOR signal pathway of podocyte in DKD mice [150], thereby improving podocyte injury in DKD mice. In addition, MSC-exos can reduce the overexpression of TGF-βto improve tubulointerstitial fibrosis in DN mice and regulate the expression of ICAM-1 for inhibiting inflammatory cell infiltration, thereby reducing diabetes kidney damage [151]. Xiang et al. found that umbilical cord mesenchymal stem cells derived EVs can reduce inflammatory factors (IL-6, TNF-α) in renal tubular cell expression, reduce inflammatory cell infiltration, interstitial fibrosis, and other pathological changes of diabetes nephropathy in renal tissue [152]. Mesenchymal stem cells derived EVs can regulate autophagy through the mTOR pathway, upregulate autophagy proteins such as LC3 and Beclin-1, and observe an increase in autophagic vesicles under electron microscopy, and the treatment of the MSC-derived EVs decreased the urine protein and serum creatinine in diabetes nephropathy mice. Renal biopsy showed that the renal pathological changes of diabetes nephropathy such as mesangium expansion and fibrosis were alleviated [153]. Jin et al. observed that the adipose stem cells derived exosomes could reverse the damage of renal function caused by high glucose environment and further found that miR-486 is a critical determiner in the reverse process, which can reduce the expression of Smad1, increase cell autophagy, and reduce podocyte apoptosis [150].

Recently, researchers injected two doses of MSC-EVs into patients with CKD phase III and IV in a single center, stochastic, placebo-contrast phase II/III clinical trial, and the results showed that the renal inflammation and function were effectively improved in the treatment group, providing worthwhile clinical evidence for the employ of MSC-EVs in the therapy of CKD. Unfortunately, this study did not investigate its mechanism and related renal pathological types [154]. Obviously, it has extremely clinical significance to further strengthening research.

4.2 Application of EVs based drug targeted delivery in the treatment of kidney disease

In 2010, Sun et al. wrapped curcumin with anti-inflammatory and antioxidant effects in EVs for the first time to treat sepsis in mice and achieved good therapeutic effects [155]. Since then, targeted drug development based on EV carriers has continued to heat up. Existing research suggests that EVs can successfully load various types of drugs such as nucleic acid drugs, protein drugs, and small molecule drugs for the treatment of kidney disease.

4.2.1 EVs as nucleic acid drug carriers

Various types of nucleic acids are the main ingredients of EVs; therefore, it has aroused widespread interest in using EVs as delivery carriers for nucleic acid drugs. Previous studies have shown that EVs can load and transport nucleic acids such as mRNA, miRNA, and small interfering RNA (siRNA) for disease treatment [156]. Among them, siRNA therapy has shown great potential in the treatment of human diseases. However, due to its instability and missing target effect, the clinical application of RNA interference technology has been limited. Recently, TANG et al. successfully developed a siRNA delivery system based on red blood cell-derived EVs, which utilizes renal injury molecule-1 targeted peptide A Kim-1-binding peptide (LTH) to modify red blood cell-derived extracellular vesicles (REVs) and successfully deliver siP65 and siSNAI1 to the injured renal tubules, effectively improving renal tubulointerstitial inflammation and fibrosis induced by ischemia-reperfusion and UUO, and blocking the chronic progression of AKI [157]. Meanwhile, they also applied muscle-targeted peptide-modified EVs with loaded miR-26a, significantly improving Sarcopenia in 5/6 nephrectomized CKD mice [158]. Combining EVs with a kidney-targeting peptide, Rabies Virus Glycoprotein (RVG), and loading miR-29a into it, can not only alleviate the myopenia in mice with UUO but also improve renal fibrosis through EVs-mediated communication between skeletal muscle and renal organs [159]. Therefore, the targeted therapy of nucleic acid drugs based on EVs can not only delay the progression of kidney disease but also improve its complications. A different strategy was applied and studies have shown that EVs from engineered MSCs overexpressing miR-let7c can shift miRNA to renal cells and inhibit RIF [160]. In addition, BM-MSC-derived EVs overexpressing miR-34a embellished by lentivirus inhibited TGF-β1 begotten epithelial-mesenchymal transition (EMT) in human renal tubular cells [161].

4.2.2 EVs as protein drug carriers

Protein deficiency and dysfunction are important causes of many diseases. Therefore, it is one of the methods for treating diseases by increasing the corresponding protein levels. However, protein drugs themselves have drawbacks, such as high molecular weight and poor stability, which limit their clinical application. The loading performance and modifiability of EVs have brought dawn to this field. Recently, Researchers constructed a cytokine IL-10 delivery system (IL-10 + EVs) using macrophage-derived EVs as a vector. The loading of EVs not only improved the stability of IL-10 but also demonstrated a unique ability to target kidney damage. Further mechanism research has found that IL-10 + EVs can promote mitochondrial autophagy in TECs by suppression the sensitization of the mTOR signaling pathway, significantly improving renal injury and chronic lesions induced by ischemia-reperfusion [162]. Kim et al. utilized a novel photogenetic engineering technique to load NF-κ inhibition protein srIκB into EVs, effectively improving the inflammatory response and cell apoptosis in the kidney after ischemia [163].

4.2.3 EVs as small molecule drug carriers

Research has found that the encapsulation and delivery of EVs can improve the targeting, cell uptake efficiency of small molecule drugs, also improve drug stability, and reduce toxic side effects [119, 164]. MSCs-EVs have been considered as another prospective acellular therapy for AKI. A Supermolecule hydrogel containing Arg-Gly-Asp (RGD) peptide has been manufactured to enhance the efficiency in the therapy of AKI. Data shows that RGD-EV hydrogel has a good rescue influence on renal function at the early stage of AKI, by dwindling tubular damage and facilitating cell proliferation through the combination of RGD and integrin [165]. Recently, researchers constructed M2 macrophage-derived EVs loaded with dexamethasone (DEX) and found that it not only targets damaged kidneys but also has effective anti-inflammatory and anti-fibrotic effects. Moreover, it significantly alleviates the adverse effects of DEX on blood glucose and the hypothalamic-pituitary renal gland axis in mice impact [166]. In the AKI model, EVs derived from MSCs ameliorative to over-express the octamer binding transcriptional factor 4 (OCT4) showed decreased expression of Snail, a trigger factor for epithelial-mesenchymal transition (EMT). Therefore, the administration of EV-ameliorated MSCs has achieved better renal tissue recovery, cell proliferation improvement, cell death elimination, and the initial fibrosis process block [132].

All in all, EVs have become a worthwhile carrier for the next generation of targeted drug delivery and have received widespread attention in recent years based on the advantages of good biocompatibility, low immunogenicity, and modifiability of EVs Cell therapy. Of course, there are still issues that need to be addressed: producing EVs that are repeatable, large-scale, high-throughput, and meeting the clinical application level; suitable parental cells selecting, culture systems such as cell factories, bioreactors, and hollowing fiber tubes establishing to achieve large-scale expansion production [167]; standardized EVs separation technology suitable for large-scale production building, a high-throughput detection platform developing, the quality of the EVs production process control and quality monitoring at the single particle level achieving, thereby further achieving standardized quality control and improving drug loading efficiency.

Advertisement

5. Conclusion

In the past 20 years, research on EVs has made rapid progress. More and more preclinical studies have shown that biomarkers and related treatment technologies based on EVs have great prospects in detecting and treating kidney disease, laying the foundation for their clinical application. Further strengthening the basic and clinical research on the role of EVs in making a diagnosis and giving treatment of kidney diseases, developing standardized, clinical-level EVs separation, purification, and quality control, and strengthening clinical queue research will offer technical support for the clinical transformation and application of electric vehicles.

References

  1. 1. Taha HB, Ati SS. Evaluation of α-synuclein in CNS-originating extracellular vesicles for Parkinsonian disorders: A systematic review and meta-analysis. CNS Neuroscience & Therapeutics. 2023;29:1-15. DOI: 10.1111/cns.14341
  2. 2. Kumar SK, Sasidhar MV. Recent trends in the use of small extracellular vesicles as optimal drug delivery vehicles in oncology. Molecular Pharmaceutics. 2023;20(8):3829-3842. DOI: 10.1021/acs.molpharmaceut.3c00363
  3. 3. Li W, Zheng Y. MicroRNAs in extracellular vesicles of alzheimer’s disease. Cell. 2023;12(10):1378. DOI: 10.3390/cells12101378
  4. 4. Zou J, Xia H, Jiang Q , Su Z, Wen S, Liang Z, et al. Exosomes derived from odontogenic stem cells: Its role in the dentin-pulp complex. Regeneration Therapy. 2023;24:135-146. DOI: 10.1016/j.reth.2023.05.008. eCollection 2023 Dec
  5. 5. Nowak M, Górczyńska J, Kołodzińska K, Rubin J, Choromańska A. Extracellular vesicles as drug transporters. International Journal of Molecular Sciences. 2023;24(12):10267. DOI: 10.3390/ijms241210267
  6. 6. Oey O, Ghaffari M, Li JJ, Hosseini-Beheshti E. Application of extracellular vesicles in the diagnosis and treatment of prostate cancer: Implications for clinical practice. Critical Reviews in Oncology/Hematology. 2021;167:103495. DOI: 10.1016/j.critrevonc.2021.103495. Epub 2021 Oct 13
  7. 7. Sohal IS, Kasinski AL. Emerging diversity in extracellular vesicles and their roles in cancer. Frontiers in Oncology. 2023;13:1167717. DOI: 10.3389/fonc.2023.1167717. eCollection 2023
  8. 8. Osna NA, Poluektova LY. Elucidating the role of extracellular vesicles in liver injury induced by HIV. Expert Review of Gastroenterology & Hepatology. 2023;3:1-8. DOI: 10.1080/17474124.2023.2230867
  9. 9. Li X, Wang Q. Platelet-derived microparticles and autoimmune diseases. International Journal of Molecular Sciences. 2023;24(12):10275. DOI: 10.3390/ijms241210275
  10. 10. Wu A, Wolley MJ, Fenton RA, Stowasser M. Using human urinary extracellular vesicles to study physiological and pathophysiological states and regulation of the sodium chloride cotransporter. Frontier in Endocrinology (Lausanne). 2022;13:981317. DOI: 10.3389/fendo.2022.981317. eCollection 2022
  11. 11. Li Q , Zhang Z, Yin M, Cui C, Zhang Y, Wang Y, et al. What do we actually know about exosomal microRNAs in kidney diseases? Frontiers in Physiology. 2022;13:941143. DOI: 10.3389/fphys.2022.941143. eCollection 2022
  12. 12. Sthl AL, Johansson K, Mossberg M, et al. Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases. Pediatric Nephrology, 2019;34(1) :11-30
  13. 13. Street JM et al. Exosomal transmission of functional aquaporin 2 in kidney cortical collecting duct cells. The Journal of Physiology. 2011;589:6119-6127
  14. 14. Gildea JJ et al. Exosomal transfer from human renal proximal tubule cells to distal tubule and collecting duct cells. Clinical Biochemistry. 2014;47:89-94
  15. 15. Hunter RW, Kumar S, Coward RJM, Buck AH, Dear JW. Extracellular RNA moves from the glomerulus to the renal tubule. Preprint at bioRxiv. 2021;7:1-29. DOI: 10.1101/2021.06.15.448584
  16. 16. Wang X, Wilkinson R, Kildey K, et al. Molecular and functional profiling of apical versus basolateral small extracellular vesicles derived from primary human proximal tubular epithelial cells under inflammatory conditions. Journal of Extracellular Vesicles. 2021;10:e12064. DOI: doi.org/10.1002/jev2.12064
  17. 17. Jella KK, Yu L, Yue Q , et al. Exosomal GAPDH from proximal tubule cells regulate ENaC activity. PLoS One. 2016;11:e0165763. DOI: 10.1371/journal.pone.0165763
  18. 18. Barros Lamus ER, Carotti V, Vries CRS, et al. Extracellular vesicles regulate purinergic signaling and epithelial sodium channel expression in renal collecting duct cells. The FASEB Journal. 2021;35:e21506. DOI: 10.1096/fj. 202002559R
  19. 19. Salomon C et al. A gestational profile of placental exosomes in maternal plasma and their effects on endothelial cell migration. PLoS One. 2014;9:e98667
  20. 20. Mazzariol M, Camussi G, Brizzi MF. Extracellular vesicles tune the immune system in renal disease: A focus on systemic lupus erythematosus, antiphospholipid syndrome, thrombotic microangiopathy and ANCA-Vasculitis. International Journal of Molecular Sciences. 2021;22(8):4194
  21. 21. Varrone E, Carnicelli D, Brigotti M. Extracellular vesicles and renal endothelial cells: A fatal attraction in Hemolytic uremic syndrome. The American Journal of Pathology. 2021;191(5):795-804. DOI: 10.1016/j.ajpath.2021.02.011
  22. 22. Jiang Y, Hong S, Zhu X, Zhang L, Tang H, Jordan KL, et al. IL-10 partly mediates the ability of MSC-derived extracellular vesicles to attenuate myocardial damage in experimental metabolic renovascular hypertension. Frontier in Immunology. 2022;13:940093. DOI: 10.3389/fimmu.2022.940093
  23. 23. Quaglia M, Dellepiane S, Guglielmetti G, Merlotti G, Castellano G, Cantaluppi V. Extracellular vesicles as mediators of cellular crosstalk between immune system and kidney graft. Frontiers in Immunology. 2020;11:74. DOI: 10.3389/fimmu.2020.00074
  24. 24. Claridge B et al. Proteome characterisation of extracellular vesicles isolated from heart. Proteomics. 2021;16:e2100026
  25. 25. Amabile N, Guérin AP, Tedgui A, Boulanger CM, London GM. Predictive value of circulating endothelial microparticles for cardiovascular mortality in end-stage renal failure: A pilot study. Nephrology Dialysis Transplantation. 2012;27:1873-1880
  26. 26. Berezin A, Zulli A, Kerrigan S, Petrovic D, Kruzliak P. Predictive role of circulating endothelial-derived microparticles in cardiovascular diseases. Clinical Biochemistry. 2015;48:562-568
  27. 27. Koide T, Mandai S, Kitaoka R, Matsuki H, Chiga M, Yamamoto K, et al. Circulating extracellular vesicle-propagated microRNA signature as a vascular calcification factor in chronic kidney disease. Circulation Research. 2023;132(4):415-431. DOI: 10.1161/CIRCRESAHA.122.321939
  28. 28. Soriano S et al. Endothelial damage and vascular calcification in patients with chronic kidney disease. American Journal of Physiology. Renal Physiology. 2014;307:F1302-F1311
  29. 29. Erdbrügger U, Le TH. Extracellular vesicles in renal diseases: More than novel biomarkers? Journal of American Society Nephrology. 2016;27:12-26
  30. 30. Dickhout A, Koenen RR. Extracellular vesicles as biomarkers in cardiovascular disease; Chances and risks. Frontier in Cardiovascular Medicine. 2018;5:113
  31. 31. Yaker L, Kamel S, Ausseil J, Boullier A. Effects of chronic kidney disease and uremic toxins on extracellular vesicle biology. Toxins (Basel). 2020;12(12):811. DOI: 10.3390/toxins12120811
  32. 32. Viegas CSB, Santos L, Macedo AL, Matos AA, Silva AP, Neves PL, et al. Chronic kidney disease circulating Calciprotein particles and extracellular vesicles promote vascular calcification: A role for GRP (Gla-rich protein). Arteriosclerosis, Thrombosis, and Vascular Biology. 2018;38(3):575-587. DOI: 10.1161/ATVBAHA.117.310578
  33. 33. Bankir L, Ahloulay M, Devreotes PN, Parent CA. Extracellular cAMP inhibits proximal reabsorption: Are plasma membrane cAMP receptors involved? American Journal of Physiology. Renal Physiology. 2002;282(3):F376-F392. DOI: 10.1152/ajprenal.00202.2001
  34. 34. Azevedo CAB, Stafim R, da Cunha C, Junho VC, Verônica J, da Silva AN, et al. Extracellular vesicles and their relationship with the heart-kidney axis, uremia and peritoneal dialysis. Toxins (Basel). 2021;13(11):778. DOI: 10.3390/toxins13110778
  35. 35. Ravi N, Ramaraju R, Vats A, Nair AR, Bandhu AK, Koirala D, et al. Role of anticoagulants for stroke prevention in low-risk population having atrial fibrillation and chronic kidney disease: A systematic review. Cureus. 2022;14(11):e31364. DOI: 10.7759/cureus.31364
  36. 36. Costantino VV, Gil Lorenzo AF, Bocanegra V, Vallés PG. Molecular mechanisms of hypertensive nephropathy: Renoprotective effect of Losartan through Hsp70. Cell. 2021;10(11):3146. DOI: 10.3390/cells10113146
  37. 37. Steegh FMEG, Keijbeck AA, de Hoogt PA, Rademakers T, Houben AJHM, Reesink KD, et al. Capillary rarefaction: A missing link in renal and cardiovascular disease? Angiogenesis. 2023;7:79-88. DOI: 10.1007/s10456-023-09883-8
  38. 38. Xia Y, Yu C, Johann Helwig E, Li Y. The role of extracellular vesicles in colorectal cancer. Technology in Cancer Research & Treatment. 2023;22:1533. DOI: 10.1177/15330338231185008
  39. 39. Zaborowski MP, Balaj L, Breakefield XO, Lai CP. Extracellular vesicles: Composition, biological relevance, and methods of study. Bioscience;65(8):783-797. DOI: 10.1093/biosci/biv084
  40. 40. Van Rooij E, Purcell AL, Levin AA. Developing microRNA therapeutics. Circulatory Research;110(3):496-507. DOI: 10.1161/ CIRCRESAHA.111.247916
  41. 41. Liu C, Bayado N, He D, et al. Therapeutic applications of extracellular vesicles for myocardial repair. Frontier in Cardiovascular Medicine. 2021;8:758050. DOI: 10.3389/fcvm.2021.758050
  42. 42. Charoenviriyakul C, Takahashi Y, Morishita M, Matsumoto A, Nishikawa M, Takakura Y. Cell type-specific and common characteristics of exosomes derived from mouse cell lines: Yield, physicochemical properties, and pharmacokinetics. European Journal of Pharmaceutical Sciences. January;96:316-322. DOI: 10.1016/j.ejps.2016.10.009
  43. 43. Mathieu M, Martin-Jaular L, Lavieu G, Thery C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nature Cell Biology. 2019;21(1):9-17. DOI: 10.1038/s41556-018-0250-9
  44. 44. Kowal J, Tkach M, Thery C. Biogenesis and secretion of exosomes. Current Opinion in Cell Biology. 2014;29:116-125. DOI: 10.1016/j.ceb.2014.05.004
  45. 45. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977. DOI: 10.1126/science.aau6977
  46. 46. Cocucci E, Meldolesi J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends in Cell Biology June 2015;25(6):364-372. eaau6977. DOI: 10.1016/j.tcb.2015.01.004
  47. 47. Anand S, Samuel M, Kumar S, Mathivanan S. Ticket to a bubble ride: Cargo sorting into exosomes and extracellular vesicles. Biochim Biophys Acta Proteins Proteom. 2019;1867(12):140203. DOI: 10.1016/j.bbapap.2019.02.005
  48. 48. Kooijmans SAA, Fliervoet LAL, van der Meel R, et al. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. Journal of Controlled Release. 2016;224:77-85. DOI: 10.1016/j.jconrel.2016.01.009
  49. 49. Han Y, Gao Z, Chen L, et al. Multifunctional oral delivery systems for enhanced bioavailability of therapeutic peptides/proteins. Acta Pharmaceutica Sinica B. 2019;9(5):902-922. DOI: 10.1016/j.apsb. 2019.01.004
  50. 50. Mastrolia I, Catani V, Oltrecolli M, Pipitone S, Vitale MG, Masciale V, et al. Chasing the Role of miRNAs in RCC: From Free-Circulating to Extracellular-Vesicle-Derived Biomarkers. Biology (Basel). 2023;12(6):877. DOI: 10.3390/biology12060877
  51. 51. Wu S, Li R, Jiang Y, Yu J, Zheng J, Li Z, et al. Liquid biopsy in urothelial carcinoma: Detection techniques and clinical applications. Biomedicine & Pharmacotherapy. 2023;165:115027. DOI: 10.1016/j.biopha.2023.115027
  52. 52. Capttaniou B, Bex A, et al. Epidemiology of renal cell carcinoma. European Urology. 2019;75(1):74-84
  53. 53. Qin ZY, Xu QW, Hu HH, et al. Extracellular vesicles in renal cell carcinoma: Multifaceted roles and potential applications identified by experimental and computational methods. Frontiers in Oncology. 2020;10:724
  54. 54. Vasconcelos MH, Caires HR, abols A, et al. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resistance Updates, 2019, 47: 100647.
  55. 55. Zieren RC, Dong L, Clark DJ, et al. Defining candidate mRNA and protein EV biomarkers to discriminate CCRCC and PRCC from non-malignant renal cells in vitro. Medical Oncology. 2021;38(9):105
  56. 56. Crentsil VC, Liu H, Sellitti DF. Comparison of exosomal microRNAs secreted by 786-O clear cell renal carcinoma cells and HK-2 proximal tubule derived cells in culture identified microRNA-205 as a potential biomarker of clear cell renal carcinoma. Oncology Letters. 2018;16(1):1285-1290
  57. 57. Xiao CT, Lai WJ, Zhu WA, et al. MicroRNA derived from circulating exosomes as noninvasive biomarkers for diagnosing renal cell carcinoma. Oncotargets and Therapy. 2020;13:10765-10774
  58. 58. Zhang W, Ni MW, Su Y, et al. MicroRNAs in serum exosomes as potential biomarkers in clear-cell renal cell carcinoma. European Urology Focus. 2018;4(3):412-419
  59. 59. Wang XG, Wang T, Chen CX, et al. Serum exosomal miR-210 as potential biomarker for clear cell renal cell carcinoma. Journal of Cellular Biochemistry. 2019;120(2):1492-1502
  60. 60. Muramatsu-Maekawa Y, Kawakami K, Fujita Y, et al. Profiling of serum extracellular vesicles reveals miRNA-4525 as a potential biomarker for advanced renal cell carcinoma. Cancer Genomics Proteomics. 2021;18(3):253-259
  61. 61. Song S, Long M, Guopeng Y, Cheng Y, Yang Q , Liu J, et al. Urinary exosome miR-30c-5p as a biomarker of clear cell renal cell carcinoma that inhibits progression by targeting HSPA5. Journal of Cellular and Molecular Medicine. 2019;23(10):6755-6765. DOI: 10.1111/jcmm.14553
  62. 62. Butz H, Nofech-Mozes R, Ding Q , et al. Exosomal microRNAs are diagnostic biomarkers and can mediate cell-cell communication in renal cell carcinoma. European Urology Focus. 2016;2(2):210-218
  63. 63. Glennon KI, Maralani M, Abdian N, et al. Rational development of liquid biopsy analysis in renal cell carcinoma. Carcers (Basel). 2021;13(22):5825
  64. 64. De Palma G et al. The three-gene signature in urinary extracellular vesicles from patients with clear cell renal cell carcinoma. Journal of Cancer. 2016;7(14):1960-1967. DOI: 10.7150/jca.16123. eCollection 2016
  65. 65. Himbert D et al. Characterization of CD147, CA9, and CD70 as tumor-specific markers on extracellular vesicles in clear cell renal cell carcinoma. Diagnostics (Basel). 2020;10(12):1034. DOI: 10.3390/diagnostics10121034
  66. 66. Vergori L et al. Circulating large extracellular vesicles carrying CA9 in the diagnosis and prognosis of clear-cell renal cell carcinoma. Clinical and Translational Medicine. 2021;11(3):e358. DOI: 10.1002/ctm2.358
  67. 67. Zhao YB, Wang YF, Zhao EY, et al. PTRF/CAVIN1, regulated by SHC1 through the EGFR oathway, is found in urine exosomes as potential biomarker of ccRCC. Carcinogenesis. 2020;41(3):274-283
  68. 68. Li J, Zhang G, Liu CG, et al. The potential role of exosomal circRNAs in the tumor microenvironment: Insights into cancer diagnosis and therapy. Theranostics. 2022;12(1):87-104
  69. 69. Jin CL, Shi LM, Li KL, et al. Mechanism of tumor? Derived extra-cellular vesicles in regulating renal cell carcinoma progression by the delivery of MALAT1. Oncology Reports. 2021;46(3):187
  70. 70. Wang L, Yang G, Zhao DF, et al. CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote miR-19b-3p. Molecular Cancer. 2019;18(1):86
  71. 71. Dias F, Teixeira AL, Nogueira I, et al. Extracellular vesicles enriched in hsa-miR-301a-3p and hsa-miR-1293 dynamics in clear cell renal cell carcinoma patients: Potential biomarkers of metastatic disease. Cancers (Basel). 2020;12(6):1450
  72. 72. Wang X-G, Zhu Y-W, Wang T, Chen B, Xing J-C, Xiao W. MiR-483-5p downregulation contributed to cell proliferation, metastasis, and inflammation of clear cell renal cell carcinoma. The Kaohsiung Journal of Medical Sciences. 2021;37:192-199
  73. 73. Kawakami K, Fujita Y, Kato T, et al. Diagnostic potential of serum extracellular vesicles expressing prostate-specific membrane antigen in urologic malignancies. Scientific Reports. 2021;11(1):15000
  74. 74. Deng H, Fan Y, Yuan F, Wang L, Hong Z, Zhan J, et al. Partial nephrectomy provides equivalent oncologic outcomes and better renal function preservation than radical nephrectomy for pathological T3a renal cell carcinoma: A meta-analysis. International Braz J Urol. 2021;47:46-60
  75. 75. Tsuruda M, Yoshino H, Okamura S, et al. Oncogenic effects of RAB27B through exosome independent functionin renal cell carcinoma including sunitinib-resistant. PLoS One. 2020;15(5):e0232545
  76. 76. Dias F, Teixeira AL, Nogueira I, et al. Plasma extracellular vesicle-derived TIMP-1 mRNA as a prognostic biomarker in clear renal cell carcinoma: A pilot study. International Journal of Molecular Sciences. 2020;21(13):4624
  77. 77. Zhang J, Wang WB, Chen SS, et al. Distinct non-invasive evaluation values of tumor-derived cell free microRNA, circulating microvesicles and exosomal microRNAs after renal carcinoma cryoablation. Biochemical and Biophysical Research Communications. 2018;503(3):1278-1283
  78. 78. Du M, Giridhar KV, Tian Y, Tschannen MR, Zhu J, Huang C-C, et al. Plasma exosomal miRNAs-based prognosis in metastatic kidney cancer. Oncotarget. 2017;8:63703-63714
  79. 79. Fujii N, Hirata H, Ueno K, Mori J, Oka S, Shimizu K, et al. Extracellular miR-224 as a prognostic marker for clear cell renal cell carcinoma. Oncotarget. 2017;8:109877-109888
  80. 80. Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, et al. Exosome-transmitted lncARSR promotes Sunitinib resistance in renal Cancer by acting as a competing endogenous RNA. Cancer Cell. 2016;29:653-668
  81. 81. Lv LL, Cao YH, Ni HF, et al. MicroRNA-29c in urinary exosome/microvesicle as a biomarker of renal fibrosis. American Journal of Physiology. Renal Physiology. 2013;305(8):F1220-F1227
  82. 82. Lv LL, Cao YH, Pan MM, et al. CD2AP mRNA in urinary exosome as biomarker of kidney disease. Clinica Chimica Acta. 2014;428:26-31
  83. 83. Feng Y, Lv LL, Wu WJ, et al. Urinary exosomes and exosomal CCL2 mRNA as biomarkers of active histologic injury in IgA nephropathy. The American Journal of Pathology. 2018;188(11):2542-2552
  84. 84. Sun IO, Santelli A, Abumoawad A, Eirin A, Ferguson CM, Woollard JR, et al. Loss of renal peritubular capillaries in hypertensive patients is detectable by urinary endothelial microparticle levels. Hypertension. 2018;72(5):1180-1188. DOI: 10.1161/hypertensionaha.118.11766
  85. 85. Trnka P, Ivanova L, Hiatt MJ, Matsell DG. Urinary biomarkers in obstructive nephropathy. Clinical Journal of the American Society of Nephrology. 2012;7:1567-1575
  86. 86. Khurana R, Ranches G, Schafferer S, Lukasser M, Rudnicki M, Mayer G, et al. Identification of urinary exosomal noncoding RNAs as novel biomarkers in chronic kidney disease. RNA. 2017;23:142-152. DOI: 10.1261/rna.058834.116
  87. 87. Furini G, Schroeder N, Huang L, Boocock D, Scarpellini A, Coveney C, et al. Proteomic profiling reveals the transglutaminase-2 externalization pathway in kidneys after unilateral ureteric obstruction. Journal of American Society Nephrology. 2018;29:880-905. DOI: 10.1681/ASN.2017050479
  88. 88. Yu Y, Bai F, Qin N, Liu W, Sun Q , Zhou Y, et al. Non-proximal renal tubule-derived urinary exosomal miR-200b as a biomarker of renal fibrosis. Nephron. 2018;139:269-282. DOI: 10.1159/000487104
  89. 89. Gudehithlu KP, Hart P, Joshi A, Garcia-Gomez I, Cimbaluk DJ, Dunea G, et al. Urine exosomal ceruloplasmin: A potential early biomarker of underlying kidney disease. Clinical and Experimental Nephrology. 2019;23:1013-1021. DOI: 10.1007/s10157-019-01734-5
  90. 90. Benito-Martin A, Ucero AC, Zubiri I, Posada-Ayala M, FernandezFernandez B, Cannata-Ortiz P, et al. Osteoprotegerin in exosome-like vesicles from human cultured tubular cells and urine. PLoS One. 2013;8:e72387
  91. 91. Asvapromtada S, Sonoda H, Kinouchi M, et al. Characterization of urinary exosomal release of aquaporin-1 and -2 after renal ischemia-reperfusion in rats. American Journal of Physiology. Renal Physiology. 2018;314(4):F584-F601
  92. 92. Qi Y, Wang X, Rose KL, et al. Activation of the endogenous renin-angiotensin-aldosterone system or aldosterone administration increases urinary exosomal sodium channel excretion. Journal of American Society Nephrology. 2016;27(2):646-656
  93. 93. Gonzales PA, Pisitkun T, Hoffert JD, et al. Large-scale proteomics and phosphoproteomics of urinary exosomes. Journal of American Society Nephrology. 2009;20(2):363-379
  94. 94. Corbetta S, Raimondo F, Tedeschi S, et al. Urinary exosomes in the diagnosis of Gitelman and Bartter syndromes. Nephrology, Dialysis, Transplantation. 2015;30(4):621-630
  95. 95. Dong L, Pietsch S, Englert C. Towards an understanding of kidney diseases associated with WT1 mutations. Kidney International. 2015;88(4):684-690
  96. 96. Zhou H, Cheruvanky A, Hu X, Matsumoto T, Hiramatsu N, Cho ME, et al. Urinary exosomal transcription factors, a new class of biomarkers for renal disease. Kidney International. 2008;74:613-621
  97. 97. Zhou H, Kajiyama H, Tsuji T, Hu X, Leelahavanichkul A, Vento S, et al. Urinary exosomal Wilms’ tumor-1 as a potential biomarker for podocyte injury. American Journal of Physiology. Renal Physiology. 2013;305:F553-F559
  98. 98. Abe H, Sakurai A, Ono H, et al. Urinary exosomal mRNA of WT1 as diagnostic and prognostic biomarker for diabetic nephropathy. The Journal of Medical Investigation. 2018;65(3.4):208-215
  99. 99. Sakurai A, Ono H, Ochi A, et al. Involvement of Elf3 on Smad3 activation-dependent injuries in podocytes and excretion of urinary exosome in diabetic nephropathy. PLoS One. 2019;14(5):e0216788
  100. 100. Moon PG, Lee JE, You S, Kim TK, Cho JH, Kim IS, et al. Proteomic analysis of urinary exosomes from patients of early IgA nephropathy and thin basement membrane nephropathy. Proteomics. 2011;11:2459-2475
  101. 101. Choi D-S. Urinary extracellular vesicles for biomarker source to monitor polycystic kidney disease. Proteomics. Clinical Applications. 2015;9(5-6):447-448. DOI: 10.1002/prca.201500053
  102. 102. Pocsfalvi G, Raj DA, Fiume I, Vilasi A, Trepiccione F, Capasso G. Urinary extracellular vesicles as reservoirs of altered proteins during the pathogenesis of polycystic kidney disease. Proteomics. Clinical Applications. 2015;9(5-6):552-567. DOI: 10.1002/prca.201400199
  103. 103. Kim SH, Choi SJ. Management of hypertension in Fabry disease. Electrolyte Blood Press. 2023;21(1):8-17. DOI: 10.5049/EBP.2023.21.1.8 Epub 2023 Jun 27
  104. 104. Rroji M, Figurek A, Spasovski G. Proteomic approaches and potential applications in autosomal dominant polycystic kidney disease and Fabry Disease. Diagnostics (Basel). 2023;13(6):1152. DOI: 10.3390/diagnostics13061152
  105. 105. Levstek T, Mlinšek T, Holcarm M, Goričar K, Lenassi M, Dolžan V, et al. Urinary extracellular vesicles and their miRNA cargo in patients with Fabry nephropathy. Genes (Basel). 2021;12(7):1057. DOI: 10.3390/genes12071057
  106. 106. Armstrong DA, Dessaint JA, Ringelberg CS, et al. Preanalytical handling conditions and small RNA recovery from urine for miRNA profiling. The Journal of Molecular Diagnostics. 2018;20(5):565-571
  107. 107. Sonoda H et al. Decreased abundance of urinary exosomal aquaporin-1 in renal ischemic-reperfusion injury. American Journal of Physiology. Renal Physiology. 2009;297:F1006-F1016
  108. 108. Zhou H et al. Exosomal Fetuin-A identified by proteomics: A novel urinary biomarker for detecting acute kidney injury. Kidney International. 2006;70:1847-1857
  109. 109. Barutta F et al. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One. 2013;8:e73798
  110. 110. Kalani A et al. Wilm’s tumor-1 protein levels in urinary exosomes from diabetic patients with or without proteinuria. PLoS One. 2013;8:e60177
  111. 111. Tao Y, Wei X, Yue Y, Wang J, Li J, Shen L, et al. Extracellular vesicle-derived AEBP1 mRNA as a novel candidate biomarker for diabetic kidney disease. Journal of Translational Medicine. 2021;19(1):326. DOI: 10.1186/s12967-021-03000-3
  112. 112. Zapała B, Kamińska A, Piwowar M, Paziewska A, Gala-Błądzińska A. Ewa Ł Stępień. miRNA signature of urine extracellular vesicles shows the involvement of inflammatory and apoptotic processes in diabetic chronic kidney disease. Pharmaceutical Research. 2023;40(4):817-832. DOI: 10.1007/s11095-023-03481-5
  113. 113. Kumari M et al. miR-451 loaded exosomes are released by the renal cells in response to injury and associated with reduced kidney function in human. Frontiers in Physiology. 2020;11:234
  114. 114. Min Q-H et al. Differential expression of urinary exosomal microRNAs in IgA nephropathy. Journal of Clinical Laboratory Analysis. 2018;32:e22226
  115. 115. Bruschi M, Granata S, Candiano G, Fabris A, Petretto A, Ghiggeri GM, et al. Proteomic analysis of urinary extracellular vesicles reveals a role for the complement system in medullary sponge kidney disease. International Journal of Molecular Sciences. 2019;20(21):5517. DOI: 10.3390/ijms20215517
  116. 116. 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. Cell. 2019;8:773
  117. 117. Dimuccio V et al. Urinary CD133+ extracellular vesicles are decreased in kidney transplanted patients with slow graft function and vascular damage. PLoS One. 2014;9:e104490
  118. 118. Alvarz S et al. Urinary exosomes as a source of kidney dysfunction biomarker in renal transplantation. Transplantation Proceedings. 2013;45:3719-3723
  119. 119. Herrmann IK, Wood M, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nature Nanotechnology. 2021;16(7):748-759
  120. 120. Varderidou-Minasian S, Lorenowicz MJ. Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: Challenges and opportunities. Theranostics. 2020;10(13):5979-5997
  121. 121. Timmers L, Lim SK, Arslan F, et al. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Research. 2007;1(2):129-137
  122. 122. Bruno S, Grange C, Deregibus MC, et al. Mesenchymal stem cellderived microvesicles protect against acute tubular injury. Journal of American Society Nephrology. 2009;20(5):1053-1067
  123. 123. Reis LA, Borges FT, Simões MJ, et al. Bone marrow-derived mesenchymal stem cells repaired but did not prevent gentamicininduced acute kidney injury through paracrine effects in rats. PLoS One. 2012;7(9):e44092
  124. 124. Bruno S, Grange C, Collino F, et al. Microvesicles derived from mesenchymal stem cells enhance survival in a lethal model of acute kidney injury. PLoS One. 2012;7(3):e33115
  125. 125. Zhu G, Pei L, Lin F, et al. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury via transferring miR-199a-3p. Journal of Cellular Physiology. 2019;234(12):23736-23749
  126. 126. Cao H, Cheng Y, Gao H, et al. In vivo tracking of mesenchymal stem cell-derived extracellular vesicles improving mitochondrial function in renal ischemia-reperfusion injury. ACS Nano. 2020;14(4):4014-4026
  127. 127. Cao JY, Wang B, Tang TT, et al. Exosomal miR125b-5p deriving from mesenchymal stem cells promotes tubular repair by suppression of p53 in ischemic acute kidney injury. Theranostics. 2021;11(11):5248-5266
  128. 128. Li X, Liao J, Su X, et al. Human urine-derived stem cells protect against renal ischemia/reperfusion injury in a rat model via exosomal miR-146a-5p which targets IRAK1. Theranostics. 2020;10(21):9561-9578
  129. 129. Ullah M, Liu DD, Rai S, et al. HSP70-mediated NLRP3 inflammasome suppression underlies reversal of acute kidney injury following extracellular vesicle and focused ultrasound combination therapy. International Journal of Molecular Sciences. 2020;21(11):4085
  130. 130. Cao H, Cheng Y, Gao H, et al. In vivo tracking of mesenchymal stem cell-derived extracellular vesicles improving mitochondrial function in renal IschemiaReperfusion injury. ACS Nano. 2020;14(4):4014-4026
  131. 131. Zhao M, Liu S, Wang C, et al. Mesenchymal stem cell-derived extracellular vesicles attenuate mitochondrial damage and inflammation by stabilizing mitochondrial DNA. ACS Nano. 2021;15(1):1519-1538
  132. 132. Zhang ZY, Hou YP, Zou XY, et al. Oct-4 enhanced the therapeutic effects of mesenchymal stem cell-derived extracellular vesicles in acute kidney injury. Kidney & Blood Pressure Research. 2020;45(1):95-108
  133. 133. Kilpinen L, Impola U, Sankkila L, et al. Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning. Journal of Extracellular Vesicles. 2013;2:21927. DOI: 10.3402/jev.v2i0.21927
  134. 134. Zhang X, Wang N, Huang Y, et al. Extracellular vesicles from three dimensional culture of human placental mesenchymal stem cells ameliorated renal ischemia/reperfusion injury. The International Journal of Artificial Organs. 2021;45(2):181-192. DOI: 10.1177/0391398820986809
  135. 135. Li X, Liao J, Su X, et al. Human urine-derived stem cells protect against renal ischemia/reperfusion injury in a rat model via exosomal miR146a-5p which targets IRAK1. Theranostics. 2020;10(21):9561-9578
  136. 136. Grange C, Papadimitriou E, Dimuccio V, et al. Urinary extracellular vesicles carrying klotho improve the recovery of renal function in an acute tubular injury model. Molecular Therapy. 2020;28(2):490-502
  137. 137. Wang Y, Guo YF, Fu GP, et al. Protective effect of miRNA-containing extracellular vesicles derived from mesenchymal stromal cells of old rats on renal function in chronic kidney disease. Stem Cell Research & Therapy. 2020;11(1):274
  138. 138. Choi HY, Lee HG, Kim BS, et al. Mesenchymal stem cell-derived microparticles ameliorate peritubular capillary rarefaction via inhibition of endothelialmesenchymal transition and decrease tubulointerstitial fibrosis in unilateral ureteral obstruction. Stem Cell Research & Therapy. 2015;6:18
  139. 139. Shi Z, Wang Q , Zhang Y, et al. Extracellular vesicles produced by bone marrow mesenchymal stem cells attenuate renal fibrosis, in part by inhibing the RhoA/ROCK pathway, in a UUO rat mode. Stem Cell Research & Therapy. 2020;11(1):253
  140. 140. Ramirez-Bajo MJ, Martin-Ramirez J, Bruno S, et al. Nephroprotective potential of mesenchymal stromal cells and their extracellular vesicles in a murine model of chronic cyclosporine nephrotoxicity. Frontiers in Cell and Development Biology. 2020;8:296. DOI: 10.3389/fcell.2020.00296 eCollection 2020
  141. 141. Renzhi H, Li X, Peng C, Gao R, Ma L, Jinbo H, et al. miR-196b-5p-enriched extracellular vesicles from tubular epithelial cells mediated aldosterone-induced renal fibrosis in mice with diabetes. BMJ Open Diabetes Research & Care. 2020;8(1):e001101. DOI: 10.1136/bmjdrc-2019-001101
  142. 142. Hong S, Qiao J, Jinxiu H, Li Y, Lin J, Qun Y, et al. Podocyte-derived extracellular vesicles mediate renal proximal tubule cells dedifferentiation via microRNA-221 in diabetic nephropathy. Molecular and Cellular Endocrinology. 2020;518:111034. DOI: 10.1016/j.mce.2020.111034
  143. 143. Kholia S, Sanchez MBH, Deregibus MC, Sassoè-Pognetto M, Camussi G, Brizzi MF. Human liver stem cell derived extracellular vesicles alleviate kidney fibrosis by interfering with the β-catenin pathway through miR29b. International Journal of Molecular Sciences. 2021;22(19):10780. DOI: 10.3390/ijms221910780
  144. 144. Ko KW, Park SY, Lee EH, et al. Integrated bioactive scaffold with polydeoxyribonucleotide and stem-cell derived extracellular vesicles for kidney regeneration. ACS Nano. 2021;15(4):7575-7585
  145. 145. Eirin A, Zhu XY, Puranik AS, et al. Mesenchymal stem cell-derived extracellular vesicles attenuate kidney inflammation. Kidney International. 2017;92(1):114-124
  146. 146. Yea J-H, Yoon YM, Lee JH, Yun CW, Lee SH. Exosomes isolated from melatonin-stimulated mesenchymal stem cells improve kidney function by regulating inflammation and fibrosis in a chronic kidney disease mouse model. Journal of Tissue Engineering. 2021;12:20417. DOI: 10.1177/20417314211059624
  147. 147. Choi HY, Kim TY, Lee M, et al. Kidney mesenchymal stem cell-derived extracellular vesicles engineered to express erythropoietin improve renal anemia mice with chronic kidney disease. Stem Cell Reviews and Reports. 2022;18(3):980-992
  148. 148. GBD Chronic Kidney Disease Collaboration. Global, regional, and national burden of chronic kidney disease, 1990-2017: A systematic analysis for the global burden of disease study 2017. Lancet. 2020;395(10225):709-733
  149. 149. Decleves AE, Sharma K. New pharmacological treatments for improving renal outcomes in diabetes. Nature Reviews. Nephrology. 2010;6(6):371-380
  150. 150. Jin J, Shi Y, Gong J, et al. Exosome secreted from adipose-derived stem cells attenuates diabetic nephropathy by promoting autophagy flux and inhibiting apoptosis in podocyte. Stem Cell Research & Therapy. 2019;10(1):95
  151. 151. Xiang E, Han B, Zhang Q , et al. Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Research & Therapy. 2020;11(1):336
  152. 152. Nagaishi K, Mizue Y, Chikenji T, et al. Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes. Scientific Reports. 2016;6:34842
  153. 153. Ebrahim N, Ahmed IA, Hussien NI, et al. Mesenchymal stem cell derived exosomes ameliorated diabetic nephropathy by autophagy induction through the mTOR signaling pathway. Cell. 2018;7(12):226
  154. 154. Nassar W et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomaterial Research. 2016;20:21
  155. 155. Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle drug delivery system: The anti-inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Molecular Therapy. 2010;18(9):1606-1614
  156. 156. O’Brien K, Breyne K, Ughetto S, et al. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nature Reviews. Molecular Cell Biology. 2020;21(10):585-606
  157. 157. Tang TT, Wang B, Li ZL, et al. Kim-1 targeted extracellular vesicles: A new therapeutic platform for RNAi to treat AKI. Journal of American Society Nephrology. 2021;32(10):2467-2483
  158. 158. Wang B, Zhang A, Wang H, et al. MiR-26a limits muscle wasting and cardiac fibrosis through ExosomeMediated microRNA transfer in chronic kidney disease. Theranostics. 2019;9(7):1864-1877
  159. 159. Wang H, Wang B, Zhang A, et al. ExosomeMediated miR-29 transfer reduces muscle atrophy and kidney fibrosis in mice. Molecular Therapy. 2019;27(3):571-583
  160. 160. Wang B, Yao K, Huuskes BM, Shen HH, Zhuang J, Godson C, et al. Mesenchymal stem cells deliver exogenous MicroRNA-let7c via exosomes to attenuate renal fibrosis. Molecular Therapy. 2016;24:1290-1301
  161. 161. He J, Jiang YL, Wang Y, Tian XJ, Sun SR. Micro-vesicles from mesenchymal stem cells over-expressing miR-34a inhibit transforming growth factor-β1-induced epithelial-mesenchymal transition in renal tubular epithelial cells in vitro. Chinese Medical Journal. 2020;133:800-807
  162. 162. Tang TT, Wang B, Wu M, et al. Extracellular vesicleencapsulated IL-10 as novel nanotherapeutics against ischemic AKI. Science Advances. 2020;6(33):eaaz0748
  163. 163. Kim S, Lee SA, Yoon H, et al. Exosome-based delivery of super-repressor IkappaBalpha ameliorates kidney ischemia-reperfusion injury. Kidney International. 2021;100(3):570-584
  164. 164. Zhang C, Shang Y, Chen X, Midgley AC, Wang Z, Zhu D, et al. Supramolecular nanofibers containing arginine-Glycine-aspartate (RGD) peptides boost therapeutic efficacy of extracellular vesicles in kidney repair. ACS Nano. 2020;14(9):12133-12147. DOI: 10.1021/acsnano.0c05681
  165. 165. Witwer KW, Wolfram J. Extracellular vesicles versus synthetic nanoparticles for drug delivery. Nature Reviews Materials. 2021;6(2):103-106
  166. 166. Tang TT, Lv LL, Wang B, et al. Employing macrophage-derived microvesicle for kidney-targeted delivery of dexamethasone: An efficient therapeutic strategy against renal inflammation and fibrosis. Theranostics. 2019;9(16):4740-4755
  167. 167. Grangier A, Branchu J, Volatron J, et al. Technological advances towards extracellular vesicles mass production. Advanced Drug Delivery Reviews. 2021;176:113843

Written By

Chunyan Lv

Submitted: 18 July 2023 Reviewed: 13 September 2023 Published: 03 November 2023