Review Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Clin Urol. Jul 24, 2014; 3(2): 66-80
Published online Jul 24, 2014. doi: 10.5410/wjcu.v3.i2.66
Employing extracellular vesicles for non-invasive renal monitoring: A captivating prospect
Gabriella Pocsfalvi, Christopher Stanly, Annalisa Vilasi, Immacolata Fiume, Institute of Biosciences and BioResources, National Research Council of Italy, 80131 Naples, Italy
Rosarita Tatè, Institute of Genetics and Biophysics, National Research Council of Italy, 80131 Naples, Italy
Giovambattista Capasso, Division of Nephrology, Department of Cardio-Vascular Sciences, Second University of Naples, 80131 Naples, Italy
Author contributions: Pocsfalvi G substantial contribution to conception, design, acquisition, analysis and interpretation of data, drafting of the article; Stanly C carried out experiments, analyzed data and contributed to drafting of the article; Vilasi A analyzed data and contributed to drafting of the article; Fiume I carried out experiments, analyzed data; Tatè R carried out experiments and analyzed data; Capasso G conceived experiments, provided access to patients’ samples; all authors contributed to the final manuscript version.
Supported by “Ricercando 2009” funded by the Italian Nephrology Society (SIN)
Correspondence to: Gabriella Pocsfalvi, PhD, Institute of Biosciences and BioResources, National Research Council of Italy, via P. Castellino, 111, 80131 Naples, Italy. gabriella.pocsfalvi@ibbr.cnr.it
Telephone: +39-81-6132585 Fax: +39-81-6132646
Received: April 29, 2014
Revised: June 6, 2014
Accepted: June 27, 2014
Published online: July 24, 2014

Abstract

Extracellular vesicles (EVs) are fascinating nano-sized subjects extensively studied over the recent years across several disparate disciplines. EVs are endlessly secreted into the extracellular microenvironment by most cell types under physiological and pathological conditions. EVs encompass a variety of molecular constituents from their cell of origin, such as lipids, cell specific proteins and RNAs, thus constituting an informative resource for studying molecular events at the cellular level. There are three main classes of EVs classified based on their size, content, biogenesis and biological functions: exosomes, shedding microvesicles and apoptotic bodies. Besides cell culture supernatants, biological fluids have also been shown to contain different types of EVs. Amongst the various body fluids, the study of urinary extracellular vesicles (uEVs) as a source of candidate biomarkers gained much attention, since: (1) urine can be non-invasively collected in large amounts; and (2) the isolated uEVs are stable for a relatively long period of time. Here, we review the important aspects of urinary extracellular vesicles which are fast gaining attention as a promising future tool for the non-invasive monitoring of urinary tract. Recent advancements in the purification and analysis of uEVs and collection of their constituents in rapidly developing public databases, allow their better exploitation in molecular diagnostics. As a result, a growing number of studies have shown that changes in expression profile at the RNA and/or protein levels of uEVs reveal the molecular architectures of underlying key pathophysiological events of different clinically important diseases with kidney involvement.

Key Words: Urine analysis, Extracellular vesicles, Biomarkers, Kidney disease, Quantitative proteomics, Exosomes

Core tip: Urinary extracellular vesicles research is a fast growing field of biomarker discovery providing new attracting prospective for monitoring tissue alteration in easily accessible clinical samples. Over the past ten years intense research has identified the various urinary vesicular cargo molecules (i.e., RNAs, proteins and lipids) and detected their alterations upon a number of renal diseases. With the number of diseases relating to kidney increasing it is essential to effectively utilize this invaluable tool for the early diagnosis. Here we provide a comprehensive overview of uEVs nicely setting the stage for their utility in future clinical diagnostics.



EXTRACELLULAR VESICLES: WHY ARE THEY FASCINATING?

Extracellular vesicles (EVs) are membrane-bound secretory vesicles which exhibit an array of proteins, bioactive lipids, nucleic acids and metabolites[1-4]. EVs are considered to play an important role in intercellular communication[5-8], regulating immune response[9-12], antigen presentation[13-15], transport and propagation of infectious cargo such as prions and retroviruses[2,16-18]. Based on the size and origin, EVs are classified into three main types: (1) “exosomes” (40-100 nm) vesicles derived from the endosomal compartment and released via fusion of the multivesicular bodies with the plasma membrane; (2) “microvesicles” (50-1000 nm) vesicles that result from the direct budding from the plasma membrane; and (3) “apoptotic bodies” (800-5000 nm) vesicles released by cells undergoing programmed cell death[19,20]. Despite some distinct features, numerous similarities exist among the different classes of EVs with respect to their physical characteristics and biochemical composition, which make the separation of different subsets challenging[21].

Nano- to micron-sized EVs have been isolated from many body fluids including urine[16,22-25], saliva[26,27], breast milk[9,27], cerebrospinal fluid[28], semen, pleural effusions and plasma[27,29]. Urinary extracellular vesicles (uEVs) are released from the renal epithelial cells, including glomerular podocytes, renal tubule cells, and the cells lining the drainage system[30]. They are promising starting material for biomarker discovery and also a great asset for non-invasive renal monitoring as they provide a full representation of the entire urinary system[3,16]. Recently developed quantitative workflows for uEVs transcriptomics, proteomics and lipidomics[31-34] including system biology approaches[35] enable researchers to study the expression profiles of the main bioactive constituents of uEVs in detail and with an increased rate and reproducibility than before. Research performed in the last 10 years highlighted that uEVs harbor 1%-3% of the total urinary proteins with a reduced dynamic range of protein concentration respect to the whole urine proteome. In this sense, the proteome of uEVs which today counts more than four thousand proteins can be considered as the urinary subproteome enclosed in double-layered vesicles. Despite the potential benefits of uEV analysis, there are barriers and limitations that must be dealt with. Foremost among these challenges is the reproducible isolation of pure vesicles suitable for downstream analysis[36]. In addition to proteins, uEVs encapsulate different RNA species amongst which messenger RNAs (mRNAs) and micro-RNAs (miRNAs) are the subjects of intense studies as possible biomarkers of renal diseases[3,37]. RNAs incorporated in uEVs are resistant to nuclease digestion and, similarly to proteins they originated from the different nephron regions[38]. For example, podocyte related mRNA expression of CD2-associated protein in uEVS was shown to correlate with renal function and level of proteinuria[39]. Moreover, miRNA-29c level in uEVs was shown to be a potential biomarker of renal fibrosis[40]. The relative abundance of different mRNAs do not necessarily correlate with cellular mRNA, suggesting a selective process of mRNA entry into vesicles[1]. As vesicle-enclosed mRNAs travel along the nephron, modification of nucleic acid cargo could permit targeted delivery of RNA to the kidney, a hypothesis which open a new area for the treatment of renal tubular disorders[41].

Here, we will focus on summarizing the current knowledge about uEVs, starting from the critical review of their isolation, biochemical characterization till their potential application in biomarker discovery and non-invasive renal monitoring.

URINARY SYSTEM ORIGIN OF uEVS

Exosomes are cell-derived secretory vesicles shed by proliferating cells through exocytosis[6,13,29]. During endosomal maturation, the formation of intraluminal vesicles (ILVs), ranging from 30-100 nm in diameter, inside the lumen of the endosome can be observed. The ILVs are generated by inward invagination of the endosomal membrane, and scission of vesicles from the limiting membrane into the endosomal lumen. The size of a fully matured late endosome, also called multivesicular body (MVB) is approximately 500 nm in diameter and, it contains several ILVs. The fate of MVBs may vary. They could fuse with lysosome and get degraded or fuse with the plasma membrane releasing the vesicles into the extracellular space. During the later process, the second inward budding of the endosome membrane results in a positive orientation of the ILVs lipid membrane. While the endosomal sorting complexes required for transport (ESCRT) machinery involved in sorting the cargo to lysosomes via MVBs within the endocytic system is well characterized (ESCRT-dependent endolysosomal pathway), protein sorting into exosomes until recently was less understood[6]. The involvement of ESCRT protein complexes and protein ubiquitination has been shown by different groups. Recently, Alvavez et al[3] revealed that syndecan-syntenin-ALIX might be a key regulator of the biogenesis of syntenin expressing exosomes. In particular, the interaction between syndecan and exosomes might thus support a novel role for proteoglycans in vesicular trafficking and cellular signaling. On the other hand, EVs displaying similar physicochemical characteristics like exosomes (density, size-distribution, presence of protein markers) have also been described to bud from the plasma membrane. For example, podocyte-positive membrane vesicles in urine has been shown to originate from tip vesiculation of podocyte microvilli[42]. These vesicles however cannot be easily distinguished from endosome-derived exosomes and therefore their separate analysis remains a problem.

Exosomes are released from cells of different tissues or organs and while they share a common group of proteins related to their biogenesis they also harbor tissue specific proteins that reflect the origin and biological functions of their parental cells[43]. In fact, EVs isolated from biological fluids have organ and tissue specific protein and RNA profiles. The urinary system consists of the two kidneys, ureters, the bladder, and the urethra. Nephrons are the kidney functional and structural units with the main role to regulate water balance and inorganic ions by filtering the blood. More than a decade ago, the key finding of the first proteomic analyses performed on isolated uEVs was the presence of proteins known to be highly and differentially expressed in various kidney tissues from the glomerular podocyte to the epithelial cells lining the various nephron segments[44,45]. Later on proteins from the transitional epithelium of the urinary bladder were also identified, confirming uEVs may be shed from cells throughout the renal[46]. Targeted proteomics applied recently to perfused isolated rat kidney model to identified kidney originated proteins in human urine[47]. Out of the 990 kidney originated proteins with human analogues 74 were present only in plasma and 240 only in urine (including uEVs) but not in plasma. Screening the 240 kidney originated urinary proteins we have found that the major part (206) is listed in the human urinary dataset of EVpedia 2.0 database. This could be a core set of kidney originated vesicle proteins with high renal pathophysiology relevance for further studies.

Figure 1 shows a schematic diagram of a nephron indicating sets of uEVs proteins highly expressed in the epithelial cells of glomerular podocyte[48] and to the different segments of renal tubule[44,45,49]. Podocalyxin (PODXL) specific for glomerular epithelial cells, aminopeptidase (AMPN) specific for proximal tubule cells, AQP2 specific for distal tubule, programmed cell death 6 interacting protein (PDCD6IP or ALIX) and uroplakin-1 and uroplakin-2 (UPK1 and UPK2) specific for bladder are frequently used as protein markers of human uEVs. Basal epithelial cells of collecting duct are hypothesized to be the major contributors toward the urinary pool of EVs including the expression of leucine-rich repeat kinase-2 (LRRK2)[50]. More recently, Benito-Martin et al[49] showed that cultured human proximal tubular cell secrete exosomes containing osteoprotegerin (OPG) and this specific sub-group of exosomes can also be found in urine. Since, most of these have kidney relevant function, proteomic analysis of uEVs hold the promise to provide an insight into the physiological or pathophysiological processes in the various cell types facing the urinary space[44].

Figure 1
Figure 1 Schematics of Nephron showing the urinary extracellular vesicle proteins identified in the different segments. A: Core; B: Glomerulus/podocyte; C: Proximal convoluted tubule (PT); D: Ascending Limb of (LoH); E: Collecting duct (CD); F: Distal convoluted tubule (DT). UniProt Acc.Number: UniProt Database Accession Number. LoH: Loop of Henle.
VESICLE ISOLATION: AN OBSTACLE TO OVERCOME

Isolation of EVs is still a major challenge in this rapidly growing field of research. The complex nature of body fluids and lack of standardized protocols makes isolation and characterization extremely difficult[34]. The research on EVs can be broadly divided into three categories depending on the end use of the vesicles: (1) discovery; (2) diagnostic; and (3) preparative, each of which demands a different level of purity, quality control and operating procedures. In most studies, ultracentrifugation is the commonly used technique for isolation of vesicles. Isolation of membrane vesicles by sequential differential centrifugations is complicated by the possibility of overlapping size distributions with microvesicles or macromolecular complexes. Recently, numerous alternate procedures were introduced, including immunoaffinity separation, filtration, microfluidic devices aided separation, and reagent based separation[20]. The choice of isolation procedure greatly depends on the source material and the goal of the EV research project.

Isolation of extracellular vesicles from urine has proven to be extremely difficult because of the presence of Tamm-Horsfall protein (THP), also known as uromodulin[36], and also due to very low vesicle load in this biofluid. Differential centrifugation is the most widely used technique for the isolation of uEVs. It includes several low-velocity centrifugation and ultracentrifugation steps with increasing centrifugal force; from 200 to 1500 g to remove cells and cellular debris, from 10000 to 20000 g to pellet microvesicles, and from 100000 to 200000 g to pellet nanometer-sized vesicles[50]. The efficiency to isolate EV depends not only on the size, shape and density of vesicles but on the volume, viscosity and temperature of the fluid in which the vesicles are present. Centrifugation time and the type of rotor used (fixed angle or swinging bucket) also influence the final yield and the purity of vesicles. Addition of dithiothreitol (DTT)[23,36,49,51] and 3-[(3-cholamidopropyl) dimethylammonio]-1​-propanesulfonate (CHAPS)[52] to the low-speed pellet has been shown to be efficient to disrupt the polymeric network formed by THP protein filaments, but do not solve the problem entirely[33,53]. Urinary EVs can be separated from non-membranous particles, such as protein aggregates by using their relatively low buoyant density, and differences in floatation velocity to separate differently sized classes of EV. Based on this, the crude uEVs containing pellet obtained by differential centrifugation can be further separated/purified using a sucrose density gradient[54,55] or sucrose cushion ultracentrifugation step[33,56,57]. It has been shown that both methods are efficient not only to separate exosomes from microvesicles but also to eliminate the interfering THP impurity[33,54-56]. The high sucrose concentration used in sucrose gradient centrifugation, however may negatively affect the biological function of EV. This can be avoided by layering the samples on top of the sucrose gradient or cushion in the tube subjected for centrifugation. Filtration through molecular filters (0.22-μm or 0.1-μm filters), which remove solvent and small molecule analytes while retaining and concentrating vesicles smaller than the pore size can also be used alone or in combination with centrifugation or ultrafiltration (MWCO 100 kDa) to isolate uEVs[25,58-60]. Among the primary advantages of the filtration method are the simplicity and easy scale-up. Though co-purification of abundant soluble urinary proteins and THP often compromises sample purity and the applied increasing forces with decreasing pore size has also been reported to result in artifacts[50]. The presence of characteristic surface proteins (CD9, CD63, CD81, Rab-5b, TSG101, Alix and A33) on certain EV classes is the basis for immunoaffinity isolation[57,61]. Immunoaffinity-based techniques employ antibodies thus have the potential for high specificity, an important consideration in the characterization of specific EV populations[57]. Because of the increasing interest in exosomes and other extracellular vesicles and their potential use in therapeutics or as biomarkers for disease, kits that allow for “easy isolation procedures” are being developed[16,62]. Commercial tests based on centrifugation (Total Exosome Isolation Reagent, Invitrogen™), filtration (Exomir™ and Exo-Spin™), affinity capturing (Exotest™ and Exosome Dynabeads®) and proprietary exosome precipitation technologies (miRCURY™, ExoQuick-TC™ and DiagExo®) are already available in the market (Table 1). Such approaches for rapid purification are welcomed but they should be only considered as for bulk isolations because they often fail to pass quality test and fail to distinguish between differently sized EV and membrane-free macromolecular aggregates[50]. A widely accepted unique standardized protocol for the reproducible isolation and purification of uEVs suitable for the downstream analysis of the various RNA and protein constituents is still awaited.

Table 1 Methods used for the isolation of urinary extracellular vesicles.
TechniqueTypeAdvantage(s)Disadvantage (s)PurityRef.
CentrifugationDifferentialEasy to performCo-purification of protein aggregates (THP) Inability to separate exosomes from microvesicles Lengthy Impractical for large-scale studies++[37,44,50,52,69,92]
Sucrose gradientNo mechanical stress and hence allows the collection of morphologically intact particles Removes THPHigh sucrose concentration may affect the biological functions of exosomes Inability to separate exosomes from particles with similar density and size Difficult and lengthy Impractical for large-scale studies+++++[23,50,54,55]
Sucrose cushionNo mechanical stress and hence allows the collection of morphologically intact particlesInability to separate exosomes from particles with similar density and size Difficult and lengthy Impractical for large-scale studies+++++[33,56,57,78,80]
FiltrationNanofiltrationEasy to performLow purity grade Co-purification of protein aggregates (THP) Low exosome yield due to their lost on the surface of the nano-membrane+++[94]
MicrofiltrationEasy to perform Rapid Maintain vesicle structureCo-purification of protein aggregates (THP) Inability to separate exosomes from microvesicles+++[59]
Immunoaffinity separationImmunobeadsAllow rapid semi quantitative characterization of the surface phenotype can be tissue-specificNot suited for large sample volumes Captured extracellular vesicles may not retain biological functionality even if successfully eluted from bead surface Co-purification of protein aggregates (THP) Low yield++[57,61,62]
Commercial kitsTotal Exosome Isolation Reagent-Invitrogen™ Exomir™ Exo-Spin™ Exotest™ Exosome Dynabeads® miRCURY™ ExoQuick-TC™ and DiagExo®Rapid and requires low sample volume PracticalImpractical for large-scale studies Reproducibility, yield and sample quality should be checked case by case Co-purification of protein aggregates (THP)++
PHYSICOCHEMICAL AND ANALYTICAL CHARACTERIZATION OF URINARY EXTRACELLULAR VESICLES

Isolated uEVs are a heterogeneous vesicle population. Exosomes have a lipid bilayer membrane and a characteristic buoyant density ranging from 1.10 to 1.19 g/mL[55]. Determination of physicochemical properties, like integrity, morphology, size and concentration is generally the first step in the characterization of uEVs. Transmission electron microscopy (TEM)[16,23,33,52,58,59,63-65], dynamic light scattering (DLS)[66], nanoparticle tracking analysis (NTA)[67,68] and flow cytometry (FC)[69] are the methods most frequently used in this process. TEM imaging of numerous uEV samples has revealed intact vesicles with round morphology[16,23,33,52,58,59,63-65]. Depending on the isolation/purification method used, uEV samples typically show a heterogeneity in size ranging from approximately 30-100 nm in diameter (Figure 2A). Besides the determination of particle concentration and size distribution, the main advantage of using transmission electron microscopy (TEM) for uEV analysis is its ability to reveal the presence of known urinary contamination, like the long polymers of THP[36,52]. TEM[16,23,33,52,58,59,63-65] and cryo-EM[68,70] based morphological characterization, including observation of vesicle heterogeneity, therefore is a great support for the quality assessment of sample preparation[64]. Orthogonal techniques, like Dynamic Light Scattering (DLS) and nanoparticle Tracking Analysis (NTA) on the other hand, measures the Brownian motion of the particles in solution and calculate the hydrodynamic radii of the particles. Owning to its rapidity and simplicity, NTA is a quickly expanding in the field of exosome research. The mean diameter of human uEVs isolated by ultracentrifugation was measured by NTA around 150 nm ± 50 nm and concentration 10E9-10E11 particles/mL (Figure 2B)[68]. It should be noted that the hydrodynamic size of nanoparticles is expected to be higher to the core size measured with TEM, however the relationship between the two still need to be determined. Subpopulation of EVs can be studied by immunoelectron microscopy using antibodies to target membrane proteins, like aquaporin-2 (AQP2), aminopeptidase-N (AMPN), podocalixin (PODXL) and CD9[37,42,65,71]. Oosthuyzen et al[66] most recently demonstrated that NTA can also monitor specific subgroups of uEVs particles using fluorescent antibodies against specific surface proteins present on uEVs.

Figure 2
Figure 2 (A) Transmission electron microscopy and (B) nanoparticle tracking analysis images of urinary extracellular vesicles isolated by the sucrose cushion ultracentrifugation method.

Analytical characterization of the components of urinary nano-vesicles (i.e., proteins, mRNAs, mi-RNAs, lipids and small molecules) can be performed by transcriptomic[31,37,40,46,63,72,73], proteomic[16,19,23,25,31,33,35,44,47,48,50,52,59,60,62,63,65,66,71,74-79], metabolomic and lipidomic[2,31,46,68,75,80] tools[34]. One of the most exciting findings is that RNA transcripts exist in exosomes and maintain their function when transferred to other cells. Vesicle mediated route for cell-cell communication within the urinary tract opens new perspectives, yet its role in kidney development, function and pathogenesis needs to be elucidated[46]. Typically, commercially available extraction kits are used for total RNA isolation. Abundance of most RNAs is low in the in uEV which makes isolation and downstream RNA analysis challenging[37]. Most of the studies so far have used microarrays[72,81] or real time quantitative PCR (qPCR) assays[39,48,72] to examine exosomal RNAs, with a focus on miRNAs. Because of the inherent limitations of these methods, unknown miRNAs or other RNA species are often overlooked. To characterize RNA profiles systemically, Cheng et al[73] performed next-generation deep sequencing providing the base to identify miRNA biomarkers in urinary exosomes.

Since the first proteomic profiling reported by Pisitkun et al[44], protein content of urinary EVs has been extensively studied[16,19,23,25,31,33,35,41,45,47-50,52-54,59,60,62,63,65,66,70,71,74-80] both under healthy and numerous disease conditions. These studies let to comprehensive datasets of the identified proteins collected in the different exosome related public databases. It should be noted, that for the generation of many of the large datasets, uEV samples isolated by differential centrifugation (crude exosome preparation) was used[23,45]. As a consequence, public databases contain a relatively high percentage of urinary and other impurities related proteins. The protein content of urinary exosomes is about 1%-3% of that of total urine. The proteomic workflow generally starts with protein concentration assay (Bradford or BCA) followed by protein profile analysis which is usually performed by visualization of proteins separated by SDS-PAGE. The band at about 90 kDa corresponds to the highly glycosylated THP monomer[36] and its relative abundance is used to assess sample quality. Western blots are performed by using a set of urinary exosomal markers, such as Alix, CD9, TSG101, PODXL, AQP2, NES and Annexin V. Chemiluminescent western blotting gives valuable information for the quality of the preparation, but its use as a quantitative method for measuring the relative expression of the target proteins in the absence of appropriate normalization method is still debatable. To measure differences in protein expression between samples, quantitative proteomics is the method of choice. Both label-free quantitation[16,25,76] and stable isotope labeling by iTRAQ[33,78] and TMT reagents[50] in combination with SCX/C18 Multidimensional Protein Identification Technology (MudPIT) have been shown to be useful in the comparative analysis of uEVs. After appropriate enrichment steps, post translational modifications (PTMs), phosphorylation[45,50,79] and glycosylation[64,69] have also been studied by mass spectrometry-based proteomics in urinary exosomes. Since PTMs may affect important physiological process and its alterations may directly reflect early pathogenic events, further studies of PTMs alteration in uEV in various disease states are expected to come.

The bioactivity of exosomes is associated not only to their protein and RNA contents but also to their lipids. Compared to their cells, exosomes have been shown to be enriched in Cholesterol and Sphingomyelin. Total exosomal lipids can be extracted by organic solvents according to Bligh and Dyer[82] or, alternatively using THF:H2O (4:1)[2]. Different lipid classes (phospholipids and glycosphingolipids) can be purified and analyzed by thin layer chromatography (TLC), gas chromatography-mass spectrometry GC-MS and liquid chromatography-mass spectrometry (LC-MS). Recent progresses in electrospray ionization-tandem mass spectrometry-(ESI-MS/MS) based high-throughput lipidomics allowed the first comparative lipid analysis uEVs isolated from healthy individuals and renal cell carcinoma patient[2].

DATABASES FOR URINARY EXTRACELLULAR VESICLES RESEARCH

Recent studies have ignited significant interest on uEVs as possible players in kidney physiology and also as potential reservoirs of biomarkers. With such a huge interest, the amount of data accumulated has increased over time. High throughput proteomics study routinely identifies more than thousand proteins in human uEVs[23,45,50] and necessitates systematic classification of the data acquired[83]. Currently, there are two integrated manually curated web-based databases publicly available: Vesiclepedia[84] and EVpedia[85]. Both dedicate separate sections for the constituents of human urinary vesicles.

Vesiclepedia 2.1 (http://www.microvesicles.org) catalogs information from published non-mammalian eukaryotic and mammalian extracellular vesicles. Based on the 15 studies published on human uEVs, the current version contains 1162 unique proteins, 20 unique mRNAs and 690 unique miRNAs. To aid biomedical scientists in assessing the quality of the preparation Vesiclepedia also contains information on the methods used for the purification as well on the biophysical and molecular characterization of EVs.

EVpedia 2.0 (http://evpedia.info) provides a comprehensive lists of proteins, mRNAs, miRNAs, and lipids identified in EVs of both eukaryotic and prokaryotic (bacteria and archaea) origin, including non-mammalian eukaryotic extracellular vesicles[31,85]. Currently, out of the 263 studies included in this database 16 deal with urinary EVs. EVpedia 2.0 is cataloging a total of 12869 proteins isolated from human uEVs out of which 6275 are unique. 10 studies are related to the study of uEVs isolated from healthy donors and the number of unique proteins in this set is 4536. The most frequently identified 100 proteins are also listed in the database. Sets of proteins identified in different disease related samples (312 from early IgA nephropathy, 552 from autosomal recessive polycystic kidney disease, 621 from basement membrane nephropathy, 689 from bladder cancer, 480 from hernia, 184 from renal cell carcinoma) are particularly valuable components of this database. In the present 2.0 release of EVpedia there are no mRNAs, miRNAs and metabolites in the human urinary EVs subset reported. Regarding lipids, 28 different lipids were identified in human uEVs from healthy donors and 26 lipids were found in patients with renal cell carcinoma. Moreover, EVpedia offers a range of analytical tools: (1) search for and browse vesicular components; (2) Gene Ontology enrichment analysis of vesicular components; (3) network analysis of vesicular components; and (4) set analysis: a comparison of vesicular datasets by ortholog identification. Detailed methods for the isolation of extracellular vesicles and publications on extracellular vesicles are also listed in this database. An overall comparison of EVpedia with Vesiclepedia is beyond the scope of this paper and has recently been published[31,85]. Regarding urinary EVs, the current release of EVpedia contains more data on proteins and lipids while miRNAs and mRNAs are more represented in Vesiclepedia. There is a concern that a relatively high percentage of the entries in the single datasets included in the databases are not native constituents of uEVs but matrix impurities related to isolation/purification[83,86]. Complete workflow solution, starting from fast and efficient purification of EVs from urine without contamination by non-vesicular components (including abundant urinary proteins, protein aggregates), is a critical prerequisite for future high-throughput analyses. Important prerequisites for the efficient use of the databases are: (1) the definition of a reliable core set of uEVs’ constituents; and (2) handling data redundancy of current databases, are still awaited.

EMPLOYING URINARY EXTRACELLULAR VESICLES IN BIOMARKER DISCOVERY

Biomarkers are defined as substances or characteristics that are objectively measured and evaluated as an indicator of normal or pathogenic processes, or pharmacologic responses to a therapeutic intervention. Amongst the different body fluid, urine is one of the most important sources of biomarkers for both urologic and non-urologic diseases. High throughput omics studies have resulted in a great number of potential urinary biomarkers which are publicly available[87]. While the characterization of the normal urinary proteome is steadily progressing, there are three major obstacles in the classical urine-based protein biomarker discovery: (1) sample instability; (2) high dynamic protein concentration range; and (3) the relatively high inter-individual and inter-gender variations of urinary proteome. Urinary EVs, on the other hand, possess some characteristics that make them particularly attractive for biomarker research, like increased stability, reduced complexity, lower dynamic range, and composition which is closely related to the cell of origin. Biomolecules enclosed in the extracellular vesicle are surrounded by a lipid bilayer which protects them against degradation by proteases and nucleases. Therefore, uEVs have been shown to be particularly stable over time[37,88]. In addition, extracellular vesicles exhibit a reduced complexity comparing that to the whole urine composition. The dynamic concentration range of proteins and RNAs was shown to be lower in uEVs when compared to whole urine[33,78]. Moreover, the content of EVs reflects the cell of urinary tract they shed from. Therefore, it was expected that biomolecules of uEVs could provide clinically more specific information for both early diagnosis of disease and also for monitoring drug responsiveness than that of urine[89]. Since the first publication of uEVs[44], the majority of the works in this field is focused on exploring the potential use of uEVs in preclinical and clinical studies[2,6,24,25,33,35,37,39-41,71,74-78,81,89-92]. Since plasma derived EVs are too large to pass through the glomerular filtration barrier, urinary tract originated vesicles (Table 2)[41,44] carry cell specific markers from the specific regions of the kidney with relevant physiological and pathophysiological information[41].

Table 2 Putative disease-associated protein and RNA biomarkers identified in urinary extracellular vesicles.
Name of the putative markerUniProt ID/RNA nameMolecular and biological functionAssociated Kidney disease(s )Disease/modelRef.
Adenine phosphoribosyltransferaseAPT_HUMANEnzyme involved in the purine nucleotide salvage pathway. Its deficiency can lead to urolithiasis and renal failureAPRT deficiencyHealthy[62]
Low-density lipoprotein receptor- related protein 2 mRNALRP2_HUMAN (megalin), LRP2Multi-ligand binding receptor; mediates endocytic uptake of complexes between the steroid 25(OH) vitamin D3 and vitamin D-binding protein in kidney proximal tubulesDonnai-Barrow syndrome; Renal aminoglycoside accumulation; NephrotoxicityHealthy[38,78]
Polycystin-1, Polycystin-2PKD1_HUMAN PKD2_HUMANIntegral membrane glycoproteins, associated with structural and/or functional defects in the primary apical cilium of epithelia and polycystic kidney diseaseADPKDHealthy[45,46,54,62]
Solute carrier family 12 member 2 (Sodium potassium chloride cotransporter-2)S12A2_HUMANMembrane transporter: aids in the active transport of sodium, potassium, and chloride into and out of cells; NKCC2 mutations lead to type I Bartter syndromeAntenatal Bartter syndrome type 1Healthy[33,41,62]
Myosin-9MYH9_HUMANMYH9 polymorphisms have been shown to associate with glomerulosclerosis and non-diabetic end stage renal diseaseFechtner syndrome and Epstein syndromeHealthy[62]
Aquaporin-2 mRNAAQP2_HUMAN AQP2Molecular water channel in the basolateral and apical plasma membranes of the collecting ductAutosomal dominant and autosomal recessive nephrogenic diabetes insipidusHealthy; Various kidney diseases[24,33,38,39,44 ,54,58,62,95]
Neprilysin (CD10)NEP_HUMANSurface metallo-endopeptidase highly expressed in the kidney brush-border membranesMembranous glomerulonephritisHealthy; Diabetic nephropathy, Renal cell carcinoma[25,33,77,95]
OsteoprotegerinTR11B_HUMANDecoy receptor of proximal tubular cellschronic kidney disease-mineral and bone disorderHealthy; ADPKD CKDD patients[49]
PodocalyxinPODXL_HUMANhe major sialoprotein of kidney glomerulus; Involved in the regulation of cell adhesion, cell morphology and cancer progressionAutosomal recessive steroid–resistant nephrotic syndromeHealthy; Glomerular disease; Renal cell carcinoma[42,58,62,77,95]
Solute carrier family 12 member 3 (Thiazide- sensitive Na-Cl cotransporter; NCC)S12A3_HUMANMembrane transporter highly expressed in the kidney: aids reabsorbing sodium and chloride ions from the tubular fluid into the cells of the distal convoluted tubule of the nephronGitelman syndrome; AldosteronismHealthy; High blood pressure; Aldosteronism[41,62,79]
Protein AMBP (alpha-1-microglobulin; bikunin precursor)AMBP_HUMANMembrane glycoprotein with serine protease inhibitor activityDiabetic nephropathyHealthy; DN patients in advanced disease stages[16,45]
Leucine-rich alpha-2-glycoprotein-1LRG1_HUMANInvolved in protein-protein interaction, signal transduction, and cell adhesion and development; Expressed during granulocyte differentiationNSCLCHealthy; NSCLC[71]
Matrix metalloproteinase 9MMP9_HUMANInvolved in extracellular matrix remodelingRenal cell carcinomaHealthy; Renal cell carcinoma[77]
Basigin , Extracellular Matrix Metallo- proteinase InducerBASI_HUMANInvolved in extracellular matrix remodelingRenal cell carcinomaHealthy; Renal cell carcinoma[77]
CeruloplasminCERU_HUMANA ferroxidase enzyme in serumRenal cell carcinomaHealthy; Renal cell carcinoma[77]
Dickkopf related protein 4DKK4_HUMANInvolved in extracellular matrix remodelingRenal cell carcinomaHealthy; Renal cell carcinoma[77]
Carbonic Anhydrase 9CAH9_HUMANA metalloenzyme that catalyzes the reversible hydration of carbon dioxideRenal cell carcinomaHealthy; Renal cell carcinoma[77]
Dipeptidase 1DPEP1_HUMANA kidney membrane enzyme that hydrolyzes a variety of dipeptides and is implicated in renal metabolism of glutathione and its conjugatesRenal cell carcinomaHealthy; Renal cell carcinoma[77]
Syntenin-1SDCB1_HUMANScaffold protein Pbp1 Syndecan-binding proteinRenal cell carcinomaHealthy; Renal cell carcinoma[77]
Leucine-rich repeatkinase2LRRK2A member of the leucine-rich repeat kinase family; Associated to risks to inflammation-linked diseases that include Crohn’s disease and mycobacterium infectionParkinson’s diseaseHealthy; Parkinson’s disease[50]
Activating transcription factor 3ATF3_HUMAN miRNA494Transcription factor with protective role in renal ischemia-reperfusion injuryAKIAKI[22]
Fetuin-AFETUA_HUMANPlasma binding glycoprotein; inhibitor of calcification, regulator of cell-dependent process of osteogenesisAKIAKI[46,96]
Wilm’s tumor proteinWT1_HUMANZinc finger protein with essential role in the normal development of the urogenital systemEarly podocyte injury in diabetic nephropathy; steroid responsiveness or renal pathological conditions in patients with idiopathic nephrotic syndromeFSGS patients; Diabetic Patients with or without Proteinuria; idiopathic nephrotic syndrome[22,30,92]
Isoform 1 of histone-lysine N-methyltransferaseMLL3_HUMANHistonemethyltransferase which methyl- ates “Lys-4” of histone H3, a specific tag for epigenetic transcriptional activationDiabetic nephropathyDN patients in advanced disease stages[16]
Voltage-dependent anion-selective channel protein 1VDAC1_HUMANPorin ion channel of the outer mitochondrial membraneDiabetic nephropathyDN patients in advanced disease stages[16]
GPRC5BGPC5B_HUMANG protein coupled receptor required for tubulogenesisAcute Kidney DiseaseAKD[94]
Aquaporin-1 mRNAAQP1_HUMAN AQP1Molecular water channel in the basolateral and apical plasma membranes of the proximal tubules.Renal ischaemia reperfusion injuryRenal transplantation; Renal cell carcinoma[38,77,97]
Neutrophil gelatinase-associated lipocalinNGAL_HUMANIron-trafficking protein involved in multiple processes such as apoptosis, innate immunity and renal development. In the kidney it is produced by distal nephronkidney transplantationkidney allograft recipients[91]
Immunoglobulin light chainIGLL1_HUMAN IGKC_HUMANImmunoglobulin light chain is the small polypeptide subunit of an antibodyLight Chain AmyloidosisLight chain amyloidosis; multiple myeloma; monoclonal gammopathy; non-paraproteinemia related kidney disease[65]
Panel of 24 proteins _HUMANSERPINA1, A2M, APOA1, APOA2, CA1, CD5L, DPEP1, FGA, FGB, FGG, FN1, GSTT1, SLC2A1, HBA1, HBB, SERPIND1, HP, HPR, ITIH1, PON1, RAB27B, S100A8, S100A9, TACSTD2-Bladder cancerBladder cancer; Hernia[77]
PSAKLK3_HUMANSerine protease with diverse physiological functions; implicated in carcinogenesisProstate cancerProstate cancer[56]
Glutamate carboxypeptidase 2 (Prostate-specific membrane antigen-PSMA)FOLH1_HUMANFolate hydrolase and N-acetylated-alpha-linked-acidic dipeptidase activity; Involved in prostate tumor progressionProstate cancerProstate cancer[56]
microRNAMicroRNA-29c-Renal fibrosisHealthy; CKD[40]
microRNAmiR-130A-Type 1 diabetestype 1 diabetic patients with and without incipient diabetic nephropathy[81]
microRNAmiR-145-Type 1 diabetestype 1 diabetic patients with and without incipient diabetic nephropathy[81]
microRNAmiR-155-Type 1 diabetestype 1 diabetic patients with and without incipient diabetic nephropathy[81]
microRNAmiR-424-Type 1 diabetestype 1 diabetic patients with and without incipient diabetic nephropathy[81]
mRNA (CD2-associated protein)CD2AP-Podocyte injuryHealthy; diabetic nephropathy; focal segmental glomerulosclerosis; IgA nephropathy;membranous nephropathy[39]
mRNA (Prostate cancer antigen 3)PCA-3-Prostate cancerHealthy; Prostate cancer[32]
mRNA (TMPRSS2/ERGa fusion transcript)TMPRSS2:ERG-Prostate cancerHealthy; Prostate cancer[32]

Proteomic analysis has revealed proteins and RNAs that have been isolated from healthy individuals but are associated with different human diseases (Table 2): Adenine phosphoribosyltransferase in APRT deficiency[62]); Low-density lipoprotein receptor-related protein 2 in Donnai-Barrow syndrome, renal aminoglycoside accumulation and Nephrotoxicity; Polycystin-1 and Polycystin-2 in ADPKD[54]; Neprilysin in Membranous glomerulonephritis[41]; Non-muscle myosin heavy chain IIA in Fechtner syndrome and Epstein syndrome[62]; Sodium potassium chloride cotransporter-2 and Thiazide-sensitive Na-Cl cotransporter in Antenatal Bartter syndrome type 1 and Gitelman syndrome, respectively[41,62]. The presence of theses disease associated analytes in uEVs is promising but their expression levels in well-defined disease cohort and conditions still need to be determined.

In disease cohorts including bladder[57,74,93], prostate[32,56,75] and renal cell carcinoma[2,77] altered expressions of vesicle derived proteins, RNAs and lipids have been demonstrated (Table 2) and panels of putative protein biomarkers have been set up for validation. Vesicle expressed aquaporin water channel proteins (AQP1 and AQP2) have been found to be differentially expressed in a number of different renal diseases too. Other putative biomarkers related to urinary system diseases, like activating transcription factor 3 and fetuin-A in acute kidney injury[22,46]; podocalyxin in autosomal recessive steroid-resistant nephrotic syndrome[62]; and Wilm’s tumor-1 in early podocyte injury[22,92] have also been described. There are only a few studies though which report putative biomarkers related to treatment, like the effect of low sodium diet and infusion of aldosterone on the phosphorylation of Thiazide-sensitive Na-Cl cotransporter and prostatin in uEVS[79], or renal transplantation[91].

The reservoir for biomarker discovery could however extend beyond diseases of the urinary tract[41]. Conde-Li et al[70] investigated a rat model of liver injury induced by galactosamine and reported a change in urinary exosomes that coincided with liver injury. Gildea et al[71] reported that leucine-rich a-2-glycoprotein (LRG-1) was increased in both human urinary exosomes from patients with lung cancer and the lung cancer tissue. High-level of LRRK2 was found in uEVs of patients affected by Parkinson’s disease[50]. These observations open a novel scenario towards the future application of uEVs also in non-renal diseases.

CONCLUSION

Chronic kidney disease (CKD) is increasingly recognized as worldwide public health problem. CKD increases the risk for many adverse health outcomes, including cardiovascular disease, end-stage renal disease, and mortality. Because CKD usually progresses asymptomatically until its advanced stages, detection of early-stage CKD requires laboratory testing. The two key markers used for the definition, classification, and monitoring of kidney function are urine albumin and estimated glomerular filtration rate (eGFR). Kidney dysfunction is indicated by eGFR of less than 60 mL/min per 1.73 m2, while kidney damage most frequently is manifested as increased urinary albumin excretion. When less invasive blood and urine tests are insufficient fine-needle aspiration (FNA) or renal mass biopsy (RMB) is performed especially to substantiate the diagnosis in renal masses. While these traditional markers are certainly of great utility they also present several limitations. Protein (albumin) concentration in urine is not very specific. Levels may rise with use of certain non-steroidal anti-inflammatory drugs, rheumatoid arthritis, lupus and cancers etc. Creatinine level used to estimate GFR, on the other hand, provides little information about the underlying cause of kidney injuries, and lacks specificity in case of low muscle mass and unusual diets.

Because of the above limitations of currently used kidney function markers intensive research is going on to find more accurate ones. Many genes have been shown to be differentially expressed in the kidney upon glomerular injury, endothelial dysfunction, inflammation, fibrosis, cardiovascular dysfunction, metabolic disorders and cancer with the corresponding protein products appearing in plasma and urine. In urine, a unique biomarker source with great potential, a number of new candidate protein biomarkers have already been proposed: cystatin, N-acetyl-β-o-glucosaminidase (NAG) and liver-type fatty acid-binding protein (L-FABP), neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule 1 (KIM-1), just to mention a few, are potential biomarkers awaiting administration (FDA) approval. NGAL is suggested to be used as a urinary biomarker of delayed graft function (DGF), a frequent complication after kidney transplantation too. Recently Alvarez et al[90] have demonstrated that NGAL is mainly secreted into urinary vesicles and that the expression level of NGAL in uEVs is a sensitive measure of DGF, findings which might support the clinical management of patients undergoing kidney transplantation.

To become a clinically approved, a putative biomarker should be validated and implemented into clinical tests. Translating a novel discovery into clinical practice is however extremely challenging, consequently there are only a few urine-based protein biomarker assays which have been developed and approved so far (i.e., BTA and NMP-22 for the diagnosis of bladder cancer and PCA3 for the diagnosis of prostate cancer). Rapidly expanding urinary extracellular vesicle research represents an interesting field relevant to the development of disease specific, non-invasive methods for clinical diagnostics as well as to the development of new therapeutic approaches. Especially, growing evidence suggest that EV-imprinted genetic and proteomic information may well reflect the state of their parental cells. In this sense, expression analysis of proteins and RNAs in circulating urinary vesicles could provide specific information about the change in the state of specific nephron segment(s) and/or of the epithelial cells of urogenital tract. Urinary EV-mediated cell-cell communication within the nephron is another interesting aspects which can have major impact in our current understanding in renal physiology. However, how to translate these captivating ideas to a non-invasive renal monitoring system, there is still a lot more to understand.

ACKNOWLEDGMENTS

The authors thank to Roberto Ghiandoni (Nanosight Ltd) for the nanoparticle tracking analysis (NTA).

Footnotes

P- Reviewer: Salvadori M S- Editor: Ji FF L- Editor: A E- Editor: Lu YJ

References
1.  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;9:654-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8246]  [Cited by in F6Publishing: 9073]  [Article Influence: 533.7]  [Reference Citation Analysis (0)]
2.  Del Boccio P, Raimondo F, Pieragostino D, Morosi L, Cozzi G, Sacchetta P, Magni F, Pitto M, Urbani A. A hyphenated microLC-Q-TOF-MS platform for exosomal lipidomics investigations: application to RCC urinary exosomes. Electrophoresis. 2012;33:689-696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 78]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
3.  Alvarez ML, Mahdieh K, Rupesh Kanchi R, Johanna KD. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney International. 2012;82:1024-1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 393]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
4.  Mathivanan S, Fahner CJ, Reid GE, Simpson RJ. ExoCarta 2012: database of exosomal proteins, RNA and lipids. Nucleic Acids Res. 2012;40:D1241-D1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 697]  [Cited by in F6Publishing: 771]  [Article Influence: 59.3]  [Reference Citation Analysis (0)]
5.  Théry C. Exosomes: secreted vesicles and intercellular communications. F1000 Biol Rep. 2011;3:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 666]  [Article Influence: 51.2]  [Reference Citation Analysis (0)]
6.  Beach A, Zhang HG, Ratajczak MZ, Kakar SS. Exosomes: an overview of biogenesis, composition and role in ovarian cancer. J Ovarian Res. 2014;7:14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 158]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
7.  Schneider A, Simons M. Exosomes: vesicular carriers for intercellular communication in neurodegenerative disorders. Cell Tissue Res. 2013;352:33-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 224]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
8.  Ludwig AK, Giebel B. Exosomes: small vesicles participating in intercellular communication. Int J Biochem Cell Biol. 2012;44:11-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 311]  [Cited by in F6Publishing: 358]  [Article Influence: 27.5]  [Reference Citation Analysis (0)]
9.  Admyre C, Johansson SM, Qazi KR, Filén JJ, Lahesmaa R, Norman M, Neve EP, Scheynius A, Gabrielsson S. Exosomes with immune modulatory features are present in human breast milk. J Immunol. 2007;179:1969-1978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 740]  [Cited by in F6Publishing: 821]  [Article Influence: 48.3]  [Reference Citation Analysis (0)]
10.  Bobrie A, Colombo M, Raposo G, Théry C. Exosome secretion: molecular mechanisms and roles in immune responses. Traffic. 2011;12:1659-1668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 686]  [Cited by in F6Publishing: 747]  [Article Influence: 57.5]  [Reference Citation Analysis (0)]
11.  Wahlgren J, Karlson Tde L, Glader P, Telemo E, Valadi H. Activated human T cells secrete exosomes that participate in IL-2 mediated immune response signaling. PLoS One. 2012;7:e49723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 103]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
12.  Anand PK. Exosomal membrane molecules are potent immune response modulators. Commun Integr Biol. 2010;3:405-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 57]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
13.  Mittelbrunn M, Gutierrez-Vazquez C, Villarroya-Beltri C, Gonzalez S, Sanchez-Cabo F, Gonzalez MA, Bernad A, Sanchez-Madrid F. Unidirectional transfer of microRNA-loaded exosomes from T cells to antigen-presenting cells. Nature communications. 2011;2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1233]  [Cited by in F6Publishing: 1352]  [Article Influence: 104.0]  [Reference Citation Analysis (0)]
14.  Qazi KR, Gehrmann U, Domange Jordö E, Karlsson MC, Gabrielsson S. Antigen-loaded exosomes alone induce Th1-type memory through a B-cell-dependent mechanism. Blood. 2009;113:2673-2683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 169]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
15.  Testa JS, Apcher GS, Comber JD, Eisenlohr LC. Exosome-driven antigen transfer for MHC class II presentation facilitated by the receptor binding activity of influenza hemagglutinin. J IMMUNOL. 2010;185:6608-6616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
16.  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. Diabetic nephropathy induces changes in the proteome of human urinary exosomes as revealed by label-free comparative analysis. J PROTEOMICS. 2014;96:92-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 101]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
17.  Porto-Carreiro I, Février B, Paquet S, Vilette D, Raposo G. Prions and exosomes: from PrPc trafficking to PrPsc propagation. Blood Cells Mol Dis. 2005;35:143-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
18.  Ashok A, Hegde RS. Prions and retroviruses: an endosomal rendezvous? EMBO Rep. 2006;7:685-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
19.  Raimondo F, Morosi L, Chinello C, Magni F, Pitto M. Advances in membranous vesicle and exosome proteomics improving biological understanding and biomarker discovery. Proteomics. 2011;11:709-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 237]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
20.  Momen-Heravi F, Balaj L, Alian S, Mantel PY, Halleck AE, Trachtenberg AJ, Soria CE, Oquin S, Bonebreak CM, Saracoglu E. Current methods for the isolation of extracellular vesicles. Biol Chem. 2013;394:1253-1262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 414]  [Article Influence: 41.4]  [Reference Citation Analysis (0)]
21.  Crescitelli R, Lässer C, Szabó TG, Kittel A, Eldh M, Dianzani I, Buzás EI, Lötvall J. Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J Extracell Vesicles. 2013;2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 570]  [Cited by in F6Publishing: 674]  [Article Influence: 61.3]  [Reference Citation Analysis (0)]
22.  Zhou H, Cheruvanky A, Hu X, Matsumoto T, Hiramatsu N, Cho ME, Berger A, Leelahavanichkul A, Doi K, Chawla LS. Urinary exosomal transcription factors, a new class of biomarkers for renal disease. Kidney Int. 2008;74:613-621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 186]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
23.  Wang Z, Hill S, Luther JM, Hachey DL, Schey KL. Proteomic analysis of urine exosomes by multidimensional protein identification technology (MudPIT). Proteomics. 2012;12:329-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 139]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
24.  Trnka P, Ivanova L, Hiatt MJ, Matsell DG. Urinary biomarkers in obstructive nephropathy. Clin J Am Soc Nephrol. 2012;7:1567-1575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
25.  Raimondo F, Corbetta S, Morosi L, Chinello C, Gianazza E, Castoldi G, Di Gioia C, Bombardi C, Stella A, Battaglia C. Urinary exosomes and diabetic nephropathy: a proteomic approach. Mol Biosyst. 2013;9:1139-1146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
26.  Michael A, Bajracharya SD, Yuen PS, Zhou H, Star RA, Illei GG, Alevizos I. Exosomes from human saliva as a source of microRNA biomarkers. Oral Dis. 2010;16:34-38.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Lasser C, Seyed Alikhani V, Ekstrom K, Eldh M, Torregrosa Paredes P, Bossios A, Sjostrand M, Gabrielsson S, Lotvall J, Valadi H. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med. 2011;9:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 594]  [Cited by in F6Publishing: 653]  [Article Influence: 50.2]  [Reference Citation Analysis (0)]
28.  Street JM, Barran PE, Mackay CL, Weidt S, Balmforth C, Walsh TS, Chalmers RT, Webb DJ, Dear JW. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J Transl Med. 2012;10:5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 335]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
29.  Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4900]  [Cited by in F6Publishing: 5527]  [Article Influence: 502.5]  [Reference Citation Analysis (0)]
30.  Lee H, Han KH, Lee SE, Kim SH, Kang HG, Cheong HI. Urinary exosomal WT1 in childhood nephrotic syndrome. Pediatr Nephrol. 2012;27:317-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
31.  Choi DS, Kim DK, Kim YK, Gho YS. Proteomics, transcriptomics and lipidomics of exosomes and ectosomes. Proteomics. 2013;13:1554-1571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 365]  [Article Influence: 33.2]  [Reference Citation Analysis (0)]
32.  Nilsson J, Skog J, Nordstrand A, Baranov V, Mincheva-Nilsson L, Breakefield XO, Widmark A. Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. Br J Cancer. 2009;100:1603-1607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 529]  [Cited by in F6Publishing: 567]  [Article Influence: 37.8]  [Reference Citation Analysis (0)]
33.  Raj DA, Fiume I, Capasso G, Pocsfalvi G. A multiplex quantitative proteomics strategy for protein biomarker studies in urinary exosomes. Kidney Int. 2012;81:1263-1272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 113]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
34.  Schageman J, Zeringer E, Li M, Barta T, Lea K, Gu J, Magdaleno S, Setterquist R, Vlassov AV. The complete exosome workflow solution: from isolation to characterization of RNA cargo. Biomed Res Int. 2013;2013:253957.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 132]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
35.  Pisitkun T, Gandolfo MT, Das S, Knepper MA, Bagnasco SM. Application of systems biology principles to protein biomarker discovery: urinary exosomal proteome in renal transplantation. Proteomics Clin Appl. 2012;6:268-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
36.  Fernández-Llama P, Khositseth S, Gonzales PA, Star RA, Pisitkun T, Knepper MA. Tamm-Horsfall protein and urinary exosome isolation. Kidney Int. 2010;77:736-742.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 209]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
37.  Lv LL, Cao Y, Liu D, Xu M, Liu H, Tang RN, Ma KL, Liu BC. Isolation and quantification of microRNAs from urinary exosomes/microvesicles for biomarker discovery. Int J Biol Sci. 2013;9:1021-1031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 147]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
38.  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;78:191-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 312]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
39.  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 Chim Acta. 2014;428:26-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 82]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
40.  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:F1220-F1227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 169]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
41.  Dear JW, Street JM, Bailey MA. Urinary exosomes: a reservoir for biomarker discovery and potential mediators of intrarenal signalling. Proteomics. 2013;13:1572-1580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 128]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
42.  Hara M, Yanagihara T, Hirayama Y, Ogasawara S, Kurosawa H, Sekine S, Kihara I. Podocyte membrane vesicles in urine originate from tip vesiculation of podocyte microvilli. Hum Pathol. 2010;41:1265-1275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
43.  Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2:569-579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3512]  [Cited by in F6Publishing: 3754]  [Article Influence: 170.6]  [Reference Citation Analysis (0)]
44.  Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci USA. 2004;101:13368-13373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1494]  [Cited by in F6Publishing: 1603]  [Article Influence: 80.2]  [Reference Citation Analysis (0)]
45.  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;20:363-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 509]  [Cited by in F6Publishing: 551]  [Article Influence: 34.4]  [Reference Citation Analysis (0)]
46.  Fang DY, King HW, Li JY, Gleadle JM. Exosomes and the kidney: blaming the messenger. Nephrology (Carlton). 2013;18:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 57]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
47.  Jia L, Li X, Shao C, Wei L, Li M, Guo Z, Liu Z, Gao Y. Using an isolated rat kidney model to identify kidney origin proteins in urine. PLoS One. 2013;8:e66911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
48.  Prunotto M, Farina A, Lane L, Pernin A, Schifferli J, Hochstrasser DF, Lescuyer P, Moll S. Proteomic analysis of podocyte exosome-enriched fraction from normal human urine. J Proteomics. 2013;82:193-229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 111]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
49.  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. Osteoprotegerin in exosome-like vesicles from human cultured tubular cells and urine. PLoS One. 2013;8:e72387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
50.  Fraser KB, Moehle MS, Daher JP, Webber PJ, Williams JY, Stewart CA, Yacoubian TA, Cowell RM, Dokland T, Ye T. LRRK2 secretion in exosomes is regulated by 14-3-3. Hum Mol Genet. 2013;22:4988-5000.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 127]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
51.  Musante L, Saraswat M, Duriez E, Byrne B, Ravidà A, Domon B, Holthofer H. Biochemical and physical characterisation of urinary nanovesicles following CHAPS treatment. PLoS One. 2012;7:e37279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
52.  Chen CY, Hogan MC, Ward CJ. Purification of exosome-like vesicles from urine. Methods Enzymol. 2013;524:225-241.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Hogan MC, Manganelli L, Woollard JR, Masyuk AI, Masyuk TV, Tammachote R, Huang BQ, Leontovich AA, Beito TG, Madden BJ. Characterization of PKD protein-positive exosome-like vesicles. J Am Soc Nephrol. 2009;20:278-288.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 248]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
54.  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;72:1095-1102.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Mitchell PJ, Welton J, Staffurth J, Court J, Mason MD, Tabi Z, Clayton A. Can urinary exosomes act as treatment response markers in prostate cancer? J Transl Med. 2009;7:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 217]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
56.  Welton JL, Khanna S, Giles PJ, Brennan P, Brewis IA, Staffurth J, Mason MD, Clayton A. Proteomics analysis of bladder cancer exosomes. Mol Cell Proteomics. 2010;9:1324-1338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 305]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
57.  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:F1657-F1661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 317]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
58.  Merchant ML, Powell DW, Wilkey DW, Cummins TD, Deegens JK, Rood IM, McAfee KJ, Fleischer C, Klein E, Klein JB. Microfiltration isolation of human urinary exosomes for characterization by MS. Proteomics Clin Appl. 2010;4:84-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 155]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
59.  Moon PG, You S, Lee JE, Hwang D, Baek MC. Urinary exosomes and proteomics. Mass Spectrom Rev. 2011;30:1185-1202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
60.  Mathivanan S, Lim JW, Tauro BJ, Ji H, Moritz RL, Simpson RJ. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol Cell Proteomics. 2010;9:197-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 415]  [Cited by in F6Publishing: 433]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
61.  Hoorn EJ, Pisitkun T, Zietse R, Gross P, Frokiaer J, Wang NS, Gonzales PA, Star RA, Knepper MA. Prospects for urinary proteomics: exosomes as a source of urinary biomarkers. Nephrology (Carlton). 2005;10:283-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 144]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
62.  Street JM, Birkhoff W, Menzies RI, Webb DJ, Bailey MA, Dear JW. Exosomal transmission of functional aquaporin 2 in kidney cortical collecting duct cells. J Physiol-London. 2011;589:6119-6127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 110]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
63.  Staubach S, Schadewaldt P, Wendel U, Nohroudi K, Hanisch FG. Differential Glycomics of Epithelial Membrane Glycoproteins from Urinary Exovesicles Reveals Shifts toward Complex-Type N-Glycosylation in Classical Galactosemia. J Proteome Res. 2012;11:906-916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
64.  Ramirez-Alvarado M, Ward CJ, Huang BQ, Gong X, Hogan MC, Madden BJ, Charlesworth MC, Leung N. Differences in immunoglobulin light chain species found in urinary exosomes in light chain amyloidosis (Al). PLoS One. 2012;7:e38061.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Musante L, Saraswat M, Ravida A, Byrne B, Holthofer H. Recovery of urinary nanovesicles from ultracentrifugation supernatants. Nephrol Dial Transpl. 2013;28:1425-1433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
66.  Oosthuyzen W, Sime NEL, Ivy JR, Turtle EJ, Street JM, Pound J, Bath LE, Webb DJ, Gregory CD, Bailey MA. Quantification of human urinary exosomes by nanoparticle tracking analysis. J Physiol-London. 2013;591:5833-5842.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 135]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
67.  Tatischeff I, Larquet E, Falcón-Pérez JM, Turpin PY, Kruglik SG. Fast characterisation of cell-derived extracellular vesicles by nanoparticles tracking analysis, cryo-electron microscopy, and Raman tweezers microspectroscopy. J Extracell Vesicles. 2012;1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 137]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
68.  Gerlach JQ, Krüger A, Gallogly S, Hanley SA, Hogan MC, Ward CJ, Joshi L, Griffin MD. Surface glycosylation profiles of urine extracellular vesicles. PLoS One. 2013;8:e74801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
69.  Conde-Vancells J, Rodriguez-Suarez E, Gonzalez E, Berisa A, Gil D, Embade N, Valle M, Luka Z, Elortza F, Wagner C. Candidate biomarkers in exosome-like vesicles purified from rat and mouse urine samples. Proteomics Clin Appl. 2010;4:416-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 102]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
70.  Li Y, Zhang Y, Qiu F, Qiu Z. Proteomic identification of exosomal LRG1: a potential urinary biomarker for detecting NSCLC. Electrophoresis. 2011;32:1976-1983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 197]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
71.  Gildea JJ, Carlson JM, Schoeffel CD, Carey RM, Felder RA. Urinary exosome miRNome analysis and its applications to salt sensitivity of blood pressure. J Clin Biochem. 2013;46:1131-1134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
72.  Cheng L, Sun X, Scicluna BJ, Coleman BM, Hill AF. Characterization and deep sequencing analysis of exosomal and non-exosomal miRNA in human urine. Kidney Int. 2013;Dec 18; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 265]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
73.  Chen CL, Lai YF, Tang P, Chien KY, Yu JS, Tsai CH, Chen HW, Wu CC, Chung T, Hsu CW. Comparative and Targeted Proteomic Analyses of Urinary Microparticles from Bladder Cancer and Hernia Patients. J Proteome Res. 2012;11:5611-5629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 157]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
74.  Hosseini-Beheshti E, Pham S, Adomat H, Li N, Tomlinson Guns ES. Exosomes as biomarker enriched microvesicles: characterization of exosomal proteins derived from a panel of prostate cell lines with distinct AR phenotypes. Mol Cell Proteomics. 2012;11:863-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 171]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
75.  Moon PG, Lee JE, You S, Kim TK, Cho JH, Kim IS, Kwon TH, Kim CD, Park SH, Hwang D. Proteomic analysis of urinary exosomes from patients of early IgA nephropathy and thin basement membrane nephropathy. Proteomics. 2011;11:2459-2475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 188]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
76.  Raimondo F, Morosi L, Corbetta S, Chinello C, Brambilla P, Della Mina P, Villa A, Albo G, Battaglia C, Bosari S. Differential protein profiling of renal cell carcinoma urinary exosomes. Mol Biosyst. 2013;9:1220-1233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 118]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
77.  Raj DAA, Fiume I, Capasso G, Pocsfalvi G. Urinary Exosomes for Protein Biomarker Research. In Tech: Proteomics-Human Diseases and Protein Functions 2012; .  [PubMed]  [DOI]  [Cited in This Article: ]
78.  van der Lubbe N, Jansen PM, Salih M, Fenton RA, van den Meiracker AH, Danser AHJ, Zietse R, Hoorn EJ. The Phosphorylated Sodium Chloride Cotransporter in Urinary Exosomes Is Superior to Prostasin as a Marker for Aldosteronism. Hypertension. 2012;60:741.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 77]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
79.  Dimov I, Jankovic Velickovic L, Stefanovic V. Urinary exosomes. ScientificWorldJournal. 2009;9:1107-1118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
80.  Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, Bruno G, Cimino D, Taverna D, Deregibus MC. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One. 2013;8:e73798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 226]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
81.  Bligh EG, Dyer WJ. A rapid method of total lipid extraction and purification. Can J Biochem Physiol. 1959;37:911-917.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29286]  [Cited by in F6Publishing: 27139]  [Article Influence: 1130.8]  [Reference Citation Analysis (0)]
82.  Choi DS, Kim DK, Kim YK, Gho YS. Proteomics of extracellular vesicles: Exosomes and ectosomes. Mass Spectrom Rev. 2014;Jan 14; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 304]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
83.  Kalra H, Simpson RJ, Ji H, Aikawa E, Altevogt P, Askenase P, Bond VC, Borràs FE, Breakefield X, Budnik V. Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation. PLoS Biol. 2012;10:e1001450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 831]  [Cited by in F6Publishing: 921]  [Article Influence: 76.8]  [Reference Citation Analysis (0)]
84.  Kim DK, Kang B, Kim OY, Choi DS, Lee J, Kim SR, Go G, Yoon YJ, Kim JH, Jang SC. EVpedia: an integrated database of high-throughput data for systemic analyses of extracellular vesicles. J Extracell Vesicles. 2013;2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 340]  [Article Influence: 30.9]  [Reference Citation Analysis (0)]
85.  Choi DS, Yang JS, Choi EJ, Jang SC, Park S, Kim OY, Hwang D, Kim KP, Kim YK, Kim S. The protein interaction network of extracellular vesicles derived from human colorectal cancer cells. J Proteome Res. 2012;11:1144-1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
86.  Shao C, Li M, Li X, Wei L, Zhu L, Yang F, Jia L, Mu Y, Wang J, Guo Z. A tool for biomarker discovery in the urinary proteome: a manually curated human and animal urine protein biomarker database. Mol Cell Proteomics. 2011;10:M111.010975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
87.  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;69:1471-1476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 407]  [Cited by in F6Publishing: 432]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
88.  Properzi F, Logozzi M, Fais S. Exosomes: the future of biomarkers in medicine. Biomark Med. 2013;7:769-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 294]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
89.  van Balkom BW, Pisitkun T, Verhaar MC, Knepper MA. Exosomes and the kidney: prospects for diagnosis and therapy of renal diseases. Kidney Int. 2011;80:1138-1145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 160]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
90.  Alvarez S, Suazo C, Boltansky A, Ursu M, Carvajal D, Innocenti G, Vukusich A, Hurtado M, Villanueva S, Carreño JE. Urinary exosomes as a source of kidney dysfunction biomarker in renal transplantation. Transplant Proc. 2013;45:3719-3723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 104]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
91.  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:e60177.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Ahn JJ, Salazar G, Seaman M, Scott A, Motamedinia P, Castaneda C, Comper W, McKiernan JM, Russo L. Evaluation of urinary exosomes in bladder cancer patients. J Urol. 2013;189:E465-E466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
93.  Kwon SH, Liu KD, Mostov KE. Intercellular transfer of GPRC5B via exosomes drives HGF-mediated outward growth. Curr Biol. 2014;24:199-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
94.  Rood IM, Deegens JK, Merchant ML, Tamboer WP, Wilkey DW, Wetzels JF, Klein JB. Comparison of three methods for isolation of urinary microvesicles to identify biomarkers of nephrotic syndrome. Kidney Int. 2010;78:810-816.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 194]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
95.  Zhou H, Pisitkun T, Aponte A, Yuen PS, Hoffert JD, Yasuda H, Hu X, Chawla L, Shen RF, Knepper MA. Exosomal Fetuin-A identified by proteomics: a novel urinary biomarker for detecting acute kidney injury. Kidney Int. 2006;70:1847-1857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 285]  [Cited by in F6Publishing: 301]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
96.  Sonoda H, Yokota-Ikeda N, Oshikawa S, Kanno Y, Yoshinaga K, Uchida K, Ueda Y, Kimiya K, Uezono S, Ueda A. Decreased abundance of urinary exosomal aquaporin-1 in renal ischemia-reperfusion injury. Am J Physiol Renal Physiol. 2009;297:F1006-F1016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 103]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
97.  Chen J, Boyle S, Zhao M, Su W, Takahashi K, Davis L, Decaestecker M, Takahashi T, Breyer MD, Hao CM. Differential expression of the intermediate filament protein nestin during renal development and its localization in adult podocytes. J Am Soc Nephrol. 2006;17:1283-1291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]