URINARY EXOSOMES AND PROTEOMICS
Pyong-Gon Moon,1,2 Sungyong You,3 Jeong-Eun Lee,1,2 Daehee Hwang,3
and Moon-Chang Baek1,2*
1
Department of Molecular Medicine
2
Cell and Matrix Biology Research Institute, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
3
School of Interdisciplinary Bioscience and Bioengineering & Department
of Chemical Eng., POSTECH, Pohang 790-784, Republic of Korea
Received 21 October 2009; revised 23 July 2010; accepted 23 July 2010
Published online in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/mas.20319
A number of highly abundant proteins in urine have been identified through proteomics approaches, and some have been considered as disease-biomarker candidates. These molecules might
be clinically useful in diagnosis of various diseases. However,
none has proven to be specifically indicative of perturbations of
cellular processes in cells associated with urogenital diseases.
Exosomes could be released into urine which flows through the
kidney, ureter, bladder and urethra, with a process of filtration
and reabsorption. Urinary exosomes have been recently suggested as alternative materials that offer new opportunities to
identify useful biomarkers, because these exosomes secreted
from epithelial cells lining the urinary track might reflect the
cellular processes associated with the pathogenesis of diseases
in their donor cells. Proteomic analysis of such urinary exosomes
assists the search of urinary biomarkers reflecting pathogenesis
of various diseases and also helps understanding the function of
urinary exosomes in urinary systems. Thus, it has been recently
suggested that urinary exosomes are one of the most valuable
targets for biomarker development and to understand pathophysiology of relevant diseases. # 2011 Wiley Periodicals, Inc.
Mass Spec Rev
Keywords: urinary exosomes; urogenital diseases; biomarkers; proteomics
I. INTRODUCTION
The attempts to identify disease biomarkers, for diagnostics
and prognostics, increase tremendously during the last 10
years as proteomics technology has been developed and optimized (Azad et al., 2006; Qian et al., 2006; Geho et al., 2007;
Tao & Lazarev, 2007). Urine could be easily used for clinical
as well as basic research because urine might be obtained in a
simple, non-invasive, sufficient and stable fashion compared to
other clinical samples (Theodorescu et al., 2006; Decramer
et al., 2008; Caubet et al., 2010). Urine contains proteins, peptides, and metabolites derived from kidney as well as blood
(Decramer et al., 2008). Therefore, urine is applicable to study
Contract grant sponsor: Korean Ministry of Education, Science and
Technology; Contract grant sponsor: Regional Technology Innovation
Program of the Ministry of Knowledge Economy; Contract grant number: RTI04-01-01.
*Correspondence to: Moon-Chang Baek, Department of Molecular
Medicine, School of Medicine, Kyungpook National University, 101
Dongin-dong 2 Ga, Jung-gu, Daegu 700-422, Republic of Korea.
E-mail: mcbaek@knu.ac.kr
Mass Spectrometry Reviews
# 2011 by Wiley Periodicals, Inc.
with regards to urogenital diseases and non-urogenital diseases
from associated organs in the body (Thongboonkerd, 2008).
The advent of the proteomics era has enormously affected
the methodology to study proteins, and has accelerated the
ability to acquire information on proteins for basic and clinical
research areas. The number of urine proteins identified during
the past 5 years exceeded by 10-fold that of urine proteins
found previously, and facilitated the search for urinary biomarkers (Spahr et al., 2001; Pieper et al., 2004; Adachi et al.,
2006; Wang et al., 2006; Moon et al., 2008; Zurbig & Mischak,
2008). Various mass spectrometry techniques including 2-D
PAGE-MS, surface-enhanced laser desorption/ionization timeof-flight mass spectrometry (SELDI-TOF MS), LC-MS/MS and
capillary electrophoresis mass spectrometry (CE-MS), have
been applied to analyze urine proteome. SELDI-TOF MS
(Cadieux et al., 2004; Khurana et al., 2006; Poon, 2007), and
CE-MS (Mischak et al., 2004; Weissinger et al., 2004; Haubitz
et al., 2005; Wittke et al., 2005; Decramer et al., 2006) have
been used for profiling approaches, and LC-MS/MS (Castagna
et al., 2005; Sun et al., 2005; Adachi et al., 2006; Ru et al.,
2006; Wang et al., 2006; Pieper, 2008; Smalley et al., 2008)
and 2-D PAGE-MS (Thongboonkerd, Klein, & Arthur, 2003;
Saito et al., 2005; Thongboonkerd, 2007) have been used for
biomarker discovery. The advantages and disadvantages of
each MS-based proteomics techniques have been reviewed in
detail elsewhere (Fliser et al., 2007; Decramer et al., 2008).
For these methods, usually the whole urine has been used.
One of the challenges for biomarker identification is to reduce
the dynamic range of protein concentrations in samples. To
overcome this difficulty, an immunosubtraction method such
as albumin depletion, affinity chromatography method such as
lectin (Yang & Hancock, 2004, 2005; Wang et al., 2006;
Moon et al., 2008) and immobilized metal-ion chromatography (IMAC) columns (Belew et al., 1987; Hoffert et al.,
2006; Villen & Gygi, 2008), and ligand library beads method
(Righetti et al., 2006) have been introduced, and the
identification efficiency of low-abundance proteins has been
improved. However, it is still difficult to select good biomarker candidates for the next validation step among proteins
with differentially expressed proteins.
Urinary exosomes are microparticles identified recently in
urine (Pisitkun, Shen, & Knepper, 2004). Proteomic research
of these urinary exosomes has become one of the newer trends
in the field of urine-biomarker discovery. Various mammalian
cells such as reticulocytes (Pan & Johnstone, 1983; Johnstone
et al., 1987), mast cells (Skokos et al., 2001a, 2003; Valadi
&
MOON ET AL.
et al., 2007), B lymphocytes cell lines (Escola et al., 1998;
Dukers et al., 2000; Wubbolts et al., 2003), and human dendritic cells (DCs) (Thery et al., 1999, 2001; Kim et al., 2005)
secrete exosomes by physiological signals. Many studies
related to exosomes have been reported, and the recent high
increase in exosome studies might be related to the growth of
proteomics research (Fig. 1). In 2004, the Knepper group first
identified exosomes in human urine, and demonstrated the
potential to use the urinary exosomes as a starting sample for
biomarker discovery in urine (Pisitkun, Shen, & Knepper,
2004). Subsequently, the number of studies of urinary exosomes has gradually increased as shown by other exosomes
studies (Fig. 1). To secrete the exosomes, an endocytic pathway and the fusion of multivesicular bodies (MVBs) with the
plasma membrane are involved (Fevrier & Raposo, 2004).
Intra-lumenal vesicles (ILVs) in MVB lumen are released into
the extracellular environment. The released small particles
(40–100 nm) were called exosomes. The exosomes obtain
cytosolic portions, including proteins and various RNAs, at
the point when endosomes are fused into MVBs, and also
contain membrane proteins and lipids from original cells
(Lin et al., 2005; Fang et al., 2007; Lakkaraju & RodriguezBoulan, 2008). There are several reports that urinary exosomes
could be excreted into urine via exocytosis from kidney
cells by a similar mechanism to other exosomes (Pisitkun,
Shen, & Knepper, 2004; Zhou et al., 2006b; Keller et al., 2007;
Lakkaraju & Rodriguez-Boulan, 2008; Gonzales et al., 2009).
Only 3% of the total urinary protein in urine is contained
in urinary exosomes, which excludes other high-abundance
proteins such as albumin (Hoorn et al., 2005). In addition, the
urinary exosomes might contain many clues regarding the
state of organs through the urinary track (Gonzales, Pisitkun,
& Knepper, 2009). Therefore, urinary exosomes could be
important sources to study urogenital diseases, especially
human diseases without proper animal models. Many
researchers have focused their current efforts on biomarkers
that are biologically relevant to the pathophysiology of diseases as reviewed recently (Knepper, 2009; Gonzales, Pisitkun, & Knepper, 2009). Therefore, urinary exosomes could be
good target materials to develop biomarkers in clinical
samples due to their attractive characteristics, including origin
from cells lining the urinary track, low complexity of proteins,
and content of cytosolic and membrane proteins of donor cells
(Hoorn et al., 2005), even if exosomal proteins are much
harder to be isolated compared to soluble urine proteins.
This review presents the characteristics of urinary exosomes, methodologies to study urinary exosomes, and proteomics studies on various urinary exosomes.
II. THE CHARACTERISTICS OF URINARY
EXOSOMES
A. Kidney Function and Urine
The kidneys are reddish, bean-shaped, paired organs, which play
a major role in the production of urine. In a human, an adult
kidney is 10–12 cm long, 5–7 cm wide, and 3 cm thick with a
mass of 115–170 g (Boron & Boulpaep, 2005; Tortora &
Grabowski, 2003). The left kidney is slightly larger than the
right. The concave surface, called the renal hilum is the point
through which the ureter emerges from the kidney along with
blood vessels, lymphatic vessels, and nerves. The kidney has two
major structures; the renal cortex at the outer region, and the
renal medullar at the inner region. Nephrons, the urine-producing
functional structures of the kidney, span the cortex and medullar
(Tortora & Grabowski, 2003; Boron & Boulpaep, 2005).
Urine is produced by the urinary system that is composed
of two kidneys, two ureters, one urinary bladder, and one urethra. The kidneys are the major part of the urinary system, and
the other parts are primarily for passageways and storage of
urine. Furthermore, the kidneys play important roles in the
regulation of ionic composition, pH, volume, pressure, and
glucose level in blood. Each nephron, the functional unit of
FIGURE 1. The relationship between exosomes research and proteomics. The high increase of publications
related to exosomes research (
) might be related to the growth of proteomics research (
). Study on
) started in 2004, and the number of publications has increased annually since
urinary exosomes (
then. The number of publications was calculated based on PubMed searches with key words ‘‘exosomes,’’
‘‘urine þ exosomes,’’ or ‘‘proteomics,’’ respectively.
2
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
&
the kidneys, consists of two parts: a renal corpuscle, where
bold plasma is filtered, and a renal tubule into which the filtered fluid passes. The two components of a renal corpuscle
are the glomerulus and the glomerular (Bowman’s) capsule, a
double-walled epithelial cup that surrounds the glomerular
capillaries. Blood plasma is filtered in the glomerular capsule,
and the filtered fluid passes into the renal tubule, which has
three main sections. In the order that fluid passes through
them, first the renal tubule consists of a (1) proximal convoluted tubule, (2) loop of Henle (nephron loop), and (3) distal
convoluted tubule. The distal convoluted tubules of several
nephrons empty into a single collecting duct. Second, collecting ducts unite and converge into several hundred large papillary ducts, which drain into the minor calyces. Third, the
collecting ducts and papillary ducts extend from the renal cortex through the renal medulla to the renal pelvis. Thus, one
kidney has about 1 million nephrons, but a much smaller number of collecting ducts and even fewer papillary ducts.
About 170–179 L of water is reabsorbed into the bloodstream, and the actual volume of urine produced is only 1–
2 L. The urine is composed of water, proteins, glucose, amino
acids, urea, and ions such as sodium, potassium, calcium,
chloride, bicarbonate, and phosphate (Tortora & Grabowski,
2003; Boron & Boulpaep, 2005) (Fig. 2A). In healthy individuals, urinary proteins consist of kidney-originated proteins
(70%) (Christensen & Birn, 2001) and plasma-derived
proteins (30%) (Thongboonkerd et al., 2002; Pieper et al.,
2004; Thongboonkerd & Malasit, 2005) (Fig. 2B). Therefore,
urine is an ideal biofluid for biomarker discovery of kidney diseases as well as non-kidney diseases (Thongboonkerd, 2008).
Urine protein is divided two parts, soluble proteins, and solidphase elements. The solid-phase elements can be divided into
sediment precipitated with low-speed centrifugation and lowdensity exosomes precipitated with ultracentrifugation. The
proteins that are present in urine are a major area of investigation for proteomics researchers. In normal urine, half of the
proteins are soluble proteins (49%), and the remaining 48% are
sediment precipitated with low-speed centrifugation and exosomes (3%) (Serafini-Cessi, Malagolini, & Cavallone, 2003;
Hoorn et al., 2005; Zhou et al., 2006b; Pisitkun, Johnstone, &
Knepper, 2006) (Fig. 2C). The exosomes are prepared with
high-speed centrifugation, and the size of the low-density
vesicle is less than 100 nm, which originates from multivesicular bodies (MVBs) in cells (Pisitkun, Shen, & Knepper, 2004).
B. Characteristics of Exosomes
1. Exosome Excretion
It has been reported that human body fluids such as urine
(Pisitkun, Shen, & Knepper, 2004; Keller et al., 2007; Gonzales
et al., 2009), plasma (Looze et al., 2009), physiological pregnancy-associated sera (Sabapatha, Gercel-Taylor, & Taylor,
2006), bronchial lavage fluid (Admyre et al., 2003), synovial
fluid (Skriner et al., 2006), saliva (Castle, Huang, & Castle,
2002; Kapsogeorgou et al., 2005; Gonzalez-Begne et al., 2009),
breast milk (Admyre et al., 2007), and malignant pleural effusion (Andre et al., 2002; Bard et al., 2004) contain exosomes.
In addition, many groups have reported that several cell types,
including B cells (Escola et al., 1998; Dukers et al., 2000;
Wubbolts et al., 2003), dendritic cells (Thery et al., 1999,
Mass Spectrometry Reviews DOI 10.1002/mas
FIGURE 2. The composition of urine and urine proteins in human. The
volume of urine excreted a day is about 1–2 L. A: The ratio of the urine
composition is; water (97.1%), urea (1.94%), ions (0.8%), creatinine
(0.1%), uric acid (0.05%), and protein (0.01%). B: Urine proteins used for
proteomics are classified into two parts based on their origin; kidney and
urinary tract (70%), filtered plasma (30%). C: Urine proteins are also divided two parts based on their solubility; Soluble (49%) and solid-phase
elements (51%). The solid-phase elements can be also divided into sediment (48%) precipitated with low-speed centrifugation and exosomes
(3%) precipitated with ultracentrifugation.
2001), mast cells (Skokos et al., 2001a, 2003; Valadi et al.,
2007), T cells (Blanchard et al., 2002; Zakharova, Svetlova, &
Fomina, 2007), platelets (Heijnen et al., 1999), neuronal cells
(Andrews & Chakrabarti, 2005; Faure et al., 2006; Smalheiser,
3
&
MOON ET AL.
2007), intestinal epithelial cells (van Niel et al., 2001; Hundorfean et al., 2007), retinal pigment epithelium (McKechnie,
Copland, & Braun, 2003; Wang et al., 2009), Schwann cells
(Beswetherick, Lane, & Allt, 1992), and tumor cells (Segura,
Amigorena, & Thery, 2005; Taylor & Gercel-Taylor, 2005,
2008; Hao et al., 2006; Choi et al., 2007), tumor cell lines
(Andre et al., 2002), and sperm (Sullivan et al., 2005) also
secrete exosomes in vitro.
A urinary exosome was first identified by the Knepper
group in 2004 (Pisitkun, Shen, & Knepper, 2004). It was predicted that some exosomes originated from epithelial cell
types that face the urinary space, because specific proteins that
correspond to various epithelial cell types are identified as
follows; podocin and podocalyxin (glomerular podocytes),
megalin, cubilin, APSN, AQP-1, type IV carbonic anhydrase,
gamma-glutamyltransferase (proximal tubule), Tamm–Horsfall
protein (THP), CD9, the type 2 Na-K-2Cl cotransporter (thick
ascending limb of Henle), NCC (distal convoluted tubule),
AQP-2, mucin-1, the RH type C glycoprotein (collecting
duct), and uroplakin-1 and -2 (Urinary bladder) (Pisitkun,
Shen, & Knepper, 2004; Hoorn et al., 2005).
2. Mechanism of Exosome Excretion
The excretion pathway of exosomes in apical membranes is
shown at Figure 3. The mechanism of exosomes excretion could
be usually described as three steps. First, the incorporation of
proteins into multivesicular body (MVB) begins when plasmamembrane proteins are monoubiquitinated. This monoubiquitination is different from polyubiquitination that targets proteins
to the proteasome (Polo et al., 2002; Shekhtman & Cowburn,
2002). Proteomic data of urinary exosomes showed that they
contain many ubiquitinated proteins, including E2 and E3 components related to the ubiquitination pathway. Second, these
ubiquitinated proteins are endocytosed, follow the endosomal
pathway (Pisitkun, Shen, & Knepper, 2004), and finally fuse
into MVBs (Futter & White, 2007). In this stage, internal
vesicles could take up proteins, mRNAs, and micro-RNAs of
cytoplasm of the cell. Some proteins identified from the urinary
exosome include members of the endosomal-sorting complex
required for transport (ESCRT) Complex, ATPase complexes,
and accessory proteins involved in MVB formation (Pisitkun,
Shen, & Knepper, 2004). Endocytosed proteins are sorted within
intralumenal vesicles (ILVs) via ESCRT component lipids and/
or tetraspanines-enriched microdomains (Gruenberg, 2003; van
Niel et al., 2006). ESCRT consist of several components.
Sorting is initiated by recognition of mono-ubiquitinated cargo
proteins by TAM, epidermal growth factor receptor substrate 15
(Eps15) and hepatocyte growth factor-regulated kinase substrate
(Hrs), a component of ESCRT-0. Hrs also recruits tumor
susceptibility gene 101 (Tsg101) of ESCRT I complex, which
binds to ESCRT II. ESCRT-II moves to the plasma membrane,
and clusters at the ubiquitinated cargo. Ubiquitin of cargo
proteins are removed by de-ubiquitylating enzyme recruited by
ESCRT-III before cargo proteins enter into ILVs. Vps4 with
ATPase activity play a role in disassembling all ESCRTs from
membranes (Babst, 2005). Alix identified from urine are class E
Vps proteins that act as a linker between various ESCRT
complexes (Morelli et al., 2004). As an alternative pathway,
ESCRT-independent mechanisms to sort proteins into ILVs
within MVBs have been identified recently that are regulated by
ceramide-mediated budding of exosome vesicles into multivesicular endosomes (Trajkovic et al., 2008). Third, exosomes are
secreted into the urinary space by fusion of MVBs into the
corresponding epithelial membrane. The mechanisms of
exosome release are similar to those involved in the fusion of
secretory lysosomes with the plasma membrane, but the mechanism was not defined (Stinchcombe, Bossi, & Griffiths, 2004;
Yu, Harris, & Levine, 2006; Proux-Gillardeaux et al., 2007;
Charette & Cosson, 2008). It has been reported that the major
factors for exosomes release are calcium ionophores (Jaiswal,
Andrews, & Simon, 2002; Khvotchev et al., 2003; Savina et al.,
2003), decrease of membrane cholesterol (Lebrand et al., 2002;
Stoeck et al., 2006; Llorente, van Deurs, & Sandvig, 2007;
Chen et al., 2008), inhibition of cholesterol biosynthesis, overexpression of citron kinases, and Rab11 (Savina, Vidal, &
Colombo, 2002) or RhoA effector (Perret et al., 2005; Rodriguez-Boulan, Kreitzer, & Musch, 2005; Chen et al., 2008).
Recent imaging techniques, such as stimulated-emission
depletion (STED) and stochastic optical reconstruction microscopy (SORM) (Willig et al., 2006; Bates et al., 2007),
increased spatial resolutions of 20–40 nm to assist the visual-
FIGURE 3. The excretion pathway of exosomes in apical membranes. The mechanism of exosomes
excretion could be usually described as three steps. First, plasma membrane-containing membrane proteins are
internalized by a signal such as ubiquitination. Second, the internal vesicles are fused into multivesicular body
(MVB) outer membrane. In this stage, internal vesicles could take up proteins, mRNAs, and micro-RNAs of
cytoplasm of the cell. MVBs contain intralumenal vesicles (ILVs). Third, vesicles, or exosomes, are secreted
to the extracellular region by fusion of MVBs into plasma membrane. [Color figure can be viewed in the
online issue, which is available at wileyonlinelibrary.com]
4
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
ization of the real-time trafficking of exosomes in cells, and
finally it will provide the opportunity to better understand the
biogenesis of various vesicles, including exosomes in living
cells.
3. Characteristics of Exosomes
There are other membrane vesicles, called apoptotic blebs,
microparticles (MPs) (Diamant et al., 2004; Lynch & Ludlam,
2007), and microvesicles (MVs) (Majno & Joris, 1995;
Aupeix et al., 1997; MacKenzie et al., 2001) in the extracellular environment. These apoptotic blebs, MPs, and MVs show
heterogeneous populations of membrane vesicles with variable
size of ranges 100–1,000 nm, compared to exosomes, which
showed homogeneous populations of membrane vesicles in
size (40–100 nm) (Mears et al., 2004; Pisitkun, Shen, & Knepper, 2004; Xiu et al., 2007; Simpson, Jensen, & Lim, 2008).
These heterogeneous membrane vesicles are shed directly
from the plasma membrane of stimulated cells. In the case of
MPs, different agents can induce release of phenotypically distinguishable particles (Baj-Krzyworzeka et al., 2002). These
membrane vesicles have the same membrane topology; cytosolic side of the lipid bilayer inside the vesicle, the luminal
part of the membrane exposed (Hugel et al., 2005).
Exosomes contain protein, lipid (Laulagnier et al., 2004;
Subra et al., 2007), and RNA (Ratajczak et al., 2006; Lotvall &
Valadi, 2007; Smalheiser, 2007; Valadi et al., 2007; Belting &
Wittrup, 2008; Rabinowits et al., 2009). The exosome composition varies depending on the cell type of origin (Schorey &
Bhatnagar, 2008). Nevertheless, all exosomes contain a number
of common protein components (van Niel et al., 2006). The
cytosolic proteins present on exosomes include Rabs, annexins,
as well as several adhesion molecules. Exosomes also contain
heat-shock proteins (Gastpar et al., 2005), and also carry some
cell-specific proteins like MHC II (Raposo et al., 1997; Denzer
et al., 2000) and CD86 present only on exosomes isolated from
antigen-presenting cells (APCs) (Raposo et al., 1996; Segura,
Amigorena, & Thery, 2005) and MFG-E8/lactadherin present
on exosomes from immature DCs (Veron et al., 2005) and
adipocytes (Aoki et al., 2007). Exosomes are also enriched in
proteins that participate in vesicle formation and trafficking
like the lysobisphosphatidic acid-binding protein, Alix. Other
proteins detected on exosomes are the metabolic enzymes. Consistent with their origin, exosomes typically do not contain
endoplasmic reticulum, mitochondria, or nuclear proteins.
The lipid compositions of exosomes are characteristic of
the cell origin. The analysis of lipid compositions have been
performed with exosomes derived from guinea pig reticulocytes (Vidal et al., 1989), rat mast cells (Laulagnier et al.,
2004), human B cell (Wubbolts et al., 2003), and human dendritic cells (Laulagnier et al., 2004). Lipids, such as lyos(bis)phosphatidic acid (LBPA) are enriched in internal
membranes of MVBs (Kobayashi et al., 1998), and play a key
role in ILV formation (Chu, Witte, & Qi, 2005) to indicate the
important roles of lipids in exosome biogenesis.
Also, exosomes derived from mast cells and dendritic cells
showed different phospholipid composition compared to the
parent cells. Especially, a high sphingomyelin level is shown in
exosomes derived from parent cells such as mast cells, dendritic
cells, and B-cells (Wubbolts et al., 2003). In addition, a nonasymmetrical distribution of phosphatidylethanolamine and a
Mass Spectrometry Reviews DOI 10.1002/mas
&
rapid flip–flop of lipids between the two membranes are
observed (Laulagnier et al., 2004). Furthermore, although most
lipids, including phosphatidylcholine, phosphatikylserine, phosphatidylethanolamine, lysophosphatidylcholine, and sphingomyelin, are also present on exosomes isolated from other cell
types, the ratios of these lipids vary.
The presence of lipid-raft domains on the exosomes has
been investigated (de Gassart et al., 2003; Kobayashi et al.,
2007). Various typical raft components, glycolipids, Src tyrosine kinases (Lyn), GPI-anchored proteins (AChE), stomatin,
and flotillin-1, were associated in a detergent-resistant membrane on the secreted exosomes (de Gassart et al., 2003;
Echarri, Muriel, & Del Pozo, 2007).
Recently, biologically active exosomal mRNAs were identified in a mouse mast cell line, a human mast cell line, and
primary bone marrow-derived mouse mast cells (Valadi et al.,
2007). With microarray and microRNA chip analysis, 1,300
mRNAs and 120 micro RNAs were detected in mast cells,
many of which were exosome-specific, and were not detected in
the cytoplasm of the donor cells. The exosomal mRNA of
mouse donor cells was transferred to human recipient cells, and
new mouse proteins were synthesized in human cells. In
addition to normal cells, tumor cells such as glioblastoma tumor
cells also release exosomes that contain mRNAs, micro-RNAs
and, angiogenic proteins (Skog et al., 2008). The mRNAs in
exosomes from tumor cells also could be delivered to normal
recipient cells in the tumor environment and were translated.
The tumor-specific mRNAs and micro-RNAs could be detected
in serum exosomes of the tumor patients; those data suggest
that the exosomes in serum could be used for diagnosis and
therapeutic decision for tumor patients (Rabinowits et al., 2009).
For urinary exosomes, recently it has been reported that
mRNA was extracted from prostate cancer-derived urinary
exosomes by a general RNA-isolation method (Nilsson et al.,
2009). In that study, two known prostate-cancer biomarkers,
PCA-3 and TMPRSS2: ERG, were detected in exosomes isolated from the urine of patients. This result suggested that urinary exosomes also contain mRNAs as exosomes secreted
from other cells, even if many studies on urinary exosomal
RNAs, including micro-RNAs, have not been reported so far.
It has been reported that exosomes secreted by tumor cell
lines induced tumor invasion (Hao et al., 2006; Segura, Amigorena, & Thery, 2005). Tumor-derived exosomes mediated
immunosuppression during tumor-infiltrating lymphocytes
processes, and exosomes secreted from tumor-cell lines played
a role as a strong angiogenic factor in vascular development
around the tumor (Kim et al., 2002; Koga et al., 2005).
It has been reported that the supernatants of primary
cortical cultures contain exosomes and GluR2/3 subunits
of AMPA (a-amino-3-hydroxy-5-methyl-4-isoxazolepropionic
acid) receptor, integral membrane proteins, are secreted into
the exosome fraction (Menager et al., 2007). Exosomes
secreted from oligodendrocytes also contains myelin proteins
(Trajkovic et al., 2008). Exosomes regulate the production and
turnover of myelin-membrane proteins, and also affect the
progression of neurodegenerative diseases (Vella et al., 2008).
Retroviruses such as HIV-1 accumulate in MVBs in
human macrophages, and escape with the intracellular machinery of MVBs for budding at plasma membranes (Fevrier &
Raposo, 2004). The HIV particles have similar components to
exosomes (Loomis et al., 2006). Exosomes can also transport
5
&
MOON ET AL.
prions (Fevrier et al., 2004), which could cause neurodegenerative disorders in humans and animals.
III. METHODOLOGY TO STUDY URINARY
EXOSOMES
Various studies with proteomics and other biochemical
approaches on exosomes from various sources (Mathivanan &
Simpson, 2009) are shown in Table 1. The preparation and
digestion method for urinary exosomes are described in detail
below.
A. Preparation of the Exosomes
There are many reports to prepare exosomes from various cells
and tissues. Usually, differential centrifugation, a combination
of differential centrifugation with gradient centrifugation, ultraTABLE 1.
PUBMED
ID
10878338
9685355
12519789
19109410
1 6302 7 29
17641064
14582906
15940614
1 7956 14 3
16446100
10545503
11390481
19367702
11487543
18452139
12147373
14975938
11231627
111 4566 2
11306949
126 2 655 8
6
filtration, and gel filtration, are used. To isolate exosomes from
a culture supernatant, cells and debris were removed with lowspeed centrifugation with 500–3,000g for 15 min. In this case,
to eliminate large-cell debris, filtration with 0.22 mm filters or
0.1 mm filters are used. To further purify exosomes, a linear
sucrose gradient (2.0–0.25 M sucrose) has been used. Recently,
an affinity capture method with magnetic beads has been introduced. With a culture supernatant of prostate cell lines (Jansen
et al., 2009), breast adenocarcinoma cell lines (Koga et al.,
2005), and ascites of ovarian cancer patient (Clayton et al.,
2001), tumor exosomes are collected with antibody-coated magnetic beads that have affinity to tumor-specific protein (KeryerBibens et al., 2006). In a study by Lamparski et al. (2002),
ultrafiltration (500 kDa membrane) and ultracentrifugation with
a 30% sucrose/deuterium oxide (98%) cushion (density,
1.210 g/cm3) were used; that method was useful to miniscale
process clinical studies with high yield.
Current studies of various exosomes, including urinary exosome
SAMPLES
B cell lymphoblastoid
cells
B cells
B cells
Brain tumor
Breast adenocarcinoma
Breast milk
Bronchoalveolar lavage
fluid
Colorectal cancer cells
Colorectal cancer cells
Cortical neurones
Dendritic cells
Dendritic cells
Hepatocytes
Intestinal epithelial cells
Keratinocytes
Malignant pleural
effusions
Malignant pleural
effusions
Mammary
adenocarcinoma
Mast cells
Mast cells
Mast cells
METHODS
REFERENCES
Western blotting
Duke rs et a l ., 2 0 00
Western blotting/ Immunoelectron Microscopy
Mass spectrometry/ Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting
Western blotting/ FACS
Mass spectrometry/ Western blotting/ Immunoelectron
Microscopy/ FACS
Escola et al., 1998
Wubbolts et al., 2003
Graner et al., 2009
Koga et al., 2005
Admyre et al., 2007
FACS/ Immunoelectron Microscopy
Admyre et al., 2003
Western blotting/ Immunoelectron Microscopy/ FACS
Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting
Mass spectrometry/ Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting
Western blotting
Huber et al., 2005
Choi et al., 2007
Faure et al., 2006
Thery et al., 1999
Thery et al., 2001
Conde-Vancells et al., 2008
van Niel et al., 2001
Chavez-Munoz et al., 2008
Western blotting
A n dr e e t a l . , 2 0 0 2
Mass spectrometry/ Western blotting
Bard et al., 2004
Western blotting/ Immunoelectron Microscopy
Wolfers et al., 2001
Skokos et al., 2001b
Skokos et al., 2001a
Skokos et al., 2003
17486113
Mast cells
15478216
15111327
16081791
Melanoma cells
Mesothelioma cells
Microglia
18309083
Oligodendrocytes
15908444
Plasma
ELISA
Western blotting
Western blotting
Mass spectrometry/ Western blotting/ Microarray/
miRCURY LNA Array
Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting
Mass spectrometry/ Western blotting/ Immunoelectron
Microscopy
Western blotting/ FACS/ Immunoelectron Microscopy
170766 7 9
Plasma
Western blotting
19028452
Plasma
10572093
630 7529
3597417
15210972
18520029
185 89 210
Platelets
Reticulocytes
Reticulocytes
Rov epithelial cells
Saliva
Serum
17133577
Synovial fluid
11907077
T cells
19190083
Tracheobronchial cells
153 26 28 9
1 7 700 6 4 0
1905 6867
19158352
Uri n e
U ri n e
Urine
Ur ine
Mass spectrometry/ Western blotting/ Immunoelectron
Microscopy
Western blotting/ FACS/ Immunoelectron Microscopy
Antibody based methods
Enzyme assay/ Antibody based methods
Western blotting/ Immunoelectron Microscopy
Mass spectrometry
Western blotting/microRNA Array
Immunoelectron Microscopy/ Mass spectrometry/ Western
blotting
Western blotting/ Immunoelectron Microscopy
Mass spectrometry/ Western blotting/ Immunoelectron
Microscopy/ FACS
Mass spectrometry/ Western blotting
Western blotting/ FACS
Mass spectrometry
Mass spectrometry
Valadi et al., 2007
Mears et al., 2004
Hegmans et al., 2004
Potolicchio et al., 2005
Trajkovic et al., 2008
Caby et al., 2005
Sabapatha, Gercel-Taylor &
Taylor 2006
Looze et al., 2009
Heijnen et al., 1999
Pan & Johnstone, 1983
Johnstone et al., 1987
Fevrier et al., 2004
Ogawa et al., 2008
Taylor & Gercel-Taylor, 2008
Skriner et al., 2006
Blanchard et al., 2002
Kesimer et al., 2009
Pisitkun, Shen & Knepper, 2004
Keller et al., 2007
Gonzales et al., 2009
Hogan et al., 2009
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
For the stability of urinary exosomes, addition of protease
inhibitor to urine sample increased the signal of Na–K–Cl
cotransporter isoform 2 in Western blotting experiment to
suggest that protease inhibitor prevented the protein from
degradation (Zhou et al., 2006b). For storage and recovery of
urinary exosomes, three different temperatures,
808C,
208C, and 48C, have been compared for periods of either
1 week or 7 months. Storage at 208C reduced the exosome
recovery compared to fresh urine. Storage at 808C with
extensive vortexing after thawing increased exosome recovery,
even in cases where samples had been frozen for 7 months.
The first and second morning urine, after normalizing with
urine creatinine, showed similar exosome recovery; those data
suggested low protein degradation in the bladder/urinary tract.
For biomarker identification, the normalization step after
obtaining biofluid from each animal or person is important
(Zhou et al., 2006a,b). Normalization with urine flow rate is
the best method. However, normalization with urine creatinine
is practical for urinary exosome with spot urine.
Briefly, the following is the method used to prepare urinary exosomes (Pisitkun, Shen, & Knepper, 2004). Protease
inhibitor (1.67 mL of 100 mM NaN3/2.5 mL of 10 mM
PMSF/50 mL of 1 mM Leupeptin per 50 mL urine) was used
to treat urine immediately after collection. To eliminate debris
such as cell-shedding membrane proteins, the urine was centrifuged at 17,000g for 15 min at 48C. The supernatant from the
first centrifugation was ultracentrifuged again at 200,000g for
1 hr at 48C, and the pellet was suspended in fresh buffer
(250 mM sucrose/10 mM triethanolamine/0.5 mM PMSF/
1 mM leupeptin) and transferred to 1.5 mL of polypropylene
tube. To remove THP, the most-abundant protein in urine, the
solution was incubated with dithiothreithol for 2 min at 958C.
Finally, the solution was re-ultracentrifuged, and the pellet
was used in the subsequent study.
As an alternative method, the ultrafiltration method with a
100 kDa MWCO filter can be used to prepare exosomes. In
this method, filter containing urine is centrifuged at 3,000g for
10–30 min at room temperature, and the upper phase is used
for urinary exosome capture. Whereas this method is rapid, it
led to a low yield of urinary exosomes compared to Pisitkun
and coworkers (Pisitkun, Shen, & Knepper, 2004; Cheruvanky
et al., 2007).
B. Digestion Methods
To digest urinary exosomes, in-gel trypsin digestion has been
used for gel slices from 1-D or 2-D SDS–PAGE. It is expected
that a general in-solution method would not work well
because the exosome is a small-vesicle structure that contains
cytosolic proteins and membrane proteins. Therefore, it will
increase the digestion efficiency of urinary exosomes by denaturation with detergent of membrane proteins on exosomes.
As a similar method of in-gel trypsin digestion, Tube-Gel
digestion (Lu & Zhu, 2005) and gel-assisted digestion (Han
et al., 2008) methods were introduced. These two methods
used detergent to denature membrane proteins in polyacrylamide gel without electrophoresis. For Tube-Gel digestion,
protein, acrylamide, ammonium persulfate, and TEMED were
mixed, and this solution was solidified in a small glass tube
for 30 min at room temperature. The gel was cut into small
pieces, and the steps that follow are similar to the in-gel
Mass Spectrometry Reviews DOI 10.1002/mas
&
trypsin digestion method. This method increased the number
of proteins and the coverage of proteins. Gel-assisted digestion
is a modified method of Tube-Gel digestion. This digestion
contains urea, EDTA, SDS, and polyacrylamide. The number
of protein identified is three-times more in this gel-assisted
digestion method than in with Tube-Gel digestion. In any
event, these Tube-Gel and gel-assisted digestion methods are
used to efficiently increase the number of proteins in several
studies: for HeLa (Han et al., 2008) and prostate-cancer cell
line (Lu & Zhu, 2005), mouse kidney (Han et al., 2008), and
mouse-brain proteome study (Yu et al., 2007).
Another approach to efficiently digest membrane protein
is the chemical-digestion method. Membrane proteins contain
transmembrane domains, which are difficult to identify with
proteomics analysis. Therefore, it is necessary to eliminate
membrane lipids for efficient digestion to identify membrane
proteins with shotgun proteomics. Some methods to extract
membrane proteins for a proteomics study were compared
with human cell membrane proteins (Speers & Wu, 2007).
Methods with acid-labile surfactant, urea, and organic solvent
have been compared (Zhang et al., 2007). Finally, a total of
729 proteins were identified, and among them 44–50% were
membrane proteins; that result suggested the necessity of lipid
solubilization for an efficient identification of membrane
proteins (Zhang et al., 2007). One of the methods to isolate
and digest membrane proteins is to use CNBr in formic acid
at room temperature (Washburn, Wolters, & Yates, 2001;
Samyn, Sergeant, & Van Beeumen, 2006). CNBr digests the
C-terminus of Methionine (M) at 48C or at room temperature.
Also, formic acid can digest at either or both sides of aspartate
(D) residues at high temperature (Li et al., 2001; Swatkoski
et al., 2007; Zhang & Basile, 2007; Swatkoski et al., 2008).
Because these chemical digestion methods produce too-large
peptides for nano-LC MS/MS analysis, they need an
additional protease treatment, such as with trypsin.
IV. PROTEOMICS APPROACHES WITH URINARY
EXOSOMES
Several proteomic studies on urinary exosomes have been performed to identify biomarkers predicative of urinary track diseases. Pisitkun, Shen, and Knepper (2004) and Pisitkun,
Johnstone, and Knepper (2006) first showed that the urine exosomes can enhance the detectability of relatively low-abundant
proteins that have potential pathophysiological significance by
isolating the exosomes (<100 nm) from urine samples collected
from normal human subjects with differential centrifugation. In
these studies, 295 proteins contained in these urinary exosomes
were measured with LC-MS/MS analysis. These 295 proteins
were suggested to be originated from epithelia that face the
urine space throughout the urinary system. Also, a majority of
these proteins were involved in (1) membrane trafficking,
mostly in the endosomal pathway, as well as in (2) the membrane proteins (cargo proteins, transporters, channels, and peripheral-membrane proteins), and GTP-binding proteins (Rab,
ARF, Rho, and Ral families). In addition to aquaporin-2, a urinary biomarker exploited in clinical studies of various waterbalance disorders, this study proposed several exosomal proteins
that can be used as potential biomarkers. These exosomal
proteins associate with kidney and systemic diseases, including
autosomal dominant polycystic kidney disease (Polucystin-1),
7
&
MOON ET AL.
Gitelman syndrome (SLC12A3, a NaCl cotrasporter), Batter
syndrome (Sodium potassium chloride cotransporter-2), autosomal recessive syndrome of osteopetrosis with renal tubular
acidosis (Carbonic anhydrase 2), and familial renal hypomagnesemia (FXYD domain-containing ion transport regulator-2).
Zhou et al. (2006a) from the same group also provided
27 proteins, whose abundances were increased (18 proteins) or
decreased (9 proteins) between the exosomes isolated from
urine samples of rats 8 hr after cisplatin injection to induce
an acute kidney injury (AKI) with 2D-DIGE followed by
MALDI-TOF-TOF analysis for the spots that showed differences in the two conditions. In this study, Fetuin-A was
suggested as a novel urinary exosomal biomarker to detect
acute kidney injury. Recently, Gonzales et al. (2009) from the
same group further provided the comprehensive proteome and
phosphoproteome profiles of urinary exosomes with LC-MS/
MS analyses. These profiles include 1,132 urinary exosomal
proteins, among which 177 proteins are associated with
various diseases according to the OMIM database, as well as
14 urinary exosomal phosphoproteins that correspond to
19 phosphorylation sites.
In addition, the bladder cells, uroepithelial cells that line
the bladder lumen, directly contact the urine space. Bladder
cancer is the fourth most common cause of cancer in men in
the United States (Jemal et al., 2006). Cytoscopy is the only
method to diagnose the disease even if it is invasive with poor
sensitivity. Therefore, it needs to identify biomarkers instead of
this cytoscopy to diagnose and monitor this disease. Recently,
the urinary exosomes originated from the bladder-cancer cells
have been used to identify biomarkers for early diagnosis
of bladder cancer (Smalley et al., 2008). They performed
LC-MS/MS analysis on the urinary exosomes collected from
five healthy individuals and four bladder cancer patients; eight
proteins had elevated abundances in the urinary exosomes from
the bladder-cancer patients. Among these eight proteins, five
proteins are associated with EGFR pathway, whereas the other
three proteins are alpha subunit of GsGTP-binding protein,
resistin, and retinoic acid-induced protein 3. As an another
study for bladder cancer, they presented proteomic analysis of
exosomes, with high quality purification, from cultured
HT1376 bladder cancer cells and identified 353 proteins with
new 72 proteins not reported previously (Welton et al.,
FIGURE 4. Proteome profiles identified from the urinary exosomes from healthy individuals. A: The Venn
diagram shows the overlaps between the urinary exosomal proteins found in two independent studies: Pisitkun
et al. and Gonzales et al. (right), and Smalley et al. (left). Among a total of 1,224 proteins, 283 proteins were
shared in the two data sets. B: The subcellular localization of the 283 shared proteins according to GO cellular
components (GOCCs). C: The GO biological processes (GOBPs) in which the 283 shared proteins are
involved. The number inside the parenthesis indicates the number of proteins that belong to the corresponding
GO annotation out of the 283 shared proteins. D: The lists of proteins that belong to several GOBPs (C) that
are likely to be closely associated with functions of urinary organs.
8
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
in press). Among them, 18 proteins were confirmed as exosomal proteins by a combination of Western blotting, flotation on
linear sucrose gradients and flow cytometry (Welton et al.,
in press). Some proteins such as CD36, CD44, 5T4, Basigin
and CD73, detected from exosomes of cultured HT1376 bladder cancer cells were confirmed positive on urinary exosomes
from a bladder cancer patient (Welton et al., in press).
In addition to kidney and bladder, prostate is a small
gland in men that is about the size and shape of a walnut
(Weiss, Kaplan, & Fair, 2004). It lies at the base of the urinary
TABLE 2.
&
bladder and surrounds the urethra which carries urine from the
bladder and out of the body. Prostate cancer occurs when cells
in the gland are damaged and start multiplying out of control
and it is the second most common cause of cancer death in
UK men (Kommu, Edwards, & Eeles, 2004; Mohammed
et al., 2007). Until now, researchers have used levels of
proteins, like prostate specific antigen (PSA), produced by
cancer cells to try to spot the aggressive tumors (Cadeddu
et al., 1993). Recently, to identify novel markers for prostate
cancer with a new approach, proteomics analysis with serum
Urogenital disease-related proteins identified from human urinary exosomes
Diseases
OMIM
Proteins
References
DIABETES INSIPIDUS
125800
aquaporin 2
a,b
FECHTNER SYNDROME
153640
myosin, heavy polypeptide 9,
non-muscle
a
EPSTEIN SYNDROME
153650
myosin, heavy polypeptide 9,
non-muscle
a
HYPERURICEMIC NEPHROPATHY
162000
CYSTINURIA
uromodulin precursor
a,b
uromodulin precursor
b
220100
solute carrier family 3,
member 1
a
RENAL GLUCOSURIA
233100
solute carrier family 5
(sodium/glucose
cotransporter), member 2
a,b
OSTEOPETROSIS WITH RENAL
TUBULAR ACIDOSIS
259730
carbonic anhydrase II
a,b
Disease description
CAUSE; ◦The inability of the renal collecting ducts to
absorb water in response to arginine vasopressin
◦type I (X-linked recessive form) : Mutation in the gene
encoding the vasopressin V2 receptor gene
◦type II (autosomal form) : Mutation in the AQP2 gene
SYMPTOM; ◦Polyuria in kidneys
◦Lower urinary tract dilatation in bladder
CAUSE; ◦Allelic mutations in the gene encoding
nonmuscle myosin heavy chain-9
SYMPTOM; ◦Nephritis
◦End stage renal disease (20-40 years)(28% of
patients) in kidneys
CAUSE; ◦Mutation in the myosin heavy chain-9
nonmuscle gene
SYMPTOM; ◦Nephritis
◦End stage renal disease (33% of patients)
◦Hypertension in kidneys
CAUSE; ◦Mutation in the gene encoding uromodulin
SYMPTOM; ◦Nephropathy
◦Renal failure
CAUSE; ◦Mutation in the SLC3A1 amino acid
transporter gene
◦Mutation in the SLC7A9 gene
SYMPTOM; ◦ Nephrolithiasis
CAUSE; ◦Mutation in the sodium/glucose
cotransporter SGLT2 encoded by the SLC5A2 gene
◦Mutation in the SLC16A12 gene
SYMPTOM; ◦Enuresis nocturna
◦Polyuria
CAUSE; ◦Mutation in the gene encoding carbonic
anhydrase II
SYMPTOM; ◦Renal tubular acidosis
CAUSE; ◦Mutation in the thiazide-sensitive Na-Cl
GITELMAN SYNDROME
263800
fibrocystin isoform 2
a
RENAL TUBULAR ACIDOSIS
267300
ATPase, H+ transporting,
lysosomal 56/58kDa, V1
subunit B1
a,b
RENAL TUBULAR
DYSGENESIS
a,b
angiotensin I converting
enzyme isoform 1 precursor
a,b
pendrin
a
RENAL TUBULAR DYSGENESIS
PENDRED SYNDROME
ENLARGED VESTIBULAR AQUEDUCT
267430
274600
600791
pendrin
a
BARTTER SYNDROME
601678
sodium potassium chloride
cotransporter 2
a,b
RENAL TUBULAR ACIDOSIS
602722
ATPase, H+ transporting,
lysosomal V0 subunit a4
a
cotransporter
SYMPTOM; ◦Polyuria
◦Renal potassium wasting
◦Renal magnesium wasting in kidneys
CAUSE; ◦Mutation in the ATP6V1B1 (ATP6B1) gene
SYMPTOM; ◦Kidney stones
CAUSE; ◦Mutations in various genes(renin,
angiotensinogen, angiotensin-converting enzyme, or
angiotensin II receptor type 1)
SYMPTOM; ◦Absence of differentiated proximal
tubules, primitive renal tubules may exist
◦Thickening of renal arterial walls
◦Anuria
CAUSE; ◦Mutation in the SLC26A4 gene
◦Heterozygous mutation in the FOXI1 gene
CAUSE; ◦Mutation in the SLC26A4 gene
◦Heterozygous mutation in the FOXI1 gene
CAUSE; ◦Mutation in the sodium-potassium-chloride
cotransporter-2 gene
SYMPTOM; ◦Renal salt wasting
◦Renal potassium wasting
◦Renal juxtaglomerular cell hypertrophy/hyperplasia
◦Polyuria ◦Nephrocalcinosis in kidneys
CAUSE; ◦Mutation in the ATP6V0A4 (ATP6N1B)
gene
CAUSE; ◦Mutation in the gene encoding uromodulin
MEDULLARY CYSTIC KIDNEY DISEASE
2
603860
uromodulin precursor
a,b
(UMOD; 191845)
SYMPTOM; ◦Impaired renal function
◦Impaired renal creatinine clearance and renal uric
acid clearance
◦Salt wasting and small kidneys
◦Tubulointerstitial nephritis and fibrosis
◦Interstitial inflammation
◦Disintegration of the tubular basement membrane
◦Progression to end stage renal failure in late
adulthood (fifth to seventh decade)
(Continued )
Mass Spectrometry Reviews DOI 10.1002/mas
9
&
MOON ET AL.
TABLE 2. (Continued )
RENAL TUBULAR ACIDOSIS
AMYLOIDOSIS
POLYCYSTIC KIDNEYS
604278
solute carrier family 4, sodium
bicarbonate cotransporter,
member 4
a
105200
lysozyme precursor
fibrinogen, alpha polypeptide
isoform alpha-E preproprotein
apolipoprotein A-I
preproprotein
a,b
protein kinase C and casein
kinase substrate in neurons 2
a
polycystin 1 isoform 1
precursor
a
uroplakin 3A precursor
a,b
a
a,b
CAUSE; ◦Mutation in the SLC4A4 gene
SYMPTOM; ◦Proximal renal tubular acidosis
◦Renal bicarbonate wasting
◦Normal distal tubule acid excretion
CAUSE; ◦Mutation in the apolipoprotein A1 gene, the
fibrinogen alpha-chain gene, or the lysozyme gene.
SYMPTOM; ◦Nephropathy with hematuria
◦Nephrotic syndrome and uremia
CAUSE; ◦Mutations in any of several human disease
173900
loci.
SYMPTOM; ◦Hypertension
◦Extrarenal cysts
◦Subarachnoid hemorrhage
◦Polycystic kidney
◦Renal failure
CAUSE; ◦Mutations in the RET gene or in the
RENAL ADYSPLASIA
191830
uroplakin IIIA gene
SYMPTOM; ◦Renal adysplasia and renal agenesis
◦Renal dysplasia
◦Megalocystis
CAUSE; ◦Contiguous gene deletion syndrome
WILLIAMS-BEUREN SYNDROME
194050
eukaryotic translation
initiation factor 4H isoform 1
a
BARDET-BIEDL SYNDROME
209900
ADP-ribosylation factor-like 6
a
resulting from the hemizygous deletion of several
genes on chromosome 7q11.23
SYMPTOM; ◦Small kidneys
◦Solitary kidney and nephrocalcinosis
◦Renal insufficiency, renal artery stenosis in kidney
◦Vesicoureteral reflux in ureters
◦Bladder diverticula
◦Urethral stenosis
◦Recurrent urinary tract infections in bladder
CAUSE; ◦Modification of CCDC28B gene
◦Mutations in MKS1 and MKS3
SYMPTOM; ◦Renal anomalies
◦Nephrogenic diabetes insipidus
DONNAI-BARROW SYNDROME
222448
low density lipoprotein-related
protein 2
a,b
CAUSE; ◦Mutation in the LRP2 gene
SYMPTOM; ◦Non-acidotic proximal tubulopathy in
kidneys
FRUCTOSE INTOLERANCE
229600
aldolase B, fructosebisphosphate
b
GM1-GANGLIOSIDOSIS, TYPE I
230500
galactosidase, beta 1 isoform
a
a,b
ACTION MYOCLONUS-RENAL FAILURE
SYNDROME
254900
scavenger receptor class B,
member 2
a,b
GLYCOGEN STORAGE DISEASE X
261670
bisphosphoglycerate mutase
2
b
SIMPSON-GOLABI-BEHMEL
SYNDROME, TYPE 2
300209
oral-facial-digital syndrome 1
protein
b
PHOSPHOGLYCERATE KINASE 1
DEFICIENCY
300653
phosphoglycerate kinase 1
b
OROFACIODIGITAL SYNDROME I
311200
oral-facial-digital syndrome 1
protein
b
CAUSE; ◦Mutation in the gene encoding aldolase B
SYMPTOM; ◦Proximal tubular acidosis in renal
CAUSE; ◦Mutation in the gene encoding betagalactosidase-1
SYMPTOM; ◦Glomerular epithelial cytoplasmic
vacuolization in kidneys
CAUSE; ◦Mutations in the SCARB2 gene
SYMPTOM; ◦Renal failure
◦Focal segmental glomerulosclerosis
◦Collapsing glomerulopathy in kidneys
CAUSE; ◦Mutation in the gene encoding muscle
phosphoglycerate mutase
SYMPTOM; ◦Renal failure
CAUSE; ◦Mutation in the CXORF5 gene in 1 affected
family
SYMPTOM; ◦Multicystic kidneys
CAUSE; ◦Mutation in the PGK1 gene
SYMPTOM; ◦Renal failure with myoglobinuria
CAUSE; ◦Mutations in the CXORF5 gene
SYMPTOM; ◦Adult onset polycystic kidney (50%) in
kidneys
(Continued )
from mice grafted with human prostate cancer xenografts
were processed and identified 44 tumor-derived proteins
(Jansen et al., 2009). Several proteins such as CD9 and proteasome b1 (PSMZB1) were identified in the exosome fraction
isolated from the prostate cancer PC346C cell line (Jansen
et al., 2009). Also, they showed that these exosomes contained
RNAs, including the gene fusion TMPRSS2-ERG product.
Furthermore, the RNAs of TMPRSS2-ERG and PCA-3 were
detected in urinary exosomes from prostate cancer patients
(Nilsson et al., 2009). This suggested that cancer-derived urinary exosomes offer possibilities for the identification of novel
biomarkers for prostate cancer.
10
Until now, proteome profiles identified from the urinary exosomes from healthy individuals are compared and analyzed in
this study (Fig. 4). Figure 4A shows the relationships between
1,081 proteins detected from Pisitkun, Shen, and Knepper
(2004) and Gonzales et al. (2009) and 426 proteins identified
from Smalley et al. (2008). Two hundred eighty three proteins
(23.1%) are shared in the data sets. Figure 4B shows that a
majority of the shared proteins are localized in vesicle-related
organelles (e.g., cytoplasmic and membrane-bound vesicles,
early and late endosome, lysosome, and ER-Golgi intermediate compartment), plasma membrane and extracellular region.
Figure 4C shows that the shared proteins are involved in the
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
&
TABLE 2. (Continued )
CAUSE; ◦Homozygous deletion on chromosome
HYPOTONIA-CYSTINURIA SYNDROME
606407
GLOMERULOCYSTIC KIDNEY DISEASE
WITH HYPERURICEMIA AND
ISOSTHENURIA
609886
solute carrier family 3,
member 1
a
uromodulin precursor
a,b
uromodulin precursor
b
2p21 that disrupts the SLC3A1 and PREPL genes
SYMPTOM; ◦Nephrolithiasis in kidney
◦Bladder cystine calculi in bladder
CAUSE; ◦Mutation in the UMOD gene
SYMPTOM; ◦Cystic dilatation of the Bowman space
◦The initial proximal convoluted tubule
CAUSE; ◦Heterozygous mutation in the APOE gene
SYMPTOM; ◦Proteinuria
LIPOPROTEIN GLOMERULOPATHY
611771
apolipoprotein E precursor
a,b
NEPHROLITHIASIS/OSTEOPOROSIS,
HYPOPHOSPHATEMIC, 2
612287
solute carrier family 9
(sodium/hydrogen
exchanger), isoform 3
regulator 1
a,b
612624
angiotensin I converting
enzyme isoform 2 precursor
angiotensin I converting
enzyme isoform 1 precursor
MICROVASCULAR COMPLICATIONS OF
DIABETES, SUSCEPTIBILITY TO, 3
a,b
a,b
607832
CD2-associated protein
a,b
HYPEROXALURIA, PRIMARY, TYPE II
260000
glyoxylate
reductase/hydroxypyruvate
reductase
a
2,8-DIHYDROXYADENINE
UROLITHIASIS
145500
angiotensin I converting
enzyme isoform 2 precursor
angiotensin I converting
enzyme isoform 1 precursor
membrane alanine
aminopeptidase precursor
dimethylarginine
dimethylaminohydrolase 1
glutamyl aminopeptidase
(aminopeptidase A)
hydroxyprostaglandin
dehydrogenase 15-(NAD)
membrane metalloendopeptidase
CAUSE; ◦Mutation in the SLC9A3R1 gene
SYMPTOM; ◦Nephrolithiasis
◦Renal phosphate wasting in kidneys
FOCAL SEGMENTAL
GLOMERULOSCLEROSIS 3
HYPERTENSION
◦Progressive kidney failure
◦Distinctive lipoprotein thrombi in glomerular
capillaries
CAUSE; ◦Variation in the gene encoding angiotensin
I-converting enzyme (ACE; 106180) on chromosome
17q23
Haploinsufficiency for CD2-associated protein
(CD2AP; 604241) increases the risk of focal
segmental glomerulosclerosis (FSGS). Focal
segmental glomerulosclerosis (FSGS) is a
histopathologic finding in several renal disorders
characterized by proteinuria and progressive decline
in renal function.
CAUSE; ◦Mutation in the glyoxylate
reductase/hydroxypyruvate reductase gene
SYMPTOM; ◦Calcium oxalate urolithiasis
a,b
a,b
a,b
Variations in many genes contribute to essential
hypertension. Examination of the biochemical
processes that effect blood pressure homeostasis
should elucidate some of the interactive physiologic
regulators that malfunction in persons with elevated
pressure and show whether single genes of large
effect are important in some.
a
a,b
a
a,b
adenine
phosphoribosyltransferase
isoform a
a
adenine
phosphoribosyltransferase
isoform b
a
102600
PROXIMAL RENAL TUBULAR ACIDOSIS
114760
carbonic anhydrase IV
precursor
a,b
RENAL CELL CARCINOMA
118955
clathrin heavy chain 1
a
Adenine phosphoribosyltransferase catalyzes the
formation of AMP from adenine and
phosphoribosylpyrophosphate. It can act as a salvage
enzyme for recycling of adenine into nucleic acids.
Complete or partial deficiency of APRT can lead to
accumulation of the insoluble purine 2,8dihydroxyadenine (DHA). Renal failure in kidney, and
urolithiasis in ureters are symptom of 2,8dihydroxyadenine urolithiasis.
Carbonic anhydrases (CAs) are a family of zinc
metalloenzymes. For background information on the
CA family. CA IV is a glycosylphosphatidylinositolanchored membrane isozyme expressed on the
luminal surfaces of pulmonary (and certain other)
capillaries and on the luminal surface of proximal renal
tubules. CA IV has ancient evolutionary status among
CA isozymes. It is functionally important in CO2 and
bicarbonate transport and has a possible role in
inherited renal abnormalities of bicarbonate transport.
Clathrin is a major protein component of the
cytoplasmic face of intracellular organelles, called
coated vesicles and coated pits. These specialized
organelles are involved in the intracellular trafficking of
receptors and endocytosis of a variety of
macromolecules. Clathrin molecules have a triskelion
structure composed of 3 noncovalently bound heavy
chains (CLTC) and 3 light chains.
(Continued )
cellular processes including vesicle-related processes (e.g.,
secretion, vesicle-mediated transport, membrane fusion, actin
cytoskeleton organization, and biogenesis), small GTPasemediated signal transduction, regulation of body-fluid levels
(vasodilation), inflammatory response, cell motility, and blood
vessel development. Figure 4D summarizes a list of the
proteins that belong to several cellular processes that might
reflect physiology of the urinary organs. Six proteins (KNG1,
SERPINA1, SERPING1, ENPEP, ANPEP, and MASP2) are
involved in regulation of body fluid levels. KNG1, a bradykinin upstream regulator, has been known to regulate the
Mass Spectrometry Reviews DOI 10.1002/mas
permeability of glomerular basement membrane (GBM) to
various molecules (Iarovaia, 2001; Pike et al., 2005). The
protein and chemical transporters (ABCB1, AQP2, SLC2A5,
SLC12A1, and SLC12A3) in the urinary exosomes might provide the state of the filtration system in the proximal tubule.
In particular, AQP2, an apical plasma-membrane protein, has
been suggested as an indicator of water-balance in the filtration system (Funayama et al., 2004).
These proteomic studies have generated comprehensive
proteome profiles of the urinary exosomes from healthy
individuals and patients with kidney disease and bladder
11
&
MOON ET AL.
TABLE 2. (Continued )
NEPHROPATHIC CYSTINOSIS
219800
cystinosis, nephropathic
isoform 1
a
HYPOMAGNESEMIA 2, RENAL
154020
FXYD domain-containing ion
transport regulator 2 isoform
1
a
podocin
a
AUTOSOMAL RECESSIVE STEROIDRESISTANT NEPHROTIC SYNDROME
600995
CAUSE; ◦Misrouting of the Na+,K(+)-ATPase gamma
subunit
CAUSE; ◦Mutations in the NPHS2 gene(podocin)
SYMPTOM; ◦Massive proteinuria
◦Hypoalbuminemia, hyperlipidemia and edema
◦Minimal glomerular changes.
CAUSE; ◦Mutation in the gene encoding fibrocystin.
SYMPTOM; ◦Enlarged kidneys
601313
polycystin 1 isoform 1
precursor
a
POLYCYSTIC KIDNEY DISEASE, ADULT,
TYPE II
173910
polycystin 2
a
DEFECTIVE KIDNEY ACID SECRETION
LEADING TO DISTAL RENAL TUBULAR
ACIDOSIS
179800
solute carrier family 4, anion
exchanger, member 1
a
GLUCOSE/GALACTOSE
MALABSORPTION
606824
solute carrier family 5
(sodium/glucose
cotransporter), member 1
a
234500
solute carrier family 6,
member 19
RENAL HYPOURICEMIA
220150
urate anion exchanger 1
isoform a
a
METACHROMATIC LEUKODYSTROPHY
DUE TO SAPOSIN B DEFICIENCY
249900
prosaposin isoform a
preproprotein
a,b
FRONTOTEMPORAL DEMENTIA,
CHROMOSOME 3-LINKED
600795
prosaposin isoform c
preproprotein
a
a
◦Renal failure,
◦Renal calculi (urate and calcium oxalate) in kidneys
SYMPTOM; ◦Renal magnesium wasting in kidneys
POLYCYSTIC KIDNEY DISEASE, ADULT,
TYPE I
HARTNUP DISORDER
CAUSE; ◦Mutation in the gene encoding cystinosin
SYMPTOM; ◦Renal tubular Fanconi syndrome
a
◦Cystic kidneys and renal failure
◦Increased echogenicity of entire parenchyma
◦Loss of corticomedullary differentiation
◦Interstitial fibrosis in kidneys
SYMPTOM; ◦Enlarged kidneys
◦Cystic kidneys acd renal failure
◦Increased echogenicity of entire parenchyma
◦Interstitial fibrosis in kidneys
◦Polycystic kidney
CAUSE; ◦Mutation in the SLC4A1 gene
SYMPTOM; ◦Nephrocalcinosis
CAUSE; ◦Mutation in the intestinal sodium/glucose
transporter
CAUSE; ◦Mutations in the SLC6A19 gene
SYMPTOM; ◦Pellagra-like light-sensitive rash
◦Cerebellar ataxia
◦Emotional instability and amino aciduria
CAUSE; ◦Mutation in the SLC22A12 gene
SYMPTOM; ◦Uric acid urolithiasis
◦Uric acid nephrolithiasis
◦Tubular defect in presecretory reabsorption of urate
◦Exercise-induced acute renal failure with acute
tubular necrosis in kidneys
CAUSE; ◦Mutation in the prosaposin gene
SYMPTOM; ◦Incontinence in bladder
CAUSE; ◦Mutation in the CHMP2B gene
SYMPTOM; ◦Urinary incontinence in bladder
Pisitkun, Shen, and Knepper (2004) and Gonzales et al. (2009).
Smalley et al. (2008).
b
cancer, identified differentially expressed proteins (DEPs)
from the detected proteome between healthy and diseased
conditions, and additionally proposed some of these
DEPs that can be validated with Western blotting (Zhou
et al., 2006a) as potential biomarkers for the investigated
diseases. Table 2 summarizes the association of all the
proteins detected from the above studies with urinary track
diseases.
V. CONCLUSION
In this review, we described the characteristics of urinary exosomes, and methods for proteomics approaches to urinary exosomes, and showed proteomic profiles of urinary exosomes
that may reflect pathophysiology of urogenital diseases. Many
exosomes were identified from various cells, tissues, and body
fluids, and urinary exosomes have also been identified recently
and added into the existing exosomes category. Exosomes
secreted from various cells contain proteins and RNAs, and
play important roles in cell–cell communications among exosomes-secreting cells and exosomes-accepting cells. However,
studies on the mechanism related to communications among
cells via urinary exosomes have been not reported. It might be
12
difficult to investigate the details of this mechanism because
interactions with other cells during urine production could be
limited. Even though natural functions of urinary exosomes
remain to be studied, the urinary exosomes have many advantages as starting materials for biomarker identification. The
identification of candidate biomarkers relevant to pathophysiology of specific diseases could be very promising to proceed
to validation steps with a large-scale human study. Urinary
exosomes might be a valuable target to identify biologically
meaningful biomarkers, finally available in clinics with assistance of systems approaches based on proteomics and/or
microarray data derived from urinary exosomes, ‘‘informative
vesicles.’’
VI. ABBREVIATIONS
2-D PAGE
APCs
AQP
CE-MS
DCs
DEPs
2 dimensional polyacrylamide gel electrophoresis
antigen-presenting cells
aquaporin
capillary electrophoresis mass spectrometry
dendritic cells
differentially expressed proteins
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
Eps15
ESCRT
GBM
Hrs
ILVs
IMAC
MHC
MPs
MVBs
MVs
MWCO
NCC
PMSF
SELDI-TOF MS
SORM
STED
THP
Tsg101
Vps
epidermal growth factor receptor substrate 15
endosomal sorting complex required for
transport
glomerular basement membraney
hepatocyte growth factor-regulated kinase
substrate
intralumenal vesicles
immobilized metal-ion chromatography
major histocompatibility complex
microparticles
multivesicular bodies
microvesicles
molecular weight cut off
thiazide-sensitive Na–Cl cotransporter
phenylmethanesulphonylfluoride
surface enhanced laser desorption/
ionization time-of-flight mass spectrometry
stochastic optical reconstruction microscopy
stimulated emission depletion
Tamm–Horsfall protein
tumor susceptibility gene 101
vacuolar protein-sorting
ACKNOWLEDGMENTS
This work was supported by the Grant of the Korean Ministry
of Education, Science and Technology (The Regional Core
Research Program/Anti-aging and Well-being Research
Center) and supported by the grant No. RTI04-01-01 from the
Regional Technology Innovation Program of the Ministry of
Knowledge Economy (MKE).
REFERENCES
Adachi J, Kumar C, Zhang Y, Olsen JV, Mann M. 2006. The human urinary proteome contains more than 1500 proteins, including a large
proportion of membrane proteins. Genome Biol 7:R80.
Admyre C, Grunewald J, Thyberg J, Gripenback S, Tornling G, Eklund A,
Scheynius A, Gabrielsson S. 2003. Exosomes with major histocompatibility complex class II and co-stimulatory molecules are present
in human BAL fluid. Eur Respir J 22:578–583.
Admyre C, Johansson SM, Qazi KR, Filen JJ, Lahesmaa R, Norman M,
Neve EP, Scheynius A, Gabrielsson S. 2007. Exosomes with immune
modulatory features are present in human breast milk. J Immunol
179:1969–1978.
Andre F, Schartz NE, Movassagh M, Flament C, Pautier P, Morice P,
Pomel C, Lhomme C, Escudier B, Le Chevalier T, Tursz T, Amigorena S, Raposo G, Angevin E, Zitvogel L. 2002. Malignant effusions
and immunogenic tumour-derived exosomes. Lancet 360:295–305.
Andrews NW, Chakrabarti S. 2005. There’s more to life than neurotransmission: The regulation of exocytosis by synaptotagmin VII. Trends
Cell Biol 15:626–631.
Aoki N, Jin-no S, Nakagawa Y, Asai N, Arakawa E, Tamura N, Tamura T,
Matsuda T. 2007. Identification and characterization of microvesicles
secreted by 3T3-L1 adipocytes: Redox- and hormone-dependent
induction of milk fat globule-epidermal growth factor 8-associated
microvesicles. Endocrinology 148:3850–3862.
Aupeix K, Hugel B, Martin T, Bischoff P, Lill H, Pasquali JL, Freyssinet
JM. 1997. The significance of shed membrane particles during programmed cell death in vitro, and in vivo, in HIV-1 infection. J Clin
Invest 99:1546–1554.
Azad NS, Rasool N, Annunziata CM, Minasian L, Whiteley G, Kohn EC.
2006. Proteomics in clinical trials and practice: Present uses and
future promise. Mol Cell Proteomics 5:1819–1829.
Mass Spectrometry Reviews DOI 10.1002/mas
&
Babst M. 2005. A protein’s final ESCRT. Traffic 6:2–9.
Baj-Krzyworzeka M, Majka M, Pratico D, Ratajczak J, Vilaire G,
Kijowski J, Reca R, Janowska-Wieczorek A, Ratajczak MZ. 2002.
Platelet-derived microparticles stimulate proliferation, survival,
adhesion, and chemotaxis of hematopoietic cells. Exp Hematol
30:450–459.
Bard MP, Hegmans JP, Hemmes A, Luider TM, Willemsen R, Severijnen
LA, van Meerbeeck JP, Burgers SA, Hoogsteden HC, Lambrecht
BN. 2004. Proteomic analysis of exosomes isolated from human
malignant pleural effusions. Am J Respir Cell Mol Biol 31:114–121.
Bates M, Huang B, Dempsey GT, Zhuang X. 2007. Multicolor superresolution imaging with photo-switchable fluorescent probes. Science
317:1749–1753.
Belew M, Yip TT, Andersson L, Ehrnstrom R. 1987. High-performance
analytical applications of immobilized metal ion affinity chromatography. Anal Biochem 164:457–465.
Belting M, Wittrup A. 2008. Nanotubes, exosomes, and nucleic acid-binding peptides provide novel mechanisms of intercellular communication in eukaryotic cells: Implications in health and disease. J Cell
Biol 183:1187–1191.
Beswetherick JT, Lane PA, Allt G. 1992. Schwann cell endocytosis: A role
in nerve regeneration? Neuropathol Appl Neurobiol 18:395–407.
Blanchard N, Lankar D, Faure F, Regnault A, Dumont C, Raposo G,
Hivroz C. 2002. TCR activation of human T cells induces the
production of exosomes bearing the TCR/CD3/zeta complex.
J Immunol 168:3235–3241.
Boron WF, Boulpaep EL. 2005. Medical physiology: A cellular and molecular approach. Philadelphia, PA: Elsevier Saunders.
Caby MP, Lankar D, Vincendeau-Scherrer C, Raposo G, Bonnerot C.
2005. Exosomal-like vesicles are present in human blood plasma. Int
Immunol 17:879–887.
Cadeddu JA, Pearson JD, Partin AW, Epstein JI, Carter HB. 1993.
Relationship between changes in prostate-specific antigen and prognosis of prostate cancer. Urology 42:383–389.
Cadieux PA, Beiko DT, Watterson JD, Burton JP, Howard JC, Knudsen
BE, Gan BS, McCormick JK, Chambers AF, Denstedt JD, Reid G.
2004. Surface-enhanced laser desorption/ionization-time of flightmass spectrometry (SELDI-TOF-MS): A new proteomic urinary test
for patients with urolithiasis. J Clin Lab Anal 18:170–175.
Castagna A, Cecconi D, Sennels L, Rappsilber J, Guerrier L, Fortis F,
Boschetti E, Lomas L, Righetti PG. 2005. Exploring the hidden human
urinary proteome via ligand library beads. J Proteome Res 4:1917–1930.
Castle AM, Huang AY, Castle JD. 2002. The minor regulated pathway, a
rapid component of salivary secretion, may provide docking/fusion
sites for granule exocytosis at the apical surface of acinar cells.
J Cell Sci 115:2963–2973.
Caubet C, Lacroix C, Decramer S, Drube J, Ehrich JH, Mischak H,
Bascands JL, Schanstra JP. 2010. Advances in urinary proteome
analysis and biomarker discovery in pediatric renal disease. Pediatr
Nephrol 25:27–35.
Charette SJ, Cosson P. 2008. Altered composition and secretion of lysosome-derived compartments in Dictyostelium AP-3 mutant cells.
Traffic 9:588–596.
Chavez-Munoz C, Morse J, Kilani R, Ghahary A. 2008. Primary human
keratinocytes externalize stratifin protein via exosomes. J Cell
Biochem 104:2165–2173.
Chen H, Yang J, Low PS, Cheng JX. 2008. Cholesterol level regulates
endosome motility via Rab proteins. Biophys J 94:1508–1520.
Cheruvanky A, Zhou H, Pisitkun T, Kopp JB, Knepper MA, Yuen PS, Star
RA. 2007. Rapid isolation of urinary exosomal biomarkers using a
nanomembrane ultrafiltration concentrator. Am J Physiol Renal
Physiol 292:F1657–F1661.
Choi DS, Lee JM, Park GW, Lim HW, Bang JY, Kim YK, Kwon KH,
Kwon HJ, Kim KP, Gho YS. 2007. Proteomic analysis of microvesicles derived from human colorectal cancer cells. J Proteome Res
6:4646–4655.
13
&
MOON ET AL.
Christensen EI, Birn H. 2001. Megalin and cubilin: Synergistic endocytic
receptors in renal proximal tubule. Am J Physiol Renal Physiol
280:F562–F573.
Chu Z, Witte DP, Qi X. 2005. Saposin C-LBPA interaction in late-endosomes/lysosomes. Exp Cell Res 303:300–307.
Clayton A, Court J, Navabi H, Adams M, Mason MD, Hobot JA, Newman
GR, Jasani B. 2001. Analysis of antigen presenting cell derived exosomes, based on immuno-magnetic isolation and flow cytometry.
J Immunol Methods 247:163–174.
Conde-Vancells J, Rodriguez-Suarez E, Embade N, Gil D, Matthiesen R,
Valle M, Elortza F, Lu SC, Mato JM, Falcon-Perez JM. 2008.
Characterization and comprehensive proteome profiling of exosomes
secreted by hepatocytes. J Proteome Res 7:5157–5166.
de Gassart A, Geminard C, Fevrier B, Raposo G, Vidal M. 2003. Lipid
raft-associated protein sorting in exosomes. Blood 102:4336–4344.
Decramer S, Wittke S, Mischak H, Zurbig P, Walden M, Bouissou F, Bascands JL, Schanstra JP. 2006. Predicting the clinical outcome of congenital unilateral ureteropelvic junction obstruction in newborn by
urinary proteome analysis. Nat Med 12:398–400.
Decramer S, Gonzalez de Peredo A, Breuil B, Mischak H, Monsarrat B,
Bascands JL, Schanstra JP. 2008. Urine in clinical proteomics. Mol
Cell Proteomics 7:1850–1862.
Denzer K, van Eijk M, Kleijmeer MJ, Jakobson E, de Groot C, Geuze HJ.
2000. Follicular dendritic cells carry MHC class II-expressing microvesicles at their surface. J Immunol 165:1259–1265.
Diamant M, Tushuizen ME, Sturk A, Nieuwland R. 2004. Cellular microparticles: New players in the field of vascular disease? Eur J Clin
Invest 34:392–401.
Dukers DF, Meij P, Vervoort MB, Vos W, Scheper RJ, Meijer CJ, Bloemena E, Middeldorp JM. 2000. Direct immunosuppressive effects of
EBV-encoded latent membrane protein 1. J Immunol 165:663–670.
Echarri A, Muriel O, Del Pozo MA. 2007. Intracellular trafficking of raft/
caveolae domains: Insights from integrin signaling. Semin Cell Dev
Biol 18:627–637.
Escola JM, Kleijmeer MJ, Stoorvogel W, Griffith JM, Yoshie O, Geuze
HJ. 1998. Selective enrichment of tetraspan proteins on the internal
vesicles of multivesicular endosomes and on exosomes secreted by
human B-lymphocytes. J Biol Chem 273:20121–20127.
Fang Y, Wu N, Gan X, Yan W, Morrell JC, Gould SJ. 2007. Higher-order
oligomerization targets plasma membrane proteins and HIV gag to
exosomes. PLoS Biol 5:e158.
Faure J, Lachenal G, Court M, Hirrlinger J, Chatellard-Causse C, Blot B,
Grange J, Schoehn G, Goldberg Y, Boyer V, Kirchhoff F, Raposo G,
Garin J, Sadoul R. 2006. Exosomes are released by cultured cortical
neurones. Mol Cell Neurosci 31:642–648.
Fevrier B, Raposo G. 2004. Exosomes: Endosomal-derived vesicles shipping extracellular messages. Curr Opin Cell Biol 16:415–421.
Fevrier B, Vilette D, Archer F, Loew D, Faigle W, Vidal M, Laude H,
Raposo G. 2004. Cells release prions in association with exosomes.
Proc Natl Acad Sci USA 101:9683–9688.
Fliser D, Novak J, Thongboonkerd V, Argilés A, Jankowski V, Girolami
MA, Jankowski J, Mischak H. 2007. Advances in urinary proteome
analysis and biomarker discovery. J Am Soc Nephrol 18:1057–1071.
Funayama H, Nakamura T, Saito T, Yoshimura A, Saito M, Kawakami M,
Ishikawa SE. 2004. Urinary excretion of aquaporin-2 water channel
exaggerated dependent upon vasopressin in congestive heart failure.
Kidney Int 66:1387–1392.
Futter CE, White IJ. 2007. Annexins and endocytosis. Traffic 8:951–958.
Gastpar R, Gehrmann M, Bausero MA, Asea A, Gross C, Schroeder JA,
Multhoff G. 2005. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer
cells. Cancer Res 65:5238–5247.
Geho DH, Luchini A, Garaci E, Belluco C, Petricoin E, Liotta LA. 2007.
Nanotechnology in clinical proteomics. Nanomedicine 2:1–5.
Gonzales PA, Pisitkun T, Hoffert JD, Tchapyjnikov D, Star RA, Kleta R,
Wang NS, Knepper MA. 2009. Large-scale proteomics and
14
phosphoproteomics of urinary exosomes. J Am Soc Nephrol 20:363–
379.
Gonzales PA, Pisitkun T, Knepper MA. 2009. Urinary exosomes: Is there
a future? Nephrol Dial Transplant 23:1799–1801.
Gonzalez-Begne M, Lu B, Han X, Hagen FK, Hand AR, Melvin JE, Yates
JR. 2009. Proteomic analysis of human parotid gland exosomes by
multidimensional protein identification technology (MudPIT).
J Proteome Res 8:1304–1314.
Graner MW, Alzate O, Dechkovskaia AM, Keene JD, Sampson JH, Mitchell DA, Bigner DD. 2009. Proteomic and immunologic analyses of
brain tumor exosomes. FASEB J 23:1541–1557.
Gruenberg J. 2003. Lipids in endocytic membrane transport and sorting.
Curr Opin Cell Biol 15:382–388.
Han CL, Chien CW, Chen WC, Chen YR, Wu CP, Li H, Chen YJ. 2008.
A multiplexed quantitative strategy for membrane proteomics:
Opportunities for mining therapeutic targets for autosomal dominant
polycystic kidney disease. Mol Cell Proteomics 7:1983–1997.
Hao S, Bai O, Yuan J, Qureshi M, Xiang J. 2006. Dendritic cell-derived
exosomes stimulate stronger CD8þ CTL responses and antitumor
immunity than tumor cell-derived exosomes. Cell Mol Immunol
3:205–211.
Haubitz M, Wittke S, Weissinger EM, Walden M, Rupprecht HD, Floege
J, Haller H, Mischak H. 2005. Urine protein patterns can serve as
diagnostic tools in patients with IgA Nephropathy. Kidney Int
67:2313–2320.
Hegmans JP, Bard MP, Hemmes A, Luider TM, Kleijmeer MJ, Prins JB,
Zitvogel L, Burgers SA, Hoogsteden HC, Lambrecht BN. 2004. Proteomic analysis of exosomes secreted by human mesothelioma cells.
Am J Pathol 164:1807–1815.
Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. 1999. Activated
platelets release two types of membrane vesicles: Microvesicles by
surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood 94:3791–3799.
Hoffert JD, Pisitkun T, Wang G, Shen RF, Knepper MA. 2006. Quantitative phosphoproteomics of vasopressin-sensitive renal cells: Regulation of aquaporin-2 phosphorylation at two sites. Proc Natl Acad Sci
USA 103:7159–7164.
Hogan MC, Manganelli L, Woollard JR, Masyuk AI, Masyuk TV, Tammachote R, Huang BQ, Leontovich AA, Beito TG, Madden BJ,
Charlesworth MC, Torres VE, LaRusso NF, Harris PC, Ward CJ.
2009. Characterization of PKD protein-positive exosome-like
vesicles. J Am Soc Nephrol 20:278–288.
Hoorn EJ, Pisitkun T, Zietse R, Gross P, Frokiaer J, Wang NS, Gonzales
PA, Star RA, Knepper MA. 2005. Prospects for urinary proteomics:
Exosomes as a source of urinary biomarkers. Nephrology (Carlton)
10:283–290.
Huber V, Fais S, Iero M, Lugini L, Canese P, Squarcina P, Zaccheddu A,
Colone M, Arancia G, Gentile M, Seregni E, Valenti R, Ballabio G,
Belli F, Leo E, Parmiani G, Rivoltini L. 2005. Human colorectal cancer
cells induce T-cell death through release of proapoptotic microvesicles:
Role in immune escape. Gastroenterology 128:1796–1804.
Hugel B, Martinez MC, Kunzelmann C, Freyssinet JM. 2005. Membrane
microparticles: Two sides of the coin. Physiology (Bethesda) 20:22–27.
Hundorfean G, Zimmer KP, Strobel S, Gebert A, Ludwig D, Buning J. 2007.
Luminal antigens access late endosomes of intestinal epithelial cells
enriched in MHC I and MHC II molecules: In vivo study in Crohn’s
ileitis. Am J Physiol Gastrointest Liver Physiol 293:G798–G808.
Iarovaia GA. 2001. Kallikrein-kinin system: novel facts and concepts
(literature review). Vopr Med Khim 47:20–42.
Jaiswal JK, Andrews NW, Simon SM. 2002. Membrane proximal lysosomes are the major vesicles responsible for calcium-dependent exocytosis in nonsecretory cells. J Cell Biol 159:625–635.
Jansen FH, Krijgsveld J, van Rijswijk A, van den Bemd GJ, van den Berg
MS, van Weerden WM, Willemsen R, Dekker LJ, Luider TM,
Jenster G. 2009. Exosomal secretion of cytoplasmic prostate cancer
xenograft-derived proteins. Mol Cell Proteomics 8:1192–1205.
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
Jemal A, Siegel R, Ward E, Murray T, Xu J, Smigal C, Thun MJ. 2006.
Cancer statistics, 2006. CA Cancer J Clin 56:106–130.
Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C. 1987. Vesicle
formation during reticulocyte maturation. Association of plasma
membrane activities with released vesicles (exosomes). J Biol Chem
262:9412–9420.
Kapsogeorgou EK, Abu-Helu RF, Moutsopoulos HM, Manoussakis MN.
2005. Salivary gland epithelial cell exosomes: A source of autoantigenic ribonucleoproteins. Arthritis Rheum 52:1517–1521.
Keller S, Rupp C, Stoeck A, Runz S, Fogel M, Lugert S, Hager HD,
Abdel-Bakky MS, Gutwein P, Altevogt P. 2007. CD24 is a marker
of exosomes secreted into urine and amniotic fluid. Kidney Int
72:1095–1102.
Keryer-Bibens C, Pioche-Durieu C, Villemant C, Souquere S, Nishi N,
Hirashima M, Middeldorp J, Busson P. 2006. Exosomes released by
EBV-infected nasopharyngeal carcinoma cells convey the viral latent
membrane protein 1 and the immunomodulatory protein galectin 9.
BMC Cancer 6:283.
Kesimer M, Scull M, Brighton B, DeMaria G, Burns K, O’Neal W,
Pickles RJ, Sheehan JK. 2009. Characterization of exosome-like
vesicles released from human tracheobronchial ciliated epithelium:
A possible role in innate defense. FASEB J 23:1858–1868.
Khurana M, Traum AZ, Aivado M, Wells MP, Guerrero M, Grall F,
Libermann TA, Schachter AD. 2006. Urine proteomic profiling of
pediatric nephrotic syndrome. Pediatr Nephrol 21:1257–1265.
Khvotchev MV, Ren M, Takamori S, Jahn R, Sudhof TC. 2003. Divergent
functions of neuronal Rab11b in Ca2þ-regulated versus constitutive
exocytosis. J Neurosci 23:10531–10539.
Kim CW, Lee HM, Lee TH, Kang C, Kleinman HK, Gho YS. 2002.
Extracellular membrane vesicles from tumor cells promote angiogenesis via sphingomyelin. Cancer Res 62:6312–6317.
Kim SH, Lechman ER, Bianco N, Menon R, Keravala A, Nash J, Mi Z,
Watkins SC, Gambotto A, Robbins PD. 2005. Exosomes derived
from IL-10-treated dendritic cells can suppress inflammation and
collagen-induced arthritis. J Immunol 174:6440–6448.
Knepper MA. 2009. Common sense approaches to urinary biomarker
study design. J Am Soc Nephrol 20:1175–1178.
Kobayashi T, Stang E, Fang KS, de Moerloose P, Parton RG, Gruenberg J.
1998. A lipid associated with the antiphospholipid syndrome
regulates endosome structure and function. Nature 392:193–197.
Kobayashi M, Katagiri T, Kosako H, Iida N, Hattori S. 2007. Global
analysis of dynamic changes in lipid raft proteins during T-cell activation. Electrophoresis 28:2035–2043.
Koga K, Matsumoto K, Akiyoshi T, Kubo M, Yamanaka N, Tasaki A,
Nakashima H, Nakamura M, Kuroki S, Tanaka M, Katano M. 2005.
Purification, characterization and biological significance of tumorderived exosomes. Anticancer Res 25:3703–3707.
Kommu S, Edwards S, Eeles R. 2004. The clinical genetics of prostate
cancer. Hered Cancer Clin Pract 2:111–121.
Lakkaraju A, Rodriguez-Boulan E. 2008. Itinerant exosomes: Emerging
roles in cell and tissue polarity. Trends Cell Biol 18:199–209.
Lamparski HG, Metha-Damani A, Yao JY, Patel S, Hsu DH, Ruegg C,
Le Pecq JB. 2002. Production and characterization of clinical grade exosomes derived from dendritic cells. J Immunol Methods 270:211–226.
Laulagnier K, Motta C, Hamdi S, Roy S, Fauvelle F, Pageaux JF, Kobayashi
T, Salles JP, Perret B, Bonnerot C, Record M. 2004. Mast cell- and
dendritic cell-derived exosomes display a specific lipid composition
and an unusual membrane organization. Biochem J 380:161–171.
Lebrand C, Corti M, Goodson H, Cosson P, Cavalli V, Mayran N, Faure J,
Gruenberg J. 2002. Late endosome motility depends on lipids via the
small GTPase Rab7. EMBO J 21:1289–1300.
Li A, Sowder RC, Henderson LE, Moore SP, Garfinkel DJ, Fisher RJ.
2001. Chemical cleavage at aspartyl residues for protein identification. Anal Chem 73:5395–5402.
Lin Y, Kimpler LA, Naismith TV, Lauer JM, Hanson PI. 2005.
Interaction of the mammalian endosomal sorting complex
Mass Spectrometry Reviews DOI 10.1002/mas
&
required for transport (ESCRT) III protein hSnf 7-1 with itself,
membranes, and the AAAþ ATPase SKD1. J Biol Chem 280:
12799–12809.
Llorente A, van Deurs B, Sandvig K. 2007. Cholesterol regulates prostasome release from secretory lysosomes in PC-3 human prostate cancer cells. Eur J Cell Biol 86:405–415.
Loomis RJ, Holmes DA, Elms A, Solski PA, Der CJ, Su L. 2006. Citron
kinase, a RhoA effector, enhances HIV-1 virion production by modulating exocytosis. Traffic 7:1643–1653.
Looze C, Yui D, Leung L, Ingham M, Kaler M, Yao X, Wu WW, Shen
RF, Daniels MP, Levine SJ. 2009. Proteomic profiling of human
plasma exosomes identifies PPARgamma as an exosome-associated
protein. Biochem Biophys Res Commun 378:433–438.
Lotvall J, Valadi H. 2007. Cell to cell signalling via exosomes through
esRNA. Cell Adh Migr 1:156–158.
Lu X, Zhu H. 2005. Tube-gel digestion: A novel proteomic approach for
high throughput analysis of membrane proteins. Mol Cell Proteomics
4:1948–1958.
Lynch SF, Ludlam CA. 2007. Plasma microparticles and vascular disorders. Br J Haematol 137:36–48.
MacKenzie A, Wilson HL, Kiss-Toth E, Dower SK, North RA, Surprenant
A. 2001. Rapid secretion of interleukin-1beta by microvesicle shedding. Immunity 15:825–835.
Majno G, Joris I. 1995. Apoptosis, oncosis, and necrosis. An overview of
cell death. Am J Pathol 146:3–15.
Mathivanan S, Simpson RJ. 2009. ExoCarta: A compendium of exosomal
proteins and RNA. Proteomics 21:4997–5000.
McKechnie NM, Copland D, Braun G. 2003. Hr44 secreted with exosomes: Loss from ciliary epithelium in response to inflammation.
Invest Ophthalmol Vis Sci 44:2650–2656.
Mears R, Craven RA, Hanrahan S, Totty N, Upton C, Young SL, Patel P,
Selby PJ, Banks RE. 2004. Proteomic analysis of melanoma-derived
exosomes by two-dimensional polyacrylamide gel electrophoresis
and mass spectrometry. Proteomics 4:4019–4031.
Menager MM, Menasche G, Romao M, Knapnougel P, Ho CH, Garfa M,
Raposo G, Feldmann J, Fischer A, de Saint Basile G. 2007.
Secretory cytotoxic granule maturation and exocytosis require the
effector protein hMunc 13-4. Nat Immunol 8:257–267.
Mischak H, Kaiser T, Walden M, Hillmann M, Wittke S, Herrmann A,
Knueppel S, Haller H, Fliser D. 2004. Proteomic analysis for the
assessment of diabetic renal damage in humans. Clin Sci (Lond)
107:485–495.
Mohammed AA, Shergill IS, Vandal MT, Gujral SS. 2007. ProstaScint
and its role in the diagnosis of prostate cancer. Expert Rev Mol
Diagn 7:345–349.
Moon PG, Hwang HH, Boo YC, Kwon J, Cho JY, Baek MC. 2008. Identification of rat urinary glycoproteome captured by three lectins using
gel and LC-based proteomics. Electrophoresis 29:4324–4331.
Morelli AE, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, Papworth GD, Zahorchak AF, Logar AJ, Wang Z, Watkins SC, Falo LD,
Jr., Thomson AW. 2004. Endocytosis, intracellular sorting, and processing of exosomes by dendritic cells. Blood 104:3257–3266.
Nilsson J, Skog J, Nordstrand A, Baranov V, Mincheva-Nilsson L, Breakefield
XO, Widmark A. 2009. Prostate cancer-derived urine exosomes: A novel
approach to biomarkers for prostate cancer. Br J Cancer 100:1603–1607.
Ogawa Y, Kanai-Azuma M, Akimoto Y, Kawakami H, Yanoshita R. 2008.
Exosome-like vesicles with dipeptidyl peptidase IV in human saliva.
Biol Pharm Bull 31:1059–1062.
Pan BT, Johnstone RM. 1983. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the
receptor. Cell 33:967–978.
Perret E, Lakkaraju A, Deborde S, Schreiner R, Rodriguez-Boulan E.
2005. Evolving endosomes: How many varieties and why? Curr
Opin Cell Biol 17:423–434.
Pieper R. 2008. Preparation of urine samples for proteomic analysis.
Methods Mol Biol 425:89–99.
15
&
MOON ET AL.
Pieper R, Gatlin CL, McGrath AM, Makusky AJ, Mondal M, Seonarain
M, Field E, Schatz CR, Estock MA, Ahmed N, Anderson NG,
Steiner S. 2004. Characterization of the human urinary proteome: A
method for high-resolution display of urinary proteins on two-dimensional electrophoresis gels with a yield of nearly 1400 distinct
protein spots. Proteomics 4:1159–1174.
Pike RN, Buckle AM, le Bonniec BF, Church FC. 2005. Control of the
coagulation system by serpins. Getting by with a little help from
glycosaminoglycans. FEBS J 272:4842–4851.
Pisitkun T, Shen RF, Knepper MA. 2004. Identification and proteomic
profiling of exosomes in human urine. Proc Natl Acad Sci USA
101:13368–13373.
Pisitkun T, Johnstone R, Knepper MA. 2006. Discovery of urinary biomarkers. Mol Cell Proteomics 5:1760–1771.
Polo S, Sigismund S, Faretta M, Guidi M, Capua MR, Bossi G, Chen H,
De Camilli P, Di Fiore PP. 2002. A single motif responsible for ubiquitin recognition and monoubiquitination in endocytic proteins.
Nature 416:451–455.
Poon TC. 2007. Opportunities and limitations of SELDI-TOF-MS in biomedical research: Practical advices. Expert Rev Proteomics 4:51–65.
Potolicchio I, Carven GJ, Xu X, Stipp C, Riese RJ, Stern LJ, Santambrogio L. 2005. Proteomic analysis of microglia-derived exosomes:
Metabolic role of the aminopeptidase CD13 in neuropeptide catabolism. J Immunol 175:2237–2243.
Proux-Gillardeaux V, Raposo G, Irinopoulou T, Galli T. 2007. Expression
of the Longin domain of TI-VAMP impairs lysosomal secretion and
epithelial cell migration. Biol Cell 99:261–271.
Qian WJ, Jacobs JM, Liu T, Camp DG II, Smith RD. 2006. Advances and
challenges in liquid chromatography-mass spectrometry-based proteomics profiling for clinical applications. Mol Cell Proteomics
5:1727–1744.
Rabinowits G, Gercel-Taylor C, Day JM, Taylor DD, Kloecker GH. 2009.
Exosomal microRNA: A diagnostic marker for lung cancer. Clin
Lung Cancer 10:42–46.
Raposo G, Nijman HW, Stoorvogel W, Liejendekker R, Harding CV,
Melief CJ, Geuze HJ. 1996. B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183:1161–1172.
Raposo G, Tenza D, Mecheri S, Peronet R, Bonnerot C, Desaymard C.
1997. Accumulation of major histocompatibility complex class II
molecules in mast cell secretory granules and their release upon
degranulation. Mol Biol Cell 8:2631–2645.
Ratajczak J, Miekus K, Kucia M, Zhang J, Reca R, Dvorak P, Ratajczak
MZ. 2006. Embryonic stem cell-derived microvesicles reprogram
hematopoietic progenitors: Evidence for horizontal transfer of
mRNA and protein delivery. Leukemia 20:847–856.
Righetti PG, Boschetti E, Lomas L, Citterio A. 2006. Protein equalizer
technology: The quest for a ‘‘democratic proteome’’. Proteomics
6:3980–3992.
Rodriguez-Boulan E, Kreitzer G, Musch A. 2005. Organization of vesicular trafficking in epithelia. Nat Rev Mol Cell Biol 6:233–247.
Ru QC, Katenhusen RA, Zhu LA, Silberman J, Yang S, Orchard TJ,
Brzeski H, Liebman M, Ellsworth DL. 2006. Proteomic profiling of
human urine using multi-dimensional protein identification technology. J Chromatogr A 1111:166–174.
Sabapatha A, Gercel-Taylor C, Taylor DD. 2006. Specific isolation of
placenta-derived exosomes from the circulation of pregnant women
and their immunoregulatory consequences. Am J Reprod Immunol
56:345–355.
Saito M, Kimoto M, Araki T, Shimada Y, Fujii R, Oofusa K, Hide M, Usui T,
Yoshizato K. 2005. Proteome analysis of gelatin-bound urinary proteins
from patients with bladder cancers. Eur Urol 48:865–871.
Samyn B, Sergeant K, Van Beeumen J. 2006. A method for C-terminal
sequence analysis in the proteomic era (proteins cleaved with cyanogen bromide). Nat Protoc 1:318–323.
Savina A, Vidal M, Colombo MI. 2002. The exosome pathway in K562
cells is regulated by Rab11. J Cell Sci 115:2505–2515.
16
Savina A, Furlan M, Vidal M, Colombo MI. 2003. Exosome release is
regulated by a calcium-dependent mechanism in K562 cells. J Biol
Chem 278:20083–20090.
Schorey JS, Bhatnagar S. 2008. Exosome function: From tumor immunology to pathogen biology. Traffic 9:871–881.
Segura E, Amigorena S, Thery C. 2005. Mature dendritic cells secrete
exosomes with strong ability to induce antigen-specific effector
immune responses. Blood Cells Mol Dis 35:89–93.
Serafini-Cessi F, Malagolini N, Cavallone D. 2003. Tamm–Horsfall glycoprotein: Biology and clinical relevance. Am J Kidney Dis 42:658–676.
Shekhtman A, Cowburn D. 2002. A ubiquitin-interacting motif from Hrs
binds to and occludes the ubiquitin surface necessary for polyubiquitination in monoubiquitinated proteins. Biochem Biophys Res Commun 296:1222–1227.
Simpson RJ, Jensen SS, Lim JW. 2008. Proteomic profiling of exosomes:
Current perspectives. Proteomics 8:4083–4099.
Skog J, Wurdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M,
Curry WT, Jr., Carter BS, Krichevsky AM, Breakefield XO. 2008.
Glioblastoma microvesicles transport RNA and proteins that promote
tumour growth and provide diagnostic biomarkers. Nat Cell Biol
10:1470–1476.
Skokos D, Le Panse S, Villa I, Rousselle JC, Peronet R, David B, Namane
A, Mecheri S. 2001a. Mast cell-dependent B and T lymphocyte activation is mediated by the secretion of immunologically active exosomes. J Immunol 166:868–876.
Skokos D, Le Panse S, Villa I, Rousselle JC, Peronet R, Namane A, David
B, Mecheri S. 2001b. Nonspecific B and T cell-stimulatory activity
mediated by mast cells is associated with exosomes. Int Arch
Allergy Immunol 124:133–136.
Skokos D, Botros HG, Demeure C, Morin J, Peronet R, Birkenmeier G,
Boudaly S, Mecheri S. 2003. Mast cell-derived exosomes induce
phenotypic and functional maturation of dendritic cells and elicit
specific immune responses in vivo. J Immunol 170:3037–3045.
Skriner K, Adolph K, Jungblut PR, Burmester GR. 2006. Association of
citrullinated proteins with synovial exosomes. Arthritis Rheum
54:3809–3814.
Smalheiser NR. 2007. Exosomal transfer of proteins and RNAs at synapses in the nervous system. Biol Direct 2:35.
Smalley DM, Sheman NE, Nelson K, Theodorescu D. 2008. Isolation and
identification of potential urinary microparticle biomarkers of bladder cancer. J Proteome Res 7:2088–2096.
Spahr CS, Davis MT, McGinley MD, Robinson JH, Bures EJ, Beierle J,
Mort J, Courchesne PL, Chen K, Wahl RC, Yu W, Luethy R,
Patterson SD. 2001. Towards defining the urinary proteome using
liquid chromatography-tandem mass spectrometry. I. Profiling an
unfractionated tryptic digest. Proteomics 1:93–107.
Speers AE, Wu CC. 2007. Proteomics of integral membrane proteins—
Theory and application. Chem Rev 107:3687–3714.
Stinchcombe J, Bossi G, Griffiths GM. 2004. Linking albinism and
immunity: The secrets of secretory lysosomes. Science 305:55–59.
Stoeck A, Keller S, Riedle S, Sanderson MP, Runz S, Le Naour F, Gutwein P, Ludwig A, Rubinstein E, Altevogt P. 2006. A role for exosomes in the constitutive and stimulus-induced ectodomain cleavage
of L1 and CD44. Biochem J 393:609–618.
Subra C, Laulagnier K, Perret B, Record M. 2007. Exosome lipidomics
unravels lipid sorting at the level of multivesicular bodies. Biochimie
89:205–212.
Sullivan R, Saez F, Girouard J, Frenette G. 2005. Role of exosomes in
sperm maturation during the transit along the male reproductive
tract. Blood Cells Mol Dis 35:1–10.
Sun W, Li F, Wu S, Wang X, Zheng D, Wang J, Gao Y. 2005. Human
urine proteome analysis by three separation approaches. Proteomics
5:4994–5001.
Swatkoski S, Gutierrez P, Ginter J, Petrov A, Dinman JD, Edwards N,
Fenselau C. 2007. Integration of residue-specific acid cleavage into
proteomic workflows. J Proteome Res 6:4525–4527.
Mass Spectrometry Reviews DOI 10.1002/mas
URINARY EXOSOMES
Swatkoski S, Gutierrez P, Wynne C, Petrov A, Dinman JD, Edwards N,
Fenselau C. 2008. Evaluation of microwave-accelerated residuespecific acid cleavage for proteomic applications. J Proteome Res
7:579–586.
Tao F, Lazarev A. 2007. Clinical proteomics: Opportunities for diagnostics, pharmaceuticals and the clinical laboratory. Expert Rev Proteomics 4:9–11.
Taylor DD, Gercel-Taylor C. 2005. Tumour-derived exosomes and their
role in cancer-associated T-cell signalling defects. Br J Cancer
92:305–311.
Taylor DD, Gercel-Taylor C. 2008. MicroRNA signatures of tumorderived exosomes as diagnostic biomarkers of ovarian cancer.
Gynecol Oncol 110:13–21.
Theodorescu D, Wittke S, Ross MM, Walden M, Conaway M, Just I,
Mischak H, Frierson HF. 2006. Discovery and validation of new
protein biomarkers for urothelial cancer: A prospective analysis.
Lancet Oncol 7:230–240.
Thery C, Regnault A, Garin J, Wolfers J, Zitvogel L, Ricciardi-Castagnoli
P, Raposo G, Amigorena S. 1999. Molecular characterization of
dendritic cell-derived exosomes. Selective accumulation of the heat
shock protein hsc73. J Cell Biol 147:599–610.
Thery C, Boussac M, Veron P, Ricciardi-Castagnoli P, Raposo G, Garin J,
Amigorena S. 2001. Proteomic analysis of dendritic cell-derived exosomes: A secreted subcellular compartment distinct from apoptotic
vesicles. J Immunol 166:7309–7318.
Thongboonkerd V. 2007. Recent progress in urinary proteomics. Proteomics Clin Appl 1:780–791.
Thongboonkerd V. 2008. Urinary proteomics: Towards biomarker
discovery, diagnostics and prognostics. Mol Biosyst 4:810–815.
Thongboonkerd V, Malasit P. 2005. Renal and urinary proteomics: Current
applications and challenges. Proteomics 5:1033–1042.
Thongboonkerd V, McLeish KR, Arthur JM, Klein JB. 2002.
Proteomic analysis of normal human urinary proteins isolated by
acetone precipitation or ultracentrifugation. Kidney Int 62:1461–
1469.
Thongboonkerd V, Klein JB, Arthur JM. 2003. Proteomic identification of
a large complement of rat urinary proteins. Nephron Exp Nephrol
95:e69–e78.
Tortora GJ, Grabowski SR. 2003. Principles of anatomy and physiology.
New York: Wiley.
Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F,
Schwille P, Brugger B, Simons M. 2008. Ceramide triggers budding
of exosome vesicles into multivesicular endosomes. Science
319:1244–1247.
Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. 2007.
Exosome-mediated transfer of mRNAs and microRNAs is a novel
mechanism of genetic exchange between cells. Nat Cell Biol 9:654–
659.
van Niel G, Raposo G, Candalh C, Boussac M, Hershberg R, CerfBensussan N, Heyman M. 2001. Intestinal epithelial cells secrete
exosome-like vesicles. Gastroenterology 121:337–349.
van Niel G, Porto-Carreiro I, Simoes S, Raposo G. 2006. Exosomes:
A common pathway for a specialized function. J Biochem
140:13–21.
Vella LJ, Sharples RA, Nisbet RM, Cappai R, Hill AF. 2008. The role of
exosomes in the processing of proteins associated with neurodegenerative diseases. Eur Biophys J 37:323–332.
Veron P, Segura E, Sugano G, Amigorena S, Thery C. 2005. Accumulation
of MFG-E8/lactadherin on exosomes from immature dendritic cells.
Blood Cells Mol Dis 35:81–88.
Vidal M, Sainte-Marie J, Philippot JR, Bienvenue A. 1989. Asymmetric
distribution of phospholipids in the membrane of vesicles released
during in vitro maturation of guinea pig reticulocytes: Evidence precluding a role for ‘‘aminophospholipid translocase’’. J Cell Physiol
140:455–462.
Mass Spectrometry Reviews DOI 10.1002/mas
&
Villen J, Gygi SP. 2008. The SCX/IMAC enrichment approach for global
phosphorylation analysis by mass spectrometry. Nat Protoc 3:1630–
1638.
Wang L, Li F, Sun W, Wu S, Wang X, Zhang L, Zheng D, Wang J, Gao
Y. 2006. Concanavalin A-captured glycoproteins in healthy human
urine. Mol Cell Proteomics 5:560–562.
Wang AL, Lukas TJ, Yuan M, Du N, Handa JT, Neufeld AH. 2009.
Changes in retinal pigment epithelium related to cigarette smoke:
Possible relevance to smoking as a risk factor for age-related macular degeneration. PLoS One 4:e5304.
Washburn MP, Wolters D, Yates JR III. 2001. Large-scale analysis of the
yeast proteome by multidimensional protein identification technology. Nat Biotechnol 19:242–247.
Weiss RE, Kaplan SA, Fair WR. 2004. Management of prostate diseases.
Caddo, OK: Professional Communications, Inc.
Weissinger EM, Wittke S, Kaiser T, Haller H, Bartel S, Krebs R, Golovko
I, Rupprecht HD, Haubitz M, Hecker H, Mischak H, Fliser D. 2004.
Proteomic patterns established with capillary electrophoresis and
mass spectrometry for diagnostic purposes. Kidney Int 65:2426–
2434.
Welton JL, Khanna S, Giles PJ, Brennan P, Brewis IA, Staffurth J, Mason
MD, Clayton A. 2010. Proteomic analysis of bladder cancer exosomes. Mol Cell Proteomics 9:1324–1338.
Willig KI, Rizzoli SO, Westphal V, Jahn R, Hell SW. 2006. STED microscopy reveals that synaptotagmin remains clustered after synaptic
vesicle exocytosis. Nature 440:935–939.
Wittke S, Haubitz M, Walden M, Rohde F, Schwarz A, Mengel M, Mischak H, Haller H, Gwinner W. 2005. Detection of acute tubulointerstitial rejection by proteomic analysis of urinary samples in renal
transplant recipients. Am J Transplant 5:2479–2488.
Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C,
Flament C, Pouzieux S, Faure F, Tursz T, Angevin E, Amigorena S,
Zitvogel L. 2001. Tumor-derived exosomes are a source of shared
tumor rejection antigens for CTL cross-priming. Nat Med 7:297–
303.
Wubbolts R, Leckie RS, Veenhuizen PT, Schwarzmann G, Mobius W,
Hoernschemeyer J, Slot JW, Geuze HJ, Stoorvogel W. 2003. Proteomic and biochemical analyses of human B cell-derived exosomes.
Potential implications for their function and multivesicular body formation. J Biol Chem 278:10963–10972.
Xiu F, Cai Z, Yang Y, Wang X, Wang J, Cao X. 2007. Surface anchorage
of superantigen SEA promotes induction of specific antitumor
immune response by tumor-derived exosomes. J Mol Med 85:511–
521.
Yang Z, Hancock WS. 2004. Approach to the comprehensive analysis of
glycoproteins isolated from human serum using a multi-lectin affinity column. J Chromatogr A 1053:79–88.
Yang Z, Hancock WS. 2005. Monitoring glycosylation pattern changes
of glycoproteins using multi-lectin affinity chromatography.
J Chromatogr A 1070:57–64.
Yu X, Harris SL, Levine AJ. 2006. The regulation of exosome secretion:
A novel function of the p53 protein. Cancer Res 66:4795–4801.
Yu H, Wakim B, Li M, Halligan B, Tint GS, Patel SB. 2007. Quantifying
raft proteins in neonatal mouse brain by ‘tube-gel’ protein digestion
label-free shotgun proteomics. Proteome Sci 5:17.
Zakharova L, Svetlova M, Fomina AF. 2007. T cell exosomes induce cholesterol accumulation in human monocytes via phosphatidylserine
receptor. J Cell Physiol 212:174–181.
Zhang S, Basile F. 2007. Site-specific pyrolysis-induced cleavage at
aspartic acid residue in peptides and proteins. J Proteome Res
6:1700–1704.
Zhang N, Chen R, Young N, Wishart D, Winter P, Weiner JH, Li L. 2007.
Comparison of SDS- and methanol-assisted protein solubilization
and digestion methods for Escherichia coli membrane proteome
analysis by 2-D LC-MS/MS. Proteomics 7:484–493.
17
&
MOON ET AL.
Zhou H, Pisitkun T, Aponte A, Yuen PS, Hoffert JD, Yasuda H, Hu X,
Chawla L, Shen RF, Knepper MA, Star RA. 2006a. Exosomal
Fetuin-A identified by proteomics: A novel urinary biomarker for
detecting acute kidney injury. Kidney Int 70:1847–1857.
Zhou H, Yuen PS, Pisitkun T, Gonzales PA, Yasuda H, Dear JW, Gross P,
Knepper MA, Star RA. 2006b. Collection, storage, preservation, and
normalization of human urinary exosomes for biomarker discovery.
Kidney Int 69:1471–1476.
Zurbig P, Mischak H. 2008. Capillary electrophoresis coupled to mass
spectrometry for biomarker discovery and diagnosis of kidney
diseases. Contrib Nephrol 160:107–126.
Pyong-Gon Moon received his M.Med. in School of Medicine from
Kyungpook National University, Republic of Korea in 2008. He is
currently a Ph.D. candidate in biomarker discovery and proteomics group
at School of Medicine of Kyungpook National University. His main
research interests are clinical proteomics, mass spectrometry-based urinary
proteomics, and urinary exosomes under supervision of Prof. Moon-Chang
Baek.
Sungyong You received his M.Sc. in Pharmacy from Seoul National University, Republic of Korea in 2005. He is currently a Ph.D. candidate at Pohang
University of Science and Technology. His research involves developing
systems medicine approaches for finding key regulators associated with
underlying mechanisms of complex human diseases under supervision of
Prof. Daehee Hwang.
Jeong-Eun Lee obtains a master at School of Medicine of Kyungpook
National University, Republic of Korea in 2009. She is currently a Ph.D.
candidate in the medical science at Kyungpook National University. She is
interested in defining the relationship between post-translational modification and signaling pathway using proteomic approach under supervision
of Prof. Moon-Chang Baek.
Daehee Hwang during the Ph.D. study in Department of Chemical Engineering at MIT, he had participated in several systems biology projects on
mortor neuron diseases. In 2003, he pursued his postdoctoral career at the
Institute for Systems Biology where he developed (1) data integration and
proteomic data analysis tools, (2) a systems medicine approach for understanding complex human diseases. In 2006, he joined POSTECH in Korea
as a junior faculty and is currently engaged in developing integrative
genomic/proteomic data analysis pipelines and various systems biology
frameworks for finding key regulators associated with underlying mechanisms in several complex human diseases.
Moon-Chang Baek received his Ph.D. degree in Pharmacy from Seoul
National University in 1997 and has held post-doctoral fellow positions in
the Department of Biological Chemistry and Molecular Pharmacology at
Harvard Medical School (1998–2002) as well as in the Department of Life
Science at POSTECH (2002–2005). During the periods, he has studied on
traditional proteomics using various mass spectrometries (MALDI-TOF,
LCQ, and Q-TOF) and also developed a fusion technology, called Ligand
Profiling and Identification (LPI) technology, briding between cell biology
and proteomics. He has been serving as an assistant professor at School of
Medicine of Kyungpook National University since 2005. He has currently
focused on the identification of biomarker candidates for kidney diseases by developing various
proteomics technologies including glycoproteomics, membrane proteomics as well as enrichment
technologies for clinic specimen analysis.
18
Mass Spectrometry Reviews DOI 10.1002/mas