JACC: BASIC TO TRANSLATIONAL SCIENCE
VOL. 2, NO. 6, 2017
ª 2017 THE AUTHORS. PUBLISHED BY ELSEVIER ON BEHALF OF THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION. THIS IS AN OPEN ACCESS ARTICLE UNDER
ISSN 2452-302X http://doi.org/10.1016/j.jacbts.2017.08.004
THE CC BY-NC-ND LICENSE (http://creativecommons.org/licenses/by-nc-nd/4.0/).
STATE-OF-THE-ART REVIEW
Endothelial- and Immune Cell-Derived Extracellular Vesicles in the Regulation of Cardiovascular Health and Disease Felix Jansen, MD,a Qian Li, MD,a,b Alexander Pfeifer, MD, PHD,c Nikos Werner, MDa
SUMMARY Intercellular signaling by extracellular vesicles (EVs) is a route of cell-cell crosstalk that allows cells to deliver biological messages to specific recipient cells. EVs convey these messages through their distinct cargoes consisting of cytokines, proteins, nucleic acids, and lipids, which they transport from the donor cell to the recipient cell. In cardiovascular disease (CVD), endothelial- and immune cell-derived EVs are emerging as key players in different stages of disease development. EVs can contribute to atherosclerosis development and progression by promoting endothelial dysfunction, intravascular calcification, unstable plaque progression, and thrombus formation after rupture. In contrast, an increasing body of evidence highlights the beneficial effects of certain EVs on vascular function and endothelial regeneration. However, the effects of EVs in CVD are extremely complex and depend on the cellular origin, the functional state of the releasing cells, the biological content, and the diverse recipient cells. This paper summarizes recent progress in our understanding of EV signaling in cardiovascular health and disease and its emerging potential as a therapeutic agent. (J Am Coll Cardiol Basic Trans Science 2017;2:790–807) © 2017 The Authors. Published by Elsevier on behalf of the American College of Cardiology Foundation. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
C
ardiovascular disease (CVD) still represents
vascular diseases associated with systemic endothe-
the leading cause of mortality worldwide.
lial damage, such as atherosclerosis, show signifi-
The underlying disease, atherosclerosis, is
cantly increased levels of circulating EVs (3,4).
initiated and propagated by continuous damage of
However, EVs are not simply inactive debris that
the vascular endothelium, leading to endothelial acti-
reflect cellular activation or injury. EVs can transfer
vation and apoptosis, the development of endothelial
proteins, cytokines, mRNA, or noncoding RNA such
dysfunction, and subsequent atherosclerotic lesion
as microRNA (miRNA) or long noncoding RNA to
formation (1). Endothelial cell (EC) injury is a key
target cells and influence their function and pheno-
element in the complex pathophysiology of athero-
type (5,6). Accordingly, the role of EVs has changed
genesis and triggers the release of EC-derived extra-
from being only a marker of vascular integrity toward
cellular
and
being relevant effectors in intercellular vascular
microvesicles (MVs) (2). Accordingly, patients with
signaling (7,8). In CVD, EVs have been shown to
vesicles
(EVs)
such
as
exosomes
From the aDepartment of Internal Medicine II, Rheinische Friedrich-Wilhelms University, Bonn, Germany; bDepartment of Cardiology, Second Hospital of Jilin University, Nanguan District, Changchun, China; and the cInstitute of Pharmacology and Toxicology, University of Bonn, Bonn, Germany. Drs. Werner, Pfeifer, and Jansen are supported by Deutsche Forschungsgemeinschaft (WE 4139/8-1, JA2352/2-1, DFG GRK1873). Dr. Jansen received support from Medical Faculty of the Rheinische Friedrich-WilhelmsUniversity Bonn, the Familie Schambach foundation and German Society of Cardiology. The authors have reported that they have no relationships relevant to the contents of this paper to disclose. Drs. Jansen and Li contributed equally to this study. All authors attest they are in compliance with human studies committees and animal welfare regulations of the authors’ institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more information, visit the JACC: Basic to Translational Science author instructions page. Manuscript received July 7, 2017; revised manuscript received August 14, 2017, accepted August 14, 2017.
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
contribute to disease development and progression
(21–23). Second, EVs are able to transfer their
ABBREVIATIONS
by promoting initial lesion formation, intravascular
biological content by membrane fusion with
AND ACRONYMS
calcifications, plaque progression, and thrombus for-
the recipient cell. The fusion process is
mation after rupture. In contrast, numerous studies
regulated by the lipid composition of EV
have demonstrated that certain subtypes of EVs can
membrane, and several reports indicate that
mediate vascular protection and endothelial regener-
the presence of phosphatidylserine contrib-
ation (9). In line with these findings, EVs released by
utes to membrane fusion (24). Third, incor-
progenitor or mesenchymal stem cells have been
poration of EVs into target cells is mediated
shown to improve cardiac function after myocardial
by endocytosis, pinocytosis, or phagocytosis
infarction in experimental studies, highlighting the
(25). Through these interaction routes, EVs
therapeutic potential of EVs in cardiovascular pathol-
transfer their biological contents containing
ogies (10). This review summarizes current knowl-
nucleic acids such as mRNA (26), noncoding
edge of EVs as regulators of cardiovascular health
RNAs (miRNAs [27], long noncoding RNAs
and disease and potential opportunities for therapeu-
[6]), proteins (28), cytokines (29), or bioactive
tic use.
lipids (30) (Figure 1).
BIOGENESIS OF EXTRACELLULAR
EXTRACELLULAR VESICLES AS
CVD = cardiovascular disease EC = endothelial cell EMV = endothelial cell-derived microvesicles
ESCRT = endosomal sorting complex required for transport
IL = interleukin miRNA = microRNA MV = microvesicles NO = nitric oxide PEG = polyethylene glycol TGF = transforming growth factor
VESICLES AND THEIR INTERACTIONS
EFFECTORS OF CARDIOVASCULAR DISEASES
WITH TARGET CELLS In cardiovascular biology, EVs have various physioEVs are membrane vesicles secreted from cells that
logical functions, including activation of platelets and
contain intracellular contents (11). Cells can release a
ECs, as well as regulation of inflammation and coag-
broad range of vesicles with diverse features. This
ulation (31–34). Therefore, EVs are emerging as key
review focuses on 2 major types: MVs and exosomes.
players in different stages of CVD development
MVs are large (>150 nm) vesicles that are released by
(31,32,35). The effects of EVs in CVD are extremely
budding from the plasma membrane, whereas exo-
complex and depend on the cellular origin, the
somes are smaller (30 to 100 nm) and originate from
functional state of the releasing cells, the intra-
the endosome (12). However, there is no strict cutoff
vesicular content, and the recipient cells (36,37). The
value that distinguishes MVs from exosomes by
following sections summarize the current knowledge
vesicle size, which can differ in diverse studies (12).
about EVs as effectors of CV disease progression or
Exosomes represent a homogeneous population of
791
Endothelial and Immune Cell EVs in CV Health and Disease
vascular repair.
vesicles that are formed by inward budding of the
DETRIMENTAL EFFECTS OF EXTRACELLULAR VESICLES
multivesicular body (MVB) membrane. Exosome
ON VASCULAR FUNCTION. Endothelial
biogenesis is mediated mainly by the endosomal
occurs as a response to cardiovascular risk factors
sorting complex required for transport (ESCRT) pro-
and represents the initial step in atherosclerosis
tein (13) or lipid ceramide and neutral sphingomyeli-
development, the underlying pathology of CVD
nase, the enzyme that converts sphingomyelin to
(38,39).
ceramide (14). Cargo sorting into exosomes involves
E n d o t h e l i a l d y s f u n c t i o n . Endothelial MVs have
ESCRT and associated proteins such as tumor sus-
been shown to impair vasorelaxation by inhibiting
ceptibility gene 101 protein (TSG101) and ALG-2-
nitric oxide (NO) production in target ECs. This
interacting protein X (ALIX) and small GTPases such
phenomenon is mediated through a decrease in
as Rab7a and Rab27b (15–17). Exosomes are liberated
endothelial NO synthase phosphorylation and activ-
into the extracellular space following fusion of MVBs
ity (40), local oxidative stress (41), or an increased
with the cell membrane, regulated by Rab27A, Rab11,
NADPH oxidase activity with MVs (33) and results in
and Rab31 (18,19). MVs represent a relatively hetero-
impaired vascular relaxation capacities. MVs in the
geneous population of vesicles formed by outward
aforementioned studies are obtained from ECs under
budding of the cell membrane. This process is regu-
physiological (40,41) or pathological (33) conditions,
lated by membrane lipid microdomains and regula-
but they all have detrimental effects on vaso-
tory proteins such as ADP-ribosylation factor 6 (ARF6)
relaxation. Regarding the relation between molecu-
(20). EVs can be regarded as intercellular messengers
lar
for various biological processes. Several routes of
investigation should be conducted to clarify and
interaction between EVs and recipient cells have been
compare the cargoes of EVs derived under different
described. First, EVs can directly activate target cell
conditions (e.g., by RNA sequencing or proteomic
surface receptors by bioactive ligands and proteins
analysis).
contents
and
function
of
dysfunction
EVs,
further
792
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
F I G U R E 1 EV Biogenesis and Interaction With Recipient Cells
Exosome formation starts with endocytosis, a process in which the cell membrane is pinched inward and captures bioactive molecules, resulting in the formation of the endosome. These molecules are sorted into smaller vesicles that bud from the perimeter membrane into the endosome lumen, forming vesicles; this leads to the multivesicular appearance of late endosomes and so they are also known as MVBs. From MSBs, exosome formation occurs by an ESCRT- and ceramide-dependent pathway. Cargo sorting into exosomes involves ESCRT and TSG101, ALIX, and Rab7a, and Rab27b. Exosomes are released into the extracellular space following the fusion of MVBs with the cell membrane, which is regulated by Rab27A, Rab11, and Rab31. Microvesicles are formed by the outward budding of the cell membrane, a process that is regulated by ARF6. Several routes of interaction between EVs and recipient cells have been described. First, EVs can directly activate target cell surface receptors. Second, EVs are able to transfer their biological content by membrane fusion with the recipient cell. Third, incorporation of EVs into target cells is mediated by endocytosis, pinocytosis, or phagocytosis. Using these interaction routes, EVs transfer their biological content containing nucleic acids such as mRNA, noncoding RNAs (microRNAs, long noncoding RNAs), proteins, cytokines, or bioactive lipids. mRNA ¼ messenger RNA; MVB ¼ multivesicular bodies.
In line with the latter findings, MVs isolated from patients with vascular (acute coronary syndrome
may have diverse biological functions need to be addressed in future studies.
[42]) or predisposing disease (chronic renal failure or
Of interest, storage of human blood under standard
metabolic syndrome [43,44]) were shown to induce
blood-banking conditions results in accumulation of
endothelial dysfunction ex vivo in rat aortic rings.
MV-encapsulated hemoglobin. These erythrocyte-
In contrast, MVs from healthy subjects did not affect
derived MVs react with and degrade NO, inducing
endothelial function (42–44), indicating that the
endothelial
pathophysiological state of the releasing cell de-
derived exosomes derived under septic conditions
termines not only the number of released MVs but
have been shown to mediate septic endothelial
also their content and biological function. Which cell-
dysfunction
specific MVs mainly influence endothelial function
involving superoxide, NO, and peroxynitrite produc-
and whether isolated MVs from different cell types
tion (46). Studying EVs derived from patients
dysfunction
by
inducing
(45).
Finally,
endothelial
platelet-
apoptosis
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
represents an important translational approach to
glucose-treated cells, but not from healthy ECs,
exploring the relationship between EVs and certain
facilitated up-regulation of ICAM-1 and VCAM-1 in
diseases. However, comorbidities as well as patients’
endothelial target cells by activating p38 in an reac-
age and sex should be taken into consideration as
tive oxygen species-dependent manner (33). In light
possible
confounders.
and
of these proinflammatory effects and the increased
exclusion criteria should be thoughtfully determined,
levels of endothelial MVs in diabetic patients, one
and control groups must be carefully selected.
may speculate that MVs released under pathological
Endothelial
adhesion/
high-glucose conditions might represent a paracrine
i n fl a m m a t i o n . Inflammation
mediator transporting proinflammatory messages to
plays a pivotal role in CVD (38,47). EVs from various
target cells and thereby foster vascular inflammation
cellular sources contribute to vascular inflammatory
(61,62). However, while studying the functional ef-
processes including endothelial activation, monocyte
fects of EVs derived under pathological conditions,
adhesion, and transmigration (48–51).
one must consider that the quantity and/or content of
infiltration,
Therefore,
activation, and
inclusion
monocyte
In vitro studies have demonstrated that MVs can
EVs may vary with different modes and durations of
induce release of the proinflammatory cytokines
stimulation. Therefore, clearly defined pathological
interleukin (IL)-6 and IL-8 from ECs and leukocytes
conditions (including standardized concentrations
(52,53); and promote expression of adhesion proteins
and duration of cell stimulations) as well as a careful
ICAM-1,
facilitating
selection of an adequate control group are mandatory
increased adhesion of monocytes (54,55) and subse-
to elaborate EV functions depending on the parent
quent transmigration, leading to vascular inflamma-
cell conditions.
tion
VCAM-1,
and
plaque
and
E-selectin,
development.
Mechanistically,
Compared with the role of MVs, the role of exo-
increased adhesion of monocytes to ECs can be
somes in vascular inflammatory processes has been
mediated by the MV-mediated transfer of proin-
less explored (63,64). However, monocyte-derived
flammatory molecules such as oxidized phospho-
exosomes seem to induce vascular inflammation and
lipids (56), caspase-3 (57), or RANTES (regulated on
cell death by transferring inflammatory miRNAs into
activation, normal T cell expressed and secreted)
ECs resulting in a significant up-regulation of ICAM-1,
protein, which is transferred from platelet MVs to
CCL2, and IL-6 levels (65) and provocation of endo-
endothelial target cells (58). Platelet MVs from
thelial apoptosis by tissue factor release (66).
apoptotic platelets also facilitate differentiation be-
Atherosclerotic plaque development, progression, and
tween
professional
rupture. Atherosclerotic plaque rupture with subse-
phagocytes (59). Vascular inflammatory processes
quent coronary thrombosis represents the ultimate
involve different cells (e.g., ECs, monocytes, and
step in atherosclerotic lesion progression, leading to
platelets [48–51]). The
aforementioned in vitro
acute myocardial ischemia. Atherosclerotic plaques
studies demonstrated that EVs from diverse parent
can release large amounts of EVs, contributing to
cells can act on various types of target cells. In sum-
plaque progression and instability through various
mary, vascular inflammation seems to be regulated by
mechanisms. Plaque EVs originate mainly from leu-
complex
routes
cocytes, reflecting the local inflammatory environ-
involving a network of cells and EVs. To gain more
ment (67). Plaque EVs express surface antigens
insight into these multifaceted mechanisms, co-
consistent with their leukocyte origin, including ma-
incubation of different EVs with diverse cell types
jor histocompatibility complex classes I and II, and
may be helpful to study this “intercellular commu-
dose-dependently induce T-cell proliferation (68).
nication network.” However, there is a lack of
Antigen-specific activation of CD4 þ T cells was also
adequate in vitro models, which should be addressed
induced by dendritic exosomes, implicating their
resident
macrophages
intercellular
and
communication
in further studies.
potential involvement in vascular inflammation and
Pathological conditions modify EV content and
plaque development (69). Furthermore, plaque MVs
biological functions (60). MVs isolated from athero-
carry catalytically active tumor necrosis factor (TNF)-
sclerotic plaques transfer ICAM-1 to ECs and recruit
a–converting enzyme (TACE/ADAM17) and signifi-
inflammatory cells, suggesting that human plaque
cantly enhance the processing of its substrates TNF-a
MVs promote atherogenesis (24).
and TNF receptor, thereby promoting an inflamma-
Moreover, oxidatively modified, but not native,
tory response (70). Importantly, plaque EVs from
EC-derived EVs contain proinflammatory oxidized
patients or in vivo models are not single-component
phospholipids that elicit specific responses in ECs,
molecules, although mainly of leucocyte origin (67),
leading to the adhesion of monocytes (56). In line
and their constitutions may also depend on the stage
with these findings, EC-derived EVs generated from
of
plaque
progression
(stable
or
unstable?).
793
794
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
Therefore, standard procedures to derive and analyze
inflammation, plaque progression, and rupture are
plaque EVs should be established.
illustrated in Figure 1.
Local inflammation is enhanced by monocytic MVs
In summary, a broad body of evidence indicates
fostering leucocyte adhesion to postcapillary venules
the active involvement of EVs in plaque develop-
and T-cell infiltration in atherosclerotic plaques
ment, progression, and thrombus formation after
in vivo (71,72). Exosomes from T cells also can
rupture. However, there is an urgent need for addi-
contribute to atherosclerotic plaque development by
tional mechanistic studies to explore how their
inducing cholesterol accumulation in human mono-
atheroprone effects can be targeted to decelerate
cytes by the phosphatidylserine-receptor (73).
atherosclerotic lesion formation and rupture.
Vascular smooth muscle cell (VSMC) proliferation plays an important role in atherosclerotic plaque development (39,74). The effect of MVs on VSMC proliferation depends on the cellular origin. In vitrogenerated platelet-derived MVs promoted VSMC proliferation in a platelet-derived growth factor (PDGF)-independent mechanism with minor effects on migratory capacity (75,76). In turn, monocytederived MVs were shown to deliver a lethal message
FAVORABLE EFFECTS OF EXTRACELLULAR VESICLES ON VASCULAR FUNCTION. Despite the various deleterious
effects of EVs in the pathogenesis of CVD, an increasing body of evidence highlights the beneficial effects of certain EVs on vascular function. The following sections summarize the impact of EV on endothelial repair, their inhibitory effect on vascular inflammation, and their role in plaque stabilization.
by encapsulated caspase-1–inducing VSMC cell death
Endothelial protection and vascular repair. Given that
(77). More
recently, calcification-competent EVs
EC injury is not only a key element in the complex
derived from smooth muscle cells, valvular intersti-
pathophysiology of atherogenesis but also in in-stent
tial cells, and macrophages have been described as
restenosis occurring after treatment of coronary ste-
mediators of vascular calcification that modulate
nosis (90,91), a mechanistic understanding of EC
heart
(78–80).
repair is pivotally important to develop therapeutic
Although EVs show protective effects against heart
strategies to preserve endothelial integrity and
valve calcification, the potential underlying mecha-
vascular health. Several studies have shown that EVs
nisms are unknown and should be addressed in
particularly of endothelial origin can act as intercel-
future studies.
lular messenger to promote endothelial regeneration
valve
disease
and
atherogenesis
Angiogenesis is a fundamental process in CVD,
and vascular protection in vitro and in vivo.
contributing to plaque instability by promoting neo-
A potential contribution of endothelial MVs in EC
vascularization. Unstable plaques are characterized
survival was shown by Abid Hussein et al. (92), who
by an increased number of vasa vasorum mediating
demonstrated that endothelial MV release is cell
intraplaque hemorrhage (81). MVs isolated from hu-
protective by exporting caspase-3 into MVs and
man atherosclerotic plaques were shown to stimulate
thereby diminishing intracellular levels of proapo-
EC proliferation in vitro after CD40 ligation and to
ptotic caspase-3. Statins seem to facilitate endothelial
enhance in vivo angiogenesis. Interestingly, the pro-
health by promoting endothelial MV release in vitro
liferative effect of MVs isolated from atherosclerotic
(93). Nevertheless, the role of statins in endothelial
plaques was more pronounced using MVs from
MV release is still a matter of debate (94,95). Our
symptomatic patients than from patients without
group has demonstrated that annexin I/phosphati-
symptoms. Therefore, MVs could represent a major
dylserine
determinant of plaque vulnerability (82). Unstable
incorporation
human plaques contain large numbers of procoagu-
endothelial-regenerating cells against apoptosis (96).
lant MVs, originating mostly from leucocytes, eryth-
Inhibition of p38 activity by annexin I-containing
rocytes, and VSMCs localized within the necrotic core
endothelial MV is possibly involved in endothelial
(83). Once plaque rupture occurs, these MVs can
MV-mediated protection. However, whether annexin
initiate the coagulation cascade through different
I also mediates endothelial EV uptake in vivo will be
mechanisms (67,84): first, by the expression of tissue
explored in additional animal models. Another study
factor (mainly on monocytic MVs), one major initiator
showed that platelet-derived MVs induced changes in
of blood coagulation (34,85,86); and second, by
the early outgrowth cell, secretome, toward a more
exposure to phosphatidylserine on their outward
proangiogenic
membrane layer (87). Procoagulatory effects of MVs
outgrowth cell-mediated induction of endothelial
have been demonstrated in vitro and in vivo, where
regeneration in vitro and in vivo (97). These studies
they facilitated thrombus formation (88,89). The
indicate that MVs may influence the endothelial
deleterious
regeneration by the following 2 mechanisms: they
effects
of
EVs
inducing
vascular
receptor-dependent by
ECs
profile
protects
and
endothelial endothelial
amplified
the
MV and
early
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
F I G U R E 2 Detrimental Effects of Extracellular Vesicles on Vascular Function
The functional effect of EVs in cardiovascular disease is extremely complex and depends on the cellular origin, the functional state of the releasing cells, the biological content, the distinct recipient cell, and the transfer capacity of intravesicular functional bioactive molecules. Here, we illustrate the role of EVs as active promoters of endothelial dysfunction, vascular calcification, atherogenesis, plaque instability and thrombosis. Endothelial EVs impair vasorelaxation through local oxidative stress or through increased NADPH oxidase activity. EVs released by erythrocytes react with and degrade NO, EVs from platelet and atherosclerotic plaque induce endothelial apoptosis, both mediating endothelial dysfunction. Monocyte-derived EVs transfer inflammatory miRNAs into endothelial cells inducing vascular inflammation. Vascular smooth muscle cell-derived EVs act as mediators of vascular calcification modulating atherogenesis. EVs isolated from atherosclerotic plaques transfer ICAM-1 to endothelial cells and recruit inflammatory cells, contributing to plaque instability by promoting neovascularization. Once plaque rupture occurs, monocyte EVs and endothelial EVs initiate the coagulation cascade by the expression of tissue factor contributing to thrombosis. EV ¼ extracellular vesicles; miRNA ¼ microRNA; NO ¼ nitric oxide.
could directly interact with ECs and promote vascular
influencing migration and proliferation capacities in
regeneration, or they may activate endothelial pro-
target cells, in addition to the already described
genitor cells, facilitating endothelial repair (96,98). In
regenerative potential of endothelial MV in interac-
line with these findings, endothelial MVs carrying
tion
endothelial protein C receptor and activated protein C
miRNA-126 plays an important role in vascular health,
(APC) could also promote cell survival by induction of
miRNA-126–mediated
cytoprotective effects (99).
processing are not clearly identified and must be
with
progenitor
cells.
However,
downstream
although
signaling
and
Among the biological contents transferred by EVs
addressed in future research. Of note, EVs could
into target cells, miRNAs seem to play a crucial role by
activate EC by mRNA transfer from endothelial pro-
affecting mRNA and protein expression in recipient
genitor cells stimulating angiogenesis (105). In line
cells (100–102). Studies by our group have shown that
with these findings, MVs from ischemic muscle pro-
endothelial MVs promote vascular endothelial repair
moted progenitor cell differentiation and subsequent
by delivering functional miRNA-126 into recipient
postnatal vasculogenesis (106). Besides MVs, exo-
endothelial
cells
somes also play an important role in cardiovascular
(103,104). Of note, endothelial MV-mediated miRNA-
regeneration. Exosomes derived from mesenchymal
126–induced endothelial repair was altered under
stem cells reduced myocardial ischemia/reperfusion
pathological hyperglycemic conditions (103). These
injury (107). Furthermore, exosomes from cardiac
findings emphasize the fact that endothelial MVs
progenitor cells increased the migratory capacity of
can stimulate endothelial repair by functionally
ECs
and
vascular
smooth
muscle
in
vitro
and
may
contribute
to
vascular
795
796
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
T A B L E 1 Detrimental and Favorable Effects of Extracellular Vesicles on Vascular Function
Effect
EV Type
Isolation Method
Donor Cell/Origin
In Vitro Experiment
In Vivo Experiment
Effects
Mechanisms
Ref. #
Detrimental effects Injured Centrifugation endothelial MVs 20,000 g
Glucose-treated HCAECs
ApoE/ mice
HCAECs
Induce EC inflammation
Up-regulate ICAM-1 and VCAM-1 in EC by activating p38
(33)
Endothelial MVs
Ultracentrifugation RMVECs 100,000 g
Aortic rings from rats
—
Impair vasorelaxation
Local oxidative stress
(41)
Circulating MVs
Centrifugation 13,000 g
Patients with MI
Aortic rings from rats
—
Vasomotor dysfunction
Impair endothelial NO transduction pathway
(42)
Erythrocyte MVs
Differential centrifugation
Human packed red blood cells under standard blood banking conditions
Reduce vasoconstrictor effects
Degrade vasodilator NO
(45)
Platelet exosomes
Ultracentrifugation Platelets from ECs 100,000 g septic patients
—
Induce ECs apoptosis
Superoxide; NO and peroxynitrite production
(46)
PMN MVs
Ultracentrifugation PMNs from 100,000 g healthy volunteers
HUVECs
—
Induce ECs activation
Stimulate EC cytokine release Induction of tissue factor
(53)
Oxidized MVs
Ultracentrifugation Oxidatively 100,000 g modified HUVECs
Monocytes
—
Stimulate monocytes adhesion to ECs
Contain oxidized phospholipids
(56)
Plaque MVs
Centrifugation 20,500 g
—
Promote inflammatory response
Carry catalytically activeTNFalpha converting enzyme (TACE/ADAM17) Enhance the processing of TNF-a and TNF receptor
(70)
Monocyte MVs
Ultracentrifugation Human 100,000 g peripheral blood monocytes
—
Induce VSMCs cell death
Deliver cell death message via encapsulated caspase-1
(77)
CD40 ligand plus plaque MPs
Centrifugation 20,500 g
Stimulate endothelial proliferation and angiogenesis
CD40L signaling
(82)
—
Rat vasoactivity models
HUVECs Human atherosclerotic plaques
VSMCs
Human HUVECs atherosclerotic plaques
Wild-type and BalbC/ Nude mice
Continued on the next page
regeneration in vivo (108). Moreover, CD34þ exo-
secrete
somes promoted angiogenesis and preserved cardiac
inflammatory release of transforming growth factor
MVs,
which
in
turn
promote
anti-
function in ischemic myocardium by delivery of sonic
(TGF)-b 1 from macrophages. These findings suggest
hedgehog (109).
MVs are potent anti-inflammatory effectors, which at
In summary, EVs derived from endothelium,
an early stage of inflammation could contribute to its
platelets, or endothelium-regenerating cells play a
resolution (112). This effect seems to be mediated by
fundamental role by facilitating regenerative pro-
annexin I expression on the surface of these EVs (113).
cesses after vascular or myocardial injury (10,110,111).
EVs are also taken up by monocytes and B cells
Anti-inflammatory
through diverse mechanisms and affect target cells
v e s i c l e s . Several
effects
studies
of
have
extracellular reported
anti-
inflammatory effects of EVs. Of interest, neutrophils
toward
an
anti-inflammatory
phenotype
(114).
Mesenchymal stem cells contribute to inflammatory
797
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
T A B L E 1 Continued
Effect
EV Type
Isolation Method
Donor Cell/Origin
In Vitro Experiment
In Vivo Experiment
Effects
Mechanisms
Ref. #
Favorable effects Endothelial apoptotic bodies
Centrifugation 16,000 g
HUVECs
HUVECs
Endothelial MVs
Centrifugation 20,000 g
HCAECs
HCAECs
Platelet MVs
Centrifugation 20,000 g
Platelets
EOCs
Endothelial MVs
Centrifugation 20,000 g
HCAECs
Endothelial MVs
Centrifugation 20,000 g
Exosomes
Mice models of Promote atherosclerosis atheroprotective effects Increase plaque stability Enhance progenitor cells recruitment —
MiRNA-126dependent inhibition of RGS16 Enhance CXCR4 and CXCL12
(27)
Prevent HCAECs apoptosis
Annexin (96) I/phosphatidylserine receptor– dependent inhibition of p38 activation
Mice models of arterial wire-induced injury
Enhance vasoregenerative potential of EOCs
Enhance EOCs recruitment, migration, differentiation Release proangiogenic factors
HCAECs
Electric injury of murine carotid artery
Promote ECs migration and proliferation Accelerate re-endothelialization
Inhibit SPRED-1 (103) via EMV-mediated transfer of miRNA-126
HCAECs
VSMCs
Wire injury of murine carotid artery
Reduce neointima formation Diminished VSMCs proliferation and migration
Inhibit LRP6 via EMPs-mediated transfer of miRNA-126-3p
(104)
Differential centrifugation
CMPCs
HMECs
—
Stimulate HMECs migration
EMMPRIN-mediated
(108)
Exosomes
HPLC
Mesenchymal stem cells
Mice models of myocardial I/R injury
Reduce local and systemic inflammation
Restore bioenergetics Reduce oxidative stress Activate pro-survival signaling
(116)
Endothelial MVs
Centrifugation 20,000 g
HCAECs
ApoE-deficient mice
Promote anti-inflammatory effects
Reduce endothelial (118) ICAM-1 expression via the transfer of functional miRNA-222
Endothelial exosomes
Centrifugation 20,500 g
KLF2-transduced HASMCs or shear-stressstimulated HUVECs
Aorta of ApoE knockout mice
Atheroprotection
EV-mediated transfer of miRNA-143/145
Circulating MVs
Ultracentrifugation Blood
ApoE-deficient mice
Penetrate the vascular wall Inhibit VSMCs proliferation and migration
miRNA-223-mediated (121) IGF-1R/PI3K-Akt pathway
—
Monocytes
VSMCs
(97)
(120)
ApoE ¼ apolipoprotein E; CMPC ¼ cardiomyocyte progenitor cell; EC ¼ endothelial cell; EMMPRIN ¼ extracellular matrix metalloproteinase inducer; EMV ¼ endothelial MV; HASMC ¼ human aortic smooth muscle cell; HCAECs ¼ human coronary artery endothelial cells; HMEC ¼ human microvascular endothelial cell; HPLC ¼ high-performance liquid chromatography; HUVEC ¼ human umbilical vein endothelial cell; I/R ¼ ischemia/reperfusion; ICAM ¼ intercellular adhesion molecule; KLF ¼ Krüppel-like factor; MI ¼ myocardial infarction; MV ¼ microvesicles; NO ¼ nitric oxide; PMNs ¼ polymorphonuclear leukocytes; RGS16 ¼ regulator of G-protein signaling; RMVEC ¼ rat renal microvascular endothelial cell; SPRED ¼ sprouty-related EVH1 domain-containing protein; TNF ¼ tumor necrosis factor; VSMC ¼ vascular smooth muscle cell.
repression by releasing exosomes that induce secre-
reperfusion injury model resulted in a significant
tion of anti-inflammatory cytokines such as IL-10 and
reduction of local and systemic inflammation after
TGF- b (115). Administration of mesenchymal stem
24 h (116). In a renal ischemia/reperfusion model in
cell-derived exosomes in a myocardial ischemia/
rats, intravenously administered mesenchymal stem
798
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
cell-derived MVs limited inflammation as well as
In conclusion, beneficial and detrimental effects of
renal fibrosis (117). Finally, endothelial MVs promoted
EVs have been described in the regulation of vascular
anti-inflammatory effects in vitro and in vivo by
health and disease. However, there are no constant
reducing endothelial ICAM-1 expression by the
rules showing a clear relationship between origin and
transfer of functional miRNA-222 into recipient cells
function of EVs. EVs derived under pathological
(118). In line with these data, ECs suppressed mono-
conditions can induce cardiovascular harm (e.g.,
cyte activation through secretion of exosomes con-
plaque EVs promote inflammatory response [70]) but
taining anti-inflammatory miRNAs (119). Importantly,
also demonstrate atheroprotective functions (e.g.,
despite a large amount of in vitro cellular studies,
MVs from ischemic muscle induce progenitor cell
more in vivo models exploring local and systemic
differentiation [106]). Even the same original EVs can
inflammation should be applied to validate and
show both detrimental and favorable effects (e.g.,
confirm the anti-inflammatory effects of EVs.
endothelial EVs impairing vasorelaxation on one
Plaque
stabilization
and
antithrombotic
e f f e c t s . miRNA-containing EVs have been shown to promote vascular protection and plaque stabilization through various mechanisms. Injection of miRNA126-3p–enriched
apoptotic
bodies
of
endothelial
origin promoted atheroprotective effects by limiting plaque
size,
increasing
plaque
stability,
and
enhancing progenitor cell recruitment. miRNA-126– dependent inhibition of regulator of G protein
hand [41] and reducing neointima formation on the other hand [104]). In order to clarify the multifaceted character of EVs and make data more comparable, additional efforts should be put into standardized EV generation techniques. Once they are established, indepth exploration of EV-incorporated and transferred biological molecules and their intracellular processing is necessary to gain more clarity in the understanding of EV function.
signaling (RGS16) and subsequent enhancement of
THERAPEUTIC POTENTIAL OF
CXCR4 and CXCL12 was elaborated as the underlying
EXTRACELLULAR VESICLES IN
mechanism (27). Exosomes from KLF-2–transduced or
CARDIOVASCULAR DISEASES
shear-stress–stimulated ECs are enriched in miRNAs 143 and 145. By transferring functional miRNAs, EVs
EXTRACELLULAR VESICLES AS NOVEL THERAPEUTIC
were shown to control target gene expression in
TOOL? EVs have emerged as vectors for transferring
vascular smooth muscle target cells and reduce
biological information by proteins or genetic mate-
atherosclerotic lesion formation in the aorta of
rial,
apolipoprotein E knockout mice. These findings sug-
favoring
gest that atheroprotective stimuli induce communi-
atherosclerosis.
thereby
maintaining
endothelial
vascular
repair,
or
homeostasis,
even
limiting
cation between ECs and vascular smooth muscle cells
Due to these beneficial effects, there has been a
through miRNA-transferring EVs (120). Similarly,
rising interest in the potential use of EVs as thera-
circulating miRNA-223–containing exosomes could
peutic vectors in the field of cardiovascular medicine
penetrate the vascular wall and inhibit vascular
and regenerative therapy. Multiple studies have
smooth muscle cell proliferation and migration,
shown that the transfer of functional miRNAs into
resulting in decreased plaque size (121). Whereas MVs
target tissue by EVs promotes vascular regeneration
are involved in enhancing blood clotting processes,
and
exosomes seem to suppress platelet aggregation and
lighting
occlusive thrombosis by inhibiting platelet CD36,
transferring EVs. In addition to miRNA-containing
inducing antithrombotic effects. However, further
EVs, many reports describing nanoparticles as a
research is needed to validate these findings in
new
adequate in vivo models and to understand the
miRNAs to recipient cells have been published
opposing roles of exosomes and MVs in this context.
recently (124–126) (Figure 4).
Finally, platelet-derived exosomes reduced CD36-
atheroprotection the
(27,103,119,120,123),
therapeutic
approach
to
potential
transport
of
miRNAs
highmiRNA-
or
anti-
Chen et al. (124) developed miRNA-34a–containing
dependent oxidized low-density lipoprotein binding
liposome-polycationhyaluronic
and macrophage cholesterol loading, potentially
particle for systemic delivery of miRNA-34a into lung
acid
(LPH)
nano-
contributing to atheroprotection (122). Figure 2 illus-
metastasis of murine melanoma, resulting in signifi-
trates the known beneficial effects of EVs in the
cant down-regulation of surviving expression in the
regulation of vascular integrity. Table 1 summarizes
metastatic
the
studies
Furthermore, biodegradable polymer nanoparticles
exploring the effect of EVs on vascular health and
coated with cell-penetrating peptides for an effective
disease.
delivery
most
important
characteristics
of
of
tumors,
as
chemically
well
as
modified
reduced
tumor.
oligonucleotide
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
F I G U R E 3 Beneficial Effects of Extracellular Vesicles on Vascular Function
An increasing body of evidence points out the beneficial influence of certain EVs of diverse cellular sources in cardiovascular biology. This figure illustrates the favorable effects of EVs on endothelial and vascular function, atherosclerosis, and plaque stabilization. Endothelial EVs reduce endothelial apoptosis by inhibition of p38 activity mediated by an annexin I/phosphatidylserine receptor-dependent mechanism, contributing to endothelial protection. Moreover, endothelial EVs decrease endothelial regenerating cell apoptosis, facilitating endothelial repair. Platelet-derived MVs induce alterations in the endothelial-regenerating cell secretome toward a more proangiogenic profile and amplify vascular protection. Among the biological content transferred by EVs into target cells, miRNAs play a crucial role. Endothelial EVs promote vascular endothelial repair by inhibition of SPRED1 by delivering functional miRNA-126. Endothelial EVs promote anti-inflammatory effects by reducing endothelial ICAM-1 expression by the transfer of functional miRNA-222 into recipient cells. Exosomes from KLF-2transduced or shear-stress-stimulated endothelial cells attribute to atheroprotection by transferring miRNA-143/145. Circulating leukocyteand platelet-derived miRNA-223-containing exosomes penetrate the vascular wall, inhibit vascular smooth muscle cell proliferation and migration, resulting in decreased plaque size. Endothelial apoptotic bodies decrease vascular smooth muscle cell proliferation, limit plaque size, and increase plaque stability by miRNA-126-dependent inhibition of G-protein signaling (RGS16) pathway. MV ¼ microvesicle; other abbreviations as in Figures 1 and 2.
analogues have been described. This nanoparticle
therapeutic tool to combat cancer, it is reasonable
system was used to block the activity of the onco-
that nanoparticles can also be used to deliver miRNAs
genic miRNA-155, as well as to attenuate the expres-
to recipient vascular cells for tackling inflammation
sion of the proto-oncogene Mcl-1, leading to reduced
and development of atherosclerosis (129). In this
cell viability and pro-apoptotic effects in the recipient
context, magnetic nanoparticle-assisted (circumfer-
cells (127). Another approach targeting miRNA-155
ential) gene transfer into the vascular endothelium
described decelerated tumor growth after applica-
has recently been described as a promising novel
tion of polymer nanoparticles containing antisense
strategy to transfer biological messages into the
peptide nucleic acids with subsequent miRNA-155
diseased vasculature (130–132).
inhibition (125). An integrin a v b3-targeted nanoparticle was used by Anand et al. (128) to deliver anti-
TRANSLATION INTO CLINICAL USE. EVs have mul-
miRNA-132 to the tumor endothelium of human
tiple advantages over currently available drug de-
breast carcinoma in mice, causing restored p120Ras-
livery vehicles, such as their ability to overcome
GAP expression in the tumor endothelium, thereby
natural barriers, their intrinsic cell-targeting proper-
suppressing
ties,
angiogenesis
and
decreasing
tumor
protection
of
their
biological
cargo
from
burden. Although these studies focused on miRNA or
degrading enzymes, and stability in the circulation
anti-miRNA delivery using nanoparticles mainly as a
(133). EV subpopulations could be used as a cargo
799
800
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
F I G U R E 4 Potential Therapeutic Use of Extracellular Vesicles
Potential therapeutic application of extracellular vesicles includes the following 4 critical steps: 1) Extracellular vesicles can be modified by using tissue- or cell-type-specific ligands present on their surface. Endogenously expressed molecules such as miRNA and noncoding RNAs can be genetically engineered for therapeutic use (e.g., genetic modification by overexpression therapeutic nucleic acids). 2) Exogenous loading permits the collection of extracellular vesicles with desired cargo molecules. The collection and purification of extracellular vesicles can be carried out by various methods, including differential ultracentrifugation, ultrafiltration, sucrose gradient centrifugation, or immunoprecipitation. 3) Extracellular vesicles, loaded by any of these strategies, can be delivered into target cells or tissues with different delivery methods (e.g., intravenously injection or intracellular injection). 4) The loaded vesicles can function as favorable effectors in intercellular vascular signaling, contributing to the cardiovascular regeneration in damaged tissue.
system for efficient and selective drug delivery to a
direct EVs to target cells or tissue, cell-specific ligands
distinct cell type within diseased tissues. This
must be stably expressed on the surface of EVs. Using
approach offers the additional advantages of low
an innovative approach of donor cell engineering
immunogenicity because patient-derived tissue could
with target cell-specific ligand expression resulted in
be used as the source of individualized and biocom-
targeted delivery of short interfering RNA (siRNA)
patible drug delivery vehicles (134,135) (Figure 3).
and miRNA-loaded EVs to target neurons (138) and
Interestingly, tumor cells incubated with chemo-
breast cancer cells (139).
therapeutic drugs are able to package these drugs into
Despite promising perspectives for the treatment
EVs, which can be collected and used to effectively
of cardiovascular pathologies, EV-based therapies
kill tumor cells in murine tumor models without
still need more investigation to translate experi-
typical side effects (136). Moreover, tumor cell-
mental data into clinical application. One challenge
derived EVs were used as a unique carrier system to
would be to control the fragile equilibrium between
deliver oncolytic adenoviruses to human tumors,
the harmful and beneficial effects reported for EVs in
leading to highly efficient cytolysis of tumor cells.
the context of CVD. Furthermore, the off-side effects
These findings highlight a novel adenovirus delivery
and clearing mechanisms of EVs need to be better
system with promising clinical applications (137).
explored before they can be seriously considered as a
An important issue regarding EV therapeutics is
novel therapeutic tool for combatting CVD (140).
the biodistribution of EVs. Intravenously injected EVs are of particular interest in the treatment of cardio-
STUDY
vascular alterations, as the entire vascular network
OBSTACLES IN EV RESEARCH. Despite the emerging
LIMITATIONS
AND
METHODOLOGICAL
would be exposed to EVs. However, to selectively
role of EVs as regulators of health and disease, there
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
T A B L E 2 Overview of Commonly Used EV Isolation Methods
Method
UC
Principle of Separation
Size and density
Advantages
Disadvantages
Widely used
Ref. #
Relatively long procedure Low throughput Depends on viscosity of biological fluids
(143)
DG
Size and density
High purity of EVs
Time-consuming
(145)
Ultrafiltration
Size
Time efficient Effective to concentrate EVs
Low purity of EVs
(146)
Precipitation kits
PEG-mediated
High yield Rapid
Low purity of EVs
(142,147)
SEC
Size
Quick procedure Reproducibility
Low purity of EVs
(143,148)
Affinity capture
Binding with EVs surface components
Production of subpopulations of EVs Relatively high purity
High cost (antibody-based) May damage surface components of EVs
(143)
DG ¼ density gradient; PEG ¼ polyethylene glycol; SEC ¼ size-exclusion chromatography; UC ¼ ultracentrifugation; other abbreviations as in Table 1.
are still general limitations in EV research. Within this
mainly on PEG. Although the PEG-mediated tech-
paragraph, we highlight relevant obstacles, which
nique provides a high-yield, rapid, and inexpensive
need to be addressed to better understand EV func-
EV isolation method from both culture media and
tions and move the EV field forward.
body fluids, some other contaminants are also cop-
M e t h o d o l o g i c a l o b s t a c l e s i n i s o l a t i n g E V s . Iso-
urified, leading to the low purity of EVs (142,147). In
lation and purification of EVs vary between different
the clinical setting, size-exclusion chromatography
research groups and also depend on the donor cells
and affinity capture are the 2 methods most often
from which they are derived. Therefore, EV classifi-
applied to isolate EVs. Size-exclusion chromatog-
cation, isolation, and purification needs to be stan-
raphy can remove most soluble components and is a
dardized to ensure that EV analysis is reproducible
relatively quick procedure with good reproducibility
and internationally comparable among different
(148), but it also faces the possible problem of protein
research groups (141). Furthermore, within EV pop-
or RNA contamination (143). Compared with other
ulations, many distinct subtypes of vesicles exist.
methods,
However, the currently used methods (differential
populations of EVs with relatively high purity, but the
ultracentrifugation,
precipitation,
cost of preparation (antibody based) may limit its
density gradients, microfiltration, size-exclusion–
applicability and may damage surface proteins and
based approaches, or polyethylene glycol [PEG]-
functionality of EVs (143). Therefore, the develop-
mediated isolation techniques) all have different
ment of new, more selective isolation techniques is
pros and cons in attempting to isolate pure EVs with a
urgently needed to increase the purity of each vesicle
distinct size and surface markers (142). The most
subpopulation (149). The pros and cons of each
commonly used isolation methods are differential
available method are summarized in Table 2.
centrifugation, followed by ultracentrifugation. Two
Size measurement techniques for EV characterization. Size
polymer-based
affinity
capture
can
produce
sub-
major problems with these techniques are the rela-
is an important defining property of EVs, and mea-
tively long procedure times and low throughput,
surement of diameter to determine a size distribution
limiting their application in the clinical setting (143).
is a critical step for EV studies. Size distribution
Moreover, the yield depends on the viscosity of bio-
measurement technologies include electron micro-
logical fluids, so the samples with a relatively high
scopy
viscosity such as plasma would significantly reduce
tracking analysis, resistive pulse sensing, and atomic
production (144). Density gradient isolation can pro-
force microscopy (AFM). Although these techniques
mote both the yield and purity of EVs compared with
are commonly used in practice (142), some issues are
ultracentrifugation; however, the procedure is time-
still unsolved. First, the ideal size measurement
consuming and hard to standardize (145). Ultrafiltra-
should detect EVs with a diameter of 50 nm and larger
tion is time-efficient and can concentrate EVs up to
(150), but most methods, except for EM, cannot detect
240-fold, but the low purity of EVs is an obstacle
the smallest EVs (151). Second, the results of size
(146). Commercial EV precipitation kits are based
distribution, even for the same EV subpopulation,
(EM),
flow
cytometry
(FC),
nanoparticle
801
802
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017
Endothelial and Immune Cell EVs in CV Health and Disease
DECEMBER 2017:790–807
C E N T R A L IL L U ST R A T I O N Extracellular Vesicles as Regulators of Vascular Health and Disease
Jansen, F. et al. J Am Coll Cardiol Basic Trans Science. 2017;2(6):790–807.
Many types of cells release EVs, such as exosomes and microvesicles, by different mechanisms. EVs have both favorable and detrimental effects on vascular integrity. The use of genetically modified EVs might represent a novel therapeutic tool in the field of cardiovascular medicine and regenerative therapy. EV ¼ extracellular vesicles; miRNA ¼ microRNA.
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
may show different results depending on the method
such as exosomes can be incorporated into recipient
used (152). Furthermore, there are no standard pro-
cells (160).
tocols (e.g., optimal EM-EV measurement protocols)
Lack
(142).
as
i n v i v o . The secretion of EVs by parent cells and
Importantly,
some
nanoparticle-tracking
methods,
analysis
or
such
resistive
of
EV
secretion
and
uptake
models
pulse
uptake by recipient cells are precisely regulated.
sensing, cannot distinguish membrane vesicles from
However, few data are available studying concrete
nonmembranous particles of similar size, so the re-
mechanisms regulating MV release and clearance.
sults should be compared by using EM, AFM, or other
Moreover, physiological models are not well estab-
microscopy technique (153).
lished (e.g., considering time-dependent release ki-
R e g u l a t i o n o f E V p a c k a g i n g . Although some key
netics of EVs from parent cells). Finally, the absence
players in sorting of cargoes into EVs are revealed
of adequate in vivo models to explore the genera-
(e.g., sumoylated hnRNPA2B1 is reported to control
tion and uptake of cell-specific EVs limits the
the location of miRNAs into exosomes through bind-
experimental opportunities to study EV functions
ing to specific motifs [154]), the underlying mecha-
in vivo.
nisms mediating the packaging and loading of
Unclear processing of EVs and their content
selected molecules into EVs remain largely unknown.
a f t e r c e l l u l a r u p t a k e . EVs interact with recipient
Therefore, additional studies exploring EV biogenesis
cells
and mechanisms regulating EV packaging are impor-
through membrane fusion in a ligand-receptor-
tant to understand cardiovascular injury and repair
mediated way or by endocytosis, pinocytosis, or
induced by EVs (155).
phagocytosis (12) (Figure 1). However, processing of
by
transferring
their
biological
contents
experiments
incorporated EVs and their intravesicular content
in vitro. EV uptake experiments are usually performed
after cellular uptake is entirely unknown. To address
Methodological
issues
on
EV
uptake
by direct visualization. Therefore, fluorescent lipid
this point, tracking experiments using fluorescence
membrane dyes, such as PKH26 (156), PKH68 (157),
labeleling represent a possible option to gain further
or rhodamine B (158), are used to stain EV mem-
insight
branes. One potential issue with membrane-binding
mechanisms of biological contents transferred by
dyes is that fluorescent molecules could potentially
EVs (161). Nevertheless, stable labeling of intra-
affect the uptake and biological behavior of EVs.
vesicular contents is highly demanding technically,
However, EV incorporation has been observed with
and further efforts are needed to improve these
many different lipid-binding dyes, suggesting that
techniques (162).
into
time-dependent
cellular
processing
such molecules do not affect internalization of vesi-
Taken together, there are still serious methodo-
cles; nevertheless, additional studies are needed to
logical issues that need to be addressed. Importantly,
verify whether the biological behavior of EVs is
recent position research papers from the Interna-
affected by dyes. Another potential limitation of the
tional Society of Extracellular Vesicles have made a
use of lipophilic dyes is leaching of the fluorescent
first step in the right direction to standardize EV
molecules
membranes,
analysis internationally among different laboratories
potentially leading to a pattern of internalization
(141,147,153). In addition, EV-TRACK (163), a novel
that is due to normal membrane recycling rather
crowd-sourcing database, has recently been imple-
than EV uptake. However, direct measurement of the
mented, centralizing knowledge about EV biology
fluorescence transfer rate between EVs and recipient
and methodology with the goal of stimulating
cells support the idea that the increased fluorescence
authors, reviewers, editors, and funders to put
in cells is due to specific uptake of EVs rather than
experimental guidelines into practice (164).
from
EVs
into
cellular
nonspecific dye leaching (159). Another issue which must be considered is the fact that most EV uptake
CONCLUSIONS
studies have relied on fluorescence microscopy, which has limited resolution because the wavelength
In the regulation of cardiovascular health and dis-
of visible light is approximately 390 to 700 nm;
ease, EVs act as urgent effectors by transferring
therefore, single EVs or aggregated vesicles, which
bioactive molecules into adjacent and distant re-
are <390 nm in diameter, cannot be distinguished.
cipients. EV-mediated intercellular vascular signaling
This should not affect the assessment of EV uptake
results in detrimental and favorable effects on
in general but may affect the visualization and dy-
vascular integrity. Studies illustrate that EVs can
namic
EVs.
contribute to atherosclerosis development and pro-
Nevertheless, the increasing use of confocal micro-
gression. In contrast, EVs also emerge as crucial
scopy has confirmed that EVs smaller than 390 nm
regulators of vascular homeostasis and mediate
localization
analysis
of
individual
803
804
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
vascular protection. Finally, the use of genetically
53105
modified EV might represent a novel therapeutic tool
ukbonn.de. OR Dr. Nikos Werner, Medizinische Kli-
in the field of cardiovascular medicine and regener-
nik und Poliklinik II, Universitätsklinikum Bonn,
ative therapy (Central Illustration).
Sigmund Freud Strasse 25, 53105 Bonn, Germany.
Bonn,
Germany.
E-mail:
[email protected]
E-mail:
[email protected] OR Dr. Qian Li, ADDRESS FOR CORRESPONDENCE: Dr. Felix Jansen,
Department of Cardiology, Second Hospital of Jilin
Medizinische
University, 218 Ziqiang Street, Nanguan District,
Klinik
und
Poliklinik
II,
Uni-
versitätsklinikum Bonn, Sigmund Freud Strasse 25,
Changchun 130000, China.
REFERENCES 1. Libby P. Changing concepts of atherogenesis. J Intern Med 2000;247:349–58. 2. Dignat-George F, Boulanger CM. The many faces of endothelial microparticles. Arterioscler Thromb Vasc Biol 2011;31:27–33. 3. Koga H, Sugiyama S, Kugiyama K, et al. Elevated levels of VE-cadherin-positive endothelial micro-
16. Nabhan JF, Hu R, Oh RS, Cohen SN, Lu Q. Formation and release of arrestin domaincontaining protein 1-mediated microvesicles (ARMMs) at plasma membrane by recruitment of
28. Skog J, Würdinger T, van Rijn S, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol 2008;10:
TSG101 protein. Proc Natl Acad Sci U S A 2012; 109:4146–51.
1470–6.
particles in patients with type 2 diabetes mellitus and coronary artery disease. J Am Coll Cardiol 2005;45:1622–30.
17. Jaé N, McEwan DG, Manavski Y, Boon RA, Dimmeler S. Rab7a and Rab27b control secretion of endothelial microRNA through extracellular vesicles. FEBS Lett 2015;589:3182–8.
4. Jansen F, Nickenig G, Werner N. Extracellular vesicles in cardiovascular disease: potential applications in diagnosis, prognosis, and epidemiology. Circ Res 2017;120:1649–57.
18. Bobrie A, Colombo M, Raposo G, Théry C. Exosome secretion: molecular mechanisms and roles in immune responses. Traffic 2011;12: 1659–68.
5. Mause SF, Weber C. Microparticles: protagonists of a novel communication network for intercellular information exchange. Circ Res 2010;107:
19. Ostrowski M, Carmo NB, Krumeich S, et al. Rab27a and Rab27b control different steps of the
1047–57. 6. Kogure T, Yan IK, Lin W-L, Patel T. Extracellular vesicle-mediated transfer of a novel long noncoding RNA TUC339: a mechanism of intercellular signaling in human hepatocellular cancer. Genes Cancer 2013;4:261–72.
exosome secretion pathway. Nat Cell Biol 2010;12 Suppl 1–13:19–30. 20. Muralidharan-Chari V, Clancy J, Plou C, et al. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr Biol 2009; 19:1875–85.
29. Wang J-G, Williams JC, Davis BK, et al. Monocytic microparticles activate endothelial cells in an IL-1b-dependent manner. Blood 2011;118: 2366–74. 30. Yáñez-Mó M, Siljander PR-M, Andreu Z, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles 2015;4:27066. 31. Ray DM, Spinelli SL, Pollock SJ, et al. Peroxisome proliferator-activated receptor gamma and retinoid X receptor transcription factors are released from activated human platelets and shed in microparticles. Thromb Haemost 2008;99: 86–95. 32. Brill A, Dashevsky O, Rivo J, Gozal Y, Varon D. Platelet-derived microparticles induce angiogenesis and stimulate post-ischemic revascularization. Cardiovasc Res 2005;67:30–8.
7. Rautou P-E, Vion A-C, Amabile N, et al. Microparticles, vascular function, and atherothrombosis. Circ Res 2011;109:593–606.
21. Lo S-C, Hung C-Y, Lin D-T, Peng H-C, Huang T-F. Involvement of platelet glycoprotein Ib in platelet microparticle mediated neutrophil activation. J Biomed Sci 2006;13:787–96.
8. Boon RA, Vickers KC. Intercellular transport of microRNAs. Arterioscler Thromb Vasc Biol 2013;
22. Dasgupta SK, Le A, Chavakis T, Rumbaut RE, Thiagarajan P. Developmental endothelial locus-1
33:186–92.
(Del-1) mediates clearance of platelet microparticles by the endothelium. Circulation 2012;125: 1664–72.
34. Owens AP, Mackman N. Microparticles in hemostasis and thrombosis. Circ Res 2011;108: 1284–97.
23. Dasgupta SK, Abdel-Monem H, Niravath P, et al. Lactadherin and clearance of plateletderived microvesicles. Blood 2009;113:1332–9.
35. Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membranederived microvesicles: important and underap-
9. Boulanger CM, Loyer X, Rautou P-E, Amabile N. Extracellular vesicles in coronary artery disease. Nat Rev Cardiol 2017;14:259–72. 10. Barile L, Moccetti T, Marbán E, Vassalli G. Roles of exosomes in cardioprotection. Eur Heart J 2017;38:1372–9. 11. Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol 2014;30:255–89. 12. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013;200:373–83. 13. Raiborg C, Stenmark H. The ESCRT machinery in endosomal sorting of ubiquitylated membrane proteins. Nature 2009;458:445–52. 14. Trajkovic K, Hsu C, Chiantia S, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science 2008;319:1244–7. 15. Baietti MF, Zhang Z, Mortier E, et al. Syndecansyntenin-ALIX regulates the biogenesis of exosomes. Nat Cell Biol 2012;14:677–85.
24. Rautou P-E, Leroyer AS, Ramkhelawon B, et al. Microparticles from human atherosclerotic plaques promote endothelial ICAM-1-dependent monocyte adhesion and transendothelial migration. Circ Res 2011;108:335–43. 25. Mulcahy LA, Pink RC, Carter DRF. Routes and mechanisms of extracellular vesicle J Extracell Vesicles 2014;3:24641.
uptake.
26. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007;9:654–9. 27. Zernecke A, Bidzhekov K, Noels H, et al. Delivery of MicroRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal 2009;2:ra81–ra81.
33. Jansen F, Yang X, Franklin BS, et al. High glucose condition increases NADPH oxidase activity in endothelial microparticles that promote vascular inflammation. Cardiovasc Res 2013;98: 94–106.
preciated mediators of cell-to-cell communication. Leukemia 2006;20:1487–95. 36. Gupta SK, Bang C, Thum T. Circulating microRNAs as biomarkers and potential paracrine mediators of cardiovascular disease. Circ Cardiovasc Genet 2010;3:484–8. 37. Bang C, Thum T. Exosomes: new players in cell-cell communication. Int J Biochem Cell Biol 2012;44:2060–4. 38. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation 2002;105: 1135–43. 39. Libby P, Ridker PM, Hansson GK, Ducluzeau PH. Transatlantic network on atherothrombosis. inflammation in atherosclerosis: from pathophysiology to practice. J Am Coll Cardiol 2009;54:2129–38.
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
40. Densmore JC, Signorino PR, Ou J, et al. Endothelium-derived microparticles induce endothelial dysfunction and acute lung injury. Shock 2006;26:464–71. 41. Brodsky SV, Zhang F, Nasjletti A, Goligorsky MS. Endothelium-derived microparticles impair endothelial function in vitro. Am J Physiol Heart Circ Physiol 2004;286:H1910–5. 42. Boulanger CM, Scoazec A, Ebrahimian T, et al. Circulating microparticles from patients with myocardial infarction cause endothelial dysfunction. Circulation 2001;104:2649–52. 43. Amabile N, Guérin AP, Leroyer A, et al. Circulating endothelial microparticles are associated with vascular dysfunction in patients with endstage renal failure. J Am Soc Nephrol 2005;16: 3381–8. 44. Agouni A, Lagrue-Lak-Hal AH, Ducluzeau PH, et al. Endothelial dysfunction caused by circulating microparticles from patients with metabolic syndrome. Am J Pathol 2008;173:1210–9. 45. Donadee C, Raat NJH, Kanias T, et al. Nitric oxide scavenging by red blood cell microparticles and cell-free hemoglobin as a mechanism for the red cell storage lesion. Circulation 2011;124: 465–76. 46. Gambim MH, do Carmo A de O, Marti L, Veríssimo-Filho S, Lopes LR, Janiszewski M. Plateletderived exosomes induce endothelial cell apoptosis through peroxynitrite generation: experimental evidence for a novel mechanism of septic vascular dysfunction. Crit Care 2007;11: R107. 47. Willerson JT, Ridker PM. Inflammation as a cardiovascular risk factor. Circulation 2004;109: II2–10. 48. Yamamoto S, Niida S, Azuma E, et al. Inflammation-induced endothelial cell-derived extracellular vesicles modulate the cellular status of pericytes. Sci Rep 2015;5:8505. 49. Buzás EI, György B, Nagy G, Falus A, Gay S. Emerging role of extracellular vesicles in inflammatory diseases. Nat Rev Rheumatol 2014;10: 356–64. 50. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat Rev Immunol 2014;14:195–208. 51. Robbins PD, Dorronsoro A, Booker CN. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J Clin Invest 2016; 126:1173–80. 52. Mesri M, Altieri DC. Endothelial cell activation by leukocyte microparticles. J Immunol 1998;161: 4382–7. 53. Mesri M, Altieri DC. Leukocyte microparticles stimulate endothelial cell cytokine release and tissue factor induction in a JNK1 signaling pathway. J Biol Chem 1999;274:23111–8. 54. Nomura S, Tandon NN, Nakamura T, Cone J, Fukuhara S, Kambayashi J. High-shear-stressinduced activation of platelets and microparticles enhances expression of cell adhesion molecules in THP-1 and endothelial cells. Atherosclerosis 2001; 158:277–87. 55. Barry OP, Praticò D, Savani RC, FitzGerald GA. Modulation of monocyte-endothelial cell
Endothelial and Immune Cell EVs in CV Health and Disease
interactions by platelet microparticles. J Clin Invest 1998;102:136–44. 56. Huber J, Vales A, Mitulovic G, et al. Oxidized membrane vesicles and blebs from apoptotic cells contain biologically active oxidized phospholipids that induce monocyte-endothelial interactions. Arterioscler Thromb Vasc Biol 2002;22:101–7. 57. Edrissi H, Schock SC, Hakim AM, Thompson CS. Microparticles generated during chronic cerebral ischemia increase the permeability of microvascular endothelial barriers in vitro. Brain Res 2016; 1634:83–93. 58. Mause SF, Hundelshausen von P, Zernecke A, Koenen RR, Weber C. Platelet microparticles: a transcellular delivery system for RANTES promoting monocyte recruitment on endothelium. Arterioscler Thromb Vasc Biol 2005;25:1512–8. 59. Vasina EM, Cauwenberghs S, Feijge MAH, Heemskerk JWM, Weber C, Koenen RR. Microparticles from apoptotic platelets promote resident macrophage differentiation. Cell Death Dis 2011;2: e211. 60. de Jong OG, Verhaar MC, Chen Y, et al. Cellular stress conditions are reflected in the protein and RNA content of endothelial cellderived exosomes. J Extracell Vesicles 2012;1. 61. Alexandru N, Badila E, Weiss E, Cochior D, E, Georgescu A. Vascular complications in Ste ˛pien diabetes: microparticles and microparticle associated microRNAs as active players. Biochem Biophys Res Commun 2016;472:1–10. 62. Wang Y, Chen L-M, Liu M-L. Microvesicles and diabetic complications–novel mediators, potential biomarkers and therapeutic targets. Acta Pharmacol Sin 2014;35:433–43. 63. Exosomes: emerging roles in communication between blood cells and vascular tissues during atherosclerosis. Curr Opin Lipidol 2015;26:412–9. 64. Liu M-L, Williams KJ. Microvesicles: potential markers and mediators of endothelial dysfunction. Curr Opin Endocrinol Diabetes Obes 2012;19:121–7. 65. Tang N, Sun B, Gupta A, Rempel H, Pulliam L. Monocyte exosomes induce adhesion molecules and cytokines via activation of NF-kB in endothelial cells. FASEB J 2016;30:3097–106. 66. Aharon A, Tamari T, Brenner B. Monocytederived microparticles and exosomes induce procoagulant and apoptotic effects on endothelial cells. Thromb Haemost 2008;100:878–85. 67. Leroyer AS, Isobe H, Leseche G, et al. Cellular origins and thrombogenic activity of microparticles isolated from human atherosclerotic plaques. J Am Coll Cardiol 2007;49:772–7. 68. Mayr M, Grainger D, Mayr U, et al. Proteomics, metabolomics, and immunomics on microparticles derived from human atherosclerotic plaques. Circ Cardiovasc Genet 2009;2:379–88. 69. Théry C, Duban L, Segura E, Véron P, Lantz O, Amigorena S. Indirect activation of naïve CD4þ T cells by dendritic cell-derived exosomes. Nat Immunol 2002;3:1156–62. 70. Canault M, Leroyer AS, Peiretti F, et al. Microparticles of human atherosclerotic plaques enhance the shedding of the tumor necrosis factor-alpha converting enzyme/ADAM17
substrates, tumor necrosis factor and tumor necrosis factor receptor-1. Am J Pathol 2007;171: 1713–23. 71. Liu M-L, Scalia R, Mehta JL, Williams KJ. Cholesterol-induced membrane microvesicles as novel carriers of damage-associated molecular patterns: mechanisms of formation, action, and detoxification. Arterioscler Thromb Vasc Biol 2012; 32:2113–21. 72. Hoyer FF, Giesen MK, Franca C, Lütjohann D, Nickenig G, Werner N. Monocytic microparticles promote atherogenesis by modulating inflammatory cells in mice. J Cell Mol Med 2012;16:2777–88. 73. Zakharova L, Svetlova M, Fomina AF. T cell exosomes induce cholesterol accumulation in human monocytes via phosphatidylserine receptor. J Cell Physiol 2007;212:174–81. 74. Yahagi K, Kolodgie FD, Otsuka F, et al. Pathophysiology of native coronary, vein graft, and instent atherosclerosis. Nat Rev Cardiol 2016;13: 79–98. 75. Weber A, Köppen HO, Schrör K. Plateletderived microparticles stimulate coronary artery smooth muscle cell mitogenesis by a PDGFindependent mechanism. Thromb Res 2000;98: 461–6. 76. Pakala R. Serotonin and thromboxane A2 stimulate platelet-derived microparticle-induced smooth muscle cell proliferation. Cardiovasc Radiat Med 2004;5:20–6. 77. Sarkar A, Mitra S, Mehta S, Raices R, Wewers MD. Monocyte derived microvesicles deliver a cell death message via encapsulated caspase-1. PLoS One 2009;4:e7140. 78. Krohn JB, Hutcheson JD, Martínez-Martínez E, Aikawa E. Extracellular vesicles in cardiovascular calcification: expanding current paradigms. J Physiol (Lond) 2016;594:2895–903. 79. Goettsch C, Hutcheson JD, Aikawa M, et al. Sortilin mediates vascular calcification via its recruitment into extracellular vesicles. J Clin Invest 2016;126:1323–36. 80. Hutcheson JD, Goettsch C, Bertazzo S, et al. Genesis and growth of extracellular-vesiclederived microcalcification in atherosclerotic plaques. Nat Mater 2016;15:335–43. 81. Virmani R, Kolodgie FD, Burke AP, et al. Atherosclerotic plaque progression and vulnerability to rupture: angiogenesis as a source of intraplaque hemorrhage. Arterioscler Thromb Vasc Biol 2005;25:2054–61. 82. Leroyer AS, Rautou P-E, Silvestre J-S, et al. CD40 ligandþ microparticles from human atherosclerotic plaques stimulate endothelial proliferation and angiogenesis a potential mechanism for intraplaque neovascularization. J Am Coll Cardiol 2008;52:1302–11. 83. Hoyer FF, Nickenig G, Werner N. Microparticles—messengers of biological information. J Cell Mol Med 2010;14:2250–6. 84. Mallat Z, Hugel B, Ohan J, Lesèche G, Freyssinet JM, Tedgui A. Shed membrane microparticles with procoagulant potential in human atherosclerotic plaques: a role for apoptosis in plaque thrombogenicity. Circulation 1999;99: 348–53.
805
806
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
Endothelial and Immune Cell EVs in CV Health and Disease
85. Geddings JE, Mackman N. Tumor-derived tissue factor-positive microparticles and venous thrombosis in cancer patients. Blood 2013;122: 1873–80.
100. Andaloussi El S, Mäger I, Breakefield XO, Wood MJA. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov 2013;12:347–57.
86. Zwicker JI, Trenor CC, Furie BC, Furie B. Tissue factor-bearing microparticles and thrombus formation. Arterioscler Thromb Vasc Biol 2011;31: 728–33.
101. Eldh M, Ekström K, Valadi H, et al. Exosomes communicate protective messages during oxidative stress; possible role of exosomal shuttle RNA. PLoS One 2010;5:e15353.
87. Zhao L, Bi Y, Kou J, Shi J, Piao D. Phosphatidylserine exposing-platelets and microparticles
102. Diehl P, Fricke A, Sander L, et al. Microparticles: major transport vehicles for distinct micro-
promote procoagulant activity in colon cancer patients. J Exp Clin Cancer Res 2016;35:54.
RNAs in circulation. Cardiovasc Res 2012;93: 633–44.
88. Chou J, Mackman N, Merrill-Skoloff G, Pedersen B, Furie BC, Furie B. Hematopoietic cellderived microparticle tissue factor contributes to fibrin formation during thrombus propagation.
103. Jansen F, Yang X, Hoelscher M, et al. Endothelial microparticle-mediated transfer of MicroRNA-126 promotes vascular endothelial cell repair via SPRED1 and is abrogated in glucose-
Blood 2004;104:3190–7.
damaged endothelial microparticles. Circulation 2013;128:2026–38.
89. Biró E, Sturk-Maquelin KN, Vogel GMT, et al. Human cell-derived microparticles promote thrombus formation in vivo in a tissue factordependent manner. J Thromb Haemost 2003;1: 2561–8. 90. Libby P, Sukhova G, Lee RT, Liao JK. Molecular biology of atherosclerosis. Int J Cardiol 1997;62 Suppl 2:S23–9. 91. King SB, Marshall JJ, Tummala PE. Revascularization for coronary artery disease: stents versus bypass surgery. Annu Rev Med 2010;61: 199–213. 92. Abid Hussein MN, Nieuwland R, Hau CM, Evers LM, Meesters EW, Sturk A. Cell-derived microparticles contain caspase 3 in vitro and in vivo. J Thromb Haemost 2005;3:888–96. 93. Diamant M, Tushuizen ME, Abid Hussein MN, et al. Simvastatin-induced endothelial cell detachment and microparticle release are prenylation dependent. Thromb Haemost 2008;100: 489–97. 94. Tramontano AF, O’Leary J, Black AD, Muniyappa R, Cutaia MV, El-Sherif N. Statin de-
104. Jansen F, Stumpf T, Proebsting S, et al. Intercellular transfer of miR-126-3p by endothelial microparticles reduces vascular smooth muscle cell proliferation and limits neointima formation by inhibiting LRP6. J Mol Cell Cardiol 2017;104: 43–52. 105. Deregibus MC, Cantaluppi V, Calogero R, et al. Endothelial progenitor cell derived microvesicles activate an angiogenic program in endothelial cells by a horizontal transfer of mRNA. Blood 2007;110:2440–8. 106. Leroyer AS, Ebrahimian TG, Cochain C, et al. Microparticles from ischemic muscle promotes postnatal vasculogenesis. Circulation 2009;119: 2808–17. 107. Lai RC, Arslan F, Lee MM, et al. Exosome secreted by MSC reduces myocardial ischemia/ reperfusion injury. Stem Cell Res 2010;4:214–22. 108. Vrijsen KR, Sluijter JPG, Schuchardt MWL, et al. Cardiomyocyte progenitor cell-derived exosomes stimulate migration of endothelial cells. J Cell Mol Med 2010;14:1064–70.
115. Mokarizadeh A, Delirezh N, Morshedi A, Mosayebi G, Farshid A-A, Mardani K. Microvesicles derived from mesenchymal stem cells: potent organelles for induction of tolerogenic signaling. Immunol Lett 2012;147:47–54. 116. Arslan F, Lai RC, Smeets MB, et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/ Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res 2013; 10:301–12. 117. Zou X, Zhang G, Cheng Z, et al. Microvesicles derived from human Wharton’s jelly mesenchymal stromal cells ameliorate renal ischemiareperfusion injury in rats by suppressing CX3CL1. Stem Cell Res Ther 2014;5:40. 118. Jansen F, Yang X, Baumann K, et al. Endothelial microparticles reduce ICAM-1 expression in a microRNA-222-dependent mechanism. J Cell Mol Med 2015;19:2202–14. 119. Njock M-S, Cheng HS, Dang LT, et al. Endothelial cells suppress monocyte activation through secretion of extracellular vesicles containing antiinflammatory microRNAs. Blood 2015;125: 3202–12. 120. Hergenreider E, Heydt S, Tréguer K, et al. Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nat Cell Biol 2012;14:249–56. 121. Shan Z, Qin S, Li W, et al. An Endocrine genetic signal between blood cells and vascular smooth muscle cells: role of microRNA-223 in smooth muscle function and atherogenesis. J Am Coll Cardiol 2015;65:2526–37. 122. Srikanthan S, Li W, Silverstein RL, McIntyre TM. Exosome poly-ubiquitin inhibits platelet activation, downregulates CD36 and inhibits pro-atherothombotic cellular functions. J Thromb Haemost 2014;12:1906–17. 123. Zhang Y, Liu D, Chen X, et al. Secreted monocytic miR-150 enhances targeted endothelial cell migration. Mol Cell 2010;39:133–44.
creases endothelial microparticle release from human coronary artery endothelial cells: implication for the rho-kinase pathway. Biochem Biophys Res Commun 2004;320:34–8.
109. Mackie AR, Klyachko E, Thorne T, et al. Sonic hedgehog-modified human CD34þ cells preserve cardiac function after acute myocardial infarction. Circ Res 2012;111:312–21.
95. Nomura S. Statin and endothelial cell-derived microparticles. Thromb Haemost 2008;100:
110. Khan M, Nickoloff E, Abramova T, et al. Embryonic stem cell-derived exosomes promote
deliver siRNA and miRNA for cancer therapy. Mol Ther 2010;18:1650–6.
377–8.
endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ Res 2015;117:52–64.
125. Babar IA, Cheng CJ, Booth CJ, et al. Nanoparticle-based therapy in an in vivo microRNA-155 (miR-155)-dependent mouse model of lymphoma. Proc Natl Acad Sci U S A 2012;109:E1695–704.
96. Jansen F, Yang X, Hoyer FF, et al. Endothelial microparticle uptake in target cells is annexin I/ phosphatidylserine receptor dependent and prevents apoptosis. Arterioscler Thromb Vasc Biol 2012;32:1925–35.
111. Sahoo S, Losordo DW. Exosomes and cardiac repair after myocardial infarction. Circ Res 2014; 114:333–44.
97. Mause SF, Ritzel E, Liehn EA, et al. Platelet microparticles enhance the vasoregenerative potential of angiogenic early outgrowth cells after vascular injury. Circulation 2010;122:495–506.
112. Gasser O, Schifferli JA. Activated polymorphonuclear neutrophils disseminate antiinflammatory microparticles by ectocytosis. Blood 2004;104:2543–8.
98. França CN, Izar MC de O, Amaral JBD, Tegani DM, Fonseca FAH. Microparticles as potential biomarkers of cardiovascular disease. Arq
113. Dalli J, Norling LV, Renshaw D, Cooper D, Leung K-Y, Perretti M. Annexin 1 mediates the rapid anti-inflammatory effects of neutrophil-
Bras Cardiol 2015;104:169–74.
derived microparticles. Blood 2008;112:2512–9.
99. Pérez-Casal M, Downey C, Cutillas-Moreno B, Zuzel M, Fukudome K, Toh CH. Microparticleassociated endothelial protein C receptor and the induction of cytoprotective and anti-inflammatory effects. Haematologica 2009;94:387–94.
114. Köppler B, Cohen C, Schlöndorff D, Mack M. Differential mechanisms of microparticle transfer toB cells and monocytes: anti-inflammatory propertiesof microparticles. Eur J Immunol 2006; 36:648–60.
124. Chen Y, Zhu X, Zhang X, Liu B, Huang L. Nanoparticles modified with tumor-targeting scFv
126. Muthiah M, Park I-K, Cho C-S. Nanoparticlemediated delivery of therapeutic genes: focus on miRNA therapeutics. Exp Opin Drug Deliv 2013;10: 1259–73. 127. Cheng
CJ,
Saltzman
WM.
Polymer
nanoparticle-mediated delivery of microRNA inhibition and alternative splicing. Mol Pharm 2012;9: 1481–8. 128. Anand S, Majeti BK, Acevedo LM, et al. MicroRNA-132-mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nat Med 2010;16:909–14. 129. Nouraee N, Mowla SJ. miRNA therapeutics in cardiovascular diseases: promises and problems. Front Genet 2015;6:232.
Jansen et al.
JACC: BASIC TO TRANSLATIONAL SCIENCE VOL. 2, NO. 6, 2017 DECEMBER 2017:790–807
130. Vosen S, Rieck S, Heidsieck A, et al. Improvement of vascular function by magnetic nanoparticle-assisted circumferential gene transfer into the native endothelium. J Control Release 2016;241:164–73. 131. Vosen S, Rieck S, Heidsieck A, et al. Vascular repair by circumferential cell therapy using magnetic nanoparticles and tailored magnets. ACS Nano 2016;10:369–76. 132. Hofmann A, Wenzel D, Becher UM, et al. Combined targeting of lentiviral vectors and positioning of transduced cells by magnetic nanoparticles. Proc Natl Acad Sci U S A 2009;106: 44–9.
Endothelial and Immune Cell EVs in CV Health and Disease
143. Stremersch S, De Smedt SC, Raemdonck K. Therapeutic and diagnostic applications of extracellular vesicles. J Control Release 2016;244: 167–83. 144. Momen-Heravi F, Balaj L, Alian S, et al. Impact of biofluid viscosity on size and sedimentation efficiency of the isolated microvesicles. Front Physiol 2012;3:162. 145. Kalra H, Adda CG, Liem M, et al. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics 2013;13:3354–64.
133. Vader P, Mol EA, Pasterkamp G, Schiffelers RM. Extracellular vesicles for drug de-
146. Lobb RJ, Becker M, Wen SW, et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles
livery. Adv Drug Deliv Rev 2016;106:148–56.
2015;4:27031.
134. Lakhal S, Wood MJA. Exosome nanotechnology: an emerging paradigm shift in drug delivery: exploitation of exosome nanovesicles for systemic in vivo delivery of RNAi heralds new horizons for drug delivery across biological barriers. Bioessays 2011;33:737–41.
147. Mateescu B, Kowal EJK, van Balkom BWM,
135. Andaloussi El S, Lakhal S, Mäger I, Wood MJA. Exosomes for targeted siRNA delivery across biological barriers. Adv Drug Deliv Rev 2013;65: 391–7. 136. Tang K, Zhang Y, Zhang H, et al. Delivery of chemotherapeutic drugs in tumour cell-derived microparticles. Nat Commun 2012;3:1282.
et al. Obstacles and opportunities in the functional analysis of extracellular vesicle RNA—an ISEV position paper. J Extracell Vesicles 2017;6: 1286095. 148. Böing AN, van der Pol E, Grootemaat AE, Coumans FAW, Sturk A, Nieuwland R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J Extracell Vesicles 2014;3: 23430. 149. Ridger VC, Boulanger CM, AngelilloScherrer A, et al. Microvesicles in vascular
Extracellular Vesicles. J Extracell Vesicles 2014;3: 26913. 154. Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez-Cabo F, et al. Sumoylated hnRNPA2B1 controls the sorting of miRNAs into exosomes through binding to specific motifs. Nat Commun 2013;4:2980. 155. Todorova D, Simoncini S, Lacroix R, Sabatier F, Dignat-George F. Extracellular vesicles in angiogenesis. Circ Res 2017;120:1658–73. 156. Atay S, Gercel-Taylor C, Taylor DD. Human trophoblast-derived exosomal fibronectin induces pro-inflammatory Il-1b production by macrophages. Am J Reprod Immunol 2011;66: 259–69. 157. Fitzner D, Schnaars M, van Rossum D, et al. Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J Cell Sci 2011;124:447–58. 158. Tian T, Zhu Y-L, Hu F-H, Wang Y-Y, Huang N-P, Xiao Z-D. Dynamics of exosome internalization and trafficking. J Cell Physiol 2013;228:1487–95. 159. Christianson HC, Svensson KJ, van Kuppevelt TH, Li J-P, Belting M. Cancer cell exosomes depend on cell-surface heparan sulfate proteoglycans for their internalization and functional activity. Proc Natl Acad Sci U S A 2013;110: 17380–5.
homeostasis and diseases. Position paper of the European Society of Cardiology (ESC) Working Group on Atherosclerosis and Vascular Biology. Thromb Haemost 2017;117:1296–316.
160. Bang C, Batkai S, Dangwal S, et al. Cardiac fibroblast-derived microRNA passenger strandenriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest 2014;124:2136–46.
150. Brisson AR, Tan S, Linares R, Gounou C, Arraud N. Extracellular vesicles from activated platelets: a semiquantitative cryo-electron microscopy and immuno-gold labeling study. Platelets
161. Qu L, Ding J, Chen C, et al. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous
2017;28:263–71.
162. Tkach M, Théry C. Communication by extra-
139. Ohno S-I, Takanashi M, Sudo K, et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol Ther 2013;21:185–91.
151. Coumans FAW, van der Pol E, Böing AN, et al.
cellular vesicles: where we are and where we need to go. Cell 2016;164:1226–32.
140. Yin M, Loyer X, Boulanger CM. Extracellular vesicles as new pharmacological targets to treat atherosclerosis. Eur J Pharmacol 2015;763:90–103.
152. van der Pol E, Coumans FAW, Grootemaat AE, et al. Particle size distribution of exosomes and microvesicles determined by transmission electron microscopy, flow cytometry, nanoparticle tracking analysis, and resistive pulse sensing. J Thromb
137. Ran L, Tan X, Li Y, et al. Delivery of oncolytic adenovirus into the nucleus of tumorigenic cells by tumor microparticles for virotherapy. Biomaterials 2016;89:56–66. 138. Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJA. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol 2011;29:341–5.
141. Witwer KW, Buzás EI, Bemis LT, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles 2013;2. 142. Coumans FAW, Brisson AR, Buzás EI, et al. Methodological Guidelines to Study Extracellular Vesicles. Circ Res 2017;120:1632–48.
Reproducible extracellular vesicle size and concentration determination with tunable resistive pulse sensing. J Extracell Vesicles 2014;3:25922.
Haemost 2014;12:1182–92.
RNA. Cancer Cell 2016;29:653–68.
163. Van Deun J. EV-TRACK: transparent reporting and centralizing knowledge in extracellular vesicle research. Nat Methods 2017;14:228–32. 164. EV-TRACK Consortium, Van Deun J, Mestdagh P, et al. EV-TRACK: transparent reporting and centralizing knowledge in extracellular vesicle research. Nat Methods 2017;14: 228–32.
153. Lötvall J, Hill AF, Hochberg F, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for
KEY WORDS cardiovascular disease, extracellular vesicles, microvesicles
807