scispace - formally typeset
Open AccessJournal ArticleDOI

Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis

Reads0
Chats0
TLDR
The results suggest that exosomal transfer of pro-angiogenic miRs plays an important role in MSC mediated angiogenesis and stem cell-to-endothelial cell communication.
Abstract
Mesenchymal stem cells (MSCs) have been found to benefit patients with a variety of ischemic diseases via promoting angiogenesis. It is also well established that exosomes secreted from MSCs deliver bioactive molecules, including microRNAs (miRs) to recipient cells. Therefore, we hypothesized that exosomes secreted from MSCs deliver miRs into endothelial cells and mediate angiogenesis. The pro-angiogenic stimulatory capacity of exosomes was investigated using tube-like structure formation and spheroid-based sprouting of human umbilical vein endothelial cells (HUVECs), and in vivo Matrigel plug assay. The secretion of pro-angiogenic miRs (pro-angiomiRs) from MSCs into culture medium and transfer of the miRs to HUVECs were confirmed using real-time quantitative PCR. Supplementation of the exosome secretion blocker GW4869 (10 μM) reduced the pro-angiomiRs in the MSC-derived conditioned medium (CdMMSC). Addition of exosomes isolated from CdMMSC could directly 1) promote HUVEC tube-like structure formation in vitro; 2) mobilize endothelial cells into Matrigel plug subcutaneously transplanted into mice; and 3) increase blood flow inside Matrigel plug. Fluorescence tracking showed that the exosomes were internalized rapidly by HUVECs causing an upregulated expression of pro-angiomiRs in HUVECs. Loss-and-gain function of the pro-angiomiRs (e.g., miR-30b) in MSCs significantly altered the pro-angiogenic properties of these MSC-derived exosomes, which could be associated with the regulation of their targets in HUVECs. These results suggest that exosomal transfer of pro-angiogenic miRs plays an important role in MSC mediated angiogenesis and stem cell-to-endothelial cell communication.

read more

Content maybe subject to copyright    Report

Oncotarget45200
www.impactjournals.com/oncotarget
www.impactjournals.com/oncotarget/ Oncotarget, 2017, Vol. 8, (No. 28), pp: 45200-45212
Mesenchymal stem cells release exosomes that transfer miRNAs
to endothelial cells and promote angiogenesis
Min Gong
1,2
, Bin Yu
1
, Jingcai Wang
1
, Yigang Wang
1
, Min Liu
1
, Christian Paul
1
, Ronald
W. Millard
1,3
, De-Sheng Xiao
4
, Muhammad Ashraf
1,5
and Meifeng Xu
1
1
Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
2
Children’s Nutrition Research Centre, Children’s Hospital of Chongqing Medical University, Chongqing, China
3
Department of Pharmacology and Cell Biophysics, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
4
Department of Preventive Medicine, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong
Province, China
5
Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
Correspondence to: Meifeng Xu, email: Meifeng.xu@uc.edu
Keywords: exosomes, miRNA transfer, mesenchymal stem cells, angiogenesis, miR-30b
Received: December 19, 2016 Accepted: March 21, 2017 Published: April 01, 2017
Copyright: Gong et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY
3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
ABSTRACT
Mesenchymal stem cells (MSCs) have been found to benet patients with a
variety of ischemic diseases via promoting angiogenesis. It is also well established
that exosomes secreted from MSCs deliver bioactive molecules, including microRNAs
(miRs) to recipient cells. Therefore, we hypothesized that exosomes secreted from
MSCs deliver miRs into endothelial cells and mediate angiogenesis. The pro-angiogenic
stimulatory capacity of exosomes was investigated using tube-like structure formation
and spheroid-based sprouting of human umbilical vein endothelial cells (HUVECs), and
in vivo Matrigel plug assay. The secretion of pro-angiogenic miRs (pro-angiomiRs)
from MSCs into culture medium and transfer of the miRs to HUVECs were conrmed
using real-time quantitative PCR. Supplementation of the exosome secretion blocker
GW4869 (10 μM) reduced the pro-angiomiRs in the MSC-derived conditioned medium
(CdM
MSC
). Addition of exosomes isolated from CdM
MSC
could directly 1) promote HUVEC
tube-like structure formation in vitro; 2) mobilize endothelial cells into Matrigel plug
subcutaneously transplanted into mice; and 3) increase blood ow inside Matrigel
plug. Fluorescence tracking showed that the exosomes were internalized rapidly by
HUVECs causing an upregulated expression of pro-angiomiRs in HUVECs. Loss-and-
gain function of the pro-angiomiRs (e.g., miR-30b) in MSCs signicantly altered the
pro-angiogenic properties of these MSC-derived exosomes, which could be associated
with the regulation of their targets in HUVECs. These results suggest that exosomal
transfer of pro-angiogenic miRs plays an important role in MSC mediated angiogenesis
and stem cell-to-endothelial cell communication.
INTRODUCTION
It has been estimated that more than 500 million
people will be beneted from various angiogenic therapies
in the coming decades for treating ischemic diseases
such as peripheral and coronary vascular disease [1],
cerebral infarction [2], and critical limb ischemia [3].
Angiogenesis is a complex biological process involving
interactions between vascular cells and the extracellular
environment. Cell-based pro-angiogenic therapies have
been increasingly utilized in the treatment of ischemic
diseases [4, 5]. Consequently, stem cells have been
extensively used to experimentally treat ischemic diseases
including myocardial infarction [6, 7] and stroke [8, 9].
Mesenchymal stem cells (MSCs) have been recognized as
a promising treatment option with the potential to generate
a variety of useful cell-based interventions [10, 11] and
pro-angiogenic therapies [12, 13]. Indeed, both in vitro
and in vivo models have shown that MSCs can increase
endothelial cell growth and enhance new blood vessel
formation [14], as a result of paracrine effects that are
considered as the predominant mechanism in addressing
Research Paper

Oncotarget45201
www.impactjournals.com/oncotarget
tissue damage [15]. We previously demonstrated that
conditioned medium (CdM) of MSCs promoted post-
infarction angiogenesis in ischemic myocardium and
global heart function recovery [16]. However, the exact
molecular mechanisms responsible for these benecial
paracrine effects of MSCs have not been identied.
Exosomes are cell-derived vesicles (diameter
30–100 nm) that exist in almost all biological uids
including blood, urine, saliva, cerebrospinal uid, and
cell preconditioned medium [17, 18]. They are initially
formed by fusion of a multi-vesicular body with a
plasma membrane, or released directly from the plasma
membrane [17, 19]. Exosomes shuttle mRNAs, miRs,
and other molecular constituents to achieve cell-to-cell
communication, and modulate the function of recipient
cells [20]. However, exosomes contents vary from different
cell types, pathological conditions and by preconditioning
or genetic manipulation of the parent MSCs [21, 22],
which might cause completely inversed fate of target cells.
Most recently, the existence of miRs in exosomes
has been reported [23–25], suggesting that exosomes
may serve as a vehicle for miR transfer and mediate
intercellular communication [26]. MiRs, a class of small
non-coding RNAs (containing about 18–22 nucleotides),
regulate gene expression on the posttranscriptional
level by binding to specic mRNA and inducing
their degradation and/or translational inhibition [27].
MiRs are recognized to participate in a wide range of
biological and pathological processes including the cell
cycle, hematopoiesis, neurogenesis, aging, cancer, and
cardiovascular disease [28]. Evidence has suggested that
miRs are key regulators of endothelial cell function and
are especially important modulators of angiogenesis [29].
For instance, it has been reported that miR-424 promoted
angiogenesis in vitro and in a mouse model by targeting
cullin 2 [30]. miR-30 family targeted DLL4 in endothelial
cells to promote angiogenesis [31]. The present study was
designed to investigate whether MSC-derived exosomes
shuttle various pro-angiogenic miRs and transfer
these miRs to endothelial cells resulting in promoting
angiogenesis.
RESULTS
Pro-angiogenic capacity of conditioned medium
derived from MSCs
MSCs line C3H10T1/2 cells were purchased from
ATCC (Manassas, VA, USA). MSCs adhered to the surface
of plastic culture dishes and exhibited a spindle-shaped
broblast-like morphology as shown in the Supplementary
Figure 1. The pro-angiogenic capacity of CdM obtained
from these cells (CdM
MSC
) was assessed using tube-like
structure formation, spheroid-based sprouting of HUVECs
and in vivo Matrigel plug assay. The cumulative tube
length was signicantly longer (31.80 ± 3.37 mm/eld) in
HUVECs treated with CdM
MSC
compared to those treated
with control medium (18.69 ± 2.83 mm/eld) following
culture for 16 h (Figure 1A). Sprout length per spheroid in
HUVECs treated with CdM
MSC
for 16 h was signicantly
longer (216.67 ± 36.29 μm/spheroid) than that treated
with control medium (82.66 ± 32.23 μm/spheroid)
(Figure 1B). The effect of CdM
MSC
on endothelial cell
invasion and hemoglobin concentration in Matrigel plug
was investigated following subcutaneous injection of
Matrigel into C57BL6 mice. The Matrigel plug contained
CdM
MSC
had a red gross appearance after transplanting
for 14 days (Figure 1C). The hemoglobin content (a sign
of increased new vessel formation) was signicantly
increased in the plugs containing CdM
MSC
(11.14 ±
5.01 μg/mg plug) compared to the Matrigel plugs without
CdM
MSC
(2.48 ± 1.19 μg/mg plug) (Figure 1D). The
neovasculature visualized by immunouorescence staining
of CD31 indicated that the number of CD31 positive cells
in the plugs containing CdM
MSC
was signicantly higher
than that without CdM
MSC
(Figure 1E).
miRs secreted from MSCs transfer to HUVECs
The expression of 26 most commonly
acknowledged pro-angiogenic miRs (pro-angiomiRs) in
CdM
MSC
was quantified using real-time PCR before and
after adding into HUVEC culture. The expression of
miR-424, miR-30c, miR-30b, and let-7f in conditioned
medium was significantly reduced after adding into
HUVECs culture for 48 h, indicating that extracellular
miRs, derived from MSCs, transferred into HUVECs.
Meanwhile, the expression of miR-21, miR-10a, miR-
126, miR-10b, miR-19a, miR-19b was significantly
increased after adding into HUVECs culture,
suggesting that HUVECs might release these miRs
(Table 1). The expression of other miRs was either
very low in CdM
MSC
or the change was not significant
(Supplementary Table 1).
Next, the transfer of miRs between MSCs and
HUVECs was determined using a non-contact co-culture
system. The CdM was collected from MSC-HUVEC
co-culture (CdM
MSC-HUVEC
) and its controls, MSC-MSC
co-culture (CdM
MSC-MSC
) and HUVEC-HUVEC co-
culture (CdM
HUVEC-HUVEC
), respectively. The expression
of miR-30b, 30c, 424 and let-7f in CdM
HUVEC-HUVEC
was
very low and in CdM
MSC-MSC
was very high. However,
the expression of these miRs in CdM
MSC-HUVEC
was
signicantly lower compared to those in CdM
MSC-MSC
,
indicating that these miRs transferred into HUVECs
(Figure 2A). Moreover, the expression of these miRs in
HUVECs co-cultured with MSCs was signicantly higher
than in those without co-cultured with MSCs (Figure 2B),
directly demonstrating a transfer of these miRs into
HUVECs.

Oncotarget45202
www.impactjournals.com/oncotarget
Exosomes derived from MSCs deliver pro-
angiomiRs and promote angiogenesis
To investigate whether exosomes mediated
miRs transfer, the expression of miRs in CdM was
measured after MSCs were treated with 10 μM GW4869
(an exosome release inhibitor) for 48 h [32]. As shown
in Figure 3A, the levels of miR-30b, -30c, -424, and
let-7f in the CdM collected from MSCs treated with
GW4869 (CdM
GW4869
) were signicantly decreased
compared with CdM obtained from control MSCs. In
addition, the expression of these miRs in HUVECs
treated with CdM
GW4869
for 48h was also signicantly
decreased compared to those treated with CdM
MSC
(Figure 3B), indicating that exosomes mediated miR
transportation between MSCs and HUVECs. Exosomes,
isolated from CdM
MSC
, exhibited the characteristic round
morphology with heterogeneous size under transmission
electron microscope (Figure 4A). The average size of
exosomes was 48.72 ± 2.7nm according to the results
of dynamic light scattering (Figure 4B). The expression
of CD9, CD63, and HSP70 was signicantly higher in
exosomes compared to their parent MSCs (Figure 4C).
The internalization of exosome pre-labeled with PKH26
by HUVECs was recorded using IncuCyte ZOOM Live
Content Imaging System every 2 h for 12 h. The PKH26
red uorescence intensity increased with the passage of
time and achieved its maximum after exosomes were
added into HUVECs culture for 10 h (Figure 4D). The
expression of miR-30b, 30c, 424, and let-7f in HUVECs
treated with exosomes for 24 h was signicantly increased
compared to those treated with BSA (Figure 4E).
Figure 1: CdM derived from MSCs promotes angiogenesis. (A) Representative images of capillary-like structures and quantitative
analysis of the total tube length (4× magnication microscopic elds); (B) Representative images of HUVEC spheroids sprouting and
quantitative analysis of the cumulative sprout length per spheroid; (C) Representative gross look of Matrigel plugs which were implanted
subcutaneously in mice for 14 days; (D) Hemoglobin content in the Matrigel plugs; (E) The neovasculature in Matrigel was visualized
by immunouorescence staining of CD31. Quantication of the CD31-positive cells in Matrigel plugs were determined by pixel density
(*P < 0.05 vs CON).

Oncotarget45203
www.impactjournals.com/oncotarget
The pro-angiogenic capacity of exosomes was
examined in vitro and in vivo. The cumulative tube length
was signicantly increased in HUVECs treated with
exosomes (100μg/ml) for 16 h (36.24 ± 3.65 mm/eld)
compared to that treated with BSA in the same protein
amount (15.73 ± 2.44 mm/eld) (Figure 5A). The Matrigel
plug test showed a more red appearance in those containing
exosomes (100μg) after being transplanted for 14 days
than those without exosomes (Figure 5B). The hemoglobin
concentration in the plugs containing exosomes
was signicantly higher (11.76 ± 5.61 μg/mg plug)
than those without exosomes (2.54 ± 1.45 μg/mg plug)
(Figure 5C). The immunouorescence staining showed
that the number of CD31 positive cells in the plugs
containing exosomes was also signicantly higher than
those without exosomes (Figure 5D).
To demonstrate the effect of transferred miRs on
angiogenesis, miR-30b was selected as a representative
miR. Exosomes were obtained from MSCs in which
miR-30b was overexpressed or knockdown, respectively.
MSCs were infected with lentivirus carrying the pre-miR-
30b fragment (MSC
miR-30b
). The expression of miR-30b in
MSC
miR-30b
and exosomes derived from MSC
miR-30b
(Exo
miR-
30b
) was 5.24-fold and 5.22-fold increase compared with
their counterpart MSC
scrambled
and Exo
scrambled
, respectively
(Figure 6A). The cumulative tube length was increased in
Table 1: The expression of pro-angiogenic miRNAs in CdM
MSC
after adding into HUVECs culture
for 48 hours
Downregulated Upregulated
miRNA
CdM
MSC
2
(−ΔCt)
CdM
MSC
with HUVECs 2
(−ΔCt)
miRNA
CdM
MSC
2
(−ΔCt)
CdM
MSC
with HUVECs 2
(−ΔCt)
miR-424
#
44.965 ± 5.542 10.725 ± 1.795
*
miR-21 89.021 ± 9.117 187.956 ± 27.620
*
miR-30c 6.420 ± 0.623 0.572 ± 0.140
*
miR-10a 0.435 ± 0.040 10.160 ± 0.985
*
miR-30b 5.877 ± 0.692 0.133 ± 0.012
*
miR-126 0.045 ± 0.014 6.988 ± 0.933
*
let-7f 4.592 ± 0.245 0.153 ± 0.003
*
miR-10b 0.008 ± 0.002 5.869 ± 0.442
*
miR-19a 1.623 ± 0.063 3.380 ± 0.316
*
miR-19b 1.540 ± 0.116 2.950 ± 0.225
*
(*P < 0.05 vs CdM
MSC
).
#
The mouse homologue of miR-424 sequence from human is miR-322-5p.
Figure 2: miRs secreted from MSCs transfer to HUVECs. (A) miRNA expression in the CdM of non-contact cell co-culture
system (*P < 0.05 vs CdM
HUVEC-HUVEC
;
&
P < 0.05 vs CdM
MSC-MSC
, respectively); (B) The expression of miRs in HUVECs after co-culture
with MSCs (*P < 0.05 vs HUVEC alone).

Oncotarget45204
www.impactjournals.com/oncotarget
Figure 4: Characterization of exosomes and their involvement in miRs transfer. (A) Morphology of exosomes under
transmission electron microscopy; (B) The exosome size was measured using a Zetasizer Nano instrument. (C) The expression of CD9,
CD63 and HSP70; (D) Representative images of time-lapse internalization of PKH26-labled exosomes (red) in HUVECs. (E) miR
expression in HUVECs following exosome treatment (*P < 0.05 vs HUVEC+ BSA).
Figure 3: Exosomes mediate the transfer of miRs from MSCs to HUVECs. (A) The expression of miRs in CdM
(*P < 0.05 vs
CdM
MSC
); (B) The expression of miRs in HUVECs treated with CdM (
&
P < 0.05 vs HUVEC + CdM
MSC
).

Citations
More filters
Journal ArticleDOI

Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering.

TL;DR: The history of MSC research from the initial discovery of their multipotency to the more recent recognition of their perivascular identity in vivo and their extraordinary capacity for immunomodulation and angiogenic signaling is summarized.
Journal ArticleDOI

MSC exosome works through a protein-based mechanism of action

TL;DR: It is proposed that MSC exosomes most probably work through the protein rather than the RNA, and this rationale beyond a physical presence to include biologically relevant concentration, biochemical functionality and the potential to elicit an appropriate timely biochemical response is expanded.
Journal ArticleDOI

The Osteosarcoma Microenvironment: A Complex But Targetable Ecosystem.

TL;DR: The different actors of the osteosarcoma microenvironment are described and an overview of the past, current, and future strategies of therapy targeting this complex ecosystem is given, with a focus on the role of extracellular vesicles and on the emergence of multi-kinase inhibitors.
Journal ArticleDOI

Intranasal Delivery of Mesenchymal Stem Cell Derived Exosomes Loaded with Phosphatase and Tensin Homolog siRNA Repairs Complete Spinal Cord Injury.

TL;DR: It is demonstrated that when given intranasally, exosomes derived from mesenchymal stem cells could pass the blood brain barrier, and migrate to the injured spinal cord area, and MSC-Exo loaded with phosphatase and tensin homolog small interfering RNA (ExoPTEN) could attenuate the expression of PTEN in thejured spinal cord region followingintranasal administrations.
Journal ArticleDOI

Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment.

TL;DR: Although MSCs have a potential to exert anti-tumor activities, they largely provide service to the tumor using the multidirectional communication system established by exosomes in the TME.
References
More filters
Journal ArticleDOI

Cardiomyocyte Protection by GATA-4 Gene Engineered Mesenchymal Stem Cells Is Partially Mediated by Translocation of miR-221 in Microvesicles

TL;DR: It is found that miR-221 translocation by microvesicles (MVs) plays an important role in cardioprotection mediated by GATA-4 overexpressed mesenchymal stem cells (MSC).
Journal ArticleDOI

Dynamic induction of pro-angiogenic milieu after transplantation of marrow-derived mesenchymal stem cells in experimental myocardial infarction.

TL;DR: An angiogenesis-promoting milieu was induced after the transplantation of marrow MSCs in the injured myocardium, and compared with the resident cells, the transplanted cells had a greater rate of cellular kinetics in the infarctedMyocardium.
Journal ArticleDOI

miR-30a Regulates Endothelial Tip Cell Formation and Arteriolar Branching

TL;DR: The data suggest that miR-30a stimulates arteriolar branching by downregulating endothelial Dll4 expression, thereby controlling endothelial tip cell behavior, which could have relevance to the rarefaction process and, therefore, to hypertension.
Journal ArticleDOI

Vascular signal transducer and activator of transcription-3 promotes angiogenesis and neuroplasticity long-term after stroke.

TL;DR: Endothelial Stat3 regulatesAngiogenesis, axon growth, and extracellular matrix remodeling and is essential for long-term recovery after stroke and might serve as a potent target for stroke treatment after the acute phase by fostering angiogenesis and neuroregeneration.
Journal ArticleDOI

The histone methyltransferase MLL is an upstream regulator of endothelial cell sprout formation

TL;DR: It is concluded that MLL regulates endothelial-cell migration via HoxA9 and EphB4, whereas sprout formation requires MLL-dependent signals beyond Hox a9 and HoxD3.
Related Papers (5)

Minimal information for studies of extracellular vesicles 2018 (MISEV2018) : a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines

Clotilde Théry, +417 more