ArticlePDF Available

miR-185/ P2Y6 Axis Inhibits Angiotensin II-Induced Human Aortic Vascular Smooth Muscle Cell Proliferation

Authors:
  • The First Hospital of Hunan University of Chinese Medicine

Abstract and Figures

The abnormal proliferation and apoptosis of human aortic vascular smooth muscle cells (HAVSMCs) play an important role in the pathogenesis of hypertension. Recent study revealed that angiotensin II (Ang II) could elicit HAVSMC dysfunction, to induce or aggravate hypertension. Purinergic receptor P2Y6, an inflammation-inducible G protein-coupled receptor, promoted Ang II-induced hypertension. In the present study, we revealed that Ang II induced HAVSMC proliferation and upregulated P2Y6 protein levels. After knockdown of P2Y6, the promotive effect of Ang II on HAVSMC proliferation was restored. microRNAs (miRNAs) involve in most biological processes. In this study, we scanned out seven candidate miRNAs, which were predicted to contain binding site of P2Y6's 3'-UTR by online tools. Among them, miR-185 was significantly downregulated by Ang II treatment. miR-185 reduced P2Y6 protein levels by direct binding to the 3'UTR of P2Y6. miR-185 overexpression suppressed HAVSMC proliferation; P2Y6 overexpression or Ang II treatment promoted HAVSMC proliferation, and restored the suppressive effect of miR-185 on HAVSMC proliferation. Besides, miR-185/P2Y6 axis also affected pERK1/2 protein levels. Taken together, the present study indicated that miR-185/P2Y6 axis might inhibit Ang II-induced HAVSMC proliferation through miR-185 negatively regulating P2Y6 expression and the downstream ERK pathway; rescuing miR-185 expression to inhibit P2Y6 may represent a therapeutic strategy against HAVSMC dysfunction and hypertension.
Content may be subject to copyright.
ORIGINAL RESEARCH ARTICLE
miR-185/P2Y6 Axis Inhibits Angiotensin II-Induced Human
Aortic Vascular Smooth Muscle Cell Proliferation
Shunmin Wang,
1,2
Lujun Tang,
1
Qian Zhou,
1
Duomei Lu,
1
Wulei Duan,
1
Cheng Chen,
1
Lu Huang,
2
and Yuansheng Tan
3,4
The abnormal proliferation and apoptosis of human aortic vascular smooth muscle cells (HAVSMCs) play an
important role in the pathogenesis of hypertension. Recent study revealed that angiotensin II (Ang II) could
elicit HAVSMC dysfunction, to induce or aggravate hypertension. Purinergic receptor P2Y6, an inflammation-
inducible G protein-coupled receptor, promoted Ang II-induced hypertension. In the present study, we revealed
that Ang II induced HAVSMC proliferation and upregulated P2Y6 protein levels. After knockdown of P2Y6,
the promotive effect of Ang II on HAVSMC proliferation was restored. microRNAs (miRNAs) involve in most
biological processes. In this study, we scanned out seven candidate miRNAs, which were predicted to contain
binding site of P2Y6’s 3¢-UTR by online tools. Among them, miR-185 was significantly downregulated by Ang
II treatment. miR-185 reduced P2Y6 protein levels by direct binding to the 3¢UTR of P2Y6. miR-185 over-
expression suppressed HAVSMC proliferation; P2Y6 overexpression or Ang II treatment promoted HAVSMC
proliferation, and restored the suppressive effect of miR-185 on HAVSMC proliferation. Besides, miR-185/
P2Y6 axis also affected pERK1/2 protein levels. Taken together, the present study indicated that miR-185/P2Y6
axis might inhibit Ang II-induced HAVSMC proliferation through miR-185 negatively regulating P2Y6 ex-
pression and the downstream ERK pathway; rescuing miR-185 expression to inhibit P2Y6 may represent a
therapeutic strategy against HAVSMC dysfunction and hypertension.
Keywords: miR-185, human aortic vascular smooth muscle cells, angiotensin II, P2Y6, proliferation, hypertension
Introduction
Human aortic vascular smooth muscle cells
(HAVSMCs), which are present under the vascular
endothelial cells, are the main cellular components that form
the structure of the vascular wall and maintain the blood
vessel. In normal adults, vascular smooth muscle cells
maintain the tension of the blood vessel wall by slow and
mild contraction. Blood pressure is largely maintained by
vascular tension. Abnormal proliferation of vascular smooth
muscle cells plays an important role in the pathogenesis of
hypertension (Voelkel and Tuder, 1997). Overproliferation of
vascular smooth muscle cells lead to smaller vascular lumen,
thickening of the vascular wall, and plays an important role in
pathogenesis of hypertension (Voelkel and Tuder, 1997).
The rennin–angiotensin system (RAS) plays an important
role in the pathogenesis of hypertension. The system in-
cludes renin, angiotensin (Ang II), angiotensin-converting
enzyme and so on. Ang II has strong capability of vaso-
constriction, and can promote vascular smooth muscle cell
proliferation, hardens the vessel wall, and causes the lumen
to narrow (Touyz, 2005). Ang II could increase blood
pressure and promote arterial reconstruction through the
strong contraction of blood vessels; on the other hand, Ang
II can directly promote vascular smooth muscle cell hy-
pertrophy and collagen fiber proliferation, leading to vas-
cular remodeling (Pueyo and Michel, 1998; Takemoto and
Egashira, 1999). Ang II can also promote many processes
of cardiovascular tissue, including cell growth, migration,
differentiation, and apoptosis. Therefore, Ang II may be
one of the important factors affecting the cardiovascu-
lar remodeling (Komuro and Kudoh, 1999). However, how
HAVSMCs determine the responsiveness to Ang II under
different developmental and region-specific conditions is
mostly unclear.
Purinergic P2Y receptors are a family of inflammation-
inducible G protein-coupled receptors that respond to extra-
cellular mononucleotides (Abbracchio et al.,2006).Multiple
1
The Graduate Institute, Hunan University of Chinese Medicine, Changsha City, China.
2
Department of Cardiovascular, The First Hospital of Hunan University of Chinese Medicine, Changsha, China.
3
Department of Cardiovascular, First College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China.
4
Department of Cardiovascular, College of Integrated Traditional Chinese and Western Medicine, Hunan Academy of Chinese Medicine,
Changsha, China.
DNA AND CELL BIOLOGY
Volume 36, Number 5, 2017
ªMary Ann Liebert, Inc.
Pp. 1–9
DOI: 10.1089/dna.2016.3605
1
P2Y receptor subtypes, including P2Y6 (Pediani et al., 1999;
Wang et al., 2002), are present in blood vessels. P2Y6 acti-
vation induces a Ca
2+
response in vascular smooth muscle
cells that results in contraction of isolated blood vessels
(Abbracchio et al., 2006). These pharmacological studies
suggest that P2Y6 plays an important role in cardiovascular
function. In addition, P2Y6 ablation attenuates vascular in-
flammation in vivo under pathological conditions (Riegel
et al., 2011; Stachon et al., 2014). These results suggest that
P2Y6 participates in the development of pathological vas-
cular remodeling. However, the functional role of P2Y6 on
HAVSMCs’ proliferation still remains to be validated.
The discovery of microRNAs (miRNAs) has opened new
avenues for studying and understanding hypertension and
hypertension-associated endothelial dysfunction, featuring a
postgenomic era of biomedical research (Nemecz et al.,
2016). These noncoding regulatory RNA molecules of *22
nucleotides have emerged as potential biomarkers, effectors,
and targets for diagnosis, prognosis, and therapy in hyper-
tension. Lately, many studies have been done on under-
standing miRNA biology and function. It seems that
miRNAs play an important role in the regulation of almost
every cellular process. Usually, a single miRNA can interact
with hundreds of mRNA molecules and a specific mRNA
molecule may be the target of multiple miRNAs. Thus,
miRNA–mRNA interactions may delineate the complex
regulatory networks with consequence on the target gene
expression and hence on some biological processes. Ad-
ditionally, the disruptions of miRNA regulation are fre-
quently associated with some pathological states, including
hypertension-associated endothelial dysfunction (Nemecz
et al., 2016).
In the present study, we validated the functional role
of Ang II in HAVSMCs proliferation and P2Y6 expres-
sion, and the effect of Ang II-induced P2Y6 expression
on HAVSMC proliferation. By using the online tools, we
scanned out some candidate miRNAs that contain the binding
site in 3¢UTR of P2Y6 and were associated with cell pro-
liferation. Among them, miR-185 could be downregulated
by Ang II treatment. As verified by performing a series of
functional and mechanistic assays, we revealed that miR-
185/P2Y6 axis could suppress HAVSMCs proliferation
through ERK signaling.
Materials and Methods
Cell lines and cell transfection
HAVSMCs were obtained from ATCC, cultured in
RPMI-1640 medium (Invitrogen, CA) adding 10% fetal calf
serum (FBS) (Gibco, CA) as the supplement, and incubated
at 37C in a humidified atmosphere with 5% CO
2
.
P2Y6-siRNA1 and P2Y6-siRNA2 were used to achieve
P2Y6 silence (GeneCopoecia, Guangzhou, China). pCMV-
P2Y6 vector was used to achieve exogenous P2Y6 expres-
sion (GeneCopoecia, Guangzhou, China). miR-185 mimics
or miR-185 inhibitor (GeneCopoecia, Guangzhou, China)
was used to achieve miR-185 overexpression or inhibition.
After being plated in 96- or 6-well plates, HAVSMCs were
then transfected with indicated siRNAs, mimics or inhibitor
with lipo2000 (Invitrogen), cultured for 24 h, and conse-
quently used for the next experiments.
RNA extraction and real-time PCR assays
We extracted total RNA from target cells by using TRIzol
reagent (Invitrogen). Mature miR-185, miR-483, miR-20a,
miR-1182, miR-3151, miR-149, and miR-877 expression in
cells was detected using the Hairpin-itmiRNAs qPCR Kit
(GenePharma, Shanghai, China). P2Y6 mRNA expression
was monitored by using SYBR Green qPCR assay. RNU6B
expression was used as an endogenous normalization. Data
were analyzed using 2
-DDCT
method.
Cell Counting Kit-8 cell proliferation assay
Cell Counting Kit-8 (CCK-8) (Beyotime, Hangzhou, China)
was used to measure HAVSMCs proliferation rates. We see-
ded 0.5 ·10
4
cells in each 96-well plate for 24 h, transfected
them with the indicated siRNA, miRNA mimics or inhibitor,
and further incubated cells for 24, 48, 72, 96, and 120 h, re-
spectively. At 1 h before the endpoint of incubation, we added
10 mL CCK-8 reagents to each well. A microplate reader was
used to determine OD
570nm
value in each well.
5-Bromo-2-deoxyUridine cell proliferation assay
By measuring 5-Bromo-2-deoxyUridine (BrdU) incorpo-
ration, the DNA synthesis in proliferating cells was deter-
mined. BrdU assays were conducted at 24 and 48 h after
HAVSMCs were transfected with the indicated vectors. Cells
were seeded in 96-well culture plates at a density of 2·10
3
cells/well, cultured for 48 h, then incubated with a final con-
centration of 10mM BrdU (BD Pharmingen, San Diego, CA)
for 2 h. When the incubation period ended, the medium was
removed, the cells were fixed for 30 min at RT, incubated with
peroxidase-coupled anti-BrdU-antibody (Sigma-Aldrich) for
60 min at RT, washed three times with PBS, incubated with
peroxidase substrate (tetramethylbenzidine) for 30 min, and
the 450 nm absorbance values were measured for each well.
Background BrdU immunofluorescence was determined in
cells not exposed to BrdU, but stained with the BrdU antibody.
Western blotting
RIPA buffer (Cell-Signaling Tech.) was used to homo-
genize the cells. The expression of P2Y6, ERK1/2, and
pERK1/2 in HAVSMC was detected by performing immu-
noblotting. After treatments, HAVSMCs were lysed in 1%
PMSF supplemented RIPA buffer. Protein were loaded onto
SDS-PAGE minigel, and then transferred onto PVDF
membrane. The blots were probed with 1:1000 diluted rabbit
polyclonal P2Y6, ERK1/2, and pERK1/2 antibody (Cat no.
ab92504, ab17942, ab50011; Abcam) at 4C overnight, and
incubated with Horseradish Peroxidase (HRP)-conjugated
secondary antibody (1:5000) (Cat nos. A0208 and A0216;
Beyotime, China). Signals were visualized using enhance
chemiluminescence (ECL) Substrates (Millipore). The pro-
tein expression was normalized to endogenous GAPDH
(AF0006; Beyotime, China).
Luciferase reporter assay
UTR luciferase reporter assays were performed in 293T
cells (ATCC). After being cultured overnight, cells were
cotransfected with the wild-type (wt-P2Y6)/mutated-type
(mut-P2Y6 3¢UTR1 and mut-P2Y6 3¢UTR2) reporter plas-
mids (purchased from Yrbio Tech, China) and miR-185
2 WANG ET AL.
mimics or miR-185 inhibitor. Luciferase assays were per-
formed 48 h after transfection using the Dual Luciferase
Reporter Assay System (Promega, WI).
Statistical analysis
Data from at least three independent experiments were
exhibited as mean standard deviation, analyzed by SPSS
17.0 statistical software (SPSS, Chicago). Paired Student’s
t-test was used to compare the differences between two
groups. One-way ANOVA was used to analyze the differ-
ences between groups in the proliferation assays. A pvalue
of <0.05 was considered statistically significant.
Results
Ang II induced HAVSMCs proliferation
and upregulated P2Y6 protein level
Ang II has been reported to induce vascular smooth muscle
cells’ proliferation. In this study, we treated HAVSMC with
different concentrations of Ang II (0, 10nM, 100 nM, 1 mM),
or for different durations (0, 12, 24, 48h), and determined the
cell proliferation by using CCK-8 assays, determined the
protein levels of P2Y6 by using western blot assays. Results
showed that the proliferation of HAVSMC was promoted by
Ang II in a concentration- or time-dependent manner (Fig. 1A,
B). P2Y6 protein levels were also upregulated by Ang II in a
concentration- or time-dependent manner (Fig. 1C, D). These
data indicated that Ang II promoted HAVSMC proliferation
and P2Y6 protein levels in HAVSMC in a concentration- and
time-dependent manner.
P2Y6 silence could partially restore the promotive
effect of Ang II on HAVSMCs proliferation
To validate the role of P2Y6 in Ang II-induced HAVSMC
proliferation, two siRNAs were used to achieve P2Y6 si-
lence. Inhibitory efficiency of two siRNAs was verified by
using real-time PCR assays, and P2Y6-siRNA1 reduced
P2Y6 expression more effectively (Fig. 2A). Protein levels
of P2Y6 were determined by using western blot assays, and
similar results were observed: P2Y6-siRNA1 reduced P2Y6
protein level more effectively (Fig. 2B). Then P2Y6-
siRNA1 was transfected into HAVSMC with the presence of
Ang II, and the cell proliferation was determined by using
CCK-8 and BrdU assays. Results showed that Ang II sig-
nificantly promoted HAVSMCs proliferation, whereas P2Y6
silence suppressed HAVSMCs proliferation, compared with
the control group (Fig. 2C, D); after being transfected with
P2Y6-siRNA1, the promotive effect of Ang II on HAVSMCs
proliferation was partially restored (Fig. 2C, D). These data
indicated that P2Y6 is involved in Ang II-induced HAVSMC
proliferation; P2Y6 silence by P2Y6-siRNA could par-
tially restore the promotive effect of Ang II on HAVSMC
proliferation.
Prediction and screening of upstream miRNAs of P2Y6
The disruptions of miRNA regulation are frequently asso-
ciated with some pathological states including hypertension-
associated endothelial dysfunction. Ang II has previously
been reported to regulate the expression of miR-29b, miR-
129-3p, and miR-212 through mechanisms depending on
Gaq/11 and ERK1/2 activation (Jeppesen et al.,2011).
FIG. 1. Ang II induced HAVSMCs pro-
liferation and upregulated P2Y6 protein le-
vel (A) The cell proliferation of HAVSMCs
in response to different concentrations of
Ang II treatment for 24 h (0, 10 nM, 100 nM,
1mM) determined by using CCK-8 assays.
(B) The cell proliferation of HAVSMC in
response to 100 nM Ang II treatment for 0,
12, 24, and 48 h determined by using CCK-8
assays. (C) P2Y6 protein levels in response
to different concentrations of Ang II treat-
ment for 24 h (0, 10 nM, 100 nM, 1 mM)
determined by using western blot assays. (D)
P2Y6 protein levels in response to100 nM
Ang II treatment for 0, 12, 24, and 48 h
determined by using western blot assays.
Data are presented as mean SD, n=8.
*p<0.05, **p<0.01. Ang II, angiotensin II;
CCK-8, Cell Counting Kit-8; HAVSMC,
human aortic vascular smooth muscle cell;
SD, standard deviation.
MIR-185/P2Y6 AXIS REGULATES HAVSMCSGROWTH 3
FIG. 2. P2Y6 silence could partially
restore the promotive effect of Ang II on
HAVSMC proliferation (A) P2Y6-siRNA1
and P2Y6-siRNA2 were constructed to
achieve P2Y6 silence. The mRNA levels
of P2Y6 were determined by using real-time
PCR assays. (B) The protein levels of P2Y6
after P2Y6-siRNA1 or P2Y6-siRNA2 trans-
fection were determined by using western
blot assays. (C) The cell viability
of HAVSMC in response to P2Y6-siRNA1
transfection with the presence of Ang II
treatment (100 nM) was determined by using
CCK-8 assays. (D) The cell proliferation
of HAVSMC in response to P2Y6-siRNA1
transfection with the presence of Ang II
treatment was determined by using BrdU
assays. Data are presented as mean SD,
n=6. *p<0.05 and **p<0.01 versus
si-NC group,
##
p<0.01 versus P2Y6-
siRNA1 group,
DD
p<0.01 versus
AngII +si-NC group.
FIG. 3. Prediction and
screening of upstream miR-
NAs of P2Y6 (A) Six candi-
date miRNAs (miR-185,
miR-483, miR-20a, miR-
1182, miR-149, and miR-
877), which had the potential
of targeting P2Y6 were
screened out by using online
tools, including miRwalk,
miRanda, RNA22, TargetS-
can, and microT-CDS, and
referring to the previous
studies. (B) The expression
levels of the indicated can-
didate miRNAs in response
to Ang II treatment (100 nM,
24 h) were determined by
using real-time PCR assays.
Data are presented as
mean SD, n=6. *p<0.05,
**p<0.01 versus DMSO
group. miRNAs, micro-
RNAs.
4 WANG ET AL.
Likewise, recent data suggested that miR-132 and miR-212
are involved in Ang II-induced hypertension (Eskildsen et al.,
2013). In this study, we used online tools, including miRwalk,
miRanda, RNA22, TargetScan, and microT-CDS to scan out
several candidate miRNAs that could potentially target P2Y6
(Fig. 3A). According to the previous literature, we chose six
miRNAs (miR-185, miR-483, miR-20a, miR-1182, miR-149,
and miR-877) to determine their expression levels in response
to Ang II treatment (100 nM, 24 h). By using real-time PCR,
we observed that the expression of miR-185, miR-483, miR-
20a, miR-1182, and miR-877 was modulated by Ang II
treatment, except miR149, among which miR-185 expression
was significantly downregulated (Fig. 3B). These data sug-
gested that miR-185 might be involved in Ang II-induced
HAVSMC proliferation and P2Y6 expression.
miR-185 inhibited P2Y6 expression by direct
binding to the 3¢UTR of P2Y6
To verify the association between miR-185 and P2Y6,
miR-185 mimics or inhibitor was transfected into HAVSMCs
to achieve miR-185 overexpression or inhibition (Fig. 4A).
The protein levels of P2Y6 were determined by using western
blot assays after transfection with miR-185 mimics or in-
hibitor. Results showed that P2Y6 protein was downregulated
by miR-185 overexpression, whereas upregulated by miR-
FIG. 4. miR-185 inhibited P2Y6 expres-
sion by direct binding to the 3¢UTR of P2Y6
(A) miR-185 mimics or inhibitor was used
to achieve miR-185 overexpression or inhi-
bition. The expression efficiency was veri-
fied by using real-time PCR. (B) The protein
levels of P2Y6 in response to miR-185
overexpression or inhibition were deter-
mined by using western blot assays. (C) A
wt-P2Y6 3¢UTR luciferase reporter gene
vector, as well as a mut-P2Y6 3¢UTR vector
was constructed by mutating the two pre-
dicted binding sites of miR-185 in the
3¢UTR of P2Y6.(D) The indicated luciferase
vectors were cotransfected into 293T cells
with miR-185 mimics or inhibitor. The lu-
ciferase activity was then determined by
using dual luciferase assays. Data are pre-
sented as mean SD, n=6. *p<0.05,
**p<0.01.
MIR-185/P2Y6 AXIS REGULATES HAVSMCSGROWTH 5
185 inhibition (Fig. 4B). We constructed a wt-P2Y6 3¢UTR
luciferase reporter gene vector, as well as a mut-P2Y6 3¢UTR
vector by mutating the two predicted binding sites of miR-
185 in the 3¢UTR of P2Y6 (Fig. 4C). The indicated vectors
were cotransfected into 293T cells with miR-185 mimics or
inhibitor, and the luciferase activity was determined by using
dual luciferase assays. Results showed that the luciferase
activity of wt-P2Y6 3¢UTR vectors was reduced by miR-185
mimics, whereas promoted by miR-185 inhibitor, compared
with the NC (negative control) group; the effect of miR-185
mimics or inhibitor on luciferase activity was offset by either
mutation in the 3¢UTR of P2Y6 (Fig. 4D). These data indi-
cated that miR-185 could reduce P2Y6 protein level by direct
binding to the 3¢UTR of P2Y6.
miR-185 regulated HAVSMC proliferation
and downstream ERK pathway
through targeting P2Y6
We revealed that miR-185 inhibits P2Y6 protein level through
direct binding to its 3¢UTR; next we validated the functional role
of miR-185 in regulating HAVSMCs proliferation. As deter-
mined by CCK-8 and BrdU assays, miR-185 overexpression
significantly inhibited HAVSMCs proliferation, P2Y6 over-
expression promoted HAVSMCs proliferation; P2Y6 could
partially restore the inhibitory effect of miR-185 on HAVSMCs
proliferation (Fig. 5A, B). According to previous studies,
ERK pathway is involved in the pathophysiology of hyper-
tension (Roberts, 2012), Moreover, P2Y receptors, including
P2Y6, could mediate ERK activation to prevent cell apo-
ptosis (Lenz et al., 2000; Kim et al., 2003). In this study, we
further monitored the protein levels of total ERK1/2 and p-
ERK1/2, as well as P2Y6 protein level in response to co-
transfection of miR-185 mimics and pCMV-P2Y6 by using
western blot assays. Results showed that miR-185 over-
expression significantly downregulated P2Y6 and pERK1/2
protein levels without affecting total ERK1/2 protein.
pCMV-P2Y6 transfection exerted an opposite function: up-
regulated both P2Y6 and pERK1/2 protein levels without
affecting total ERK1/2 protein. In addition, P2Y6 partially
restored the effect of miR-185 on the indicated factors
(Fig. 5C). These data indicated that miR-185 inhibits
FIG. 5. miR-185 regulated HAVSMCs
proliferation and downstream ERK pathway
through targeting P2Y6. (A) The cell via-
bility of HAVSMCs in response to co-
transfection of miR-185 mimics and pCMV-
P2Y6 was determined by using CCK-8 as-
says. (B) The cell proliferation of HAVSMC
in response to cotransfection of miR-185
mimics and pCMV-P2Y6 was determined
by using BrdU assays. (C) The protein levels
of ERK1/2 and pERK1/2 in HAVSMC in
response to cotransfection of miR-185
mimics and pCMV-P2Y6 was determined
by using western blot assays. Data are pre-
sented as mean SD, n=6. *p<0.05,
**p<0.01 versus NC mimics +Vector
group;
##
p<0.01 versus miR-185 mimics +
Vector group;
DD
p<0.01 versus NC mimics
+pCMV-P2Y6 group. BrdU, 5-Bromo-2-
deoxyUridine.
6 WANG ET AL.
HAVSMC proliferation through inhibiting P2Y6 expression
and regulating the downstream ERK pathway.
miR-185 restored the promotive effect of Ang II
on P2Y6 expression and HAVSMCs proliferation
We have revealed that miR-185 inhibits HAVSMC pro-
liferation through inhibiting P2Y6 expression; we further
investigated whether ectopic miR-185 expression could re-
store the effect of Ang II on P2Y6 and HAVSMC prolifer-
ation. In HAVSMCs, miR-185 expression was significantly
upregulated by miR-185 mimics; when Ang II treatment was
conducted, miR-185 mimics-induced miR-185 expression
was partially reduced (Fig. 6A). Then the protein levels of
P2Y6 were monitored. As shown by western blot assays,
P2Y6 protein was reduced by ectopic miR-185 expression,
increased by Ang II treatment; the promotive effect of Ang II
treatment on P2Y6 protein could be partially reversed by
ectopic miR-185 expression (Fig. 6B). Furthermore, the cell
proliferation of HAVSMC was monitored by using CCK-8
and BrdU assays. Similarly, the promotive effect of Ang II
treatment on HAVSMC proliferation could be partially re-
versed by miR-185 (Fig. 6C, D). These data further indicated
that miR-185 restores the promotive effect of Ang II on P2Y6
expression; miR-185/P2Y6 axis regulates Ang II-induced
HAVSMC proliferation.
Discussion
Excessive proliferation of vascular smooth muscle cells is
a crucial event in the pathogenesis of several cardiovascular
diseases, including hypertension. In the present study, we
found that Ang II induced HAVSMC proliferation and P2Y6
protein levels in a concentration- or time-dependent manner.
After silence of P2Y6 by P2Y6-siRNA, HAVSMC prolif-
eration was downregulated; in addition, P2Y6 silence could
also partially restore the effect of Ang II on HAVSMC
proliferation. Through online tools and previous literatures,
we chose several candidate miRNAs that might potentially
target P2Y6 and were associated with cell proliferation.
Among them, miR-185 expression could be significantly
downregulated by Ang II treatment. By direct binding to the
FIG. 6. miR-185 restored the promotive effect of Ang II on P2Y6 expression and HAVSMCs proliferation (A) miR-185
mimics was transfected into HAVSMC under Ang II treatment. The expression levels of miR-185 were determined by using
real-time PCR assays. (B) The protein levels of P2Y6 in response to ectopic miR-185 expression under Ang II treatment
were determined by using western blot assays. (C) and (D) The cell proliferation of HAVSMC in response to ectopic miR-
185 expression under Ang II treatment was determined by using CCK-8 and BrdU assays. Data are presented as mean SD,
n=6. *p<0.05, **p<0.01 versus NC mimics group;
#
p<0.05,
##
p<0.01, versus miR-185 mimics group,
DD
p<0.01, versus
Ang II +NC mimics group.
MIR-185/P2Y6 AXIS REGULATES HAVSMCSGROWTH 7
3¢UTR of P2Y6, miR-185 inhibited P2Y6 expression and
HAVSMCs proliferation. Moreover, downstream ERK
pathway might be involved in this process.
The excessive proliferation of vascular smooth muscle cell
can be induced by various cytokines and growth factors, such as
Ang II, insulin, and platelet-derived growth factors (Touyz and
Schiffrin, 1996). Ang II has been frequently reported to be
associated with cardiovascular smooth muscle cell proliferation
and apoptosis (Pueyo and Michel, 1998; Takemoto and Ega-
shira, 1999). Ang II could increase human brain vascular
smooth muscle cell migration, proliferation, and apoptosis, and
increased the blood pressure, neurological deficit score, middle
cerebral artery (MCA) remodeling, and hemorrhage volume in
intracerebral hemorrhage (ICH) mice (Bihl et al., 2015). One
micromole of Ang II significantly promoted the proliferation of
A7r5 cell, an aortic smooth muscle cell line, from 100% to
131% (Li et al., 2015). In the present study, we treated
HAVSMCs with different concentrations of Ang II, for differ-
ent durations. HAVSMCs proliferation was promoted by Ang II
in a concentration- or time-dependent manner. Besides, Ang II
treatment could also upregulate P2Y6 protein levels in a con-
centration- or time-dependent manner.
P2Y receptors are G-protein-coupled receptors activated
by extracellular nucleotides. The P2Y6 receptor is selec-
tively activated by UDP, and its transcript has been detected
in numerous organs, including the spleen, thymus, intestine,
blood leukocytes, and aorta. Nishimura et al. reported that
through heterodimerization with the angiotensin type 1 re-
ceptor, P2Y6 could promote Ang II-induced hypertension
(Nishimura et al., 2016). By generating P2Y6
-/-
mice, Bar
et al. (2008) revealed that the P2Y6 receptor is indeed re-
sponsible for the contractile action of both UDP and UTP in
aorta, and P2Y6 is expressed and functional in vascular
smooth muscle cells. In the present study, we investigated
the detailed role of P2Y6 in HAVSMCs proliferation. After
P2Y6-siRNA transfection, HAVSMCs proliferation was
suppressed. Moreover, Ang II-induced HAVSMC prolifer-
ation could be partially restored by P2Y6 silence. However,
the mechanism by which P2Y6 affects Ang II-induced
HAVSMC proliferation and Ang II-upregulated P2Y6 ex-
pression still remains to be validated.
The RAS has an essential role in blood pressure regula-
tion by affecting cardiac contractility, vascular resistance,
and blood volume. RAS overactivation is a major patho-
genetic factor in hypertension. Several miRNAs have been
shown to interact with the RAS. miR-155 has been found to
regulate AGTR1 expression (Martin et al., 2006). miR-124
and miR-135a independently repressed the translation of
NR3C2, a ligand-dependent transcription factor that regu-
lates water and ion transporter expression, without affecting
mRNA levels. The authors proposed that miR-124 and miR-
135a may contribute to the modulation of the RAAS and
thereby to blood pressure regulation (Sober et al., 2010). In
this study, we used online tools to screen out several can-
didate miRNAs, which were predicted to have the potential
of binding to P2Y6. These candidate miRNAs were also
reported to be associated with cell proliferation: miR-185,
miR-483, miR-20a, miR-1182, miR-149, and miR-877
(Bertero et al., 2011; Fan et al., 2015; Zhang et al., 2015;
Chen et al., 2016; Li et al., 2016; Zhu et al., 2016). After
Ang II treatment, the expression of all candidate miRNAs was
modulated by Ang II, except miR-149, among which miR-185
expression was more strongly downregulated. We then veri-
fied that miR-185 could bind to the 3¢UTR of P2Y6 on the two
predicted binding sites, and inhibit P2Y6 expression through
direct targeting. These data suggested the involvement of miR-
185 in regulation of HAVSMCs proliferation; so we per-
formed a series of functional assays to investigate whether
miR-185 and P2Y6 constitute a regulatory axis to affect
HAVSMC proliferation together.
We observed a significant inhibitory effect of miR-185 on
HAVSMC proliferation by CCK-8 and BrdU assays; in ad-
dition, exogenous P2Y6 expression could partially restore the
inhibitory effect of miR-185 on HAVSMCs proliferation,
indicating that miR-185/P2Y6 axis affected HAVSMC pro-
liferation. A similar result has been reported by Shan et al.
that miR-185-5p inhibitor transfection increased the viability
and proliferation of HUVECs; lncRNA-RNCR3, miR-185-
5p, and KLF2 constitute a regulatory network to regulate the
viability and proliferation of HUVECs (Shan et al., 2016).
Recent studies reported that miRNA–mRNA interactions
may delineate the complex regulatory networks with conse-
quence on the target gene expression and hence on some
biological processes. Due to its major role in pathophysiology
of hypertension (Guo et al., 2014; Li et al.,2017),wethen
validated whether ERK pathway was involved in regulation
of HAVSMC proliferation by miR-185. As shown by western
blot, miR-185 reduced pERK1/2 protein level; P2Y6 in-
creased pERK1/2 protein level, and partially restored the
effect of miR-185 on pERK protein. These data suggested
that ERK pathway was involved in the process of miR-185/
P2Y6 axis inhibiting Ang II-induced HAVSMC proliferation.
Taken together, we indicated that miR-185/P2Y6 axis
inhibits Ang II-induced HAVSMC proliferation through the
downstream ERK pathway; rescuing miR-185 expression to
inhibit P2Y6 expression may thus represent a therapeutic
strategy against HAVSMCs dysfunction and hypertension.
Acknowledgments
This study was supported by the National Natural Science
Foundation of China (program no.: 81473616), Science and
Technology Program Major Project of China Hunan Pro-
vincial Science & Technology Department (2016DK2002),
and Hunan Traditional Chinese Medicine Scientific Re-
search Key Project (201753).
Disclosure Statement
No competing financial interests exist.
References
Abbracchio, M.P., Burnstock, G., Boeynaems, J.M., Barnard,
E.A., Boyer, J.L., Kennedy, C., Knight, G.E., Fumagalli, M.,
Gachet, C., and Jacobson, K.A. et al. (2006). International
Union of Pharmacology LVIII: update on the P2Y G protein-
coupled nucleotide receptors: from molecular mechanisms
and pathophysiology to therapy. Pharmacol Rev 58, 281–341.
Bar, I., Guns, P.J., Metallo, J., Cammarata, D., Wilkin, F.,
Boeynams, J.M., Bult, H., and Robaye, B. (2008). Knockout
mice reveal a role for P2Y6 receptor in macrophages, endo-
thelial cells, and vascular smooth muscle cells. Mol Phar-
macol 74, 777–784.
8 WANG ET AL.
Bertero, T., Gastaldi, C., Bourgetponzio, I., Imbert, V., Loubat,
A., Selva, E., Busca, R., Mari, B., Hofman, P., and Barbry, P.
(2011). miR-483-3p controls proliferation in wounded epi-
thelial cells. FASEB J 25, 3092–3105.
Bihl, J.C., Zhang, C., Zhao, Y., Xiao, X., Ma, X., Chen, Y., Chen, S.,
and Zhao, B. (2015). Angiotensin-(1-7) counteracts the effects of
AngIIonvascularsmoothmusclecells, vascular remodeling and
hemorrhagic stroke: Role of the NFsmall ka, CyrillicB inflam-
matory pathway. Vascul Pharmacol 73, 115–123.
Chen, Y., Wang, X., Cheng, J., Wang, Z., Jiang, T., Hou, N.,
Liu, N., Song, T., and Huang, C. (2016). MicroRNA-20a-5p
targets RUNX3 to regulate proliferation and migration of
human hepatocellular cancer cells. Oncol Rep 36, 3379–3386.
Eskildsen, T.V., Jeppesen, P.L., Schneider, M., Nossent, A.Y.,
Sandberg, M.B., Hansen, P.B., Jensen, C.H., Hansen, M.L.,
Marcussen, N., and Rasmussen, L.M., et al. (2013). Angio-
tensin II regulates microRNA-132/-212 in hypertensive rats
and humans. Int J Mol Sci 14, 11190–11207.
Fan, S.J., Li, H.B., Cui, G., Kong, X.L., Sun, L.L., Zhao, Y.Q.,
Li, Y.H., and Zhou, J. (2015). miRNA-149* promotes cell
proliferation and suppresses apoptosis by mediating JunB in
T-cell acute lymphoblastic leukemia. Leuk Resh 41, 62–70.
Guo, W., Hanying, M.A., Zhao, T., Sijing, W.U., Hou, W., Yinhua,
L.I., Dong, F., Zhao, Y., Zhou, Y., and Zhang, W. (2014). The
expression of AT_1R,TGF-b1 and ERK1/2 on pulmonary arte-
rial hypertension related right ventricular remodeling in
monocreotaline-induced rats. J Cardiovasc Pulm Dis 33, 296–299.
Jeppesen, P.L., Christensen, G.L., Schneider, M., Nossent, A.Y.,
Jensen, H.B., Andersen, D.C., Eskildsen, T., Gammeltoft, S.,
Hansen, J.L., and Sheikh, S.P. (2011). Angiotensin II type 1
receptor signalling regulates microRNA differentially in car-
diac fibroblasts and myocytes. Br J Pharmacol 164, 394–404.
Kim, S.G., Gao, Z.G., Soltysiak, K.A., Chang, T.S., Brodie, C.,
and Jacobson, K.A. (2003). P2Y6 nucleotide receptor activates
PKC to protect 1321n1 astrocytoma cells against tumor necrosis
factor-induced apoptosis. Cell Mol Neurobiol 23, 401–418.
Komuro, I., and Kudoh, S. (1999). [The role of angiotensin II in
the development of cardiovascular remodeling]. Nihon Rin-
sho 57, 1090–1095.
Lenz, G., Gottfried, C., Luo, Z., Avruch, J., Rodnight, R., Nie,
W.J., Kang, Y., and Neary, J.T. (2000). P(2Y) purinoceptor
subtypes recruit different mek activators in astrocytes. Br J
Pharmacol 129, 927–936.
Li, C., Liu, Y., Xie, Z., Lu, Q., and Luo, S. (2015). Stigmasterol
protects against Ang II-induced proliferation of the A7r5
aortic smooth muscle cell-line. Food Funct 6, 2266–2272.
Li, H., Wang, Y., Chen, L., Han, L., Li, L., He, H., Li, Y.,
Huang, N., Ren, H., and Pei, F. (2017). The role of MIF,
cyclinD1 and ERK in the development of pulmonary hyper-
tension in broilers. Avian Pathol 446, 202–208.
Li, S., Zhu, Y., Liang, Z., Wang, X., Meng, S., Xu, X., Xu, X.,
Wu, J., Ji, A., and Hu, Z. (2016). Up-regulation of p16 by
miR-877-3p inhibits proliferation of bladder cancer. Onco-
target 9, 51773–51783.
Martin, M.M., Lee, E.J., Buckenberger, J.A., Schmittgen, T.D.,
and Elton, T.S. (2006). MicroRNA-155 regulates human an-
giotensin II type 1 receptor expression in fibroblasts. J Biol
Chem 281, 18277–18284.
Nemecz, M., Alexandru, N., Tanko, G., and Georgescu, A.
(2016). Role of MicroRNA in endothelial dysfunction and
hypertension. Curr Hypertens Rep 18, 87.
Nishimura, A., Sunggip, C., Tozaki-Saitoh, H., Shimauchi, T.,
Numaga-Tomita, T., Hirano, K., Ide, T., Boeynaems, J.M.,
Kurose, H., Tsuda, M., et al. (2016). Purinergic P2Y6 receptors
heterodimerize with angiotensin AT1 receptors to promote
angiotensin II-induced hypertension. Sci Signal 9, ra7.
Pediani, J.D., McGrath, J.C., and Wilson, S.M. (1999). P2Y
receptor-mediated Ca2+signalling in cultured rat aortic
smooth muscle cells. Br J Pharmacol 126, 1660–1666.
Pueyo, M., and Michel, J.B. (1998). [Effects of angiotensin ii on
vascular remodeling]. Nephrologie 19, 443–449.
Riegel, A.K., Faigle, M., Zug, S., Rosenberger, P., Robaye, B.,
Boeynaems, J.M., Idzko, M., and Eltzschig, H.K. (2011).
Selective induction of endothelial P2Y6 nucleotide receptor
promotes vascular inflammation. Blood 117, 2548–2555.
Roberts, R.E. (2012). The extracellular signal-regulated kinase
(ERK) pathway: a potential therapeutic target in hyperten-
sion. J Exp Pharmacol 4, 77–83.
Shan, K., Jiang, Q., Wang, X.Q., Wang, Y.N., Yang, H., Yao,
M.D., Liu, C., Li, X.M., Yao, J., Liu, B., et al. (2016). Role of
long noncoding RNA-RNCR3 in atherosclerosis-related vas-
cular dysfunction. Cell Death Dis 7, e2248.
Sober, S., Laan, M., and Annilo, T. (2010). MicroRNAs miR-
124 and miR-135a are potential regulators of the mineralo-
corticoid receptor gene (NR3C2) expression. Biochem Bio-
phys Res Commun 391, 727–732.
Stachon,P.,Peikert,A.,Michel,N.A.,Hergeth,S.,Marchini,T.,
Wolf, D., Dufner, B., Hoppe, N., Ayata, C.K., Grimm, M., et al.
(2014). P2Y6 deficiency limits vascular inflammation and ath-
erosclerosis in mice. Arterioscler Thromb Vasc Biol 34, 2237–
2245.
Takemoto, M., and Egashira, K. (1999). [Vascular remodeling
and angiotensin II]. Nihon Rinsho 57, 1158–1163.
Touyz, R.M. (2005). Intracellular mechanisms involved in
vascular remodelling of resistance arteries in hypertension:
role of angiotensin II. Exp Physiol 90, 449–455.
Touyz, R.M., and Schiffrin, E.L. (1996). Tyrosine kinase sig-
naling pathways modulate angiotensin II-induced calcium
([Ca2+]i) transients in vascular smooth muscle cells. Hy-
pertension 27, 1097–1103.
Voelkel, N.F., and Tuder, R.M. (1997). Cellular and molecular
biology of vascular smooth muscle cells in pulmonary hy-
pertension. Pulm Pharmacol Ther 10, 231–241.
Wang, L., Karlsson, L., Moses, S., Hultgardh-Nilsson, A., An-
dersson, M., Borna, C., Gudbjartsson, T., Jern, S., and Er-
linge, D. (2002). P2 receptor expression profiles in human
vascular smooth muscle and endothelial cells. J Cardiovasc
Pharmacol 40, 841–853.
Zhang, D., Xiao, Y.F., Zhang, J.W., Xie, R., Hu, C.J., Tang, B.,
Wang, S.M., Wu, Y.Y., Hao, N.B., and Yang, S.M. (2015). miR-
1182 attenuates gastric cancer proliferation and metastasis by
targeting the open reading frame of hTERT. Cancer Lett 360, 151–
159.
Zhu, S.M., Chen, C.M., Jiang, Z.Y., Yuan, B., Ji, M., Wu, F.H., and
Jin, J. (2016). MicroRNA-185 inhibits cell proliferation and
epithelial-mesenchymal transition in hepatocellular carcinoma by
targeting Six2. Eur Rev Med Pharmacol Sci 20, 1712.
Address correspondence to:
Yuansheng Tan, MD
Hunan University of Chinese Medicine
Hanpu science and Technology Park
Changsha 410208
China
E-mail: tanyuansheng_hn@163.com
Received for publication December 14, 2016; received in
revised form February 6, 2017; accepted February 6, 2017.
MIR-185/P2Y6 AXIS REGULATES HAVSMCSGROWTH 9
... Stimulation of cardiomyocyte P2Y6R can promote the development of myocardial fibrosis [26]. It has been revealed that the purinergic receptor P2Y6 promotes Ang II-induced hypertension [27][28][29]. Pressure (such as hypertension) stimulate cardiomyocytes, UDP, ATP and other nucleotide released to the extracellular via Pnx channels, stimulate P2Y6R in cardiomyocytes, regulates the expression of fibrosis factors (such as CTGF and TGF-β) through activated Rho pathway by coupled with G12/13 protein, thereby promoting the expression of downstream Type 1 collagen, and promoting the production of myocardial fibrosis [26]. The release of nucleotides coupled to specific autocrine/paracrine activation of the P2Y6 uracil nucleotide receptor could impair tissue perfusion in cardiovascular diseases [9]. ...
... Whether P2Y6R is connected with the pathological mechanism of AD is still under debate. The pathologically features of AD are the occurrence of β-amyloid plaques, amyloid vascular lesions and neurofibrillary tangles [37], accompanied by inflammation, neuromal nutrition, pathological gliosis as well as nerve loss [27][28][29][30][31][32][33]. UDP released by degraded or damaged neurons, can activate P2Y6R, enhance chemotaxis, and then promote its phagocytosis [38]. ...
... A growing body of evidence suggests that aberrant vascular smooth muscle cells (VSMCs) proliferation, migration and apoptosis are major pathological phenomena in the development of hypertension [6,7]. Angiotensin (Ang) II, a vasoactive peptide in the renin-angiotensin system, constricts blood vessels and increases blood pressure through its structural and functional effects, thereby provoking the pathological development of hypertension via increasing blood pressure [8]. ...
Article
Full-text available
Background Hypertension is known as a major factor for global mortality. We aimed to investigate the role of Cullin3 (CUL3) in the regulation of hypertension. Material and methods Human vascular smooth muscle cells (VSMCs) were treated with Angiotensin II (Ang II) to establish a hypertension in vitro model. Cell viability was detected by a cell counting kit-8 (CCK-8) assay. The content of reactive oxygen species (ROS) was evaluated by kit. Transwell assay and TUNEL staining were, respectively, used to assess cell migration and apoptosis. Additionally, the expression of sonic hedgehog (SHH) signaling-related proteins (SHH, smoothened homolog (Smo) and glioblastoma (Gli)) and CUL3 was tested with western blotting. Following treatment with Cyclopamine (Cycl), an inhibitor of SHH signaling, in Ang II-induced VSMCs, cell viability, migration, apoptosis and ROS content were determined again. Then, VSMCs were transfected with CUL3 plasmid or/and treated with sonic hedgehog signaling agonist (SAG) to explore the impacts on Ang II-induced VSMCs damage. In vivo, a hypertensive mouse model was established. Systolic blood pressure and diastolic blood pressure were determined. The histopathologic changes of abdominal aortic tissues were examined using H&E staining. The expression of SHH, Smo, Gli and CUL3 was tested with western blotting. Results Significantly increased proliferation, migration and apoptosis of VSMCs were observed after Ang II exposure. Moreover, Ang II induced upregulated SHH, Smo and Gli expression, whereas limited increase in CUL3 expression was observed. The content of ROS in Ang II-stimulated VSMCs presented the same results. Following Cycl treatment, the high levels of proliferation and migration in Ang II-treated VSMCs were notably remedied while the apoptosis and ROS concentration were further increased. Moreover, Cycl downregulated SHH, Smo, Gli and CUL3 expression. Above-mentioned changes caused by Ang II were reversed following SAG addition. Indeed, SAG treatment combined with restoration of CUL3 expression inhibited proliferation, migration, apoptosis and ROS level in Ang II-stimulated VSMCs. In vivo, SAG aggravated the histopathological changes of the aorta and with a worse tendency after both SAG intervention and CUL3 silencing. By contrast, SAG treatment and rebound in CUL3 expression alleviated the vascular damage. Conclusions Collectively, restoration of CUL3 gene expression protected against hypertension through enhancing the effects of SHH activation in inhibition of apoptosis and oxidative stress for hypertension and alleviating the dysfunction of VSMCs.
... While the plasma levels of miRNA-181b-5p were decreased, Ang II and HMGB1 levels are increased in hypertensive elderly (60.83 ± 1.43) patients [136••]. Ang II also induced HAVSMC proliferation, upregulation of P2Y6, an inflammation inducible G protein-coupled purinergic receptor, in addition to downregulating miRNA-185, which was shown to be capable of directly binding to the 3'UTR of P2Y6 and, in an overexpression condition, suppressing HAVSMC proliferation [137]. Together, these results pointed out that rescuing miRNA-181b-5p and miRNA-185 expression represents a therapeutic approach against HAVSMCs dysfunction in hypertension. ...
Article
Full-text available
Purpose of Review This review summarizes the involvement of inflammaging in vascular damage with focus on the epigenetic mechanisms by which inflammaging-induced hypertension is triggered. Recent findings Inflammaging in hypertension is a complex condition associated with the production of inflammatory mediators by the immune cells, enhancement of oxidative stress, and tissue remodeling in vascular smooth muscle cells and endothelial cells. Cellular processes are numerous, including inflammasome assembly and cell senescence which may involve mitochondrial dysfunction, autophagy, DNA damage response, dysbiosis, and many others. More recently, a series of noncoding RNAs, mainly microRNAs, have been described as possessing epigenetic actions on the regulation of inflammasome-related hypertension, emerging as a promising therapeutic strategy. Although there are a variety of pharmacological agents that effectively regulate inflammaging-related hypertension, a deeper understanding of the epigenetic events behind the control of vessel deterioration is needed for the treatment or even to prevent the disease onset.
... It was revealed that P2Y6 receptors promote Ang II-induced hypertension [56]. A miR-185/P2Y6 axis was found to inhibit Ang II-induced human aortic vascular smooth muscle cells (HAVSMCs) proliferation through miR-185, negatively regulating the P2Y6 receptor expression and the downstream ERK pathway [57]. Rescuing miR-185 expression to inhibit P2Y6 may represent a therapeutic strategy against HAVSMC dysfunction and hypertension. ...
Article
Full-text available
Both microRNAs (miRNAs) and purinergic signalling are widely and respectively expressed in various tissues of different organisms and play vital roles in a variety of physiological and pathological processes. Here, we reviewed the current publications contributed to the relationship of miRNAs and purinergic signalling in cardiovascular diseases, gastrointestinal diseases, neurological diseases, and ophthalmic diseases. We tried to decode the miRNAs-purinergic signalling network of purinergic signalling involved diseases. The evidence indicated that more than 30 miRNAs (miR-22, miR-30, miR-146, miR-150, miR-155, miR-187, etc.) directly or indirectly modulate P1 receptors (A1, A2A, A2B, A3), P2 receptors (P2X1, P2X3, P2X4, P2X7, P2Y2, P2Y6, P2Y12), and ecto-enzymes (CD39, CD73, ADA2); P2X7 and CD73 could be modulated by multiple miRNAs (P2X7: miR-21, miR-22, miR-30, miR-135a, miR-150, miR-186, miR-187, miR-216b; CD73: miR-141, miR-101, miR-193b, miR-340, miR-187, miR-30, miR-422a); miR-187 would be the common miRNA to modulate P2X7 and CD73.
... Angiogenesis is inhibited in human microvascular ECs by miR-185, through the targeting and down-regulation of stromal interaction molecule 1 (STIM1) (Hou et al., 2016). The negative regulation of P2Y6 by miR-185, and the downstream ERK pathway, inhibits angiotensin II-induced human aortic vascular SMC (VSMC) proliferation (Wang et al., 2017). Furthermore, circulating miR-185 was proposed to be a novel biomarker for clinical outcome in patients with dilated cardiomyopathy as high miR-185 levels were associated with a more favorable prognosis as a consequence of repressing B cell function (Yu et al., 2016). ...
Article
Full-text available
Dysfunction of endothelial cells (ECs) contributes to restenosis after vascular reconstruction for patients with coronary artery disease (CAD). The intercellular communication between ECs and vascular smooth muscle cells (VSMCs) might be critical in the development of restenosis and can be mediated by exosomes carrying functional microRNAs. miR-185 is reported to be associated with atherosclerosis, whether it plays a similar role in restenosis is unknown. In this study, we observed an elevated level of extracellular miR-185 in platelet-derived growth factor (PDGF)-stimulated VSMCs. The medium from PDGF-stimulated VSMCs promoted miR-185 expression in rat aortic ECs and inhibited EC angiogenesis. PDGF-stimulated VSMCs transferred miR-185 into ECs via exosomes. Furthermore, we found that the CXCL12 gene, a target of miR-185, is essential for the angiogenic potential of ECs. Exosomes derived from miR-185 mimic transfected VSMCs attenuated re-endothelialization after vascular injury. Moreover, we show that exosome-mediated miR-185 transfer is modulated by hnRNPA2B1. We also observed that hnRNPA2B1 is up-regulated during neointima formation and hnRNPA2B1 inhibition accelerates re-endothelialization and attenuates neointima formation following carotid injury. Taken together, our results indicate that exosomal miR-185 transfer from VSMCs to ECs is controlled by hnRNPA2B1 and impairs re-endothelialization after vascular injury.
Article
The kidney is an important target organ in the treatment of hypertension, but the effect of peptide QIGLF with antihypertensive activity on kidneys remains unknown. In the work, we aimed to further understand the hypotensive effects of QIGLF in spontaneously hypertensive rats (SHRs) using widely targeted metabolomics technology to investigate the kidney metabolic profiling variations. After four weeks of oral administration, the results showed different renal metabolomics profiles between QIGLF and model groups. Besides, a total of 10 potential biomarkers were identified, that is, 3-hydroxybutanoate, 20-hydroxyeicosatetraenoic acid, 19(S)-hydroxyeicosatetraenoic acid, 15-oxoETE, L-ornithine, malonate, uridine, uridine 5'-monophosphate, argininosuccinic acid, and N-carbamoyl-L-aspartate. These metabolites might exhibit antihypertensive activity of QIGLF by regulating synthesis and degradation of ketone bodies, arachidonic acid metabolism, pyrimidine metabolism, and arginine biosynthesis. These findings suggest that QIGLF might alleviate hypertension by inhibiting renal inflammation, promoting natriuresis, and regulating renal nitric oxide production.
Article
Objective Extracellular vesicles (EVs) are vital mediators of transferring microRNAs (miRNAs). We focused on effect of miR-185-3p that mediated by macrophages-derived EVs on atherosclerosis (AS) by targeting small mothers against decapentaplegic 7 (Smad7). Methods EVs were extracted from M1 macrophages and identified. ApoE-/- mice were treated with EVs, EVs containing miR-185-3p inhibitor or mimic, then the pathological changes of mouse aorta were observed. The levels of blood lipid, cell adhesion molecules, oxidative stress factors, inflammatory factors, and proliferation and apoptosis of vascular endothelial cells were assessed. Expression of miR-185-3p and Smad7 was detected and the targeting relationship between miR-185-3p and Smad7 was validated. Results MiR-185-3p was upregulated while Smad7 was downregulated in atherosclerotic mouse aorta. M1 macrophages-derived EVs elevated miR-185-3p to promote development of AS pathology and levels of blood lipid, endothelial cellular adhesion, oxidative stress factors and inflammatory factors, suppressed cell proliferation and promoted cell apoptosis of vascular endothelial cells in atherosclerotic mice through downregulating Smad7. Smad7 was a target gene of miR-185-3p and miR-185-3p could inhibit expression of Smad7. Conclusion M1 macrophages-derived EVs and upregulated miR-185-3p aggravated the development of AS in ApoE-/- mice by negatively regulating Smad7. This research may further the understanding of AS mechanism.
Article
Hypertension is a multifactorial disorder that involves complex genetic and environmental factors. Vascular smooth muscle cells (VSMCs) are important components of blood vessels, and their dysregulation has been shown to be involved in vascular remodeling during the development of systemic hypertension and pulmonary arterial hypertension (PAH) via multiple mechanisms, such as aberrant apoptosis, phenotype conversion, proliferation, and migration of VSMCs. With increasing advances in microarrays and next-generation sequencing, nonprotein-coding RNAs (ncRNAs) have attracted much attention due to their numerous functions in health and diseases. Among ncRNAs, microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs are emerging as novel modulators in the biological behaviors of VSMCs, especially in systemic hypertension and PAH. Studies have recommended miRNAs, lncRNAs, and circular RNAs as predictive biomarkers and therapeutic targets for systemic hypertension and PAH. In this review, we summarize the current studies focusing on the roles of VSMC-derived miRNAs, lncRNAs and circular RNAs in the pathologies of systemic hypertension and PAH. MiRNAs, lncRNAs, and circular RNAs might serve as attractive targets for the prevention and treatment of VSMC dysfunction-linked systemic hypertension and PAH.
Article
Aim: The co-expression network of long non-coding RNA ROR (lncRNA-ROR) and microRNA-185-3p (miR-185-3p) has not been focused on osteosarcoma. Therein, this work was initiated to uncover lncRNA-ROR and miR-185-3p functions in osteosarcoma. Methods: LncRNA-ROR, miR-185-3p and Yes-associated protein 1 (YAP1) expression in osteosarcoma tissues and cells were detected. The screened cells (MG63 and U2OS) were transfected with decreased and/or increased lncRNA-ROR and miR-185-3p to explore osteosarcoma progression. Tumor growth was detected by tumor xenografts in mice. Results: Up-regulated lncRNA-ROR and YAP1 and down-regulated miR-185-3p were found in osteosarcoma. LncRNA ROR knockdown or miR-185-3p overexpression inhibited osteosarcoma cell progression while lncRNA ROR elevation or miR-185-3p inhibition presented the opposite effects. Function of lncRNA ROR was rescued by miR-185-3p and regulated the growth and metastasis of osteosarcoma cells via modulating YAP1, the target gene of miR-185-3p. Conclusion: This work illustrates that lncRNA-ROR down-regulation or miR-185-3p up-regulation inhibits osteosarcoma progression via YAP1 repression.
Article
Full-text available
Background Extracellular vesicles (EVs) are produced during abnormal and normal physiological conditions. Understanding the expression profile of microRNA (miRNA) in plasma-derived small extracellular vesicles (sEVs) and their roles in subarachnoid hemorrhage (SAH) that cause cerebral vasospasm (CVS) is imperative.Methods Sprague Dawley rats (250–300 g) were allocated to sham or SAH groups established using endovascular perforation method. miRNA expression profiles of plasma sEVs in both groups (each n = 4) were evaluated using next-generation sequencing (NGS).ResultsThere were 142 microRNAs (miRNAs) significantly expressed differently between the two groups, of which 73 were up-regulated while 69 were down-regulated in SAH sEVs compared with those of sham (p < 0.05; fold change ≥ 2). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) analyses of differently expressed (DE) miRNAs revealed signaling pathways and target genes (TGs) in the SAH group. rno-miR-185-5p, rno-miR-103-3p, rno-miR-15b-3p, rno-miR-93-5p, and rno-miR-98-5p were the top five most up-regulated sEVs miRNAs.Conclusion Our results suggest that miRNA can be selectively packaged into sEVs under SAH, and this could help develop potential targets for the prevention, diagnosis, and treatment of CVS after this condition.
Article
Full-text available
Objective: Epithelial-mesenchymal transition (EMT) plays an important role in the malignant transformation of cancer. MicroRNAs are a group of small non-coding RNA molecules that down-regulate the expression of genes involved in tumorigenesis. Although microRNA-185 (miR-185) participates in the pathogenesis of several types of cancer, its relationship with EMT in human hepatocellular carcinoma (HCC) has not been investigated. The present study aims to elucidate the regulatory effects of miR-185 on EMT in HCC cells. Materials and methods: MTT and an in vitro wound-healing assay were performed to determine cell growth and metastasis potential, respectively. Real-time PCR was used to measure the mRNA expression of miR-185 and Six2. In addition, protein expression levels of Six2 and EMT-related markers were determined by western blot. Results: Our study showed that miR-185 was significantly down-regulated in HCC cells. Also, a luciferase reporter gene assay confirmed Six2 as a direct target of miR-185. Functional analyses indicated that miR-185 up-regulation remarkably suppressed cell growth and the metastatic potential of HCC cells. We also found that ectopic expression of miR-185 reversed EMT via the up-regulation of E-cadherin and down-regulation of vimentin in epithelial and mesenchymal HCC cells. Conclusions: miR-185 suppresses cell growth and EMT progression by targeting Six2, providing a novel target for the molecular treatment of liver malignancies.
Article
Full-text available
Purpose of review: Hypertension is either a cause or a consequence of the endothelial dysfunction and a major risk factor for cardiovascular disease (CVD). In vitro and in vivo studies established that microRNAs (miRNAs) are decisive for endothelial cell gene expression and function in various pathological conditions associated with CVD. This review provides an overview of the miRNA role in controlling the key connections between endothelial dysfunction and hypertension. Recent findings: Herein we summarize the present understanding of mechanisms underlying hypertension and its associated endothelial dysfunction as well as the miRNA role in endothelial cells with accent on the modulation of renin-angiotensin-aldosterone-system, nitric oxide, oxidative stress and on the control of vascular inflammation and angiogenesis in relation to endothelial dysfunction in hypertension. In particular, latest insights in the identification of endothelial-specific microRNAs and their targets are added to the understanding of miRNA significance in hypertension. This comprehensive knowledge of the role of miRNAs in endothelial dysfunction and hypertension and of molecular mechanisms proposed for miRNA actions may offer novel diagnostic biomarkers and therapeutic targets for controlling hypertension-associated endothelial dysfunction and other cardiovascular complications.
Article
Full-text available
Growing evidence indicates that some abnormally expressed microRNAs (miRNAs) influence tumorigenesis and progression. Previous studies reported that miR-20a is among the frequently altered miRNAs in human hepatocellular carcinoma (HCC), but its expression pattern and role in HCC remain controversial. In the present study, we demonstrated that miR-20a-5p exhibited aberrant expression in HCC tissues compared with paired non-tumor tissues: 52% of the tumor samples showed a greater increase. Overexpression of miR-20a contributed to HCC cell proliferation and migration in vitro, and treatment with anti-miR20a-5p caused the opposite effects. Further studies revealed RUNX3, an important tumor-suppressor, as a direct target of miR-20a-5p. We observed that the level of RUNX3 was sharply reduced in both mRNA and protein in HCC tissues compared with paired non-tumor tissues. Collectively, our results support the viewpoint that miR-20-5p has an oncogenic property, miR-20a overexpression contributed to HCC cell proliferation and migration through reducing the translation of RUNX3. The data provide a new mechanism of miR-20a regulating RUNX3 in HCC.
Article
Full-text available
Despite the recent studies which have shown that microRNA (miRNA) negatively regulates gene expression by silencing the expression of target genes, here we reported the new evidence of microRNA-mediated gene activation by targeting specific promoter sites. We identified a miR-877-3p binding site on the promoter site of tumor suppressor gene p16 which alters frequently in bladder cancer. Enforced expression of miR-877-3p could increase the expression of p16, which inhibit the proliferation and tumorigenicity of bladder cancer through cell cycle G1-phase arrest. Further evidences confirmed that the correlation between p16 activation and miR-877-3p was due to the direct binding. These findings demonstrate the anti-tumor function of miR-877-3p in bladder cancer cells and reveal a new pattern of miRNA involved gene regulation.
Article
Pulmonary hypertension (PH) is a major disease in the broiler breeding industry. During PH, the pulmonary artery undergoes remodelling, which is caused by pulmonary vascular smooth muscle cell proliferation. CyclinD1 regulates cell proliferation. This study investigated the role of cyclinD1 in the development of PH in broilers, and which bioactivators and signalling pathway are involved in the pathological process. The PH group contained 3–4 week-old broilers with clinical PH, and the healthy group broilers from the same flock without PH. Histopathology indicated pulmonary arterial walls were thicker in the PH group compared with the healthy group. Target gene expressions of MIF, ERK, and cyclinD1 detected by quantitative real-time PCR were up-regulated in the PH group compared with the healthy group. Immunohistochemistry showed MIF, p-ERK and cyclinD1 were present on pulmonary vascular walls; MIF was present in the cytoplasm of arterial endothelial cells and smooth muscle cells; p-ERK and cyclinD1 were present in smooth muscle cell cytoplasm. Western blotting demonstrated MIF, phosphorylated ERK and cyclinD1 levels were significantly higher (P<0.01) in the PH group compared with the healthy group. In summary, increased MIF in PH broiler pulmonary arteries upregulated cyclinD1 via the ERK signalling pathway to induce pulmonary vascular smooth muscle cell proliferation, causing pulmonary artery remodelling and hypertension.
Article
Atherosclerosis is one of the most common vascular disorders. Endothelial cell (EC) dysfunction and vascular smooth muscle cell (VSMC) proliferation contributes to the development of atherosclerosis. Long non-coding RNAs (lncRNAs) have been implicated in several biological processes and human diseases. Here we show that lncRNA-RNCR3 is expressed in ECs and VSMCs. RNCR3 expression is significantly upregulated in mouse and human aortic atherosclerotic lesions, and cultured ECs and VSMCs upon ox-LDL treatment in vitro. RNCR3 knockdown accelerates the development of atherosclerosis, aggravates hypercholesterolemia and inflammatory factor releases, and decreases EC and VSMC proliferation in vivo. RNCR3 knockdown also reduces the proliferation and migration, and accelerates apoptosis development of EC and VSMC in vitro. RNCR3 acts as a ceRNA, and forms a feedback loop with Kruppel-like factor 2 and miR-185-5p to regulate cell function. This study reveals that RNCR3 has an atheroprotective role in atherosclerosis, and its intervention is a promising strategy for treating atherosclerosis-related vascular dysfunction.
Article
The angiotensin (Ang) type 1 receptor (AT1R) promotes functional and structural integrity of the arterial wall to contribute to vascular homeostasis, but this receptor also promotes hypertension. In our investigation of how Ang II signals are converted by the AT1R from physiological to pathological outputs, we found that the purinergic P2Y6 receptor (P2Y6R), an inflammation-inducible G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor (GPCR), promoted Ang II-induced hypertension in mice. In mice, deletion of P2Y6R attenuated Ang II-induced increase in blood pressure, vascular remodeling, oxidative stress, and endothelial dysfunction. AT1R and P2Y6R formed stable heterodimers, which enhanced G protein-dependent vascular hypertrophy but reduced b-arrestin-dependent AT1R internalization. Pharmacological disruption of AT1R-P2Y6R heterodimers by the P2Y6R antagonist MRS2578 suppressed Ang II-induced hypertension in mice. Furthermore, P2Y6R abundance increased with age in vascularsmoothmuscle cells. The increased abundance of P2Y6R converted AT1R-stimulated signaling in vascular smooth muscle cells from β-arrestin-dependent proliferation to G protein- dependent hypertrophy. These results suggest that increased formation of AT1R-P2Y6R heterodimers with age may increase the likelihood of hypertension induced by Ang II.
Article
MicroRNA-149* (miRNA-149*) functions as an oncogenic regulator in human melanoma. However, the effect of miRNA-149* on T-cell acute lymphoblastic leukemia (T-ALL) is unclear. Here we aimed to analyze the effects of miRNA-149* on in vitro T-ALL cells and to uncover the target for miRNA-149* in these cells. The miRNA-149* level was determined in multiple cell lines and bone marrow cells derived from patients with T-ALL, B acute lymphoblastic leukemia (B-ALL), acute myelocytic leukemia (AML), and healthy donors. We found that miRNA-149* was highly expressed in T-ALL cell lines and T-ALL patients' bone marrow samples. JunB was identified as a direct target of miR-149*. miRNA-149* mimics downregulated JunB levels in Molt-4 and Jurkat cells, while miRNA-149* inhibitors dramatically upregulated JunB expression in these cells. miRNA-149* mimics promoted proliferation, decreased the proportion of cells in G1 phase, and reduced cell apoptosis in T-ALL cells, while miRNA-149* inhibitors prevented these effects. miRNA-149* mimics downregulated p21 and upregulated cyclinD1, 4EBP1, and p70s6k in Molt-4 and Jurkat cells. Again, inhibitors prevented these effects. Our findings demonstrate that miRNA-149* may serve as an oncogenic regulator in T-ALL by negatively regulating JunB.
Article
Angiotensin (Ang)-(1-7) is a potential vasoprotective peptide. In the present study, we investigated its counteractive effects to Ang II on vascular smooth muscle cells (VSMCs) and intracerebral hemorrhagic stroke (ICH) through inflammatory mechanism. In in vitro experiments, human brain VSMCs (HBVSMCs) were treated with vehicle, Ang II, Ang II+Ang-(1-7), Ang II+A-779 or Ang II+Ang-(1-7)+A-779 (Mas receptor antagonist). HBVSMC proliferation, migration and apoptosis were determined by methyl thiazolyltetrazolium, wounding healing assay and flow cytometry, respectively. In in vivo experiments,C57BL/6 mice were divided into vehicle, Ang II, Ang II+Ang-(1-7), Ang II+A-779 or Ang II+Ang-(1-7)+A-779 groups before they were subjected to collagenase-induced ICH or sham surgery. Hemorrhage volume and middle cerebral artery (MCA) remodeling were determined by histological analyses. Levels of NFκB, Inhibitor of κBα (IκBα), tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein 1 (MCP-1) and interleukin (IL-8) were measured by western blot or ELISA. We found that 1) Ang II increased HBVSMC migration, proliferation and apoptosis, and increased the blood pressure (BP), neurological deficit score, MCA remodeling and hemorrhage volume in ICH mice. 2) Ang-(1-7) counteracted these effects of Ang II, which was independent of BP, with the down-regulation of NFκB, up-regulation of IκBα, and decreased levels of TNF-α, MCP-1 and IL-8. 3) The beneficial effects of Ang-(1-7) could be abolished by A-779. In conclusion, Ang-(1-7) counteracts the effects of Ang II on ICH via modulating NFκB inflammation pathway in HBVSMCs and cerebral microvessels. Copyright © 2015. Published by Elsevier Inc.
Article
Excessive proliferation of vascular smooth muscle cells is a crucial event in the pathogenesis of several cardiovascular diseases, including atherosclerosis and restenosis. In this study, we reported that stigmasterol was effective in inhibiting vascular cell proliferation exemplified using A7r5 cells stimulated by Ang II. Mechanism analysis showed that this inhibiting effect is mainly occurred by cell-cycle arrest and promotion of apoptosis. In addition, stigmasterol inhibition effects were detected to associate with decreased ROS production, enhanced SOD and CAT activity, decreased abundance of cyclin A, CDK2, PCNA, bax and bcl-2, and increased levels of p53 protein. Our study provided implications for the development of therapeutic strategies to protect against certain cardiovascular pathologies, such as atherosclerosis and restenosis.