Invest Clin 62(4): 295 - 306, 2021 https://doi.org/10.22209/IC.v62n4a01
Corresponding author: Yong Tang. Department of Cardiology, Affiliated Nanjing Hospital of Nanjing University of
Chinese Medicine, Nanjing, China. Email: tangyong86@126.com
Thymosin β4 regulates endothelial cell
function via activating the AKT pathway.
Yong Tang1, Hao Dong1, Wenbin Lu2, Xiaofeng Zhang1, Xiao Shen1 and Peizhe Zhang1
1Department of Cardiology, Affiliated Nanjing Hospital of Nanjing University
of Chinese Medicine, Nanjing, China.
2Department of Cardiology, Zhongda Hospital Affiliated to Southeast University,
Zhongda, China.
Key words: thymosin β4; HUVECs; proliferation; migration; apoptosis; AKT signaling.
Abstract. The vascular eendothelial cells are highly heterogeneous and
associated with numerous diseases. Thymosin β4 (Tβ4) plays pleiotropic roles
in endothelial cell differentiation, migration and angiogenesis. However, the
underlying mechanisms played by Tβ4 in the regulation of endothelial cells
have not yet been well investigated. In the present study, Tβ4 -GFP adenovirus,
transfected into human umbilical vein endothelial cells (HUVECs), and cell
morphology were analyzed by fluorescence microscopy. ELISA was used to de-
termine the concentration of Tβ4 expression. Furthermore, the effects of Tβ4
overexpression on HUVECs proliferation, apoptosis and migration were investi-
gated. Real-time quantitative PCR and western blot were conducted to examine
mRNA and protein expression in HUVECs with Tβ4 overexpression. Moreover,
the underlying molecular mechanism of Tβ4 in HUVECs function was tested
through treatment with LY294002, a PI3K/AKT inhibitor. Overexpression of
Tβ4 increased the cell ability of HUVECs, and up-regulated the expression of
the proliferation markers PCNA and Cyclin D1. In addition, overexpression of
Tβ4 reduced HUVECs apoptosis, both under normoxic and hypoxic conditions.
Moreover, overexpression of Tβ4 increased the ability of HUVECs to migrate
through the membrane and up-regulated levels of MMP-2 and MMP-9. The use of
LY294002 decreased the p-AKT (Ser473) level, which was induced by Tβ4 over-
expression. Importantly, LY294002 reduced Tβ4-induced HUVECs proliferation
and migration. In conclusion, our results suggest that Tβ4 is a major regulator
of HUVECs function by activating the AKT signaling pathway.
296 Tang et al.
Investigación Clínica 62(4): 2021
La timosina β4 regula la función de las células endoteliales,
activando la vía AKT.
Invest Clin 2021; 62 (4): 295-306
Palabras clave: timosina β4; HUVEC; proliferación; migración; apoptosis; señalización
de AKT.
Resumen. Las células endoteliales vasculares son muy heterogéneas y es-
tán asociadas con numerosas enfermedades. La Tβ4 desempeña papeles pleio-
trópicos en la diferenciación, migración y angiogénesis de células endoteliales.
Sin embargo, los mecanismos fundamentales que realiza la Tβ4 en la regulación
de las células endoteliales aún no han sido bien investigados. En el presente es-
tudio se analizaron el adenovirus Tβ4 –GFP transfectado en las células endote-
liales de vena umbilical humana (HUVEC) y la morfología celular, mediante mi-
croscopía de fluorescencia. ELISA se utilizó para determinar la concentración
de la expresión de Tβ4. También, se investigaron los efectos de la sobrexpresión
de la Tβ4, sobre la proliferación de las HUVEC, la apoptosis y la migración. Se
realizaron la PCR cuantitativa en tiempo real y el Western blot para examinar
el mRNA y la expresión de proteínas en las HUVEC con sobreexpresión de Tβ4.
Además, se probó el mecanismo molecular subyacente de Tβ4 sobre la función
de las HUVEC mediante el tratamiento con LY294002, un inhibidor de la PI3K/
AKT. La sobreexpresión de Tβ4 aumentó la capacidad celular de las HUVEC, re-
gulando al alza la expresión de los marcadores de proliferación PCNA y Ciclina
D1. Además, la sobreexpresión de Tβ4 redujo la apoptosis de las HUVEC, tanto
en condiciones normóxicas como hipóxicas. Por otra parte, la sobreexpresión
de Tβ4 aumentó la capacidad de las HUVEC a migrar a través de la membrana
y reguló hacia arriba los niveles de MMP-2 y MMP-9. El uso del LY294002 dis-
minuyó el nivel de p-AKT (Ser473), que fue inducido por la sobreexpresión de
Tβ4. Es importante destacar que LY294002 bajó la proliferación y la migración
de las HUVEC inducidas por Tβ4. En conclusión, nuestros resultados sugieren
que Tβ4 es un regulador principal de la función de las HUVEC mediante la acti-
vación de la vía de señalización de AKT.
Received: 06-04-2021 Accepted: 30-05-2021
INTRODUCTION
The vascular endothelial cells are highly
heterogeneous and associated with numer-
ous diseases, such as atherosclerosis (1).
Atherosclerosis causes stroke and coronary
heart disease (2), aging induced cardiovas-
cular disease (3), diabetes mellitus (4), vas-
cular injuries induced by limb ischemia and
reperfusion (5), etc. However, the underly-
ing mechanism of endothelial cell prolifera-
tion, apoptosis and migration has not been
fully elucidated.
Thymosin beta 4 (Tβ4) was first isolated
from calf thymus and it is a highly conserved
G-actin-sequestering peptide (6). It has been
Thymosin β4 regulates endothelial cell function through AKT pathway 297
Vol. 62(4): 295 - 306, 2021
reported that Tβ4 is expressed in various cell
types, such as endothelial cells, and plays
pleiotropic roles in endothelial cell differen-
tiation, migration, and angiogenesis (7-9).
Smart et al. (10) found that Tβ4 knockdown
reduced the coronary vasculogenesis and an-
giogenesis by a significant reduction in the
pro-angiogenic cleavage product N-acetyl-
seryl-aspartyl-lysyl-proline (AcSDKP) in mice
heart. Tβ4, which was secreted from the
myocardium, promoted epicardium-derived
cells inward migration and differentiation
into endothelial cells, and further to form
the coronary vasculature (11). Tβ4 could
activate mast cells to produce angiogenesis
associated factors, such as VEGF, and stimu-
late endothelial cell migration and differen-
tiation (12). Although the pieces of evidence
mentioned above demonstrated benecial
roles of Tβ4 for cardiac disease treatment by
regulating endothelial cell function, the un-
derlying mechanisms played by Tβ4 have not
yet been well considered. In this study, we
aimed to investigate the role of Tβ4 in the
regulation of HUVECs proliferation, apopto-
sis, and migration through the AKT signal-
ing pathway.
MATERIALS AND METHODS
Cell culture
Human HUVECs were obtained from
Nanjing KeyGen Biotech Co. Ltd. (Nanjing,
China) and cultured in RPMI-1640 (Gibco,
Grand Island, NY, USA) supplemented with
10% fetal bovine serum (FBS; Gibco, Grand
Island, NY, USA), streptomycin and penicillin
(Nanjing KeyGen Biotech Co. Ltd.) at 37°C
under a humidied 5% CO2 atmosphere.
The hypoxic condition was made in a sealed
chamber with a gas mixture containing 5%
CO2, 92% N2, and 3% O2.
Cell transfection
TB4-GFP adenovirus (Ad-Tβ4) and its
negative control (Ad-NC) were obtained
from GenePharma Co., Ltd. (Shanghai,
China). Logarithmic growth phase HUVECs
were seeded into 6-well plates (8×103 cells/
well) and then infected with Ad-Tβ4 or Ad-NC
at an MOI of 25. The efciency of infection
was assessed by green fluorescence protein
(GFP), and expression of GFP in HUVECs
was confirmed by uorescence microscopy.
The enzyme-linked immunosorbent
assay (ELISA)
The concentration of Tβ4 in superna-
tant fluid of cultured HUVECs were analyzed
by a human Tβ4 ELISA kit (Jingkang Bio-
engineering Co., Ltd., Shanghai, China) ac-
cording to the manufacturer’s instructions.
Cell proliferation assay
To investigate the effect of Tβ4 up-reg-
ulation on HUVECs, an MTT assay was de-
signed. In brief, 5×104 HUVECs were seeded
into 96-well plates and were infected with
Ad-T β4 or Ad-NC for 24 h. Then 20 μL of 5
mg/mL [3-(4,5-dimethylthiazol-2-yl)-2,5-
diphenyl tetrazolium bromide (MTT)] (Am-
resco, Washington, USA) in PBS was added
and incubated at 37˚C for 4 h. Subsequently,
150 μL dimethylsulfoxide (DMSO; Sigma,
USA) was added to each well to dissolve the
formazan product. The absorbance was mea-
sured at 490 nm using a microplate reader
(Bio-Rad, Hercules, CA, USA).
Cell apoptosis assay
To explore the effect of Tβ4 on HUVECs
apoptosis, 1×106 HUVECs cells were collect-
ed, washed with PBS, and resuspended in 100
μL binding buffer, containing 5 μL Annexin V-
APC and 5 μL 7-AAD (BD Biosciences, Frank-
lin Lakes, NJ, USA) at room temperature.
Then, the cell apoptosis was tested using a
flow cytometer (BD FACSCalibur, BD Biosci-
ences, CA, USA). Four subpopulations were
divided in figure from flow-cytometric analy-
sis: normal cells (Annexin V-APC−/7-AAD−),
necrotic cells (Annexin V-APC−/7-AAD+),
early apoptotic (Annexin V-APC+/7-AAD−)
and late apoptotic (Annexin V-APC+/7-
AAD+). Apoptosis index was the total rates
of early apoptotic and late apoptotic cells.
298 Tang et al.
Investigación Clínica 62(4): 2021
Cell migration assay
For cell migration assay, the Transwell
cell culture chambers with 8-μm pore polycar-
bonate membrane filters (Millipore, Billerica,
MA, USA) were used. Approximately 5×105
HUVECs, which mixed in FBS-free RPMI-1640,
were seeded into the upper chamber, whereas
the lower chamber was filled with RPMI-1640
containing 5% FBS. After 24 h, non-migrated
cells from the upper chamber were removed;
and migrated cells to the bottom side of the
membrane were fixed with 90% alcohol and
stained with crystal violet. The migrated cells
were counted under a light microscope with
200-fold magnification.
RNA isolation and real-time quantitative
PCR
Total RNA from cultured HUVECs was
extracted using a miRNeasy kit (Qiagen,
Hilden, Germany) according to the manu-
facturer’s protocol. Total RNA (1 μg) was
reverse transcribed into cDNA by MuLV re-
verse transcriptase (NEB, USA). Real-time
quantitative PCR (RT-qPCR) analysis was
performed using Fast SYBR Green (Applied
Biosystems, Forster City, CA, USA). Details
of the specific RT-qPCR primers to deter-
mine relative levels of gene expression are
shown in Table I.
Western Blot
HUVECs were lysed with the RIPA ly-
sis buffer (Sigma, USA) according to the
manufacturer’s instruction. Equal protein
was separated by using the 10% sodium
dodecyl sulfate-polyacrylamide gel electro-
phoresis (SDS-PAGE) and transferred onto
a polyvinylidene difluoride (PVDF) mem-
brane (Millipore, MA, USA). The membranes
were blocked with 5% skim milk, and probed
overnight at 4°C using the following pri-
mary antibodies: anti-PCNA (Abcam, MA,
USA), anti-Cyclin D1 (Abcam, MA, USA),
anti-MMP-2 (Cell Signaling Technology, Bev-
erly, MA, USA), anti-MMP-9 (Cell Signaling
Technology, Beverly, MA, USA), anti-cleaved
caspase-3 (Abcam, MA, USA), anti-AKT (Cell
Signaling Technology, Beverly, MA, USA), an-
ti-p-AKT (Ser473) (Cell Signaling Technolo-
gy, Beverly, MA, USA), and GAPDH (Abcam,
MA, USA). Then, the membrane was incu-
bated with HRP-labelled secondary antibody
(Abcam, MA, USA) and visualized using an
enhanced chemiluminescence system (GE
Healthcare, Piscataway, NJ, USA).
Statistical analysis
The data in this study were analyzed by
Graphpad Prism 5.0 software and displayed
as mean ± standard deviation (SD). The Stu-
TABLE I
PRIMER SEQUENCES USED IN RT-qPCR EXPERIMENTS.
Gene name Primer sequence (5’ 3’)
PCNA Forward: CCTGCTGGGATATTAGCTCCA
Reverse: CAGCGGTAGGTGTCGAAGC
Cyclin D1 Forward: GCTGCGAAGTGGAAACCATC
Reverse: CCTCCTTCTGCACACATTTGAA
MMP-2 Forward: TGACTTTCTTGGATCGGGTCG
Reverse: AAGCACCACATCAGATGACTG
MMP-9 Forward: TGTACCGCTATGGTTACACTCG
Reverse: GGCAGGGACAGTTGCTTCT
GAPDH Forward: GGAGCGAGATCCCTCCAAAAT
Reverse: GGCTGTTGTCATACTTCTCATGG
Thymosin β4 regulates endothelial cell function through AKT pathway 299
Vol. 62(4): 295 - 306, 2021
dent’s t-test was used to compare the means
between the two groups. A value of p<0.05
was considered to indicate a statistically sig-
nificant difference.
RESULTS
Tβ4 overexpression promotes HUVECs
proliferation
To explore the role of Tβ4 in the reg-
ulation of HUVECs function, we infected
HUVECs with Ad-Tβ4 or Ad-NC (Fig. 1A).
An ELISA analysis was conducted to verify
the transfection efficiency, and the results
showed that Ad-Tβ4 significantly elevated
Tβ4 expression in HUVECs (Fig. 1B).
The overexpression of Tβ4 significantly
promoted HUVECs growth (Fig. 2A). More-
over, RT-qPCR and western blot analysis indi-
cated that the mRNA and protein expression
of PCNA and Cyclin D1, which are cell pro-
liferation markers, significantly increased in
HUVECs infected with Ad-Tβ4 (Fig. 2B-D).
These data confirmed that Tβ4 promoted
the growth of HUVECs.
Tβ4 overexpression reduces HUVECs
apoptosis
To investigate whether Tβ4 could affect
HUVECs apoptosis, HUVECs were stained
with Annexin V, and followed by flow cytom-
etry. As shown in Fig. 3A, HUVECs infected
with Ad-Tβ4 showed less rate of cell apopto-
sis compared with cells infected with Ad-NC.
The level of cleaved caspase-3 in HUVECs
infected with Ad-Tβ4 was down-regulated
when compared with HUVECs infected with
Ad-NC (Fig. 3B). Also, the effect of Tβ4 on
HUVECs apoptosis under hypoxic conditions
was examined by flow cytometry. The apop-
tosis results showed that hypoxia significant-
ly increased the rate of HUVECs apoptosis,
whereas overexpression of Tβ4 decreased
HUVECs apoptosis after exposure to hypoxic
conditions (Fig. 3C). Moreover, overexpres-
sion of Tβ4 reduced the level of cleaved cas-
Fig.1. The level of Tβ4 was increased in HUVECs infected with Ad-Tβ4. (A) Morphology of HUVECs observed
under a fluorescence microscope upper panel, bright field, 200×; lower panel, fluorescence field,
200×). (B) Tβ4 concentration determined by ELISA. Data are presented as means ± S.D. from 3
independent experiments. *p<0.05.
300 Tang et al.
Investigación Clínica 62(4): 2021
pase-3 in HUVECs after exposure to hypoxic
conditions (Fig. 3D). Our data indicated that
Tβ4 could inhibit HUVECs apoptosis both
under normoxic and hypoxic conditions.
Tβ4 overexpression promotes HUVECs
migration
To demonstrate the role of Tβ4 in HU-
VECs migration, a Transwell migration assay
was conducted. As shown in Fig.4.A, overex-
pression of Tβ4 increased the ability of HU-
VECs to migrate through the membrane.
MMPs are the main proteolytic enzymes that
contribute to the degradation of the extra-
cellular matrix and serve critical roles in the
invasion process (13). RT-qPCR determined
that Tβ4 overexpression up-regulated the
mRNA and protein levels of MMP-2 and MMP-
Fig. 2. Tβ4 overexpression promoted HUVECs proliferation. (A) CCK-8 analysis of the cell proliferation in
HUVECs infected with Ad-NC or Ad-Tβ4. (B, C) The relative mRNA level of PCNA (B) and Cyclin D (C)
in HUVECs infected with Ad-NC or Ad-Tβ4. (D) The protein level of PCNA and Cyclin D in HUVECs
infected with Ad-NC or Ad-Tβ4. Data are presented as means ± S.D. from 3 independent experiments.
*p<0.05, **p<0.01.
Thymosin β4 regulates endothelial cell function through AKT pathway 301
Vol. 62(4): 295 - 306, 2021
9 (Fig. 4B and 4C). These data confirmed
that Tβ4 remarkably promoted the HUVECs
migration.
Tβ4 overexpression activates AKT
signaling pathway
It has been reported that Tβ4 could
regulate the migratory and proliferative ac-
tivity of high glucose-treated HUVECs by ac-
tivation of the AKT signaling pathway (14).
Therefore, we hypothesized that overexpres-
sion of Tβ4 might regulate HUVECs function
via the AKT signaling pathway. As shown in
Fig. 5, western blot analysis indicated that
the p-AKT (Ser473) level in HUVECs infect-
ed with Ad-Tβ4 was significantly increased.
To evaluate the possibility that Tβ4 exerts
its function through the AKT pathway, HU-
VECs were treated with LY294002, a PI3K/
AKT inhibitor (15-18). After exposure to
LY294002 (50 μmol/L (19)), the expression
of the p-AKT (Ser473) level was significantly
decreased, which was induced by Tβ4 overex-
pression (Fig. 5).
Inhibition of AKT signaling reduces
Tβ4-induced HUVECs proliferation and
migration
To demonstrate that the AKT signaling
regulated Tβ4-induced proliferation and mi-
gration, the Ad-Tβ4 infected HUVECs were
treated with LY294002. As shown in Fig.
6A, LY294002 reduced the cell numbers of
HUVECs induced by Tβ4 overexpression.
Fig. 3. Tβ4 overexpression reduced HUVECs apoptosis. (A) Scatter diagram and Histogram of apoptosis in
HUVECs infected with Ad-NC or Ad-Tβ4. (B) The protein level of cleaved caspase-3 in HUVECs infec-
ted with Ad-NC or Ad-Tβ4. (C) Scatter diagram and Histogram of apoptosis in HUVECs infected with
Ad-NC or Ad-Tβ4 under hypoxic conditions. (D) The protein level of cleaved caspase-3 in HUVECs
infected with Ad-NC or Ad-Tβ4 under hypoxic conditions. Data are presented as means ± S.D. from 3
independent experiments. *p<0.05.
302 Tang et al.
Investigación Clínica 62(4): 2021
Fig. 4. Tβ4 overexpression promoted HUVECs migration. (A) The cell migratory ability was investigated when
HUVECs were infected with Ad-NC or Ad-Tβ4 (200x). (B) The relative mRNA level of MMP-2 and MMP-
9 in HUVECs infected with Ad-NC or Ad-Tβ4. (C) The protein level of MMP-2 and MMP-9 in HUVECs
infected with Ad-NC or Ad-Tβ4. Data are presented as means ± S.D. from 3 independent experiments.
*p<0.05.
Fig. 5. Tβ4 enhanced AKT signaling. (A) The protein level of p-AKT (Ser473) and AKT in HUVECs infected
with Ad-NC or Ad-Tβ4. (B) The density of the western blots bands shown in (A) was quantified using
ImageJ software. Data are presented as means ± S.D. from 3 independent experiments. *p<0.05.
Thymosin β4 regulates endothelial cell function through AKT pathway 303
Vol. 62(4): 295 - 306, 2021
LY294002 enhanced the apoptosis rate of
HUVECs, which was decreased by Tβ4 overex-
pression (Fig. 6. B). Furthermore, LY294002
could reverse the Tβ4 overexpression-medi-
ated increase in the ability of HUVECs to mi-
grate through the membrane (Fig. 6C).
DISCUSSION
The novelty of our findings was as fol-
lows: (1) gene expression modalities of Tβ4
alongside its mechanism of actions in endo-
thelial cells, which clarifies the Tβ4 function
in biomolecular levels, (2) the role of Tβ4 in
endothelial cells under both, normoxic and
hypoxic conditions.
Vascular endothelial cells, which cover
the intima of the vascular wall, play an es-
sential role in maintaining vascular wall ten-
sion, repairing vascular wall inflammation
and promoting vascular proliferation, by
secreting a variety of vasoactive substances
(20,21). The dysfunction of vascular endo-
thelial cells plays a central role in the patho-
genesis of vascular-associated diseases (20).
Therefore, controlling endothelial cell func-
tion may be a potential novel therapeutic
strategy against vascular-associated diseas-
es. The results of this study conrmed that
Tβ4 plays an important role in the regula-
tion of HUVECs proliferation, apoptosis, and
migration. According to the above findings,
this study revealed the beneficial effects of
Tβ4 on endothelial cells, which may provide
novel insights into the potential application
of Tβ4 for vascular protection and therapy in
Fig. 6. Tβ4 regulated HUVECs proliferation and migration via AKT signaling (A) Tβ4-induced HUVECs proli-
feration was abolished by the AKT signaling inhibitor Ly294002. (B) Tβ4-reduced HUVECs apoptosis
was abolished by the AKT signaling inhibitor Ly294002. (C) Tβ4-induced HUVECs migration was
abolished by the AKT signaling inhibitor Ly294002. Data are presented as means ± S.D. from 3 inde-
pendent experiments. *p<0.05.
304 Tang et al.
Investigación Clínica 62(4): 2021
vascular-related diseases, including cardio-
vascular, cerebrovascular, coronary artery,
and diabetes diseases.
Tβ4 has been shown to be secreted from
embryonic endothelial progenitor cells, en-
dothelial cells, and cardiomyocytes (10,22),
suggesting that it may play an important
role in endothelial cell function. It has been
demonstrated that HUVECs transfected with
Tβ4 increased the rate of tube formation on
Matrigel while silencing of Tβ4 abrogated
tube formation (23). The addition of exog-
enous Tβ4 can enhance vascular sprouting
in cultured HUVECs by inducing several bio-
logical responses (24). Ho et al. found that
exogenous Tβ4 protected bovine corneal
endothelial cells from low-dose ultraviolet-
induced oxidative stress and apoptosis (25).
Tβ4 also mediated the inhibitory effect on
endothelial progenitor cells apoptosis in-
duced by serum deprivation (26). Qiu et al.
(27) reported that Tβ4 could induce circu-
lating endothelial progenitor cell directional
migration, which is essential for re-endothe-
lialization and neovascularization. Moreover,
Tβ4 could increase telomerase activity and
inhibit the senescence of endothelial pro-
genitor cells (28). In our study, we used Tβ4-
expressing adenovirus to demonstrate the
effect of Tβ4 on HUVECs proliferation, apop-
tosis, and migration. Our results showed
that Tβ4 overexpression promoted HUVECs
proliferation and migration. Furthermore,
Tβ4 overexpression could effectively reduce
the HUVECs apoptosis under normoxic and
hypoxic conditions. Hence, both previous
studies and our results indicate that Tβ4
plays an important role in the function of
HUVECs.
It has been investigated that the molec-
ular mechanism of Tβ4-regulating endothe-
lial cell function involved various known reg-
ulatory pathways (7). It has been reported
that Tβ4 could promote the migration and
proliferation of embryonic endothelial cells
via activating protein kinase C (PKC) activ-
ity (29). Moreover, Tβ4 induced the migra-
tion of endothelial progenitor cells via the
PI3K/AKT/eNOS signaling pathway (14). Lv
et al. found that Tβ4 significantly reduced
VE-cadherin expression levels in HUVECs
through the Notch signaling pathway (30).
In high-glucose-exposed vascular endothelial
cells, Tβ4 protects against hyperglycemia-in-
duced damage of endothelial cells via up-reg-
ulating the expression of insulin-like growth
factor-1 (IGF-1) (31). In the present study,
overexpression of Tβ4 significantly increased
the p-AKT (Ser473) level in HUVECs, while
LY294002, a PI3K/AKT inhibitor, decreased
the p-AKT (Ser473) level, which was induced
by Tβ4 overexpression. LY294002 reduced
Tβ4-induced HUVECs proliferation and mi-
gration. Thus, Tβ4 could regulate the cell
function of HUVECs via the AKT signaling
pathway. Except for AKT signaling pathway,
we couldn’t rule out the possibility that Tβ4
regulated HUVECs function by an alterna-
tive signaling pathway.
In conclusion, the results of the pres-
ent study indicate that Tβ4 is a major reg-
ulator of HUVECs function. Tβ4 increased
proliferation and migration, while reduced
apoptosis of HUVECs. The underline mecha-
nism is the increase of p-AKT (Ser473) level
by Tβ4, which is demonstrated by the PI3K/
AKT inhibitor LY294002. These results pro-
vide novel insights into the role of Tβ4 in the
pathogenesis of vascular-associated diseases.
ACKNOWLEDGMENTS
The present study was supported by the
Special Fund for Medical Science Develop-
ment of Health Committee of Nanjing (No.
YKK18152).
REFERENCES
1. Wu X, Zheng X, Cheng J, Zhang K, Ma C.
LncRNA TUG1 regulates proliferation and
apoptosis by regulating miR-148b/IGF2
axis in ox-LDL-stimulated VSMC and HU-
VEC. Life Sci 2020; 243: p. 117287.
2. Chen H, Liu X, Wu Y, Wu X, Wen X, Lu Y,
Zhao X. Apoptosis in HUVECs induced by
Thymosin β4 regulates endothelial cell function through AKT pathway 305
Vol. 62(4): 295 - 306, 2021
microRNA-616-3p via X-linked inhibitor of
apoptosis protein targeting. Exp Ther Med
2021;21(6):661.
3. Jiang F, Xu XR, Li WM, Xia K, Wang LF,
Yang XC. Monotropein alleviates H2O2in-
duced inflammation, oxidative stress and
apoptosis via NF-κB/AP-1 signaling. Mol
Med Rep 2020;22(6):4828-4836.
4. Nie J. inhibits high glucose induced apop-
tosis in HUVEC by targeting Rab4. Biomed
Pharmaother 2020;131: p. 110662.
5. Mi L, Zhang Y, Xu Y, Zheng X, Zhang X,
Wang Z, Xue M, Jin X. HMGB1/RAGE pro-
inflammatory axis promotes vascular en-
dothelial cell apoptosis in limb ischemia/
reperfusion injury. Biomed Pharmacother.
2019; 116:109005.
6. Low T, Hu S-K, Goldstein AL. Complete
amino acid sequence of bovine thymosin
beta 4: a thymic hormone that induces ter-
minal deoxynucleotidyl transferase activity
in thymocyte populations. Proc Natl Acad
Sci 1981;78(2):1162-1166.
7. Dubé KN, Smart N. Thymosin β4 and the
vasculature: multiple roles in development,
repair and protection against disease. Ex-
pert Opin Biol Ther 2018;18(sup1):131-139.
8. Kupatt C, Horstkotte J, Vlastos GA, Pfos-
ser A, Lebherz C, Semisch M, Thalgott M,
Büttner K, Browarzyk C, Mages J. Embr-
yonic endothelial progenitor cells expres-
sing a broad range of proangiogenic and
remodeling factors enhance vascularization
and tissue recovery in acute and chronic
ischemia. The FASEB J 2005;19(11):1576-
1578.
9. Smart N, Risebro CA, Clark JE, Ehler E,
Miquerol L, Rossdeutsch A, Marber MS,
Riley PR. Thymosin β4 facilitates epicar-
dial neovascularization of the injured adult
heart. Ann N Y Acad Sci 2010;1194(1):97-
104.
10. Smart N, Risebro CA, Melville AA, Moses
K, Schwartz RJ, Chien KR, Riley PR. Thy-
mosin β4 induces adult epicardial proge-
nitor mobilization and neovascularization.
Nature 2007;445(7124):177-182.
11. Smart N, Risebro CA, Melville AA, Moses
K, Schwartz RJ, Chien KR, Riley PR. Thy-
mosin β4 is essential for coronary vessel
development and promotes neovasculariza-
tion via adult epicardium. Ann N Y Acad Sci
2007;1112(1):171-188.
12. Wyczółkowska J, Walczak-Drzewiecka A,
Wagner W, Dastych J. Thymosin β4 and thy-
mosin β4-derived peptides induce mast cell
exocytosis. Peptides 2007;28(4):752-759.
13. Sun C, Feng SB, Cao ZW, Bei JJ, Chen Q,
Xu XJ, Zhou Z, Yu ZP, Hu HY. Up-regulated
expression of matrix metalloproteinases in
endothelial cells mediates platelet micro-
vesicle-induced angiogenesis. Cell Physiol
Biochem 2017;41(6):2319-2332.
14. Qiu F, Song J, Bi X, Wang M, Zhao Y, Fu
G. Thymosin β4 promotes glucose-impaired
endothelial progenitor cell function via Akt/
endothelial nitric oxide synthesis signaling
pathway. Exp Ther Med 2018;16(4):3439-
3444.
15. Dong W, Luo B, Qiu C, Jiang X, Shen B,
Zhang L, Liu W, Zhang W. TRIM3 attenua-
tes apoptosis in Parkinson’s disease via ac-
tivating PI3K/AKT signal pathway. Aging
(Albany NY) 2020;13(1):735-749.
16. Wang Y, Lin Y, Wang L, Zhan H, Luo X,
Zeng Y, Wu W, Zhang X, Wang F. TREM2
ameliorates neuroinflammatory response
and cognitive impairment via PI3K/AKT/
FoxO3a signaling pathway in Alzheimer’s
disease mice. Aging (Albany NY)
2020;12(20):20862-20879.
17. Xu L, Chen J, Jia L, Chen X, Awaleh
Moumin F, Cai J. SLC1A3 promotes gas-
tric cancer progression via the PI3K/AKT
signalling pathway. J Cell Mol Med 2020
;24(24):14392-14404.
18. Zhu S, Liu X, Xue M, Li Y, Cai D, Wang
S, Zhang L. 20(S)-ginsenoside Rh2 induces
caspase-dependent promyelocytic leuke-
mia-retinoic acid receptor A degradation in
NB4 cells via Akt/Bax/caspase9 and TNF-α/
caspase8 signaling cascades. J Ginseng Res
2021;45(2):295-304.
19. Zhou P, Xie W, Luo Y, Lu S, Dai Z, Wang
R, Sun G, Sun X. Protective effects of total
saponins of Aralia elata (Miq.) on endothe-
lial cell injury induced by TNF-α via modu-
lation of the PI3K/Akt and NF-κB signalling
pathways. Int J Mol Sci 2018;20(1):36.
20. Rajendran P, Rengarajan T, Thangavel
J, Nishigaki Y, Sakthisekaran D, Sethi
G, Nishigaki I. The vascular endothe-
306 Tang et al.
Investigación Clínica 62(4): 2021
lium and human diseases. Int J Biol Sci
2013;9(10):1057.
21. Palmer RM, Ashton D, Moncada S. Vascular
endothelial cells synthesize nitric oxide from
L-arginine. Nature 1988;333(6174):664-
666.
22. Kupatt C, Bock-Marquette I, Boekste-
gers P. Embryonic endothelial progeni-
tor cell-mediated cardioprotection requi-
res Thymosin β4. Trends Cardiovasc Med
2008;18(6):205-210.
23. Grant D, Kinsella J, Kibbey M, LaFlamme
S, Burbelo P, Goldstein A, Kleinman, HK.
Matrigel induces thymosin beta 4 gene in
differentiating endothelial cells. J Cell Sci
1995;108(12):3685-3694.
24. Grant DS, Rose W, Yaen C, Goldstein A,
Martinez J, Kleinman H. Thymosin β4 en-
hances endothelial cell differentiation and
angiogenesis. Angiogenesis 1999;3(2):125-
135.
25. Ho J, Su Y, Chen KH, Lee O. Protection
of thymosin beta-4 on corneal endothelial
cells from UVB-induced apoptosis. Chin J
Physiol 2010;53(3):190-195.
26. Zhao Y, Qiu F, Xu S, Yu L, Fu G. Thymosin
β4 activates integrin-linked kinase and de-
creases endothelial progenitor cells apopto-
sis under serum deprivation. J Cell Physiol
2011;226(11):2798-2806.
27. Qiu FY, Song XX, Zheng H, Zhao YB, Fu
GS. Thymosin β4 induces endothelial pro-
genitor cell migration via PI3K/Akt/eNOS
signal transduction pathway. J Cardiovasc
Pharmacol 2009;53(3):209-214.
28. Li J, Qiu F, Yu L, Zhao Y, Fu G, Zhou B.
Thymosin β4 reduces senescence of en-
dothelial progenitor cells via the PI3K/Akt/
eNOS signal transduction pathway. Mol
Med Rep 2013;7(2):598-602.
29. Bock-Marquette I, Shrivastava S, Pipes GT,
Thatcher JE, Blystone A, Shelton JM, Ga-
lindo CL, Melegh B, Srivastava D, Olson
EN. Thymosin β4 mediated PKC activation
is essential to initiate the embryonic coro-
nary developmental program and epicardial
progenitor cell activation in adult mice in
vivo. J Mol Cell Cardiol 2009;46(5):728-
738.
30. Lv S, Cheng G, Zhou Y, Xu G. Thymosin
beta4 induces angiogenesis through Notch
signaling in endothelial cells. Mol Cell Bio-
chem 2013;381(1):283-290.
31. Kim S, Kwon J. Effect of thymosin beta 4 in
the presence of up-regulation of the insulin-
like growth factor-1 signaling pathway on
high-glucose-exposed vascular endothelial
cells. Mol Cell Endocrinol 2015;401:238-
247.