You are on page 1of 9

Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Mechanical stretch aggravates aortic dissection by regulating MAPK T


pathway and the expression of MMP-9 and inflammation factors

Yunnan Hu1, Lin Lu1, Zhihuang Qiu, Qiuyu Huang, Yinhai Chen, Liangwan Chen
Department of Cardiac Surgery, Fujian Medical University Union Hospital, Xinquan Road No. 29, Fuzhou, 350001, Fujian, China

A R T I C LE I N FO A B S T R A C T

Keywords: This study aimed to explore whether mechanical stretch aggravated aortic dissection through regulating MAPK
Mechanical stretch pathway, MMP-9, and inflammation factors. We first established aortic dissection model rats. Mechanical stretch
Aortic dissection (3 g) was exerted on vascular ring of aortic dissection which was also treated by inhibitors of MAPK pathway
MAPK (SB203580, SP600125, and U0126). HE and Masson staining showed that aortic dissection severity with 3 g
MMP-9
tension was worse than that without tension (0 g); after the treatments of diverse inhibitors, the fracture and
Inflammation factor
breakage of the elastic fibers decreased. The expression of MMP-9, TNF-α, IL-1β) p38/p-p38, JNK1/p-JNK1, and
ERK1/2/p-ERK1/2 were determined by immunohistochemical analysis, RT-PCR, and western blot. No matter
whether tension was exerted or inhibitors were added, there was no change in the expression of p38, JNK1, and
ERK1/2. However, compared to the 0 g group, the expression of MMP-9, TNF-α, IL-1β, p-p38, p-JNK1, and p-
ERK1/2 was significantly upregulated in the 3 g group (P < 0.05). In both 0 g and 3 g groups, the expression of
MMP-9, TNF-α, IL-1β, p-p38, p-JNK1, and p-ERK1/2 was remarkably downregulated after inhibitors treatment
(P < 0.05). In conclusion, mechanical stretch aggravated aortic dissection by regulating the MAPK pathway and
the consequent expression of MMP-9 and inflammation factors.

1. Introduction adventitia [5]. Therefore, various pathological factors such as in-


flammatory reaction will increase the expression of MMP-9 in the
Aortic dissection (AD) is a kind of critical aortic diseases. Local media and adventitia, especially in the smooth muscle cells around the
aortic intimal crevasse and intimal/medial exfoliation and expansion degenerated media [5].
caused by high velocity blood flow separate the middle layer of the Mechanical stretch is an important factor in regulating the structure
artery wall along the long axis, consequently producing true and false and function of mammalian cells and tissues, but excessive mechanical
lumens [1]. At present, effective drugs are lacked and surgery is the stretch will cause vascular remodeling [6]. The increase of MMP-9
most commonly used therapeutics. However, the surgery is compli- expression degrades the elastin and collagen fibers in the media and
cated, hugely traumatic, and accompanied with high risk. The surgery subsequently weakens the vascular wall [7]. Under the action of me-
may injure recurrent laryngeal nerve and phrenic nerve. Moreover, chanical stretch, the intima is torn and separated from the media, and
intraoperative and postoperative bleeding frequently happens in the then AD happens [7]. The mechanical stretch is derived from the
suture. Therefore, drug adjunctive therapy shall be refocused. pulling and shearing stress caused by blood pressure and blood flow
Matrix metalloproteinase (MMP) is a protease family being capable [8].
of lysing protein substrates [2]. MMP contains type II fibronectin-like Therefore, this study aims to explore whether mechanical stretch
repeating segments in its catalytic domain and has strong ability of aggravates AD through regulating mitogen-activated protein kinase
hydrolyzing elastin and gelatin [3]. MMP-9 belongs to one of two kinds (MAPK) pathway, MMP-9, and inflammation factors, and then provide
of gelatinases in MMP family and it also has the common structural promising targets in the drug development for AD therapy.
characteristics of MMP [4]. Inflammatory cytokines can induce mul-
tiple cells to produce MMP-9, while as the enzymolysis substrate of
MMP-9, extracellular matrix (ECM) is mainly located in the media and


Corresponding author.
E-mail addresses: huyunnan-105@163.com (Y. Hu), lulinusa@163.com (L. Lu), qzhflm@126.com (Z. Qiu), terryhqy@163.com (Q. Huang),
798956199@qq.com (Y. Chen), chenliangwan@tom.com (L. Chen).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biopha.2018.09.129
Received 27 June 2018; Received in revised form 22 September 2018; Accepted 24 September 2018
0753-3322/ © 2018 Elsevier Masson SAS. All rights reserved.
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

2. Materials and methods 2.4. Hematoxylin eosin (HE) staining

2.1. Materials and animals Samples were taken out, dehydrated in ethanol solution, and
transparentized in dimethylbenzene. After paraffin embedding, they
Scott bluing buffer (G1865), Masson trichrome staining solution were cut into slices. The slices were baked, dewaxed, hydrated, and
(G1340), 5% bovine serum albumin (BSA) blocking buffer (SW3015), incubated in hematoxylin solution for 3 min. Then they were differ-
and β-aminopropionitrile (20170627) were purchased from Solarbio entiated in alcoholic hydrochloric acid for 15 s and blued in Scott
(Beijing, China). SB203580 (22898), SP600125 (13701), and U0126 bluing buffer for 15 s. The slices were washed and incubated in eosin
(19826) were bought from MedChem Express (NJ, USA). DAB kit solution for 3 min. Finally, they were dehydrated, transparentized,
(CW0125), TRIzon reagent (CW0580), HiFiScript cDNA synthesis kit mounted, and observed under a microscope (CKX41, Olympus, Japan).
(CW2569), and UltraSYBR mixture (CW0957) were gotten from CWBIO
(Beijing, China). Rabbit monoclonal antibody ERK1/2 (MAPK P44/42) 2.5. Masson staining
was from Cell Signaling Technology (1:1000, 4695, MA, USA). Rabbit
monoclonal antibodies MMP-9 (1:10000, ab76003), JNK1 (1:2000, Samples were taken out, dehydrated in ethanol solution, and
ab110724) and rabbit polyclonal antibodies p-JNK1 (1:4000, ab47337), transparentized in dimethylbenzene. After paraffin embedding, they
p-p38 (1:10000, ab47363), p-ERK1/2 (1:1000, ab214362) were ob- were cut into slices. The slices were incubated in Weigert solution for
tained from Abcam (MA, USA). Rabbit polyclonal antibodies p38 10 min. Then they were differentiated in alcoholic hydrochloric acid for
(1:1000, A0227) and interleukin-1β (IL-1β, 1:1000, A11369) were 10 s and blued in Masson bluing buffer for 4 min. The slices were wa-
bought from ABclonal (MA, USA). Rabbit polyclonal antibody tumor shed and incubated in ponceau S-fuchsin solution for 7 min.
necrosis factor-α (TNF-α) was from Bioss (1:400, bs-2150R, Beijing, Subsequently, they were washed with weak acid solution and phos-
China). Peroxidase-conjugated goat anti-rabbit IgG(H + L) was pur- phomolybdic acid solution before staining in aniline blue solution for
chased from ZSGB-BIO (1:2000, ZB-2301, Beijing, China). 1 min. Eventually, they were washed, dehydrated, transparentized,
Forty-five male Spraque-Dawley (SD) rats (three weeks, mounted, and observed under a microscope (CKX41, Olympus, Japan).
51.5 ± 1.2 g) were purchased from Shanghai SLAC Laboratory Animal
Co., Ltd (License No. SCXK(HU)2017-0005). All rats were housed in a 2.6. Immunohistochemical analysis
temperature-controlled room (22–25 °C) with a 12-h light/dark cycle
and were given free access to food and water. Study protocols were in Paraffin sections of the samples were made and baked at 75 °C for
compliance with the NIH Guide for Care and Use of Laboratory 1.5 h. The slices were immersed in dimethylbenzene for 10 min and in
Animals. fresh dimethylbenzene for another 10 min. After successively bathing in
100% ethanol, 100% ethanol, 95% ethanol, 80% ethanol, and water for
each 5 min, they were incubated in citrate buffer solution and washed
2.2. AD model establishment and vascular management by phosphate buffer solution (PBS). Subsequently, the slices were in-
cubated in fresh 3% hydrogen peroxide at room temperature for
β-aminopropionitrile (0.8%) was administrated into the rats for 5 10 min. After PBS washing, goat serum buffer was dropwise added onto
weeks through water drinking. Then the rats were anesthetized by in- the slices and incubated at room temperature for 30 min. The slices
traperitoneally injecting 1% pentobarbital sodium at 45 mg/kg. Hearts were then incubated in primary antibody buffer at 4 °C overnight. After
and aortae (to kidney) were collected and immediately immersed in KH washing with PBS, they were incubated in secondary antibody buffer at
solution pre-aerated by 95%O2-5%CO2 at 4 °C. The rats without un- room temperature for 1 h. Finally, they were stained, dehydrated,
dergoing these treatments served as control (n = 5). Dissection was transparentized, mounted, and microscopically examined.
evaluated under a microscope (CX41, Olympus, Japan). Vascular strips
were cut into a number of 3 mm vascular rings, and then the intima was 2.7. RT-PCR
carefully removed. Each vascular ring was fixed by two stainless steel
triangular scaffolds through the vascular lumen. It was vertically sus- Total RNA of the samples was collected by Trizon reagent and re-
pended in the bath containing 25 ml KH solution. The downward side versely transcribed to cDNA by HiFiScript cDNA synthesis kit according
was fixed and the upward side was connected to a tension sensor. The to the instruction for use. Sequences of primers were listed in Table 1.
tension of the vascular rings was regulated and recorded by a bio- PCR system (25 μl) included RNase free dH2O 9.5 μl, cDNA/DNA 1 μl,
function experiment system (BL-420S, Chengdu Techman Software, forward primer 1 μl, reverse primer 1 μl, and 2×ULtraSYBR Mixture
China). The perfusion chamber was kept at 37 °C and 95%O2-5%CO2 12.5 μl. Reaction parameters were shown as follows: Pre-denaturation
was aerated continuously (HV1403, Chengdu Techman Software, for 10 min at 95 °C, Denaturation for 10 s at 95 °C, annealing for 30 s at
China). The KH solution was changed every 15 min. Balance tension 57.5 °C, elongation for 30 s at 72 °C, and 40 circles. Dissociation curve
was 0.2 g. After balancing for 1 h, the artery was constricted with was analyzed as follows: 15 s at 95 °C, 1 min at 57.5 °C, 15 s at 95 °C,
60 mmol/L KCl. The vascular ring was washed to restore the tension to 15 s at 57.5 °C, 15 s at 57.5 °C, and measured stepwise from 92 °C, every
the baseline before follow-up experiments. According to the require- 0.5 °C. Eventually, the product was analyzed on a RT-PCR Detection
ments of the following eight groups, inhibitors were added. After System (CFX Connect™, Bio-Rad, USA). GAPDH served as internal
30 min, the vascular ring was immediately stored at −80 °C for the control.
follow-up tests.
2.8. Western blot

2.3. Experimental grouping Samples were incubated in lysis buffer for 30 min in an ice bath. The
lysate was centrifugated at 10,000 rpm and 4 °C for 10 min. Total pro-
Eight groups (n = 5): 1) 0 g (modeling only); 2) 3 g (3 g tension after tein was obtained by carefully collecting the supernatant. Protein
modeling); 3) 3 g + SB203580 (40 μmol/L); 4) 3 g + SP600125 concentration was examined by BCA kit. Protein was thereafter dena-
(40 μmol/L); 5) 3 g + U0126 (40 μmol/L); 6) 0 g + SB203580 tured and loaded to perform SDS-PAGE. The membrane was transferred
(40 μmol/L); 7) 0 g + SP600125 (40 μmol/L); 8) 0 g + U0126 for 40 min through a wet method and subsequently incubated with
(40 μmol/L). SP600125, SB203580 and U0126 inhibited JNK, p38 and diluted primary antibodies at 4 °C overnight. It was rinsed and in-
ERK1/2 pathways, respectively [9–11]. cubated in secondary antibody buffer at room temperature for 1.5 h.

1295
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

Table 1
PCR primers.
Primer Sequence Length of primer (bp) Length of product (bp) annealing temperature (°C)

MMP-9 For:CAAGGATGGTCTACTGGCACACG 23 158 57.5


Rev:AGGTGAAGGGAAAGTGACATGGG 23

IL-1β For:AGGAGAGACAAGCAACGACA 20 148 60


Rev:CTTTTCCATCTTCTTCTTTGGGTAT 25

TNF-α For:GCGTGTTCATCCGTTCTCTACC 22 196 60


Rev:TACTTCAGCGTCTCGTGTGTTTCT 24

GAPDH For:TACCCACGGCAAGTTCAA 18 111 57.5


Rev:ACCAGCATCACCCCATTT 18

Finally, enhanced chemiluminescence (ECL) solution was dropwise U0126 could reduce the breakage of aortic collagenous fibers.
added onto the membrane and the membrane was evaluated on a gel
imaging system (ChemiDoc™ XRS+, Bio-Rad, USA). Gray values were 3.4. Levels of IL-1β and TNF-α
analyzed with “Quantity one” software (v4.62, Bio-Rad, USA). GAPDH
served as the internal control. The mRNA and protein levels of IL-1β and TNF-α which were de-
termined by RT-PCR and western blot, respectively were depicted in
2.9. Statistical analysis Fig. 2A. Similar results were found between RT-PCR and western blot.
Their levels in the 3 g group were much higher than those in the 0 g
Data were presented as mean ± standard deviation (SD). Statistical group (P < 0.05). Interestingly, their levels were sharply down-
analysis was conducted with one way analysis of variance (ANOVA) regulated by the addition of inhibitors SB203580, SP600125, and
followed by Tukey post‑hoc test via SPSS software (19.0, SPSS, USA). U0126, which was true for both 0 g and 3 g groups (P < 0.05). It was
Significant difference was considered at P < 0.05. implied that stronger tension promoted the production of IL-1β and
TNF-α while inhibitors SB203580, SP600125, and U0126 suppressed
3. Results their production.

3.1. AD model establishment 3.5. Level of MMP-9

The establishment of AD model in rats was evaluated by HE and The mRNA and protein levels of MMP-9 which were evaluated by
Masson staining, which was shown in Fig. 1A. Normal aortae were RT-PCR, western blot, and immunohistochemical analysis were shown
found in the control rats. However, in the model rats, there were elastic in Fig. 2B and C. Similar results were found among RT-PCR, western
fibers’ damages in the aortic media. The blood in the aortic lumen blot, and immunohistochemical analysis. In comparison with the 0 g
entered the media. These resulted in the longitudinal tearing and group, the expression of MMP-9 was remarkably elevated in the 3 g
stripping of the media, which further induced the formation of true and group (P < 0.05). As for both 0 g and 3 g groups, its expression was
false lumens. It was indicated that AD model establishment in rats was sharply downregulated after inhibitors treatment (P < 0.05). It was
successful. suggested that tension exertion upregulated the expression of MMP-9
while inhibitors SB203580, SP600125, and U0126 downregulated its
3.2. HE staining expression.

Fig. 1B demonstrated the HE staining results of aortae in eight 3.6. Levels of p-ERK1/2 and ERK1/2
groups. In the groups without tension (0 g), the intima and the elastic
fibers in the media were ruptured. After the treatments of diverse in- The protein expression of p-ERK1/2 and ERK1/2 which was as-
hibitors (0 g + SB203580, 0 g + SP600125, 0 g + U0126), the sessed by immunohistochemical analysis and western blot was shown in
breakage of elastic fibers in the media was slightly reduced. In the Fig. 3. Similar results were found between immunohistochemical ana-
groups with tension (3 g), the intima rupture, the media tearing, and lysis and western blot. No matter whether tension exertion or inhibitors
the breakage and reduction of elastic fibers were found. After the addition, there was no obvious change in the level of ERK1/2. However,
treatments of diverse inhibitors (3 g + SB203580, 3 g + SP600125, in comparison with the 0 g group, the ratio of p-ERK1/2 to ERK1/2 (p-
3 g + U0126), the fracture of the elastic fibers in the media was ob- ERK1/2/ERK1/2) was remarkably elevated in the 3 g group
viously decreased. (P < 0.05). As for both 0 g and 3 g groups, the p-ERK1/2/ERK1/2 was
notably reduced after inhibitors exposure (P < 0.05). It was demon-
3.3. Masson staining strated that stronger tension enhanced the production of p-ERK1/2
while inhibitors SB203580, SP600125, and U0126 depressed its pro-
Fig. 1C showed the Masson staining results of aortae in eight groups. duction.
In the Masson staining, collagenous fibers were blue, and muscle fibers,
red cells, and cellulose were orange red. As compared with the group 3.7. Levels of p-p38&p38 and p-JNK1&JNK1
without tension and inhibitors (0 g), there was no obvious difference in
collagenous fibers after the treatments of diverse inhibitors The protein expression of p-p38&p38 and p-JNK1&JNK1 which was
(0 g + SB203580, 0 g + SP600125, 0 g + U0126). The content of col- investigated by immunohistochemical analysis and western blot was
lagenous fibers under 3 g tension seemed to be lower than that under shown in Fig. 4A and B. Similar results were also found between the
0 g tension. Comparing to the group with tension and without inhibitors immunohistochemical analysis and western blot. No matter whether
(3 g), the content of collagenous fibers increased after the treatments of tension exert or inhibitors addition, there was no obvious change in the
diverse inhibitors (3 g + SB203580, 3 g + SP600125, 3 g + U0126). expression of p38 and JNK1. However, comparing to the 0 g group, the
These results suggested that inhibitors SB203580, SP600125, and expression of p-p38 and p-JNK1 was significantly augmented in the 3 g

1296
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

Fig. 1. Pathological examination of aortae. (A) HE and Masson staining of aortae in the control and model rats to evaluate the success of the aortic dissection model
establishment. (B) HE staining of aortae in various groups with different tensions (0 g and 3 g) and inhibitors (SB203580, SP600125, and U0126). (C) Masson staining
of aortae in various groups with different tensions (0 g and 3 g) and inhibitors (SB203580, SP600125, and U0126).

group (P < 0.05). As for both 0 g and 3 g groups, inhibitors exposure through MAPK signaling pathway. We employed β-aminopropionitrile
remarkably lowered their expression (P < 0.05). It was suggested that to induce AD in SD rats and exerted different mechanical tensions to
stronger tension facilitated the generation of p-p38 and p-JNK1 but stretch the vascular rings of aortae with dissection. HE and Masson
inhibitors SB203580, SP600125, and U0126 reduced their expression. staining had validated the success of AD model establishment and re-
vealed that the AD was more severe after mechanical stretch.
Some studies have shown that mechanical stretch can activate a
4. Discussion
large number of calcium channels on the vascular wall and intracellular
multiple signal transduction pathways including MAPK, reactive
As one of the major diseases that endanger the patients’ lives, AD
oxygen species (ROS), protein kinase B (AKT), and nitric oxide (NO)
has the characteristics of acute onset, rapid progress, and high mor-
signaling pathways [14–16]. MAPK signaling pathway is closely related
tality. Its specific pathogenesis is not clear, and now it is considered to
to cell proliferation, differentiation, metastasis, apoptosis, and the pa-
be related to a variety of factors such as hypertension and inflammation
thogenesis of aortic diseases [17,18]. Five different MAPK signal
[12,13].
transduction pathways have been found in mammalian organisms.
In this study, we explored whether mechanical stretch aggravated
Thereinto, ERK1/2 signaling pathway regulates cell growth and
AD by regulating the expression of MMP-9 and inflammation factors

1297
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

Fig. 2. Levels of IL-1β, TNF-α and MMP-9 (n = 5). (A) The mRNA and protein levels of IL-1β and TNF-α which were determined by RT-PCR and western blot,
respectively. (B) The mRNA and protein levels of MMP-9 which were determined by RT-PCR and western blot, respectively. (C) The protein level of MMP-9 which
was evaluated by immunohistochemical analysis. The target in the immunohistochemical image was shown in brown. *P<0.05 vs. 0 g, #P<0.05 vs. 3 g.

differentiation. JNK and p38 pathways play an important role in stress stimuli such as stress, injury, and inflammation activate the upstream
response such as inflammation and apoptosis. Advanced glycation end bispecific sites (Thrl83 and Tyrl8) of JNK to induce its active form
products participate in the proliferation of human aortic smooth muscle through phosphorylation [20]. P38 is mainly involved in the signal
cells via activating ERK1/2 pathway [19]. A variety of extracellular transduction of injury, stress, and inflammation processes. It can be

1298
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

Fig. 3. The protein expression of p-ERK1/2 and ERK1/2 which was assessed by immunohistochemical analysis and western blot (n = 5). The target in the im-
munohistochemical image was shown in brown. *P<0.05 vs. 0 g, #P<0.05 vs. 3 g.

activated by some factors such as proinflammatory cytokines and toxins be secreted only if they are converted into synthetic phenotypes
and its abnormal excessive activation may lead to growth arrest and [28–30]. The MMP-9 level in the plasma of patients with AD is higher
apoptosis of cells [21–25]. We revealed that after tension exertion, the than that in the plasma of normal population [31–33]. This study found
expression of p-JNK, p-ERK1/2, and p-p38 was remarkably upregu- that the expression of MMP-9 was significantly upregulated after me-
lated, but the expression of JNK, ERK1/2, and p38 was not changed. It chanical stretch in the AD model, which was consistent with a previous
was demonstrated that mechanical stretch aggravated AD through ac- report [34].
tivating JNK, ERK1/2, and p38 pathways in a phosphorylation way. TNF-α is mainly produced by monocytes and lymphocytes. It can
MMP and its inhibitors have been reported to take part in the pa- stimulate the generation of inflammatory factors and plays a direct role
thological process of aortic diseases and have a correlation with the in promoting inflammation, thereby affecting the thrombosis formation
pathogenesis of AD [26,27]. The MMP-9 secreted by aortic smooth and plaque stability [35,36]. It plays an important role in the devel-
muscle cells plays an important role in the occurrence of AD. Normal opment and progression of AD [35,36]. IL-1β plays an important role in
aortic smooth muscle cells are contractile phenotypes and MMP-9 can the signaling transduction, the activation and regulation of immune

1299
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

Fig. 4. The protein expression of p-p38&p38 (A) and p-JNK1&JNK1 (B) which was investigated by immunohistochemical analysis and western blot (n = 5). The
target in the immunohistochemical image was shown in brown. *P<0.05 vs. 0 g, #P<0.05 vs. 3 g.

cells, the mediation of T cells and B cells activation, proliferation, and can inhibit the phosphorylation of c-Jun induced by TNF-α activation,
differentiation, and the inflammatory response [37]. This study showed and thereby prevent apoptosis in a variety of cell types [39–42]. A
that the expression of TNF-α and IL-1β was elevated after the exertion number of in vivo experiments using SP600125 have demonstrated the
of mechanical stretch. It was indicated that mechanical stretch medi- potential of directly inhibiting JNK for therapeutic intervention
ated the production of AD inflammation. [43,44]. SB203580 is also a well-known selective inhibitor of p38
In order to prove that the expression of MMP-9, TNF-α, and IL-1β pathway [45–47]. SB203580 directly inhibits p38 pathway by binding
was related to MAPK signaling pathways such as ERK1/2, JNK, and to the ATP-binding pocket at an ATP-competitive manner [46]. U0126
p38, we performed inhibition tests by utilizing ERK1/2 inhibitor is a highly selective inhibitor of both MEK1 and MEK2, a type of MAPK/
U0126, JNK inhibitor SP600125, and p38 inhibitor SB203580. ERK kinase [48,49]. U0126 functionally antagonizes transcriptional
Interestingly, we revealed that the expression of MMP-9, TNF-α, and IL- activity of activator protein 1 (AP-1) and prevents the activation of
1β was also inhibited after the treatment of these inhibitors. These MAP kinase p42 and p44 encoded by ERK2 and ERK1 genes respec-
results suggested that the increase in the expression of MMP-9, TNF-α, tively via non-competitive inhibition of the dual specificity kinase MEK
and IL-1β induced by mechanical stretch might be mediated by MAPK [48,49]. The activation of ERK1/2 is significantly blocked by the
signaling pathways. Moreover, the inhibitors of MAPK signaling path- MEK1/2 inhibitor U0126 [47,50]. The effects of U0126 on the growth
ways might alleviate the influence of mechanical stretch on AD by of eight human breast cancer cell lines show that U0126 selectively
downregulating the expression of MMP-9 and inflammation factors. repressed anchorage-independent growth of MDA-MB231 and HBC4
When mechanical stretch was exerted on the aortae with dissection, cells, two lines with constitutively activated ERK [51]. In sum,
stretch-activated channel on vascular smooth muscle cells might be SP600125, SB203580 and U0126 selectively inhibit JNK, p38 and
opened, and then the ERK1/2, p38, and JNK pathways were activated, ERK1/2 pathways, respectively, by majorly direct inhibition and min-
consequently upregulating the expression of MMP-9 and inflammation orly suppressing the ATP activity (Fig. 5).
factors at transcriptional and expression levels.
SP600125, SB203580 and U0126 inhibited JNK, p38 and ERK1/2
pathways through suppressing the activity of adenosine triphosphate
(ATP). SP600125 also showed some ability to inhibit p38 pathway [9].
SB203580 could also inhibit the expression of JNK to some extent [10].
Therefore, no matter what branch of the MAPK pathway was inhibited
all markers seemed to be downregulated. For example, ERK1/2 in-
hibition affected both JNK and p38 when they were not downstream of
ERK. Although all three inhibitors have the ability to inhibit ATP and
one of them will make moderate effect on the other two pathways, all of
them are selective inhibitors for the corresponding pathway. SP600125
is one of the most extensively used ATP-competitive JNK inhibitors
[38]. It binds reversibly and competitively to the anthrapyrazolone
domain of JNK [38]. A number of studies have shown that SP600125 Fig. 5. SP600125, SB203580 and U0126 inhibited JNK, p38 and ERK1/2
pathways through suppressing the activity of adenosine triphosphate (ATP).

1300
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

5. Conclusions and apoptosis, PLoS One 10 (2015) e0120440.


[21] L. Shi, X. Yu, H. Yang, et al., Advanced glycation end products induce human
corneal epithelial cells apoptosis through generation of reactive oxygen species and
Mechanical stretch aggravated AD by regulating the MAPK sig- activation of JNK and p38 MAPK pathways, PLoS One 8 (2013) e66781.
naling pathway and the consequent expression of MMP-9 and in- [22] K. Palanivel, V. Kanimozhi, B. Kadalmani, Verrucarin A alters cell-cycle regulatory
flammation factors. These results might provide novel and promising proteins and induces apoptosis through reactive oxygen species-dependent
p38MAPK activation in the human breast cancer cell line MCF-7, Tumour Biol. 35
insights into the mechanism understanding of AD and into the targeting (2014) 10159–10167.
drug development for AD treatment. [23] M.M. Yurinskaya, M.G. Vinokurov, S.V. Grachev, et al., Actovegin reduces the hy-
drogen peroxide-induced cell apoptosis of SK-N-SH neuroblastoma by means of
p38MAPK and PI-3K inhibition, Dokl. Biol. Sci. 456 (2014) 215–217.
Funding [24] N. Wu, X. Lin, X. Zhao, et al., MiR-125b acts as an oncogene in glioblastoma cells
and inhibits cell apoptosis through p53 and p38MAPK-independent pathways, Br. J.
This work was supported by the National Natural Science Cancer 109 (2013) 2853–2863.
[25] H.L. Chang, C.Y. Chen, Y.F. Hsu, et al., Simvastatin induced HCT116 colorectal
Foundation of China [grant number 81370414].
cancer cell apoptosis through p38MAPK-p53-survivin signaling cascade, Biochim.
Biophys. Acta 1830 (2013) 4053–4064.
Conflict of interest [26] Y. Gao, W. Wu, C. Yu, et al., A disintegrin and metalloproteinase with thrombos-
pondin motif 1 (ADAMTS1) expression increases in acute aortic dissection, Sci.
China Life Sci. 59 (2016) 59–67.
None. [27] F. Giachino, M. Loiacono, M. Lucchiari, et al., Rule out of acute aortic dissection
with plasma matrix metalloproteinase 8 in the emergency department, Crit. Care 17
Appendix A. Supplementary data (2013) R33.
[28] L. Wang, J. Zhang, W. Fu, et al., Association of smooth muscle cell phenotypes with
extracellular matrix disorders in thoracic aortic dissection, J. Vasc. Surg. 56 (2012)
Supplementary material related to this article can be found, in the 1698–1709.
online version, at doi:https://doi.org/10.1016/j.biopha.2018.09.129. [29] J. Zhang, L. Wang, W. Fu, et al., Smooth muscle cell phenotypic diversity between
dissected and unaffected thoracic aortic media, J. Cardiovasc. Surg. (Torino) 54
(2013) 511–521.
References [30] V. Lesaukaite, P. Tanganelli, C. Sassi, et al., Smooth muscle cells of the media in the
dilatative pathology of ascending thoracic aorta:morphology, immunoreactivity for
osteopontin,matrix metalloproteinases, and their inhibitors, Hum. Pathol. 32
[1] M.D. Dake, N. Kato, R.S. Mitchell, et al., Endovascular stent-graft placement for the
(2001) 1003–1011.
treatment of acute aortic dissection, N. Engl. J. Med. 340 (1999) 1546–1552.
[31] D. Wen, X.L. Zhou, J.J. Li, et al., Plasma concentrations of interleukin-6, C-reactive
[2] T. Sorsa, L. Tjäderhane, T. Salo, Matrix metalloproteinases (MMPs) in oral diseases,
protein, tumor necrosis factor-α and matrix metalloproteinase-9 in aortic dissec-
Oral Dis. 10 (2004) 311–318.
tion, Clin. Chim. Acta 413 (2012) 198–202.
[3] L.J. Xiao, P. Lin, F. Lin, et al., ADAM17 targets MMP-2 and MMP-9 via EGFR-MEK-
[32] G. Sangiorgi, S. Trimarchi, A. Mauriello, et al., Plasma levels of metalloproteinases-
ERK pathway activation to promote prostate cancer cell invasion, Int. J. Oncol. 40
9 and-2 in the acute and subacute phases of type A and type B aortic dissection, J.
(2012) 1714–1724.
Cardiovasc. Med. (Hagerstown) 7 (2006) 307–315.
[4] R.J. Turner, F.R. Sharp, Implications of MMP-9 for blood brain barrier disruption
[33] G.T. Karapanagiotidis, P. Antonitsis, N. Charokopos, et al., Serum levels of matrix
and hemorrhagic transformation following ischemic stroke, Front. Cell. Neurosci.
metalloproteinase-1,-2,-3,and-9 in thoracic aortic diseases and acute myocardial
10 (2016) 56.
ischemia, J. Cardiothorac. Surg. 4 (2009) 59.
[5] T. Kurihara, R. Shimizu-Hirota, M. Shimoda, et al., Neutrophil-derived matrix
[34] Z. Qiu, L. Chen, H. Cao, et al., Mechanical strain induced expression of matrix
metalloproteinase 9 triggers acute aortic dissection, Circulation 126 (2012)
Metalloproteinase-9 via stretch-activated channels in rat abdominal aortic dissec-
3070–3080.
tion, Med. Sci. Monit. 23 (2017) 1268–1276.
[6] B.W. Benhampyle, B.L. Pruitt, W.J. Nelson, Mechanical strain induces E-cadherin-
[35] G.S. Hotamisligil, P. Peraldi, A. Budavari, et al., IRS-1-mediated inhibition of insulin
dependent Yap1 and E-catenin activation to drive cell cycle entry, Science 348
receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin re-
(2015) 1024–1027.
sistance, Science 271 (1996) 665–668.
[7] J.M. Ruiz-Morales, R. Dorantes-Heredia, O. Arrieta, et al., Neutrophil gelatinase-
[36] C.D. Dumitru, J.D. Ceci, C. Tsatsanis, et al., TNF-α induction by LPS is regulated
associated lipocalin (NGAL) and matrix metalloproteinase-9 (MMP-9) prognostic
posttranscriptionally via a Tpl2/ERK-Dependent pathway, Cell 103 (2000)
value in lung adenocarcinoma, Tumour Biol. 36 (2015) 3601–3610.
1071–1083.
[8] G. Huang, A. Wang, X. Li, et al., Change in high-sensitive C-reactive protein during
[37] F.R. Greten, M.C. Arkan, J. Bollrath, et al., NF-κB is a negative regulator of IL-1β
abdominal aortic aneurysm formation, J. Hypertens. 27 (2009) 1829–1837.
secretion as revealed by genetic and pharmacological inhibition of IKKβ, Cell 130
[9] M.A. Bogoyevitch, I. Boehm, A. Oakley, et al., Targeting the JNK MAPK cascade for
(2007) 918–931.
inhibition: basic science and therapeutic potential, Biochim. Biophys. Acta 1697
[38] B.L. Bennett, D.T. Sasaki, B.W. Murray, et al., SP600125, an anthrapyrazolone in-
(2004) 89–101.
hibitor of Jun N-terminal kinase, Proc. Natl. Acad. Sci. U. S. A. 98 (2001)
[10] M.C. Bagley, T. Davis, P.G.S. Murziani, et al., Use of p38 MAPK inhibitors for the
13681–13686.
treatment of Werner syndrome, Pharmaceuticals (Basel) 3 (2010) 1842–1872.
[39] E.L. Marderstein, B. Bucher, Z. Guo, et al., Protection of rat hepatocytes from
[11] X. Wang, S. Pesakhov, A. Weng, et al., ERK 5/MAPK pathway has a major role in
apoptosis by inhibition of c-Jun N-terminal kinase, Surgery 134 (2003) 280–284.
1α,25-(OH)2 vitamin D3-induced terminal differentiation of myeloid leukemia
[40] L. Minutoli, D. Altavilla, H. Marini, et al., Protective effects of SP600125 a new
cells, J. Steroid Biochem. Mol. Biol. 144PA (2014) 223–227.
inhibitor of c-jun N-terminal kinase (JNK) and extracellular-regulated kinase
[12] L.W. Chen, X.J. Wu, X.F. Dai, et al., Repair of acute type A aortic dissection with
(ERK1/2) in an experimental model of cerulean-induced pancreatitis, Life Sci. 75
ascending aorta replacement combined with open fenestrated stent graft placement,
(2004) 2853–2866.
Ann. Thorac. Surg. 101 (2016) 644–649.
[41] N. Sharma, H. Suzuki, Q. He, et al., Tumor necrosis factor-α-mediated activation of
[13] L.W. Chen, X.J. Wu, X.F. Dai, et al., Total arch repair for acute type A aortic dis-
c-Jun NH(2)terminal kinase as a mechanism for fumonisin B(1) induced apoptosis
section with open placement of a modified triple-branched stent graft and the arch
in murine primary hepatocytes, J. Biochem. Mol. Toxicol. 19 (2005) 359–367.
open technique, J. Cardiothorac. Surg. 9 (2014) 135.
[42] K. Assi, R. Pillai, A. Gómez‐Muñoz, et al., The specific JNK inhibitor SP600125
[14] C.M. Ghantous, F.H. Kobeissy, N. Soudani, et al., Mechanical stretch-induced vas-
targets tumour necrosis factor-α production and epithelial cell apoptosis in acute
cular hypertrophy occurs through modulation of leptin synthesis-mediated ROS
murine colitis, Immunology 118 (2006) 112–121.
formation and GATA-4 nuclear translocation, Front. Pharmacol. 6 (2015) 240.
[43] C. Davies, C. Tournier, Exploring the function of the JNK (c-Jun N-terminal kinase)
[15] K.W. Seo, S.J. Lee, B.H. Ye, et al., Mechanical stretch enhances the expression and
signalling pathway in physiological and pathological processes to design novel
activity of osteopontin and MMP-2 via the AKT1/AP-1 pathways in VSMC, J. Mol.
therapeutic strategies, Biochem. Soc. Trans. 40 (2012) 85–89.
Cell. Cardiol. 85 (2009) 13–24.
[44] W. Wang, L. Shi, Y. Xie, et al., SP600125, a new JNK inhibitor, protects dopami-
[16] L. Du, P. Empery, J. Ji, et al., Probenecid and N-acetylcysteine prevent loss of in-
nergic neurons in the MPTP model of Parkinson’s disease, Neurosci. Res. 48 (2004)
tracellular glutathione and inhibit neuronal death after mechanical stretch injury in
195–202.
vitro, J. Neruotrauma 33 (2016) 1913–1917.
[45] M. Baranclk, V. Bohacova, J. Kvackajova, et al., SB203580, a specific inhibitor of
[17] E.K. Kim, E.J. Choi, Compromised MAPK signaling in human diseases: an update,
p38-MAPK pathway, is a new reversal agent of P-glycoprotein-mediated multidrug
Arch. Toxicol. 89 (2015) 867–882.
resistance, Eur. J. Pharm. Sci. 14 (2001) 29–36.
[18] Y. Sun, W.Z. Liu, T. Liu, et al., Signaling pathway of MAPK/ERK in cell prolifera-
[46] P.R. Young, M.M. McLaughlin, S. Kumar, et al., Pyridinyl imidazole inhibitors of
tion, differentiation, migration, senescence and apoptosis, J. Recept. Signal
p38 mitogen-activated protein kinase bind in the ATP site, J. Biol. Chem. 272
Transduct. Res. 35 (2015) 600–604.
(1997) 12116–12121.
[19] G. Yuan, Y. Liu, H. Zeng, et al., Effect of ERK1/2 phosphorylation on proliferation of
[47] E. Corcelle, N. Djerbi, M. Mari, et al., Control of the autophagy maturation step by
human aortic vascular smooth muscle cells stimulated by advanced glycation end
the MAPK ERK and p38: lessons from environmental carcinogens, Autophagy 3
products, J. Pract. Med. 32 (2016) 2430–2433.
(2007) 57–59.
[20] J. Li, Y. Xia, T. Liu, et al., Protective effects of astaxanthin on ConA-induced au-
[48] M.F. Favata, K.Y. Horiuchi, E.J. Manos, et al., Identification of a novel inhibitor of
toimmune hepatitis by the JNK/p-JNK pathway-mediated inhibition of autophagy
mitogen-activated protein kinase kinase, J. Biol. Chem. 273 (1998) 18623–18632.

1301
Y. Hu et al. Biomedicine & Pharmacotherapy 108 (2018) 1294–1302

[49] J.V. Duncia, J.B. Santella 3rd, C.A. Higley, et al., MEK inhibitors: the chemistry and signaling cascades, Cell. Mol. Biol. Lett. 22 (2017) 13.
biological activity of U0126, its analogs, and cyclization products, Bioorg. Med. [51] H. Fukazawa, K. Noguchi, Y. Murakami, et al., Mitogen-activated protein/extra-
Chem. Lett. 8 (1998) 2839–2844. cellular signal-regulated kinase kinase (MEK) inhibitors restore anoikis sensitivity
[50] J. Qian, Y. Gu, C. Wu, et al., Agonist-induced activation of human FFA1 receptor in human breast cancer cell lines with a constitutively activated extracellular-
signals to extracellular signal-regulated kinase 1 and 2 through Gq- and Gi-coupled regulated kinase (ERK) pathway, Mol. Cancer Ther. 1 (2002) 303–309.

1302

You might also like