Open Access

EOFAZ inhibits endothelial‑to‑mesenchymal transition through downregulation of KLF4

  • Authors:
    • Yanyan Zhang
    • Chen Li
    • Yongpan Huang
    • Shuang Zhao
    • Yini Xu
    • Yan Chen
    • Feng Jiang
    • Ling Tao
    • Xiangchun Shen
  • View Affiliations

  • Published online on: April 9, 2020     https://doi.org/10.3892/ijmm.2020.4572
  • Pages: 300-310
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Essential oil from Alpinia zerumbet rhizome (EOFAZ), which is termed Yan shanjiang in China, is extensively used as an herbal medicine in the Guizhou area and has been shown to protect against the damaging effects of cardiovascular injury in vitro and in vivo. In the present study, it was hypothesized that the protective effects of EOFAZ on transforming growth factor (TGF)‑β1‑induced endothelial‑to‑mesenchymal transition (EndMT) in human umbilical vein endothelial cells (HUVECs) were mediated by inhibition of Krüppel‑like factor 4 (KLF4). Cell motility was assessed using wound healing and Transwell assays. The expression of endothelial markers and mesenchymal markers were determined by reverse transcription‑quantitative PCR, immunofluorescence staining and western blotting, and additionally, phosphorylated NF‑κB p65 expression was determined by western blotting. Furthermore, the involvement of KLF4 in EndMT was determined using RNA interference to knockdown the expression of KLF4. TGF‑β1 treatment significantly promoted EndMT, as evidenced by downregulation of vascular endothelial‑cadherin and upregulation of α‑smooth muscle actin in HUVECs, and by enhancing cell migration. Small interfering RNA‑mediated knockdown of KLF4 reversed TGF‑β1‑induced EndMT. Additionally, treatment with EOFAZ inhibited TGF‑β1‑induced EndMT in a dose‑dependent manner. These results suggest that TGF‑β1 may induce EndMT through upregulation of KLF4, and this may be reversed by EOFAZ. Therefore, EOFAZ was shown to inhibit TGF‑β1‑induced EndMT through regulation of KLF4.

Introduction

The vascular wall consists of endothelial cells, smooth muscle cells, fibroblasts and extracellular matrix, and is considered an active and integrated organ (1). The vasculature is sensitive to a range of stimuli, several of which may contribute to pathophysiological changes in response to stresses that result in proliferation of the vasculature (2). The initiating event in vascular remodeling is endothelial cell damage. Alterations in the presence of vasoactive substances following endothelial cell dysfunction are involved in the development and progress of cardiovascular diseases. A vicious circle is caused by further injury of endothelial cells following the onset of cardiovascular diseases (3). This dynamic process characterizes vascular remodeling, and is defined as any enduring change in the size and/or composition of an adult blood vessel that allows blood vessels to adapt and heal (4). Vascular remodeling underlies the pathogenesis of major cardiovascular disorders, including pulmonary arterial hypertension (PAH) and atherosclerosis, as well as wound healing (5). Vascular remodeling is involved in cells progressing through four cellular processes: Differentiation, death, migration, and production or degradation of extracellular matrix (3,6). Endothelial cells are pivotal in the initiation of vascular remodeling. The increase in mesenchymal or smooth muscle (SM)-like phenotypes originates from increased endothelial cell transformation (7). Endothelial cells are the first barrier of blood vessels and recently they have been regarded as contributors to vascular remodeling through endothelial-to-mesenchymal transition (EndMT) (8).

EndMT is the process by which endothelial cells undergo phenotypic changes and acquire a mesenchymal/SM-like phenotype (9). Under pathophysiological conditions, such as transplant atherosclerosis and restenosis, endothelial-derived mesenchymal cells may accelerate the development of neointimal lesions (10). Evidence for EndMT has been demonstrated in the context of cardiac and vascular development, wound healing and various dysfunctions, including tissue fibrosis and diabetic nephropathy (11). Thus, EndMT is regarded as an important mechanism for the generation of SM-like cells. Endothelial cells facilitate vascular development and dysfunction, which act through direct and indirect roles in the vascular remodeling process (12). A previous study has demonstrated that EndMT is involved in the pathogenesis of vascular remodeling (13). EndMT contributes to neointimal formation in interpositional vein grafts (14). Similarly, EndMT contributes to intimal hyperplasia and early-lesion formation in atherosclerosis (15). Furthermore, EndMT contributes to progression of PAH, atherosclerosis and wound healing (16). Therefore, in the present study, it was hypothesized that EndMT may accelerate the pathogenesis of vascular remodeling. However, the role of EndMT and the mechanisms regulating cell phenotype adaption during vascular remodeling are poorly understood. Studies have reported that mouse and human endothelial cells are induced by transforming growth factor (TGF)-β1 to transform into SM-like cells in which the expression of α-smooth muscle actin (α-SMA) is upregu-lated (17,18). Under the synergistic action of TGF-β1 and the Wnt signaling pathway, EndMT may be induced. Therefore, in the present study, TGF-β1 was used to develop an in vitro model of EndMT (19).

Zingiberaceae belongs to the ginger family, which consists of a large number of aromatic perennial species shown to exhibit therapeutic potential in a number of cardiac-related therapies (20). Alpinia zerumbet (Pers.) Burtt et Smith, termed Yan Shanjiang in China, is an aromatic plant originating in the East Indies and is widely used as an herbal medicine in South America, Oceania and Asia (21,22). It contains several bioactive constituents and possesses a broad spectrum of pharmacological properties, such as antihypertensive, anti-inflammatory and cardiovascular protective effects, as well as antioxidant effects (23,24), and is widely used as a local Miao herbal medicine in the Guizhou province. Previous studies have shown that essential oil from Alpinia zerumbet rhizome (EOFAZ) is effective against the vasoconstriction induced by release of norepinephrine and KCl (25). The mechanisms mediating the beneficial effects of EOFAZ may include inhibition of oxidative stress and inflammation, and induction of apoptosis (26). However, the effects of EOFAZ on EndMT in a range of stress situations has not been investigated, particularly in relation to TGF-β1 signaling (22).

In the present study, a cell based in vitro model treated with TGF-β1 was established to verify the hypothesis that EOFAZ protects against TGF-β1-induced EndMT, and to determine the underlying molecular mechanism by which EOFAZ exerts its beneficial effects. The present study identified a novel molecular mechanism by which EOFAZ exerts its effects and also provides a theoretical basis for use of EOFAZ treatment for cardiac disorders.

Materials and methods

Extraction of EOFAZ

The essential oil was extracted from the fruit of Alpinia zerumbet, which was collected in Zhenfeng County, Guiyang, China, in October 2018. The fruit was identified by Professor Qingde Long (Department of Pharmacognosy and Medico-botany, Guizhou Medical University, Guiyang, China) and a voucher specimen (no. 20181029) was deposited to the Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University. The method of extraction/isolation and the compounds of EOFAZ were identified according to our previous study (22).

Cell culture

Human umbilical vein endothelial cells (HUVECs) were purchased from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. Cells were cultured in Endothelial Cell Medium (ScienCell Research Laboratories, Inc.) supplemented with 5% FBS (ScienCell Research Laboratories, Inc.) and 100 U/ml penicillin and streptomycin, and maintained in an incubator at 37°C with 5% CO2. Cells were sub-cultured and seeded into 6- or 24-well plates for subsequent experiments as required. The cells were pretreated with EOFAZ and LY2109761 (Selleck Chemicals) for 2 h at 37°C, and TGF-β1 (Peprotech EC Ltd.) was added for 72 h. HUVECs were trypsinized with 0.25% trypsin and collected for analysis.

Cell morphological assessment

The HUVECs were plated in 6-well plates at a density of 3×105 cells/well and were incubated overnight. Following treatment with 10 ng/ml TGF-β1 for 72 h at 37°C, the cultured plates were examined and photographs were obtained using an inverted light microscope (magnification, ×100).

Western blot analysis

Cells were lysed in RIPA lysis buffer containing protease inhibitors (Beyotime Institute of Biotechnology). The protein concentration was determined by BCA assay. For western blotting, a total of 40 μg protein/lane from lysed cells was separated by 10% SDS-PAGE. The proteins were transferred to a PVDF membrane and the membrane was blocked with 5% non-fat dry milk at room temperature (25°C) for 2 h. The membrane was then incubated with the following primary antibodies overnight at 4°C: Rabbit anti-Krüppel-like factor 4 (KLF4; 1:1,000; cat. no. 12173S), anti-vascular endothelial (VE)-cadherin (1:1,000; cat. no. 2500), anti-α-SMA (1:1,000; cat. no. 19245), anti-snail (1:1,000; cat. no. 5276) and anti-NF-κB phosphorylated (p)-p65 (1:1,000; cat. no. 3033T). All primary antibodies were from Cell Signaling Technology, Inc. and all were diluted in TBS and 0.2% Tween-20. Subsequently, the membranes were washed and incubated with a horseradish peroxidase-conjugated secondary antibody (Cell Signaling Technology, Inc.; 1:10,000; cat. no. 7076) for 90 min at room temperature. Signals were visualized using an ECL kit (GE Healthcare). The expression levels of protein were calculated by using ImageJ V1.8.0.112 software (National Institutes of Health). Protein signals were normalized to β-actin (Cell Signaling Technology, Inc.; 1:1,000; cat. no. 3700).

Reverse transcription-quantitative (RT-q)PCR

Total RNA was extracted using the TransZol Up Plus RNA kit (Sangon Biotech Co., Ltd.). Total RNA was purified with 75% ethanol and its concentration was determined using spectrophotometry. cDNA was synthesized from the purified RNA (200 ng per sample) using a reverse transcription kit (cat. no. DRR037A; Takara Bio, Inc.), according to the manufacturer's protocol. Subsequently, qPCR was performed on an ABI 7300 Real-Time PCR system SYBR Premix ExTaq (Takara Bio, Inc.). The thermocycling conditions were as follows: Pre-incubation at 94°C for 30 sec followed by 39 cycles of amplification at 94°C for 5 sec and 56°C for 30 sec. The sequences of the primers were: Vascular endothelial (VE)-cadherin forward, 5′-CTTCACCCAGACCAAGTACACA-3′ and reverse, 5′-AGGGCTCATGTATCGGAGGT-3′; α-SMA forward, 5′-ACCATCGGGAATGAACGCTT-3′ and reverse, 5′-CTGTCAGCAATGCCTGGGTA-3′; snail forward, 5′-GAAGATGCACATCCGAAGCC-3′ and reverse, 5′-TTCACATCCGAGTGGGTCTG-3′; and GAPDH forward, 5′-TGTGAACGGATTTGGCCGTA-3′ and reverse, 5′-GATGGTGATGGGTTTCCCGT-3′. GAPDH was used as the loading control.

Immunofluorescence staining

Cells at a density of 1×104/ml were seeded onto slides and fixed with 4% paraformaldehyde (200 μl/well) for 10 min, and washed twice with PBS, 10 min each time. The cell membrane was treated with 0.1% Triton X-100 in PBS (200 μl/well) at room temperature for 10 min, and then washed twice with PBS, 10 min each time. The plates were agitated gently several times by hand to drain the liquid and cells were then blocked with 4% goat serum in PBS at room temperature for 30 min. The liquid was gently removed, anti-VE-cadherin and anti-α-SMA primary antibodies were added, and the cells were incubated at 37°C for 1 h or at 4°C overnight. The primary antibodies used were: Anti-VE-cadherin (1:100; Cell Signaling Technology, Inc.; cat. no. 2500) and anti-α-SMA (1:100, Cell Signaling Technology, Inc.; cat. no. 19245). DAPI (1:1,000; Sigma-Aldrich; Merck KGaA; cat. no. AC23192) was used to stain the nuclei at room temperature for 10 min. Subsequently, the cells were washed three times with PBS and then incubated with fluorescein-labeled secondary antibodies (Cell Signaling Technology, Inc.; 1:100; cat. nos. 4416 and 12877) at 37°C for 60 min. Following washing with 0.01 M PBS three times, 5 min per wash, anti-fade reagent was used to mount the slides at 4°C. Finally, cells were imaged using a fluorescence microscope (magnification, ×200).

Wound healing assay

HUVECs were seeded in 24-well plates and grown overnight to confluence. For EndMT induction, cells were treated with 10 ng/ml TGF-β1, 2 μM LY21097612 and 0.25, 1 or 4 μg/l EOFAZ for 72 h at 37°C. The monolayer of cells was scratched using a 20-μl pipette tip to create the wound. Floating cells were removed by washing twice with PBS and serum-free Endothelial Cell Medium was added. Wound closure rate was assessed using images captured with a light microscope (Leika DM3000k magnification, ×100) after 24 h. The motility index=(Mean of 0 h migration distance-mean of 12 h migration distance)/mean of 0 h migration distance.

Transwell migration assay

Cells were trypsinized, centrifuged at 150 × g for 5 min at room temperature, washed with PBS 1-2 times and resuspended in serum-free culture medium containing BSA. A cell suspension of 100 μl (2×104 cells) was added to the upper chamber of the Transwell insert, and 400 μl medium containing 10% FBS was added to the lower chamber. The cells were cultured for 12 h, after which the Transwell chamber was removed, the medium was discarded, and cells were washed twice with PBS and fixed with paraformaldehyde for 15 min at room temperature. Subsequently, the cells were stained for 10 min with 0.1% crystal violet at room temperature. The unmigrated cells in the upper layer were gently removed with a cotton swab and the chamber was then washed three times with PBS. Cells were observed under an inverted light microscope (magnification, ×100) and the number of cells in five randomly chosen fields were counted. The number of migrated cells was quantified by manual counting, and the motility index was determined using the following formula: Motility index=the number of migrated cells in the experimental group/the number of cells that migrated in the control group.

Immunoprecipitation

HUVECs were harvested 72 h after treatment with TGF-β1 or EOFAZ at 37°C for 2 h, the supernatant was removed, the cells were lysed with NP-40 lysis buffer (Beyotime Institute of Biotechnology) containing protease inhibitors, and the lysate was centrifuged on ice at 12,000 × g for 30 min at 4°C. A small quantity of lysate was used for western blotting, and 1 μg antibody (anti-KLF4; 1:200; Cell Signaling Technology, Inc.; cat. no. 12173S) was added to the remaining cell lysate and incubated overnight at 4°C with gentle agitation. A total of 10 μl protein A agarose beads were washed with lysis buffer and centrifuged at 300 × g for 3 min at 4°C. The washed protein A agarose beads were then added to the cell lysate-antibody solution and incubated at 4°C with gentle agitation for 2-4 h. Following immunoprecipitation, agarose beads were pelleted by centrifugation at 300 × g for 3 min at 4°C. The supernatant was aspirated, and the agarose beads were washed 3-4 times with 1 ml lysis buffer. Finally, 15 μl 2x SDS sample buffer was added, and the samples were boiled for 5 min. Samples were subsequently subjected to western blotting, as previously described.

Small interference (si)RNA transfection

HUVECs were plated in 6-well plates at a density of 2×106 cells/well and transfected with siRNA. Briefly, 5 μl KLF4 and control siRNA was mixed with RPMI-1640 media (Wisent, Inc.). Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) was mixed with the RPMI-1640 media and the mixture was left to stand for 20 min at room temperature. The Lipofectamine®-siRNA mixture was then added to each well containing cells and the cells were incubated for 6 h. Medium was then replaced with antibiotic-free Endothelial Cell Medium supplemented with 5% FBS for 24 h, after which cells were used for subsequent experiments. The KLF4 siRNA sequences were: Sense 5′-AGAGTTCCCATCTCAAGGCA-3′ and antisense, 5′-GTCAGTTCATCTGAGCGGG-3′. The sequences of the control non-specific siRNA were: Sense 5′-CGUUUGUUCGCUUCCUGAGTT-3′ and antisense, 5′-CUCAGGAAGCGAACAAACGTG-3′.

Statistical analysis

All data are expressed as the mean ± standard deviation of at least three repeats. SPSS v.21.0 (IBM, Corp.) was used for statistical analysis. Comparisons among multiple groups were analyzed using one-way analysis of variance followed by Bonferroni's post hoc test. Comparisons between two groups were analyzed using a two-tailed Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

EndMT is induced by TGF-β1 in HUVECs

TGF-β1 is a powerful factor for induction of EndMT, although the effect depends of the treatment time and dose in different models (17). Relative VE-cadherin and α-SMA expression levels were determined by western blotting. After cells were treated in Endothelial Cell Medium with or without 10 ng/ml of TGF-β1, the expression levels of VE-cadherin and α-SMA changed in a time-dependent manner. As presented in Fig. 1A, the HUVECs were treated with 10 ng/ml TGF-β1 for various durations, and the VE-cadherin and α-SMA levels were most significantly changed at 72 h, thus treatment with 10 ng/ml TGF-β1 for 72 h was selected to establish the EndMT model in following experiments. In addition, the morphological EndMT-associated changes in HUVECs by TGF-β1 treatment for 72 h were observed using an inverted microscope (Fig. 1B). Cells in the control group demonstrated a cobble-stone like morphology and the junctions between the cells were dense. The cells induced by TGF-β1 were primarily fibroblast in morphology, the distance between cells was increased, and additionally the presence of filamentous pseudopod protrusions was observed. Taken together, TGF-β1 induced morphological and phenotypic changes in HUVECs to a more mesenchymal-like phenotype, suggesting the cells had undergone EndMT and an EndMT model had been established.

EOFAZ inhibits TGF-β1-induced EndMT in HUVECs

Subsequently, HUVECs were exposed to 10 ng/ml TGF-β1 for 72 h for EndMT induction. Additionally, the cells were pretreated with EOFAZ and LY2109761 (a TGF-β1 inhibitor) for 2 h and then stimulated with TGF-β1. The results of RT-qPCR and western blotting analysis demonstrated that EOFAZ, like LY2109761, significantly reversed the TGF-β1-iduced increase in α-SMA and Snail expression and the TGF-β1-induced decrease in VE-cadherin expression (Fig. 2A and B). Similar changes were observed by immunofluorescence (Fig. 2C).

EOFAZ inhibits the expression of EndMT markers in cell migration

During the EndMT process initiated in endothelial cells, the cellular migratory ability is promoted (14). A wound healing assay was used to observe the effects of EOFAZ on the migration of endothelial cells treated with TGF-β1. The results demonstrated that the cell migration rate significantly increased in the TGF-β1-treated group compared with the control group. In addition, scratch immunofluorescence staining demonstrated that VE-cadherin was downregulated, while α-SMA was upregulated in the migrating cells after TGF-β1 treatment. The number of migrated cells was significantly reduced in cells pre-treated with LY2109761 and EOFAZ, and the expression of VE-cadherin was increased and the expression of α-SMA was decreased compared with the cells only treated with TGF-β1 (Fig. 3A and B). These results suggest that the TGF-β1-induced EndMT is associated with the enhanced migratory capacity of mesenchymal cells, whereas EOFAZ inhibits this migratory capacity.

Effect of EOFAZ on the expression of KLF4 and NF-κB p-p65, and on the binding between KLF4 and NF-κB induced by TGF-β1 in HUVECs

As presented in Fig. 3C and D, KLF4 and NF-κB p-p65 expression levels were upregulated in the TGF-β1-induced cells compared with the control. The p-p65/p65 ratio is appropriate to evaluate the activation of NF-κB (27). The results demonstrated that EOFAZ and LY2109761 significantly decreased the TGF-β1-induced increase in p-p65/p65 ratio, which indicated the inhibitory effects of EOFAZ on NF-κB nuclear translocation and activation. The TGF-β1-induced increase in KLF4 was also decreased in the presence of EOFAZ or LY2109761 (Fig. 3C and D). These results suggested that EOFAZ inhibits TGF-β1-induced KLF4 and NF-κB activation. A previous study reported that NF-κB is one of key regulators during EndMT in endothelial cells; activation or inhibition of the NF-κB signaling pathway resulted in EndMT reversal (28). In addition, it has been reported that NF-κB is essential for both the induction and maintenance of EndMT (29).

A previous study suggested that KLF4 inhibits TNF-α-mediated induction of vascular cell adhesion protein 1 (VCAM1) expression by blocking the binding of p65 to the VCAM1 promoter through the physical association between KLF4 and p65 (30-32). Therefore, the present study hypothesized that KLF4 similarly regulates the activation of NF-κB in the TGF-β1-induced EndMT process. To confirm binding between KLF4 and NF-κB in HUVECs treated with TGF-β1 in the absence or presence of EOFAZ, co-immunoprecipitation studies were performed. The results demonstrated that TGF-β1 enhanced the binding between KLF4 and NF-κB, However, EOFAZ treatment reduced the TGF-β1-induced interaction between KLF4 and NF-κB, although it was not determined if this effect was direct or indirect (Fig. 3E).

Effect of KLF4 siRNA transfection on KLF4 expression

Previously, it has been suggested that KLF4 serves an important role in EndMT, which is involved in the development and progression of cerebral cavernous malformations. However, to the best of our knowledge, whether the effect of EOFAZ on EndMT is mediated by KLF4 in HUVECs has not been assessed. To investigate the relationship between EOFAZ and KLF4, a KLF4-specific siRNA was utilized to knockdown the expression of KLF4 (Fig. 4A and B). The results demonstrated that the green fluorescence-labeled siRNA was successfully transferred into HUVECs, and western blotting analysis demonstrated that KLF4 expression was significantly reduced following transfection with KLF4 siRNA.

Effect of EOFAZ on TGF-β1-induced HUVEC migration following KLF4-knockdown

To evaluate HUVEC migration following transfection with KLF4 siRNA, wound healing and Transwell assays were performed. The KLF4 expression was significantly downregulated by siRNA interference (Fig. 4A and B). As shown in Fig. 4C, KLF4-knockdown significantly reduced TGF-β1-induced cell migration. Transwell assays revealed that the number of migrated cells was significantly decreased in the KLF4-knockdown cells compared with the TGF-β1 group, and EOFAZ treatment decreased the number of migrated cells under TGF-β1 stimulation (Fig. 4D). These results suggest that EOFAZ inhibition of TGF-β1-mediated EndMT induction in HUVECs may occur through downregulation of KLF4 expression.

EOFAZ inhibits TGF-β1-induced EndMT via KLF4

To determine the effect of KLF4 siRNA on EOFAZ-mediated inhibition of TGF-β1-induced EndMT, the expression of α-SMA and VE-cadherin was determined by immunofluorescence analysis. As presented in Fig. 5A, VE-cadherin expression was downregulated and α-SMA expression was upregulated in the TGF-β1 group. However, following KLF-knockdown and EOFAZ treatment, the effects on VE-cadherin and α-SMA expression were reversed compared with the TGF-β1-treated group. Furthermore, the expression levels of α-SMA, Snail, VE-cadherin and NF-κB p-p65/p65 ratio were determined by western blotting, and α-SMA, Snail, VE-cadherin were analyzed by RT-qPCR. Similar results were observed for the changes in VE-cadherin and α-SMA expression. In addition, the Snail expression and NF-κB p-p65/p65 ratio levels were significantly repressed when exposed to siKLF and EOFAZ (Fig. 5B). Alterations in mRNA expression levels of VE-cadherin, α-SMA and Snail were consistent with the western blotting (Fig. 5C). Together, these results demonstrated that KLF4-knockdown inhibited EndMT, and there were no significant differences between EOFAZ treatment and siKLF4 treatment. These results suggest that EOFAZ regulates EndMT via KLF4.

Discussion

Endothelial cells are highly sensitive to stimuli and physiological cues in the immediate environment (33). Thus, injury of the vascular endothelium is an initial step in the pathogenesis of vascular remodeling and contributes to vascular remodeling as a source of SM-like cells, as endothelial cells can acquire a fibro-proliferative mesenchymal phenotype through EndMT under specific pathological conditions, including shear stress, oxidative stress and atherosclerosis (34). The present study demonstrated that endothelial cells differentiate towards a SM-like phenotype via EndMT trans-differentiation when treated with TGF-β1, and that the KLF4 signaling pathway was involved in this process. Additionally, it was shown that EOFAZ could inhibit TGF-β1-induced EndMT via downregulation of KLF4.

EndMT is a newly recognized, fundamental biological process involved in development and tissue regeneration, as well as pathological processes, including certain complications of diabetes, fibrosis, PAH, atherosclerosis (35-38), tumor metastasis and development of fibrotic lesions in certain vital organs (32). EndMT is tightly controlled by a series of signaling networks, similar to the epithelial-to-mesenchymal transition (38). EndMT is involved in intima formation and pulmonary vascular angiogenesis (16) and may contribute to cardiac fibrosis (39); similar results have been observed in diabetes mellitus-induced cardiac fibrosis (40) and further studies confirmed that EndMT contributes to fibrogenesis associated with exposure to TGF-β1 (41).

Consistent with these previous results, the present study demonstrated that HUVECs underwent phenotypic and biological behavioral transitions following TGF-β1 exposure consistent with fibrogenesis. TGF-β1 is involved in a wide range of pathological conditions, including a number of different types of cancer (42), myocardial infarction (43), cerebral cavernous malformations (44), vascular remolding and different types of organ fibrosis (40,41). During this process, TGF-β1 activates EndMT via multiple pathways including the canonical Smad-dependent and non-canonical Smad-independent pathways, and the bFGF, Wnt and Notch signaling pathways (45). A previous study has demonstrated the contribution of TGF-β1 to EndMT in the regulation of cardiovascular diseases (46). Therefore, any potential target to inhibit or reduce TGF-β1/Snail signaling may exhibit potential as an anti-proliferative reagent. The present study demonstrated that TGF-β1 treatment increased the expression of Snail, α-SMA and NF-κB p-p65/p65 ratio, and reduced the expression of VE-cadherin, suggesting that TGF-β1 can induce EndMT. Treatment with EOFAZ inhibited TGF-β1-mediated induction of EndMT. Additionally, the increases in Snail and α-SMA expression and NF-κB p-p65/p65 ratio in TGF-β1-treated cells were significantly decreased following treatment with EOFAZ. These results suggest that EOFAZ may be a promising agent for ameliorating EndMT-related diseases.

KLF4 is an evolutionarily conserved zinc finger-containing transcription factor and is widely expressed in a range of tissues where it regulates a range of cellular processes, including cell growth, proliferation and differentiation (47). KLF4 has a critical regulatory effect on a range of functions, including maintaining intestinal epithelial homeostasis, proliferation, migration and tube formation in human retinal microvascular endothelial cells, and inhibiting NF-κB transcriptional activity and the expression of downstream prothrombotic, proinflam-matory genes (48-51). KLF4 mediates an increase in the lesions and the pseudo-normal vasculature of endothelial-specific CCM1-knockout mice (32). KLF4 also regulates VE-cadherin expression and endothelial barrier function, angiogenesis via the Notch signaling pathway, vascular tone and permeability, apoptosis, inflammation and atherothrombosis (50). KLF4 can contribute to epithelial-mesenchymal transition during the development and progression of cancer (51). Additionally, previous studies have shown that KLF4 is involved in EndMT in cerebral cavernous malformations (47). In the present study, the results demonstrated that KLF4 was excessively activated during TGF-β1 exposure, which resulted in the increase in the degree of EndMT in HUVECs. KLF4-knockdown inhibited EndMT and decreased the protein and mRNA expression levels of Snail and α-SMA. whilst increased VE-cadherin expression. Following treatment with EOFAZ, the processes associated with EndMT were reversed. These results suggest that EOFAZ may inhibit TGF-β1-induced EndMT through inhibition of KLF4.

In conclusion, EOFAZ significantly inhibited the extent of EndMT following TGF-β1 exposure, a protective effect that was mediated through reduction in KLF4 activity. In sum, a graphical abstract is proposed and presented in Fig. 6, which summarizes the whole investigation. The results of the present study contribute to the understanding of the molecular mechanisms underlying EndMT and reveal novel targets, which may be the basis of potential therapeutic strategies.

Funding

This study was supported by the National Natural Science Foundation of China (grant nos. 81560811, 81760725 and U1812403-4-4), the Guizhou Provincial Key Technology R&D Program [grant no. (2020)4Y093], the Scientific and Technological Founding of Guizhou Province [grant no. QKHJ (2016)1128], the Foundation of Science and Technology Department Cooperation Program of Guizhou Province (grant no. 2016-7358) and Guizhou Graduate Research (grant no. 0711007020259).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

YZ performed part of the experiments and wrote the manuscript. CL performed the experiments. YH designed the study and analyzed the data. SZ performed the experiments. YX and YC analyzed the data. FJ and LT extracted the essential oil. XS designed the experiment and revised the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors wish to thank Dr Di Pan (The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, China) for their kind linguistic advice and technical assistance.

References

1 

Chen Q, Zhang H, Liu Y, Adams S, Eilken H, Stehling M, Corada M, Dejana E, Zhou B and Adams RH: Endothelial cells are progenitors of cardiac pericytes and vascular smooth muscle cells. Nat Commun. 7:124222016. View Article : Google Scholar : PubMed/NCBI

2 

Gibbons GH and Dzau VJ: The emerging concept of vascular remodeling. N Engl J Med. 330:1431–1438. 1994. View Article : Google Scholar : PubMed/NCBI

3 

Davis GE and Senger DR: Endothelial extracellular matrix: Biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ Res. 97:1093–1107. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Karimi Galougahi K, Ashley EA and Ali ZA: Redox regulation of vascular remodeling. Cell Mol Life Sci. 73:349–363. 2016. View Article : Google Scholar

5 

Zhang B, Niu W, Dong HY, Liu ML, Luo Y and Li ZC: Hypoxia induces endothelial-mesenchymal transition in pulmonary vascular remodeling. Int J Mol Med. 42:270–278. 2018.PubMed/NCBI

6 

Jackson AO, Zhang J, Jiang Z and Yin K: Endothelial-to-mesenchymal transition: A novel therapeutic target for cardiovascular diseases. Trends Cardiovasc Med. 27:383–393. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Zhang L, Li YM, Zeng XX, Wang XY, Chen SK, Gui LX and Lin MJ: Galectin-3-mediated transdifferentiation of pulmonary artery endothelial cells contributes to hypoxic pulmonary vascular remodeling. Cell Physiol Biochem. 51:763–777. 2018. View Article : Google Scholar

8 

Wesseling M, Sakkers TR, de Jager SCA, Pasterkamp G and Goumans MJ: The morphological and molecular mechanisms of epithelial/endothelial-to-mesenchymal transition and its involvement in atherosclerosis. Vascul Pharmacol. 106:1–8. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Zhang Y, Wu X, Li Y, Zhang H, Li Z, Zhang Y, Zhang L, Ju J, Liu X, Chen X, et al: Endothelial to mesenchymal transition contributes to arsenic-trioxide-induced cardiac fibrosis. Sci Rep. 6:337872016. View Article : Google Scholar : PubMed/NCBI

10 

Tian D, Zeng X, Wang W, Wang Z, Zhang Y and Wang Y: Protective effect of rapamycin on endothelial-to-mesenchymal transition in HUVECs through the Notch signaling pathway. Vascul Pharmacol. 113:20–26. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Kanasaki K, Taduri G and Koya D: Diabetic nephropathy: The role of inflammation in fibroblast activation and kidney fibrosis. Front Endocrinol (Lausanne). 4:72013. View Article : Google Scholar

12 

Xiong J, Kawagishi H, Yan Y, Liu J, Wells QS, Edmunds LR, Fergusson MM, Yu ZX, Rovira II, Brittain EL, et al: A metabolic basis for endothelial-to-mesenchymal transition. Mol Cell. 69:689–698.e7. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Yu QC, Song W, Wang D and Zeng YA: Identification of blood vascular endothelial stem cells by the expression of protein C receptor. Cell Res. 26:1079–1098. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Cooley BC, Nevado J, Mellad J, Yang D, St Hilaire C, Negro A, Fang F, Chen G, San H, Walts AD, et al: TGF-β signaling mediates endothelial-to-mesenchymal transition (EndMT) during vein graft remodeling. Sci Transl Med. 6:227ra342014. View Article : Google Scholar

15 

Vanchin B, Offringa E, Friedrich J, Brinker MG, Kiers B, Pereira AC, Harmsen MC, Moonen JA and Krenning G: MicroRNA-374b induces endothelial-to-mesenchymal transition and early lesion formation through the inhibition of MAPK7 signaling. J Pathol. 247:456–470. 2019. View Article : Google Scholar :

16 

Suzuki T, Carrier EJ, Talati MH, Rathinasabapathy A, Chen X, Nishimura R, Tada Y, Tatsumi K and West J: Isolation and characterization of endothelial-to-mesenchymal transition cells in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol. 314:L118–L126. 2018. View Article : Google Scholar :

17 

Li J, Xiong J, Yang B, Zhou Q, Wu Y, Luo H, Zhou H, Liu N, Li Y, Song Z and Zheng Q: Endothelial cell apoptosis induces TGF-β signaling-dependent host endothelial-mesenchymal transition to promote transplant arteriosclerosis. Am J Transplant. 15:3095–3111. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Wang J, Yan G, Guo H, Zhu Y, Shui X, He Y, Chen C and Lei W: ITE promotes hypoxia-induced transdifferentiation of human pulmonary arterial endothelial cells possibly by activating transforming growth factor-β/Smads and MAPK/ERK pathways. J Cell Biochem. 120:19567–19577. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Zuo XX, Yang Y, Zhang Y, Zhang ZG, Wang XF and Shi YG: Platelets promote breast cancer cell MCF-7 metastasis by direct interaction: Surface integrin α2β1-contacting-mediated activation of Wnt-β-catenin pathway. Cell Commun Signal. 17:1422019. View Article : Google Scholar

20 

Shen XC, Tao L, Li WK, Zhang YY, Luo H and Xia YY: Evidence-based antioxidant activity of the essential oil from fructus A. Zerumbet on cultured human umbilical vein endothelial cells' injury induced by ox-LDL. BMC Complement Altern Med. 12:1742012. View Article : Google Scholar : PubMed/NCBI

21 

Tao L, Hu HS and Shen XC: Endothelium-dependent vasodilatation effects of the essential oil from Fructus alpiniae zerumbet (EOFAZ) on rat thoracic aortic rings in vitro. Phytomedicine. 20:387–393. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Chen Y, Li D, Xu Y, Zhang Y, Tao L, Li S, Jiang Y and Shen X: Essential oils from fructus A. Zerumbet protect human aortic endothelial cells from apoptosis induced by Ox-LDL in vitro. Evid Based Complement Alternat Med. 2014:9568242014. View Article : Google Scholar

23 

Yob NJ, Jofrry SM, Affandi MM, The LK, Salleh MZ and Zakaria ZA: Zingiber zerumbet (L.) Smith: A review of its ethnomedicinal, chemical, and pharmacological uses. Evid Based Complement Alternat Med. 2011:5432162011. View Article : Google Scholar : PubMed/NCBI

24 

Chompoo J, Upadhyay A, Fukuta M and Tawata S: Effect of Alpinia zerumbet components on antioxidant and skin diseases-related enzymes. BMC Complement Altern Med. 12:1062012. View Article : Google Scholar : PubMed/NCBI

25 

de Moura RS, Emiliano AF, de Carvalho LC, Souza MA, Guedes DC, Tano T and Resende AC: Antihypertensive and endothelium-dependent vasodilator effects of Alpinia zerumbet, a medicinal plant. J Cardiovasc Pharmacol. 46:288–294. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Linghu K, Lin D, Yang H, Xu Y, Zhang Y, Tao L, Chen Y and Shen X: Ameliorating effects of 1,8-cineole on LPS-induced human umbilical vein endothelial cell injury by suppressing NF-κB signaling in vitro. Eur J Pharmacol. 789:195–201. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Yang H, Fang Z, Qu X, Zhang X and Wang Y: Procyanidin compound (PC) suppresses lipopolysaccharide-induced cervical cancer cell proliferation through blocking the TLR4/NF-κB pathway. Cancer Manag Res. 12:497–509. 2020. View Article : Google Scholar :

28 

Shu Y, Liu Y, Li X, Cao L, Yuan X, Li W and Cao Q: Aspirin-triggered resolvin D1 inhibits TGF-β1-induced EndMT through increasing the expression of Smad7 and is closely related to oxidative stress. Biomol Ther (Seoul). 24:132–139. 2016. View Article : Google Scholar

29 

Mahler GJ, Farrar EJ and Butcher JT: Inflammatory cytokines promote mesenchymal transformation in embryonic and adult valve endothelial cells. Arterioscler Thromb Vasc Biol. 33:121–130. 2013. View Article : Google Scholar :

30 

Yoshida T, Yamashita M, Iwai M and Hayashi M: Endothelial Krüppel-like factor 4 mediates the protective effect of statins against ischemic AKI. J Am Soc Nephrol. 27:1379–1388. 2016. View Article : Google Scholar

31 

Cuttano R, Rudini N, Bravi L, Corada M, Giampietro C, Papa E, Morini MF, Maddaluno L, Baeyens N, Adams RH, et al: KLF4 is a key determinant in the development and progression of cerebral cavernous malformations. EMBO Mol Med. 8:6–24. 2016. View Article : Google Scholar :

32 

Zhou Z, Tang AT, Wong WY, Bamezai S, Goddard LM, Shenkar R, Zhou S, Yang J, Wright AC, Foley M, et al: Cerebral cavernous malformations arise from endothelial gain of MEKK3-KLF2/4 signalling. Nature. 532:122–126. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Huang N, Xu Y, Zhou H, Lin D, Zhang B, Zhang Y, Pan D, Tao L, Liu X and Shen X: Essential oil from fructus alpiniae zerumbet protects human umbilical vein endothelial cells in vitro from injury induced by high glucose levels by suppressing nuclear transcription factor-kappa B signaling. Med Sci Monit. 23:4760–4767. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Wu M, Liu L, Xing Y, Yang S, Li H and Cao Y: Roles and mechanisms of hawthorn and its extracts on atherosclerosis: A review. Front Pharmacol. 11:1182020. View Article : Google Scholar : PubMed/NCBI

35 

Yu W, Liu Z, An S, Zhao J, Xiao L, Gou Y, Lin Y and Wang J: The endothelial-mesenchymal transition (EndMT) and tissue regeneration. Curr Stem Cell Res Ther. 9:196–204. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Cruz-Solbes AS and Youker K: Epithelial to mesenchymal transition (EMT) and endothelial to mesenchymal transition (EndMT): Role and implications in kidney fibrosis. Results Probl Cell Differ. 60:345–372. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Pardali E, Sanchez-Duffhues G, Gomez-Puerto MC and Ten Dijke P: TGF-β-induced endothelial-mesenchymal transition in fibrotic diseases. Int J Mol Sci. 18:pii: E2157. 2017. View Article : Google Scholar

38 

Hao YM, Yuan HQ, Ren Z, Qu SL, Liu LS, Dang H, Yin K, Fu M and Jiang ZS: Endothelial to mesenchymal transition in atherosclerotic vascular remodeling. Clin Chim Acta. 490:34–38. 2019. View Article : Google Scholar

39 

Geng H and Guan J: MiR-18a-5p inhibits endothelial-mesen-chymal transition and cardiac fibrosis through the Notch2 pathway. Biochem Biophys Res Commun. 491:329–336. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Chen XY, Lv RJ, Zhang W, Yan YG, Li P, Dong WQ, Liu X, Liang ES, Tian HL, Lu QH and Zhang MX: Inhibition of myocyte-specific enhancer factor 2A improved diabetic cardiac fibrosis partially by regulating endothelial-to-mesenchymal transition. Oncotarget. 7:31053–31066. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Guan S and Zhou J: CXCR7 attenuates the TGF-β-induced endothelial-to-mesenchymal transition and pulmonary fibrosis. Mol Biosyst. 13:2116–2124. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Akhurst RJ: Targeting TGF-β signaling for therapeutic gain. Cold Spring Harb Perspect Biol. 9:pii: a022301. 2017. View Article : Google Scholar

43 

Bakhta O, Blanchard S, Guihot AL, Tamareille S, Mirebeau-Prunier D, Jeannin P and Prunier F: Cardioprotective role of colchicine against inflammatory injury in a rat model of acute myocardial infarction. J Cardiovasc Pharmacol Ther. 23:446–455. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Wetzel-Strong SE, Detter MR and Marchuk DA: The pathobiology of vascular malformations: Insights from human and model organism genetics. J Pathol. 241:281–293. 2017. View Article : Google Scholar

45 

Fu Y, Chang A, Chang L, Niessen K, Eapen S, Setiadi A and Karsan A: Differential regulation of transforming growth factor beta signaling pathways by Notch in human endothelial cells. J Biol Chem. 284:19452–19462. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Cunha SI, Magnusson PU, Dejana E and Lampugnani MG: Deregulated TGF-β/BMP signaling in vascular malformations. Circ Res. 121:981–999. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Ghaleb AM and Yang VW: Krüppel-like factor 4 (KLF4): What we currently know. Gene. 611:27–37. 2017. View Article : Google Scholar : PubMed/NCBI

48 

McConnell BB, Ghaleb AM, Nandan MO and Yang VW: The diverse functions of Krüppel-like factors 4 and 5 in epithelial biology and pathobiology. Bioessays. 29:549–557. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Zhang Y, Lam O, Nguyen MT, Ng G, Pear WS, Ai W, Wang IJ, Kao WW and Liu CY: Mastermind-like transcriptional co-activator-mediated Notch signaling is indispensable for maintaining conjunctival epithelial identity. Development. 140:594–605. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Wang Y, Yang C, Gu Q, Sims M, Gu W, Pfeffer LM and Yue J: KLF4 promotes angiogenesis by activating VEGF signaling in human retinal microvascular endothelial cells. PLoS One. 10:e01303412015. View Article : Google Scholar : PubMed/NCBI

51 

Ji L, Zhao G, Zhang P, Huo W, Dong P, Watari H, Jia L, Pfeffer LM, Yue J and Zheng J: Knockout of MTF1 inhibits the epithelial to mesenchymal transition in ovarian cancer cells. J Cancer. 9:4578–4585. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 46 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Li C, Huang Y, Zhao S, Xu Y, Chen Y, Jiang F, Tao L and Shen X: EOFAZ inhibits endothelial‑to‑mesenchymal transition through downregulation of KLF4. Int J Mol Med 46: 300-310, 2020
APA
Zhang, Y., Li, C., Huang, Y., Zhao, S., Xu, Y., Chen, Y. ... Shen, X. (2020). EOFAZ inhibits endothelial‑to‑mesenchymal transition through downregulation of KLF4. International Journal of Molecular Medicine, 46, 300-310. https://doi.org/10.3892/ijmm.2020.4572
MLA
Zhang, Y., Li, C., Huang, Y., Zhao, S., Xu, Y., Chen, Y., Jiang, F., Tao, L., Shen, X."EOFAZ inhibits endothelial‑to‑mesenchymal transition through downregulation of KLF4". International Journal of Molecular Medicine 46.1 (2020): 300-310.
Chicago
Zhang, Y., Li, C., Huang, Y., Zhao, S., Xu, Y., Chen, Y., Jiang, F., Tao, L., Shen, X."EOFAZ inhibits endothelial‑to‑mesenchymal transition through downregulation of KLF4". International Journal of Molecular Medicine 46, no. 1 (2020): 300-310. https://doi.org/10.3892/ijmm.2020.4572