Open Access

SESN1 attenuates the Ox‑LDL‑induced inflammation, apoptosis and endothelial‑mesenchymal transition of human umbilical vein endothelial cells by regulating AMPK/SIRT1/LOX1 signaling

  • Authors:
    • Feng Gao
    • Yongcheng Zhao
    • Bin Zhang
    • Chunwei Xiao
    • Zhanfa Sun
    • Yuan Gao
    • Xueyong Dou
  • View Affiliations

  • Published online on: March 11, 2022     https://doi.org/10.3892/mmr.2022.12678
  • Article Number: 161
  • Copyright: © Gao 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

Endothelial cells are an important component of the heart and vasculature and form a crucial link between the cardiovascular system and the immune system. Sestrin 1 (SESN1) has an important role in atherosclerosis by inhibiting NOD‑like receptor family pyrin domain containing 3 inflammasome activation. However, whether SESN1 is involved in human umbilical vein endothelial cell (HUVEC) injury caused by atherosclerosis has remained to be elucidated. The present study aimed to investigate the functions of SESN1 in the inflammatory response, apoptosis and endothelial‑mesenchymal transition (EndMT) of HUVECs following stimulation with oxidized low‑density lipoprotein (Ox‑LDL). SESN1 expression at the mRNA and protein levels was detected using reverse transcription‑quantitative PCR (RT‑qPCR) and western blot analysis. Following SESN1 overexpression in Ox‑LDL‑stimulated HUVECs, cell viability was determined using a Cell Counting Kit‑8 assay. Terminal deoxynucleotidyl transferase‑mediated nick‑end labeling staining was employed to detect cell apoptosis and western blot analysis was used to determine the levels of apoptosis‑related proteins. RT‑qPCR, ELISA and western blot were utilized to determine the levels of inflammatory factors. Immunofluorescence staining, RT‑qPCR and western blot analysis were employed to assess the EndMT of Ox‑LDL‑stimulated HUVECs. The results revealed that SESN1 exhibited a low expression in HUVECs following Ox‑LDL stimulation. SESN1 overexpression suppressed inflammation, apoptosis and EndMT in Ox‑LDL‑induced HUVECs. In addition, SESN1 stimulated adenosine monophosphate‑activated protein kinase catalytic subunit α1/sirtuin 1 signaling to suppress Ox‑LDL receptor‑1 expression. An AMPK and SIRT1 inhibitor reversed the effects of SESN1 overexpression on the inflammatory response, apoptosis and EndMT of HUVECs exposed to Ox‑LDL. Taken together, the present study demonstrated that SENS1 exerts a suppressive effect on Ox‑LDL‑induced inflammation, apoptosis and EndMT of HUVECs, suggesting that SENS1 may be used as a novel biomarker for endothelial injury‑related disorders.

Introduction

Atherosclerosis, the leading cause of cardiovascular disease, is a chronic inflammatory disorder, during which the dysfunction of endothelial cells is the first step (13). Accumulating evidence has confirmed that endothelial cells have a major role in the development of atherosclerosis and that endothelial dysfunction and damage have an initial role (4,5). Under pathological conditions, injured endothelial cells display great plasticity by differentiating into healthy and functional endothelial cells (6). Atherosclerosis is a persistent inflammatory condition and vascular endothelial cell apoptosis is considered the initiation factor of the occurrence, development and pathogenesis of atherosclerosis (7,8). Furthermore, endothelial-mesenchymal transition (EndMT) helps endothelial cells to acquire a mesenchymal phenotype (9). Oxidized low-density lipoprotein (Ox-LDL) is a common factor in the establishment of experimental models of atherosclerosis, which may induce endothelial cell apoptosis and act as an essential risk factor for the formation of atherosclerosis (10,11).

Sestrins (SESNs) are a family of highly conserved stress-inducible proteins that regulate multiple cell homeostatic mechanisms (12). A deficiency in endogenous SESNs may lead to metabolic disorders, including insulin resistance, fat accumulation, mitochondrial dysfunction and oxidative damage (13). In addition, the SESN family are antioxidant enzymes and transcriptional targets of tumor suppressor protein p53 (14). As a primary member of the SESN family, SESN1 functions as a vital mediator in multiple human diseases, including human maxillary cancer, myoblast differentiation and diabetes (1517). Furthermore, SESN1 has been reported to activate the adenosine monophosphate-activated protein kinase catalytic subunit α1 (AMPK) signaling pathway and to function as a suppressor of the mechanistic target of rapamycin complex 1 kinase (18,19). Recent evidence has demonstrated that SESN1 suppresses the inflammation of macrophages in a murine atherosclerosis model (20). In addition, Zhang et al (21) revealed that SESN1 is expressed, at low levels, in endothelial cells subjected to mild shear stress. However, whether SESN1 is involved in human umbilical vein endothelial cell (HUVEC) injury induced by atherosclerosis has remained largely elusive.

SESN1 has been indicated to reduce myocardial hypertrophy and activate AMPK signaling (22). As metabolic sensors, AMPK and sirtuin 1 (SIRT1) have been identified as master regulators of metabolism (23). In a previous study, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that AMPK is able to regulate the expression of downstream SIRT1 genes, while SIRT1 may inhibit the expression of Ox-LDL receptor-1 (LOX1) (24). It is well established that endothelial cell injury is induced by Ox-LDL to drive the progression of atherosclerosis and LOX1 is the primary OxLDL receptor of endothelial cells, which may be activated by Ox-LDL, and it mediates cell damage in atherosclerosis (25). Therefore, blocking LOX1 expression is considered to be a major strategy in the management of atherosclerosis.

In the present study, the effects of SENS1 on the inflammation, apoptosis and EndMT of HUVECs exposed to Ox-LDL were investigated. In addition, the latent regulatory mechanisms among SESN1, AMPK/SIRT1 signaling and LOX1 were identified. The present results may highlight a novel target for the treatment of atherosclerosis.

Materials and methods

Bioinformatics analysis

The Human Protein Atlas (https://www.proteinatlas.org/) was used to assess SESN1 expression in HUVECs.

Cells and cell treatment

HUVECs (cat. no. KCB2012087YJ) obtained from the Kunming Cell Bank of the Type Culture Collection of the Chinese Academy of Sciences were maintained in Dulbecco's modified Eagle's medium Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (RWDLS) in a humidified atmosphere with 5% CO2 at 37°C. For the subsequent experiments, various concentrations (0, 25, 50, 75 and 100 µg/ml) of Ox-LDL (cat. no. 20605ES05; Yeasen Biotechnology Co., Ltd.) were used to pre-treat the HUVECs for 24 h according to a previous study (26). In addition, 15 µM SIRT1 inhibitor, nicotinamide (NAM; MilliporeSigma) (27) and 8 µM AMPK inhibitor (compound C; MilliporeSigma) were respectively utilized to pre-treat the cells for 30 min (28).

Cell Counting Kit-8 (CCK-8) assay

Cell viability was evaluated by the CCK-8 reagent (Beyotime Institute of Biotechnology) according to manufacturer's protocol. The HUVECs were seeded into 96-well plates at the density of 4×104 cells/well and cultured at 37°C overnight. Following the indicated treatments, CCK-8 solution (10 µl) was added to each well. Following incubation for 2 h at 37°C, cell proliferation was detected using a microplate reader (Bio-Rad Laboratories, Inc.) at an absorbance wavelength of 450 nm.

Reverse transcription-quantitative PCR (RT-qPCR)

Isolation of total RNA was performed using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) in compliance with the manufacturer's instructions. The temperature protocol for this step was as follows: 70°C for 5 min, 37°C for 5 min and 42°C for 1 h. Subsequently, complementary DNA was synthesized using the PrimeScript RT reagent (Takara Bio, Inc.). An ABI 7500 Fast Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) with a SYBR Green Master Mix Kit (Toyobo Co. Ltd.) were used for PCR analysis. The following thermocycling conditions were used: Initial denaturation at 95°C for 7 min; and 40 cycles of 95°C for 15 sec and 60°C for 30 sec; and a final extension at 72°C for 30 sec. The primers sequences were as follows: SESN1 forward, 5′-GCGACCAGGACGAGGAACTT-3′ and reverse, 5′-TGCATCTGTGCGTCTTCACT-3′; tumor necrosis factor (TNF)-α forward, 5′-CTGGGCAGGTCTACTTTGGG-3′ and reverse, 5′-CTGGAGGCCCCAGTTTGAAT-3′; interleukin (IL)-6 forward, 5′-TCCACAAGCGCCTTCGGTC-3′ and reverse, 5′-GGTCAGGGGTGGTTATTGCAT-3′; IL-1β forward, 5′-GCTCGCCAGTGAAATGATGG-3′ and reverse, 5′-TCGTGCACATAAGCCTCGTT-3′; α smooth muscle actin (α-SMA) forward, 5′-AAAGCAAGTCCTCCAGCGTT-3′ and reverse, 5′-TTAGTCCCGGGGATAGGCAA-3′; vimentin forward, 5′-AACTTAGGGGCGCTCTTGTC-3′ and reverse, 5′-ATTCAAGTCTCAGCGGGCTC-3′; CD31 (also known as platelet endothelial cell adhesion molecule 1) forward, 5′-AGAGAGGCTGCTGTCATTGC-3′ and reverse, 5′-GGCCCCTCAGAAGACAACAT-3′; von Willebrand factor (vWF) forward, 5′-CAACACCTGCATTTGCCGAA-3′ and reverse, 5′-ATGCGGAGGTCACCTTTCAG-3′; GAPDH forward, 5′-AATGGGCAGCCGTTAGGAAA-3′ and reverse, 5′-GCGCCCAATACGACCAAATC-3′. Relative gene expression was calculated using the 2−ΔΔCq method using GAPDH as an internal control (29).

Western blot analysis

Total protein was extracted from the HUVECs using lysis buffer (Beyotime Institute of Biotechnology). Total protein was quantified using a bicinchoninic acid kit (Beyotime Institute of Biotechnology). Subsequently, 40 µg of total protein was loaded per lane on a 10% SDS-PAGE gel, electroblotted onto polyvinylidene difluoride membranes (Thermo Fisher Scientific, Inc.) and blocked using 5% skimmed milk (MilliporeSigma) for 1 h at room temperature. The membranes were then incubated overnight at 4°C with the following primary antibodies: Anti-SESN1 (1:1,000 dilution; cat. no. ab134091), anti-inducible nitric oxide synthase (iNOS) (1:1,000 dilution; cat. no. ab178945), anti-total NF-κB p65 (t-NF-κB p65) (1:1,000 dilution; cat. no. ab32536), anti-phosphorylated NF-κB p65 (p-NF-κB p65) (1:2,000 dilution; cat. no. ab86299), anti-BCL2 associated X, apoptosis regulator (Bax) (1:1,000 dilution; cat. no. ab182733), anti-cleaved caspase-3 (1:400 dilution; cat. no. ab32042), anti-caspase-3 (1:5,000 dilution; cat. no. ab32351), anti-poly(adenosine diphosphate ribose) polymerase (PARP) (1:1,000 dilution; cat. no. ab191217) anti-cleaved PARP (1:1,000 dilution; cat. no. ab32064), anti-α-SMA (1:50 dilution; cat. no. ab150301), anti-CD31 (1:1,000 dilution; cat. no. ab9498), anti-vimentin (1:1,000 dilution; cat. no. ab92547), anti-vWF (1:1,000 dilution; cat. no. ab287962), anti-p-AMPK (1:500 dilution; cat. no. ab131357), anti-AMPK (1:500 dilution; cat. no. ab3759), anti-SIRT1 (1:1,000 dilution; cat. no. ab189494), anti-LOX1 (1:1,000 dilution; cat. no. ab214427) and anti-β-actin (1:1,000 dilution; cat. no. ab8227; all from Abcam) and anti-B cell lymphoma-2 (Bcl-2) (1:1,000 dilution; cat. no. 15071, Cell Signaling Technology, Inc.). Following primary antibody incubation, the membranes were incubated with horseradish peroxidase-conjugated secondary antibody (1:3,000 dilution; cat. no. 7074S; Cell Signaling Technology, Inc.) for 1 h at room temperature and washed three times with PBS. Proteins bands were visualized using enhanced chemiluminescence (Thermo Fisher Scientific, Inc.) and ImageJ software (v6; National Institutes of Health) was used to analyze the protein bands.

Cell transfection

To overexpress SESN1, transfection of the pcDNA3.1 vector (Shanghai GenePharma, Co., Ltd.) containing the SESN1 gene (Ov-SESN1; 4 µg) or the empty vector plasmid (Ov-NC; 4 µg) was performed using Lipofectamine 2000® (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C in accordance with the manufacturer's protocol. After 48 h, the transfected cells were collected for use in subsequent experiments.

Test for inflammatory factors contents

The cell culture medium was collected and the supernatant was obtained via centrifugation. The ELISA sandwich method was employed to evaluate the concentrations of TNF-α (cat. no. F02810), IL-6 (cat. no. F01310) and IL-1β (cat. no. F01220) in culture medium supernatant in accordance with the manufacturer's protocols. These kits were acquired from Shanghai Xitang Biotechnology. The optical density at 450 nm was measured with a microplate reader (Bio-Rad Laboratories, Inc.).

Terminal deoxynucleotidyl transferase-mediated nick-end labeling (TUNEL) staining

A TUNEL staining kit (Beyotime Institute of Biotechnology) was utilized according to the manufacturer's instructions. In brief, following incubation with 4% paraformaldehyde at 4°C for 20 min, the cells were treated with 0.5% Triton X-100. They were then incubated with 50 µl TUNEL reaction buffer for 1 h at 37°C. Cell nuclei were stained with 2 µg/ml DAPI solution for 10 min at room temperature in the dark. Images were acquired under a fluorescence microscope (Olympus Corporation) and cells were counted in five randomly selected microscopic fields.

Immunofluorescence staining

HUVECs were first cultured on sterilized coverslips. Subsequently, the transfected HUVECs were fixed with 4% paraformaldehyde for 20 min at room temperature, permeabilized with 0.5% Triton X-100 and blocked with 1% bovine serum albumin (MilliporeSigma) for 1 h at room temperature. The cells were then incubated with primary antibodies against CD31 (1:1,000 dilution; cat. no. ab9498; Abcam) or α-SMA (1:50 dilution; cat. no. ab150301; Abcam) overnight at 4°C. Subsequently, cells were incubated with Alexa Fluor® 488-conjugated secondary antibody (1:400 dilution; cat. no. A11008; Molecular Probes; Thermo Fisher Scientific, Inc.) for 1 h at room temperature. After washing with PBS three times, DAPI was used to stain the coverslips for 5 min at room temperature. The results were imaged using a fluorescence microscope (Olympus Corporation).

Statistical analysis

Values are expressed as the mean ± standard deviation and analyzed using GraphPad Prism software (version 8.0; GraphPad Software, Inc.). All experiments were performed as three independent replicates. An unpaired Student's t-test was used to compare differences between two groups, while differences between more than two groups were analyzed using a one-way ANOVA followed by Tukey's post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

SESN1 expression is downregulated in Ox-LDL-stimulated HUVECs

To evaluate the role of SESN1 in HUVECs, SESN1 expression was first examined in HUVECs. Using the Human Protein Atlas, SESN1 was indicated to be highly expressed in HUVECs (Fig. 1A). Subsequently, various concentrations (0, 25, 50, 75 and 100 µg/ml) of Ox-LDL were utilized to stimulate the HUVECs and the viability was examined using a CCK-8 assay. The results suggested that the viability of HUVECs was decreased upon exposure to Ox-LDL in a concentration-dependent manner (Fig. 1B). In addition, RT-qPCR and western blot analysis demonstrated that SESN1 expression at both the mRNA and protein level was decreased as the concentration of Ox-LDL increased; when the Ox-LDL concentration was 100 µg/ml, SESN1 expression was the lowest (Fig. 1C and D). Therefore, 100 µg/ml Ox-LDL was used in the subsequent experiments. Taken together, the results demonstrated that SESN1 exhibits low expression in Ox-LDL-stimulated HUVECs.

SESN1 overexpression mitigates the decrease in the viability and inflammation of HUVECs stimulated with Ox-LDL

For the purpose of assessing the function of SESN1 in HUVECs, SESN1 was first overexpressed by transfection with the Ov-SESN1 plasmid and the transfection efficiency was examined using RT-qPCR and western blot analysis (Fig. 2A and B). Subsequently, CCK-8 assays demonstrated that SESN1 overexpression led to an increase in cell viability following stimulation with Ox-LDL (Fig. 2C). Furthermore, RT-qPCR revealed that the enhanced mRNA levels of inflammatory factors, including TNF-α, IL-6 and IL-1β, in the HUVECs stimulated with Ox-LDL were decreased following the overexpression of SESN1 (Fig. 2D-F). Consistently, Ox-LDL stimulation led to significant increases in the contents of TNF-α, IL-6 and IL-1β compared to the control group, which were restored by overexpression of SESN1 (Fig. 2G). Furthermore, western blot analysis indicated that overexpression of SESN1 attenuated the iNOS and p/t-NF-κB p65 protein levels triggered by Ox-LDL (Fig. 2H). In summary, these results demonstrated that SESN1 exerted protective effects against the Ox-LDL-induced decrease in the viability and inflammation of HUVECs.

SESN1 overexpression suppresses the apoptosis of Ox-LDL-stimulated HUVECs

The detection of cell apoptosis was performed using a TUNEL assay. The results revealed that the Ox-LDL-induced apoptosis of HUVECs was inhibited by SESN1 (Fig. 3A and B). In addition, western blot analysis was used to examine the protein levels of Bax, cleaved caspase-3, caspase-3, cleaved PARP, PARP and Bcl-2, all of which are apoptosis-related factors. It was noted that following stimulation with Ox-LDL, the Bax, cleaved caspase-3/caspase-3 and cleaved PARP/PARP protein levels were increased, while the Bcl-2 protein level was decreased, indicating that Ox-LDL induced apoptosis of HUVECs. However, when SESN1 was overexpressed, HUVEC apoptosis was impeded (Fig. 3C). These results suggested that SESN1 exerted a suppressive effect against HUVEC apoptosis following stimulation with Ox-LDL.

SESN1 attenuates the Ox-LDL-induced EndMT of HUVECs

Using immunofluorescence staining, it was demonstrated that Ox-LDL decreased the expression of the endothelial marker CD31 (Fig. 4A) and increased that of the mesenchymal marker α-SMA (Fig. 4B) in HUVECs; these effects were mitigated by SESN1 overexpression. In addition, the increased expression of mesenchymal markers (α-SMA and vimentin) and the decreased expression of endothelial markers (CD31 and vWF) in Ox-LDL-stimulated HUVECs were both reversed by SESN1 overexpression (Fig. 4C and D). Thus, SESN1 prevented the Ox-LDL-induced EndMT of HUVECs.

SESN1 mediates AMPK/SIRT1/LOX1 signaling in Ox-LDL-stimulated HUVECs

It is well established that SESN1 is capable of stimulating the AMPK signaling pathway (16). Using KEGG pathway enrichment analysis, it was indicated that SIRT1 was a critical regulator of AMPK activation and it inhibited the expression of LOX1, which has been identified as a major receptor for Ox-LDL in endothelial cells (30,31). Hence, whether SESN1 was associated with the activation of AMPK/SIRT1 signaling to suppress LOX1, thus participating in the inflammation, apoptosis and EndMT of HUVECs, was next assessed. Western blot analysis revealed that Ox-LDL stimulation decreased the protein levels of p-AMPK/AMPK and SIRT1, whereas it increased the protein levels of LOX1. However, these effects were reversed by SESN1 overexpression (Fig. 5A). Of note, the addition of the AMPK inhibitor compound C notably downregulated p-AMPK/AMPK and SIRT1 expression and upregulated LOX1 expression when compared with the Ox-LDL + Ov-SESN1 group (Fig. 5B). In addition, the SIRT1 inhibitor NAM decreased SIRT1 expression and increased LOX1 expression. However, the inhibitors had no effect on p-AMPK/AMPK expression. In conclusion, SESN1 activated AMPK/SIRT1 signaling to suppress LOX1 in HUVECs following stimulation with Ox-LDL.

SESN1 exerts protective effects on the viability and inflammation of Ox-LDL-stimulated HUVECs via activation of AMPK/SIRT1/LOX1 signaling

Subsequently, the results of a CCK-8 assay demonstrated that the promotion of the viability of Ox-LDL-stimulated HUVECs by SESN1 overexpression was attenuated by the inhibitors of AMPK or SIRT1 (Fig. 6A). Similarly, the decreased expression of the inflammatory factors, TNF-α, IL-6 and IL-1β [tested by RT-qPCR (Fig. 6B) and ELISA (Fig. 6C)], as well as iNOS and p-p65/p65 of NF-κB [detected by western blot analysis (Fig. 6D)] due to SESN1 overexpression was abrogated by AMPK or SIRT1 inhibitor. These results suggested that AMPK/SIRT1 signaling reversed the protective effects of SESN1 against the decrease in the viability and inflammation of HUVECs mediated by Ox-LDL.

Overexpression of SESN1 prevents Ox-LDL-mediated HUVEC apoptosis by regulating AMPK/SIRT1/LOX1 signaling

Analysis of cell apoptosis using the TUNEL assay revealed that the suppression of apoptosis of Ox-LDL-stimulated HUVECs associated with SESN1 overexpression was abrogated by AMPK or SIRT1 inhibitor (Fig. 7A and B). Western blot analysis indicated that the decrease in the protein levels of Bax, cleaved caspase-3/caspase-3 and cleaved PARP/PARP, and the increase in the protein level of Bcl-2 in Ox-LDL-stimulated HUVECs induced by SESN1 overexpression were all reversed by treatment with an AMPK or SIRT1 inhibitor; these observations were consistent with the results of TUNEL assay (Fig. 7C). Taken together, it was indicated that SESN1 impeded Ox-LDL-induced HUVEC apoptosis via the activation of AMPK/SIRT1/LOX1 signaling.

Inhibition of AMPK/SIRT1/LOX1 signaling attenuates the protective effects of SESN1 against the Ox-LDL-induced EndMT of HUVECs

Immunofluorescence staining confirmed that SESN1 enhanced CD31 expression and decreased α-SMA expression upon exposure to Ox-LDL; this effect was reversed by AMPK or SIRT1 inhibitor (Fig. 8A and B). Furthermore, AMPK or SIRT1 inhibitor reversed the promoting effects of SESN1 on the expression of CD31 and vWF, and abrogated the suppressive effects of SESN1 on α-SMA and vimentin expression in Ox-LDL-stimulated HUVECs (Fig. 8C and D). Collectively, SESN1 impeded the Ox-LDL-mediated EndMT of HUVECs via the activation of AMPK/SIRT1/LOX1 signaling.

Discussion

Endothelial cells are considered active metabolic components of biological tissue that have crucial physiological functions (32). It is widely acknowledged that endothelial cell injury is highly associated with various human diseases, including chronic cardiovascular, renal and metabolic diseases (33). With regard to cardiovascular diseases, endothelial cell injury is considered to be an early and enduring feature, preceding the development of cardiovascular disease (34). As a risk factor for atherosclerosis, Ox-LDL contributes to atherosclerotic plaque formation and progression via several mechanisms, including endothelial cell dysfunction (25). Under pathological conditions, Ox-LDL is known to induce endothelial cell injury during atherosclerosis (35). SESN1 has been reported to be involved in cell metabolism and cardiovascular and age-related diseases (36). Furthermore, SESN1 has been demonstrated to inhibit NOD-like receptor family pyrin domain containing 3 inflammasome activation in lipopolysaccharide-primed macrophages induced by Ox-LDL via the inactivation of NF-κB signaling (20). The present study confirmed the low expression of SESN1 in Ox-LDL-stimulated HUVECs and Ox-LDL stimulation decreased cell viability in a concentration-dependent manner.

Endothelial cell apoptosis has a key role in the process of atherosclerosis, which is hallmarked by the inflammatory response (37). For instance, iNOS, a pro-inflammatory cytokine, is considered to be a major contributor to atherosclerosis (38). NF-κB transcription factors formed by the dimerization of Rel proteins [RelA (p65), c-Rel, RelB, p50, p52] have been confirmed to have crucial roles in inflammation, immunity, cell proliferation and apoptosis (39,40). In addition, as has been demonstrated, EndMT is the process through which endothelial cells undergo a series of molecular events, which leads to the adoption of a mesenchymal-like phenotype (41). Furthermore, endothelial cells undergoing EndMT lose expression of endothelial cell-specific proteins, including CD31 and vWF, and this initiates the expression of mesenchymal cell-specific genes and the production of their encoded proteins, including α-SMA and vimentin (42). For instance, α-SMA, an EndMT-related marker, has been reported to be involved in cardiogenesis and cardiovascular diseases (43). As an EndMT-related marker, CD31 has also been confirmed as a potential therapeutic target in atherosclerosis (44). Furthermore, apoptosis, inflammation and EndMT induced by Ox-LDL are vital for the development of atherosclerosis (45). Previous studies have indicated that SESN1 has a crucial role in cell apoptosis and the inflammatory response. For instance, SESN1 is targeted by microRNA-16-5p and influences myoblast proliferation and apoptosis (16). SESN1 also inhibits macrophage-mediated inflammation of the aorta (20). SESN1 has a crucial role in aerobic exercise and suppresses the activation of inflammatory signaling (46). In the present study, functional experiments demonstrated that stimulation with 100 µg/ml Ox-LDL promoted apoptosis of HUVECs. Furthermore, Ox-LDL stimulation elevated the TNF-α, IL-6 and IL-1β mRNA levels, as well as the iNOS, p/t-NF-κB p65 protein levels. In addition, Ox-LDL increased α-SMA and vimentin expression, whereas it decreased CD31 and vWF expression. All these aforementioned effects were reversed by SESN1 overexpression.

AMPK is a central regulator of endothelial cell metabolism and has a crucial role in diabetes, cancers and vascular diseases (47,48). SIRT1 protein, the product of the longevity gene, is involved in a wide variety of cellular processes (49). It has been reported that SIRT1 mediates endothelial functions (50). Furthermore, SIRT1 is an important activator of AMPK (30). Of note, SIRT1 suppresses the expression of LOX1, which is a 50-kDa transmembrane glycoprotein that serves as a receptor for Ox-LDL (30,51). As has been previously reported, SESNs are the primary regulators of AMPK (52). SESN1 has been indicated to directly interact with AMPK, thereby participating in several human diseases. For instance, Li et al (53) demonstrated that SESN1 protected cardiomyocytes from doxorubicin-induced damage by upregulating AMPK expression. The SESN1-2/AMPK/mTOR axis is regulated by AMPK phosphorylation, which has a crucial role in pancreatic cancer cell autophagy (54). In the present study, it was observed that the overexpression of SESN1 enhanced the decreased protein levels of p-AMPK/AMPK and SIRT1, and decreased the elevated protein levels of LOX1 following exposure to Ox-LDL. This indicated that SESN1 led to the activation of AMPK/SIRT1 signaling to inhibit LOX1 expression in Ox-LDL-stimulated HUVECs. At the same time, it was noted that AMPK or SIRT1 inhibitor attenuated the protective effects of SESN1 against the Ox-LDL-induced apoptosis, inflammation and EndMT of HUVECs.

In conclusion, the present study demonstrated that SESN1 suppressed Ox-LDL-induced endothelial EndMT, inflammation and apoptosis by regulating the AMPK/SIRT1/LOX1 signaling pathway. SESN1 may thus prove to be an effective biomarker for protection from endothelial cell injury, thus highlighting a novel target for the treatment of atherosclerosis. The lack of elucidation of upstream mechanisms of SESN1 and an in vivo animal study are limitations of the present study and comprehensive analysis is required in the future.

Acknowledgements

Not applicable.

Funding

Funding: Not applicable.

Availability of data and materials

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

Authors' contributions

FG and YZ conceived and designed the study. BZ and CX performed the experiments. ZS, YG and XD analyzed and interpreted the experimental data. FG and XD wrote and revised the manuscript. YZ and BZ confirmed the authenticity of all the raw data. 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.

References

1 

Getz GS and Reardon CA: Animal models of atherosclerosis. Arterioscler Thromb Vasc Biol. 32:1104–1115. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Frostegård J: Immunity, atherosclerosis and cardiovascular disease. BMC medicine. 11:1172013. View Article : Google Scholar : PubMed/NCBI

3 

Jensen HA and Mehta JL: Endothelial cell dysfunction as a novel therapeutic target in atherosclerosis. Expert Rev Cardiovasc Ther. 14:1021–1033. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Falk E: Pathogenesis of atherosclerosis. J Am Coll Cardiol. 47 (8 Suppl):C7–C12. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Bai X, Wang X and Xu Q: Endothelial damage and stem cell repair in atherosclerosis. Vascul Pharmacol. 52:224–229. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Lao KH, Zeng L and Xu Q: Endothelial and smooth muscle cell transformation in atherosclerosis. Curr Opin Lipidol. 26:449–456. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Qin B, Yang H and Xiao B: Role of microRNAs in endothelial inflammation and senescence. Mol Biol Rep. 39:4509–4518. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Winn RK and Harlan JM: The role of endothelial cell apoptosis in inflammatory and immune diseases. J Thromb Haemost. 3:1815–1824. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Souilhol C, Harmsen MC, Evans PC and Krenning G: Endothelial-mesenchymal transition in atherosclerosis. Cardiovasc Res. 114:565–577. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Zhu L, Gong X, Gong J, Xuan Y, Fu T, Ni S, Xu L and Ji N: Notoginsenoside R1 upregulates miR-221-3p expression to alleviate ox-LDL-induced apoptosis, inflammation, and oxidative stress by inhibiting the TLR4/NF-κB pathway in HUVECs. Braz J Med Biol Res. 53:e93462020. View Article : Google Scholar : PubMed/NCBI

11 

Xu K, Liu X, Ren G, Yin D, Guo S and Zhao Y: Depletion of CPEB1 protects against oxidized LDL-induced endothelial apoptosis and inflammation though SIRT1/LOX-1 signalling pathway. Life Sci. 239:1168742019. View Article : Google Scholar : PubMed/NCBI

12 

Kim M, Kowalsky AH and Lee JH: Sestrins in physiological stress responses. Annu Rev Physiol. 83:381–403. 2021. View Article : Google Scholar : PubMed/NCBI

13 

Wang M, Xu Y, Liu J, Ye J, Yuan W, Jiang H, Wang Z, Jiang H and Wan J: Recent insights into the biological functions of sestrins in health and disease. Cell Physiol Biochem. 43:1731–1741. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Sanchis-Gomar F: Sestrins: Novel antioxidant and AMPK-modulating functions regulated by exercise? J Cell Physiol. 228:1647–1650. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Narita N, Ito Y, Takabayashi T, Okamoto M, Imoto Y, Ogi K, Tokunaga T, Matsumoto H and Fujieda S: Suppression of SESN1 reduces cisplatin and hyperthermia resistance through increasing reactive oxygen species (ROS) in human maxillary cancer cells. Int J Hyperthermia. 35:269–278. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Cai B, Ma M, Chen B, Li Z, Abdalla BA, Nie Q and Zhang X: MiR-16-5p targets SESN1 to regulate the p53 signaling pathway, affecting myoblast proliferation and apoptosis, and is involved in myoblast differentiation. Cell Death Dis. 9:3672018. View Article : Google Scholar : PubMed/NCBI

17 

Dong XC: The potential of sestrins as therapeutic targets for diabetes. Expert Opin Ther Targets. 19:1011–1015. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Lee JH, Cho US and Karin M: Sestrin regulation of TORC1: Is Sestrin a leucine sensor? Sci Signal. 9:re52016. View Article : Google Scholar : PubMed/NCBI

19 

Budanov AV: SESTRINs regulate mTORC1 via RRAGs: The riddle of GATOR. Mol Cell Oncol. 2:e9971132015. View Article : Google Scholar : PubMed/NCBI

20 

Keping Y, Yunfeng S, Pengzhuo X, Liang L, Chenhong X and Jinghua M: Sestrin1 inhibits oxidized low-density lipoprotein-induced activation of NLRP3 inflammasome in macrophages in a murine atherosclerosis model. Eur J Immunol. 50:1154–1166. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Zhang J, Wang Z, Zhang J, Zuo G, Li B, Mao W and Chen S: Rapamycin attenuates endothelial apoptosis induced by low shear stress via mTOR and sestrin1 related redox regulation. Mediators Inflamm. 2014:7696082014. View Article : Google Scholar : PubMed/NCBI

22 

Xue R, Zeng J, Chen Y, Chen C, Tan W, Zhao J, Dong B, Sun Y, Dong Y and Liu C: Sestrin 1 ameliorates cardiac hypertrophy via autophagy activation. J Cell Mol Med. 21:1193–1205. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Fulco M and Sartorelli V: Comparing and contrasting the roles of AMPK and SIRT1 in metabolic tissues. Cell Cycle. 7:3669–3679. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Wang H, Yang G, Zhang Q, Liang X, Liu Y, Gao M, Guo Y and Chen L: Apremilast ameliorates ox-LDL-induced endothelial dysfunction mediated by KLF6. Aging (Albany NY). 12:19012–19021. 2020.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

25 

Pirillo A, Norata GD and Catapano AL: LOX-1, OxLDL, and atherosclerosis. Mediators Inflamm. 2013:1527862013. View Article : Google Scholar : PubMed/NCBI

26 

Ou HC, Chou WC, Hung CH, Chu PM, Hsieh PL, Chan SH and Tsai KL: Galectin-3 aggravates ox-LDL-induced endothelial dysfunction through LOX-1 mediated signaling pathway. Environ Toxicol. 34:825–835. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Hwang ES and Song SB: Nicotinamide is an inhibitor of SIRT1 in vitro, but can be a stimulator in cells. Cell Mol Life Sci. 74:3347–3362. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Zheng S, Li W, Xu M, Bai X, Zhou Z, Han J, Shyy JY and Wang X: Calcitonin gene-related peptide promotes angiogenesis via AMP-activated protein kinase. Am J Physiol Cell Physiol. 299:C1485–C1492. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Lu J, Mitra S, Wang X, Khaidakov M and Mehta JL: Oxidative stress and lectin-like ox-LDL-receptor LOX-1 in atherogenesis and tumorigenesis. Antioxid Redox Signal. 15:2301–2333. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Cetrullo S, D'Adamo S, Tantini B, Borzi RM and Flamigni F: mTOR, AMPK, and Sirt1: Key players in metabolic stress management. Crit Rev Eukaryot Gene Expr. 25:59–75. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Lawal AO, Davids LM and Marnewick JL: Diesel exhaust particles and endothelial cells dysfunction: An update. Toxicol In Vitro. 32:92–104. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Zhang X, Sun D, Song JW, Zullo J, Lipphardt M, Coneh-Gould L and Goligorsky MS: Endothelial cell dysfunction and glycocalyx-A vicious circle. Matrix Biol. 71-72:421–431. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Monteiro JP, Bennett M, Rodor J, Caudrillier A, Ulitsky I and Baker AH: Endothelial function and dysfunction in the cardiovascular system: The long non-coding road. Cardiovasc Res. 115:1692–1704. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Su Q, Sun Y, Ye Z, Yang H, Kong B and Li L: Pinocembrin protects endothelial cells from oxidized LDL-induced injury. Cytokine. 111:475–480. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Sun W, Wang Y, Zheng Y and Quan N: The emerging role of sestrin2 in cell metabolism, and cardiovascular and age-related diseases. Aging Dis. 11:154–163. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Zhang T, Tian F, Wang J, Jing J, Zhou SS and Chen YD: Atherosclerosis-associated endothelial cell apoptosis by MiR-429-mediated down regulation of Bcl-2. Cell Physiol Biochem. 37:1421–1430. 2015. View Article : Google Scholar : View Article : Google Scholar : PubMed/NCBI

38 

Lind M, Hayes A, Caprnda M, Petrovic D, Rodrigo L, Kruzliak P and Zulli A: Inducible nitric oxide synthase: Good or bad? Biomed Pharmacother. 93:370–375. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Lawrence T and Fong C: The resolution of inflammation: Anti-inflammatory roles for NF-kappaB. Int J Biochem Cell Biol. 42:519–523. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Viatour P, Merville MP, Bours V and Chariot A: Phosphorylation of NF-kappaB and IkappaB proteins: Implications in cancer and inflammation. Trends Biochem Sci. 30:43–52. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Hulshoff MS, Del Monte-Nieto G, Kovacic J and Krenning G: Non-coding RNA in endothelial-to-mesenchymal transition. Cardiovasc Res. 115:1716–1731. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Piera-Velazquez S and Jimenez SA: Endothelial to mesenchymal transition: Role in physiology and in the pathogenesis of human diseases. Physiol Rev. 99:1281–1324. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Anbara T, Sharifi M and Aboutaleb N: Endothelial to mesenchymal transition in the cardiogenesis and cardiovascular diseases. Curr Cardiol Rev. 16:306–314. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Caligiuri G: CD31 as a therapeutic target in atherosclerosis. Circ Res. 126:1178–1189. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Zhao H, Liu M, Liu H, Suo R and Lu C: Naringin protects endothelial cells from apoptosis and inflammation by regulating the Hippo-YAP pathway. Biosci Rep. 40:BSR201934312020. View Article : Google Scholar : View Article : Google Scholar : PubMed/NCBI

46 

Sun Y, Wu Y, Jiang Y and Liu H: Aerobic exercise inhibits inflammatory response in atherosclerosis via sestrin1 protein. Exp Gerontol. 155:1115812021. View Article : Google Scholar : PubMed/NCBI

47 

Qi D and Young LH: AMPK: Energy sensor and survival mechanism in the ischemic heart. Trends Endocrinol Metab. 26:422–429. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Mount PF, Lane N, Venkatesan S, Steinberg GR, Fraser SA, Kemp BE and Power DA: Bradykinin stimulates endothelial cell fatty acid oxidation by CaMKK-dependent activation of AMPK. Atherosclerosis. 200:28–36. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Quiñones M, Al-Massadi O, Fernø J and Nogueiras R: Cross-talk between SIRT1 and endocrine factors: Effects on energy homeostasis. Mol Cell Endocrinol. 397:42–50. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Hwang JW, Yao H, Caito S, Sundar IK and Rahman I: Redox regulation of SIRT1 in inflammation and cellular senescence. Free Radic Biol Med. 61:95–110. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Hung CH, Chan SH, Chu PM, Lin HC and Tsai KL: Metformin regulates oxLDL-facilitated endothelial dysfunction by modulation of SIRT1 through repressing LOX-1-modulated oxidative signaling. Oncotarget. 7:10773–10787. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Shen T, Alvarez-Garcia O, Li Y, Olmer M and Lotz MK: Suppression of Sestrins in aging and osteoarthritic cartilage: Dysfunction of an important stress defense mechanism. Osteoarthritis Cartilage. 25:287–296. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Li EZ, Sun Y, Lv GX, Li Y, Zhang Z, Hu Z and Cao W: Sinoporphyrin sodium based sonodynamic therapy induces anti-tumor effects in hepatocellular carcinoma and activates p53/caspase 3 axis. Int J Biochem Cell Biol. 113:104–114. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Fiorini C, Menegazzi M, Padroni C, Dando I, Dalla Pozza E, Gregorelli A, Costanzo C, Palmieri M and Donadelli M: Autophagy induced by p53-reactivating molecules protects pancreatic cancer cells from apoptosis. Apoptosis. 18:337–346. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2022
Volume 25 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y and Dou X: SESN1 attenuates the Ox‑LDL‑induced inflammation, apoptosis and endothelial‑mesenchymal transition of human umbilical vein endothelial cells by regulating AMPK/SIRT1/LOX1 signaling. Mol Med Rep 25: 161, 2022
APA
Gao, F., Zhao, Y., Zhang, B., Xiao, C., Sun, Z., Gao, Y., & Dou, X. (2022). SESN1 attenuates the Ox‑LDL‑induced inflammation, apoptosis and endothelial‑mesenchymal transition of human umbilical vein endothelial cells by regulating AMPK/SIRT1/LOX1 signaling. Molecular Medicine Reports, 25, 161. https://doi.org/10.3892/mmr.2022.12678
MLA
Gao, F., Zhao, Y., Zhang, B., Xiao, C., Sun, Z., Gao, Y., Dou, X."SESN1 attenuates the Ox‑LDL‑induced inflammation, apoptosis and endothelial‑mesenchymal transition of human umbilical vein endothelial cells by regulating AMPK/SIRT1/LOX1 signaling". Molecular Medicine Reports 25.5 (2022): 161.
Chicago
Gao, F., Zhao, Y., Zhang, B., Xiao, C., Sun, Z., Gao, Y., Dou, X."SESN1 attenuates the Ox‑LDL‑induced inflammation, apoptosis and endothelial‑mesenchymal transition of human umbilical vein endothelial cells by regulating AMPK/SIRT1/LOX1 signaling". Molecular Medicine Reports 25, no. 5 (2022): 161. https://doi.org/10.3892/mmr.2022.12678