CircRNA‑0044073 is upregulated in atherosclerosis and increases the proliferation and invasion of cells by targeting miR‑107

  • Authors:
    • Lin Shen
    • Yanyan Hu
    • Jianwei Lou
    • Sen Yin
    • Weiling Wang
    • Yuanyan Wang
    • Yong Xia
    • Wei Wu
  • View Affiliations

  • Published online on: March 6, 2019     https://doi.org/10.3892/mmr.2019.10011
  • Pages: 3923-3932
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Circular RNAs (circRNAs) are endogenous non‑coding RNAs implicated in atherosclerosis. The aim of the present study was to explore the function of circRNA‑0044073 in atherosclerosis. Reverse transcription quantitative polymerase chain reaction assays were used to measure the expression levels of circRNA‑0044073, microRNA (miRNA/miR)‑107, janus kinase 1 (JAK1), signal transducer and activator of transcription 3 (STAT3), B‑cell lymphoma 2 (Bcl‑2) and v‑myc avian myelocytomatosis viral oncogene homolog (c‑myc) in in blood cells from patients with atherosclerosis. RNA pull‑down and luciferase reporter assays were then used to determine the association between circRNA and miR expression, and miR and gene expression, respectively. Matrigel invasion assay and flow cytometry were used to analyze cell invasion and cell cycle. Western blot analysis and ELISA were used to evaluate the expression levels of proteins. It was identified that the expression of circRNA‑0044073 was upregulated and the expression of miR‑107 was downregulated in atherosclerotic blood cells. Overexpression of circRNA‑0044073 promoted the proliferation of human vascular smooth muscle cells (HUVSMCs) and human vascular endothelial cells (HUVECs), while overexpression of miR‑107 inhibited their proliferation. In addition, circRNA‑0044073 suppressed the levels of miR‑107 via a sponge mechanism. Lipopolysaccharide (LPS) affected the proliferation of HUVSMCs and HUVECs, and also resulted in changes in circRNA‑0044073 expression levels. CircRNA‑0044073 promoted the proliferation and invasion of HUVSMCs and HUVECs in spite of the opposite effect observed with LPS treatment. The JAK/STAT signaling pathway was activated in patients with atherosclerosis. CircRNA‑0044073 favored the activation of the JAK/STAT signaling pathway and inflammation in HUVSMCs and HUVECs. These data indicate that circRNA‑0044073 is upregulated in atherosclerosis and promotes the proliferation and invasion of cells by targeting miR‑107 and activating the JAK/STAT signaling pathway, potentially offering a target for novel treatment strategies against atherosclerosis.

Introduction

Atherosclerosis is a chronic inflammatory disorder of the arterial vessel walls characterized by lipid deposition and fibrous cap formation in the arterial intima. Atherosclerosis contributes to cardiovascular disease (CVD), a major cause of morbidity and mortality in Western countries. A total of ~80 million Americans have one or more forms of CVD (1). Growing evidence suggests that dysfunction of endothelial cells (ECs) and vascular smooth muscle cells (VSMC) is critical in the formation of atherosclerosis. Abnormal proliferation and migration of these cells contribute to the progression of atherosclerosis (24). VSMCs migrate and proliferate to form a stabilizing fibrous cap that encapsulates atherosclerotic plaques (5). During the initiation and development of inflammation, ECs and VSMCs produce numerous types of cytokines, including tumor necrosis factors (TNF), interleukin (IL), adhesion molecules, interferon, and adventitium-derived relaxing factors that are implicated in atherosclerosis (69).

Circular RNAs (circRNAs) are evolutionarily conserved non-coding RNAs produced by the scrambling of exons at the time of splicing. They are primarily produced in the brain and are present inside the cell. Previous data have suggested that the circRNA antisense non-coding RNA in the INK locus modulates ribosomal RNA maturation and atherosclerosis in humans, indicating that circularization of long non-coding RNAs may alter RNA function and therefore protect against disease (10). The great potential of circRNAs as biomarkers for the early detection of cardiovascular diseases has been highlighted in several patents and previous studies (11). Certain circRNAs, including circRNA-0003575 and circRNA-000595, are significantly associated with atherosclerosis (12,13). Furthermore, the role of circRNAs in atherosclerosis is associated with the regulation of the physiology of ECs and smooth muscle cells (SMCs) (10). MicroRNAs (miRNAs) are a group of highly conserved small non-coding RNA molecules measuring 18–22 nucleotides in length. By binding to the 3′-untranslated region of their target mRNAs, miRNAs regulate expression levels of downstream genes (14,15). In addition to participating in regulating cell-cycle and cell proliferation (16,17), miRNAs are associated with inflammatory processes involved in atherosclerosis (18). miR-107 is implicated in the pathogenesis of diseases, including renal disease, Alzheimer's disease, gastric cancer, colon cancer and hepatocellular carcinoma (1922). Previous studies suggested that miR-107 is a regulator of atherosclerosis (23,24). In addition, binding of miR-107 to the Clock circadian rhythm gene resulted in the deregulation of the circadian rhythm of the cells (25,26). However, the exact mechanism is largely unknown.

Certain circRNAs have been described as regulatory transcripts as they behave like sponges to bind miRNAs. For example, a previous study examining the role of circular RNA-7 demonstrated that the circRNA was an inhibitor of miR-7, an miRNA known to regulate various diseases including, cancer, neurodegenerative diseases, diabetes and atherosclerosis (27). In this context, the present study aimed to investigate the role of circRNA in the regulation of atherosclerosis and to understand the underlying mechanism associated with miRNAs and signaling pathways.

Materials and methods

Patients and samples

Human blood cells were collected from 20 patients with atherosclerosis and healthy controls at the Qi-Lu Hospital of Shandong University (Jinan, China). In total, 13 males and seven females (age range, 48–68 years) were enrolled in the study. Following sonographic examination, the patients with a carotid intima-media thickness ≥ 1.3 mm were considered to exhibit an atherosclerotic plaque. Informed consent was obtained from all patients and ethical approval was granted by The Ethics Committee of the Qi-Lu Hospital of Shandong University.

Cell culture and treatment

All reagents for cell culture were obtained from Invitrogen; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Human umbilical vein endothelial cells (HUVECs) were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA), and human umbilical vein smooth muscle cells (HUVSMCs) were purchased from iXCells Biotechnologies (San Diego, CA, USA). HUVECs and HUVSMCs cells were maintained in minimum essential medium-L-glutamine (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), smooth muscle cell growth medium and RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.). The medium was supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin-amphotericin. The cells were maintained in a humidified incubator with 5% CO2 at 37°C. The culture medium was changed twice weekly. HUVECs and HUVSMCs were sub-cultured subsequent to reaching 80% confluency. For the following assay, the cells initially received either 100 ng/ml lipopolysaccharide (LPS) or vehicle. The template DNA of circRNA-0044073 was synthesized and then inserted into the multiple cloning site in the pAd-Track-cmv vector (Addgene, Inc., Cambridge, MA, USA). A total of 500 ng of plasmid DNA was subsequently transfected into HUVECs or HUVSMCs in 24-well plates using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) to increase circRNA-0044073 expression, whereas, the empty vector was transfected as the negative control (NC). Following 48 h of incubation, the transfected cells were used in the following experiments.

Reverse transcription quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from cells using TRIzol® reagent (Thermo Fisher Scientific, Inc.) and was reverse transcribed using the Bestar qPCR RT kit (DBI Bioscience, Ludwigshafen, Germany) at 65°C for 5 min. qPCR amplification was performed in triplicate using DBI Bestar® SybrGreen qPCR MasterMix (DBI Bioscience, Ludwigshafen, Germany) in a total volume of 20 µl containing 2 µl each cDNA. The amplification protocol consisted of initial denaturation for 2 min at 94°C, followed by 40 cycles of denaturation (94°C for 20 sec), annealing for 20 sec at 58°C and elongation for 20 sec at 72°C. The primers for circRNA-0044073, miR-107, U6, janus kinase 1 (JAK1), signal transducer and activator of transcription 3 (STAT3), B-cell lymphoma 2 (Bcl-2), v-myc avian myelocytomatosis viral oncogene homolog (c-myc) and GAPDH are summarized in Table I. Using GAPDH or U6 as endogenous control genes, the relative transcript levels were calculated using the 2−ΔΔCq quantification method (28).

Table I.

Primers used in the reverse transcription quantitative polymerase chain reaction assay.

Table I.

Primers used in the reverse transcription quantitative polymerase chain reaction assay.

GenesSequence (5′-3′)
hsa_circ_0044073F ACAGGGGTGTTTGTGTGTGT
hsa_circ_0044073R CTTCACGTTGCAGGTGTAGC
miR-107 F ACACTCCAGCTGGGAGCAGCA UUGUACAGGG
miR-107 R CTCAACTGGTGTCGTGGA
U6 F CTCGCTTCGGCAGCACA
U6 R AACGCTTCACGAATTTGCGT
JAK1 F CACCAGGATGCGGATAAATAAT
JAK1 R CAAAGTTTCCAAGGTAGCCAAG
STAT3 F TGGTGTCTCCACTGGTCTATCTC
STAT3 R CATCAATGAATCTAAAGTGCGG
Bcl-2 F ATCGCCCTGTGGATGACTG
Bcl-2 R AGACAGCCAGGAGAAATCAAAC
c-myc F CATACATCCTGTCCGTCCA
c-myc R CGCACAAGAGTTCCGTAGC
GAPDH F TGTTCGTCATGGGTGTGAAC
GAPDH R ATGGCATGGACTGTGGTCAT

[i] F, forward; R, reverse; hsa, Homo sapiens; miR, microRNA; JAK1, janus kinase 1; STAT3, signal transducer and activator of transcription 3; Bcl-2, B-cell lymphoma 2; c-myc, v-myc avian myelocytomatosis viral oncogene homolog.

Cell viability assay

Cell viabilities were assessed by the Cell Counting Kit-8 (Dojindo Molecular Technologies, Inc., Kumamoto, Japan). In brief, cells were added into a 96-well plate at a density of 2×103 cells/well and incubated overnight at 37°C. Then, LPS was added to each well and incubated at 37°C for 24, 48 and 72 h at the final concentration of 100 ng/ml. Thereafter, 10 µl CCK-8 solution was added into each well and incubated for 2 h at 37°C. The absorbance was detected at 450 nm with the microplate reader. Optical density was defined as the relative number of viable cells. The assay was performed in triplicate.

Western blot analysis

Following treatments with LPS for 24 h, cells were washed with PBS and subsequently harvested. Total cellular proteins were extracted with radioimmunoprecipitation assay lysis buffer containing 1% phenylmethanesulfonyl fluoride (Sigma-Aldrich; Merck KGaA). Protein concentrations were determined with a BCA assay (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Subsequent to denaturation, a total of 20 µg protein per lane was separated by electrophoresis by 10% SDS-PAGE and analyzed. Proteins were transferred onto a polyvinylidene difluoride membrane (Bio-Rad Laboratories, Inc.) and then blocked with 5% dry milk in TBST (TBS with 0.1% Tween 20) for 1 h at room temperature, washed, and incubated overnight at 4°C with primary antibodies against JAK1 (1:1,000; cat. no. ab47435; Abcam, Cambridge, MA, USA), STAT3 (1:1,000; cat. no. ab68153; Abcam), phosphorylated (p)-STAT3 (1:800; cat. no. ab30647; Abcam), Bcl-2 (1:1,000; cat. no. 15071; Cell Signaling Technology, Inc., Danvers, MA, USA), c-myc (1:1,000; cat. no. 13987; Cell Signaling Technology, Inc.) and GAPDH (1:1,000; cat. no. 2118; Cell Signaling Technology, Inc.). The membranes were then washed with TBST and then incubated with the secondary antibodies anti-rabbit and anti-mouse immunoglobulin G conjugated with horseradish peroxidase (HRP; 1:10,000; cat. no. 7074 and 7076; Cell Signaling Technology, Inc.). Finally, the immunoreactive bands were visualized by using an enhanced chemiluminescent substrate (ECL-Plus; GE Healthcare, Chicago, IL USA). Using ImageJ software (version 1.8.0; National Institutes of Health, Bethesda, MD, USA; http://imagej.nih.gove/ij/), densitometric analysis of bands was performed.

ELISA

The IL-1β (cat. no. CSB-E08053h), IL-6 (cat. no. CSB-E04638h) and TNF-α (cat. no. CSB-E04740h) ELISA assay kits were purchased from CUSABIO (Wuhan, China). After centrifugation at 1,000 × g for 15 min at 4°C, the concentrations in the supernatants were assessed according to the manufacturer's protocols. In brief, the samples were added to wells in duplicate and then incubated at room temperature for 2 h. The liquid was removed, followed by the addition of biotin-antibody and then the plates were incubated at room temperature for 1 h. Following removal of the liquid and washing, HRP-avidin was added and incubated at room temperature for 1 h. Following additional wash steps, visualization was achieved by adding the TMB substrate. The absorbance was detected at 450 nm with a microplate reader (Thermo Fisher Scientific, Inc.). The experiments were performed in triplicate.

Cell cycle analysis by flow cytometry

The cell cycle distribution was analyzed as described previously (29). Following treatment with LPS for 24 h, the cells were collected and washed with PBS and then stained with 50 mg/ml propidium iodide (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China) for 30 min at 4°C. The data were collected with a FACSCalibur flow cytometer (Becton, Dickinson and Company, Franklin Lakes, NJ, USA) for cell-cycle distribution analysis using Cell Quest Software (version 5.1; Becton, Dickinson and Company).

Matrigel invasion assay

The cells were collected and suspended in medium without serum. Transwell inserts (24-well insert; pore size, 8 mm; Becton, Dickinson and Company) were inserted in the chamber and incubated at 37°C overnight with serum-free RPMI-1640 with matrigel (0.6 mg/ml; Becton, Dickinson and Company). Subsequently, 4×104 cells in 0.2 ml serum-free medium were seeded in the top chambers of the transwell inserts, and 10% FBS was used as an attractant. The cells were incubated with 1 mg/ml LPS or control treatment for 48 h at 37°C. The cells that did not invade through the pores were removed, and the filters were stained at room temperature with hematoxylin for 5 min, and with eosin for 1 min (cat. no. C0105; Beyotime Institute of Biotechnology, Haimen, China). Samples were visualized under a light microscope (magnification, ×200) and counting.

RNA pull-down assay

The RNA pull-down assay was performed as previously described (30). In brief, either the pAd-Track-cmv-circRNA-0044073 or the control vectors were transfected into 293 cells (ATCC). Subsequently, 100 µg total RNA was extracted with TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) from the cells and incubated with 500 µg streptavidin magnetic beads (cat. no. S1421S; New England BioLabs, Inc., Ipswich, MA, USA), which were previously incubated for 2 h at 37°C with either 200 pmol biotin-miR-107 or 200 pmol biotin-miR-107-mut (Shanghai GenePharma Co., Ltd., Shanghai, China). Following RNA elution, RT-qPCR was performed to detect circRNA-0044073 as aforementioned.

DNA construct and luciferase reporter assay

As described previously (18), the psiCHECKTM-2 firefly luciferase reporter plasmids (Promega Corporation, Madison, WI, USA) were recombined with the potential miR-107 binding site sequences of JAK1 or its mutated sequence. 293 cells were seeded into a 12-well plate (3×105 cells/well) and transfected with 200 ng recombinant luciferase reporter plasmid using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Following transfection with 10 nM miR-107 mimics (5′-AGCAGCAUUGUACAGGGCUAUCA-3′) or NC miRNA (5′UUCUCCGAACGUGUCACGUTT-3′) for 24 h, the cells were collected, and luciferase activity was determined using the Dual-Luciferase Reporter Gene Assay kit (Beyotime Institute of Biotechnology). The relative luciferase activity was obtained by normalizing the firefly luciferase activity to the internal control Renilla luciferase activity.

Statistical analysis

Data in the present study are presented as the mean ± standard deviation of three independent tests. Using GraphPad Prism (version 5.0; GraphPad Software, Inc., La Jolla, CA, USA), two-tailed Student's t-tests were performed to compare data between two groups, whereas, one-way analysis of variance followed by Tukey's post hoc test was performed to compare data among three or more groups.

Results

Levels of circRNA-0044073 are upregulated in blood cells of patients with atherosclerosis and are associated with the proliferation of HUVSMCs and HUVECs

The present study identified that the expression of circRNA-0044073 in blood cells was significantly increased in patients with atherosclerosis compared with that of healthy controls (P<0.001; Fig. 1A). Therefore, we hypothesized that circRNA-0044073 serves a key role in atherosclerosis. As the abnormal proliferation of ECs, SMCs and macrophages contribute to the progression of atherosclerosis, the in vitro protocols of the present study included HUVECs and HUVSMCs to determine the effect of circRNA-0044073 on cell growth. It was observed that the overexpression of circRNA-0044073 significantly increased the proliferation of HUVECs and HUVSMCs, compared with control groups, at 48 and 72 h (P<0.01; Fig. 1B and C). These results suggest a potential association between the upregulation of circRNA-0044073 and proliferation of atherosclerosis-associated cells.

Levels of miR-107 are downregulated in blood cells of patients with atherosclerosis and is targeted by circRNA-0044073

A previous study has suggested the involvement of miR-107 in the development of atherosclerosis (26). Therefore, the levels of miR-107 were detected and compared between patients with atherosclerosis and healthy individuals. Notably, the expression of miR-107 was significantly decreased in the blood cells of patients with atherosclerosis compared with the controls (P<0.05; Fig. 2A). In addition, miR-107 mimics significantly inhibited the proliferation of HUVECs and HUVSMCs, compared with the control groups, at 48 and 72 h, which was in contrast to the observed effects of circRNA-0044073 (P<0.01; Fig. 2B and C). The association between circRNA-0044073 and miR-107 was then investigated; the data suggested that the overexpression of circRNA-0044073 significantly downregulated the levels of miR-107 in HUVECs and HUVSMCs (P<0.01; Fig. 2D and E). Furthermore, RNA pull down assay demonstrated that miR-107 was a target of circRNA-0044073 (P<0.01; Fig. 2D-F). These results indicate that circRNA-0044073 counteracts the effect of miR-107 via a sponge mechanism.

CircRNA-0044073 increases the proliferation and invasion of cells by regulating miR-107

The regulatory mechanism of circRNA-0044073 on the invasive activity of HUVSMCs and HUVECs was then explored. As an endotoxin, LPS is a potentially important stimulator and atherosclerosis risk factor. Therefore, an in vitro atherosclerosis model was established by treating cells with LPS. Compared with the control groups, treatment with LPS significantly increased the number of invasive HUVSMCs and inhibited the proliferation of HUVECs, respectively (P<0.01; Fig. 3A and B). Additionally, LPS treatment significantly increased the levels of circRNA-0044073 in HUVSMCs but downregulated the levels of circRNA-0044073 in HUVECs (P<0.001; Fig. 3C). It was additionally identified that in HUVSMCs, the combined use of LPS and circRNA-0044073 significantly increased the proliferation of cells, compared with a single treatment of LPS or circRNA-0044073 overexpression vector (P<0.01; Fig. 3D and E). By contrast, miR-107 mimics significantly alleviated the effect of LPS + circRNA-0044073 treatment on the promotion the proliferation of cells (P<0.01; Fig. 3D-E). Although the invasive activity of HUVECs was suppressed by LPS, circRNA-0044073 transfection partially reversed the effect of LPS and induced the invasion of HUVECs. Similar to the effect in HUVSMCs, miR-107 impeded the role of circRNA-0044073 in promoting cell proliferation. The effect of circRNA-0044073 on the cell cycle distribution was also examined. Despite the observation that LPS treatment affected the percentages of G2/M and S phase cells, with the addition of circRNA-0044073 transfection, the percentage of cells in G2/M phase was additionally increased, and was partially impaired by using miR-107 mimics (Fig. 4). These results support the hypothesis that circRNA-0044073 facilitates the proliferation and invasion of cells by inhibiting miR-107 in the development of atherosclerosis.

CircRNA-0044073 activates the JAK/STAT signaling pathway of HUVSMCs and HUVECs

JAK1 is a critical contributor to the pathogenesis of a number of inflammatory diseases and has been identified as a potential therapeutic target (31,32). The expression levels of JAK1 and STAT3 were significantly increased in blood cells in patients with atherosclerosis, in addition to an increase in Bcl-2 and c-myc levels, compared with those in the healthy controls (P<0.001; Fig. 5A-D). Notably, in the LPS treatment group, the Bcl-2 and c-myc levels were markedly increased in HUVSMCs and decreased in HUVECs, which is consistent with the distinct effect of LPS on the proliferation of these two types of cells (33,34). However, the overexpression of miR-107 significantly inhibited the levels of JAK1 in HUVSMCs and HUVECs (Fig. 5E-F). The luciferase reporter assay additionally confirmed that JAK1 was a target of miR-107 (Fig. 5G). In contrast to the inhibitory role of miR-107 on JAK1 expression, overexpression of circRNA-0044073 activated the JAK/STAT signaling pathway, along with upregulation of Bcl-2, c-myc, IL-1β, IL-6 and TNF-α levels, compared with those in control group, which is in accordance with the in vivo data concerning the changes in expression of JAK1, p-STAT3, Bcl-2 and c-myc (Fig. 5H-N).

Discussion

As the primary cause of myocardial infarction, heart failure, myocardial ischemia and stroke, atherosclerosis accounts for high mortality and morbidity rates worldwide (35). Although a number of treatments have been developed and widely applied for atherosclerosis, certain subgroups of patients remain at high risk for the development and progression of atherosclerosis, creating an urgent requirement to explore novel therapeutic targets or/and more effective treatments for atherosclerosis. A previous study demonstrated that circRNAs are crucial contributors to the pathogenesis of atherosclerosis (36). Therefore, increasing interest in the treatment of atherosclerosis is being focused on circRNAs at present. In the present study, to the best of our knowledge, it was observed for the first time that circRNA-0044073 levels were negatively associated with atherosclerosis and that cirRNA-0044073 may directly target miR-107 and thereby lead to the upregulation of JAK1/STAT3 signaling.

As a chronic inflammatory disease, atherosclerosis constitutes a dysregulation of a variety of cytokines. A previous study demonstrated that the JAK/STAT signaling pathway promotes vascular cell inflammation, proliferation, migration and adhesion (37). JAK1 is expressed in all cell types, including ECs and SMCs (38,39). Among the STAT proteins, STAT3 is an activator of systemic inflammatory genes and is present in the inflammatory regions of human atherosclerotic lesions in an activated form. Previous data demonstrated an important role for the JAK/STAT pathway in oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine-induced IL-8 transcription in vitro and in atherosclerosis in vivo (1). It has also been indicated that suppressors of cytokine signaling modulate the JAK/STAT-dependent responses in vascular cells, which are associated with atherosclerotic plaque development (40). In the present study, it was observed that the expression levels of JAK1 and STAT3 were significantly increased in the blood cells from patients with atherosclerosis, which was consistent with previous studies (18,41). Bcl-2 proteins represent the major regulators of extrinsic and intrinsic apoptosis signaling pathways and target apoptosis of vascular cells in atherosclerotic lesions. The decrease in c-myc oncogene levels alleviates the proliferation in rat VSMCs and induces apoptosis (42). Therefore, Bcl-2 and c-myc serve important roles in the deregulation of cell apoptosis and proliferation, contributing to the pathogenesis of atherosclerosis (43). The data presented in the present study indicated that the expression levels of Bcl-2 and c-myc were significantly increased in the blood cells from patients with atherosclerosis compared with the healthy controls. Consistent with these results, a previous study revealed that c-myc was overexpressed in plaque SMCs (44). However, a small number of studies contradict the Bcl-2 data from the present study (45,46). This may be due to differences in the test samples and the stage of atherosclerosis investigated.

HUVECs have been used as in vitro models for the study of atherosclerosis (47). As an endotoxin, LPS is a potentially important stimulator and risk factor for atherosclerosis (48). LPS has been implicated in endothelial injury and has induced apoptosis and proliferation in endothelial and VSMCs, respectively (34,35,49). Consistently, the present study observed that LPS exposure resulted in the apoptosis of HUVECs and the proliferation of HUVSMCs, and the increased expression of circRNA-0044073 in the cells. With the proliferative effect of circRNA-0044073, the apoptosis of HUVECs by LPS was significantly attenuated, and the proliferation of HUVSMCs was significantly improved. Furthermore, it was also observed that the invasion levels of HUVECs and HUVSMCs were promoted on treatment with LPS. Therefore, we hypothesized that circRNA-0044073 is associated with the initiation and development of the disease. However, additional studies are required to validate this.

miR-107 is associated with inflammation (23). Previously, it was demonstrated that the expression of miR-107 is decreased in macrophages following LPS stimulation (23,50). A consistent change in miR-107 expression in the blood cells from patients with atherosclerosis was observed in the present study. However, it was also demonstrated that the miR-107 expression was upregulated in HUVECs and HUVSMCs following LPS treatment. This variation may be due to the different cells examined. Based on the actual pro-inflammation function of miR-107, the decrease in miR-107 expression may be a result of the regulative feedback (23). Consistent with a previous study, the present study confirmed that miR-107 directly targeted JAK1 (51). Additionally, it was demonstrated that miR-107 is also a direct target of circRNA-0044073. Furthermore, it was observed that in HUVSMCs and HUVECs, circRNA-0044073 overexpression decreased miR-107 levels and resultantly increased the levels of JAK1 and p-STAT3, and the levels of downstream proteins including Bcl-2 and c-myc. As JAK1/STAT3 serve critical roles in inflammation, we hypothesized that cirRNA-0044073 may be associated with inflammation, which is a key contributor to atherosclerosis. In the present study, it was observed that circRNA-0044073 significantly induced the levels of IL-1β, IL-6 and TNF-α. These pro-inflammatory cytokines are pro-atherogenic. However, the underlying mechanisms by which circRNA-0044073 regulates these inflammatory cytokines requires additional investigation.

Taken together, the data from the present study suggests that circRNA-0044073 is a potential therapeutic target for the treatment of atherosclerosis, as it directly targets miR-107 and resultantly increases STAT3 activation, and upregulates the expression of the downstream proteins that contribute to atherosclerosis.

Acknowledgements

Not applicable.

Funding

The present study was supported by Key Research and Develop-ment Plan of Shandong Province (grant no. 2017GSF218012) and Shandong Provincial Natural Science Foundation (grant no. ZR2015HM052).

Authors' contributions

LS, YH and WWu designed the study. LS, YH, JL, SY, WWa and YW performed the experiments. LS, YX and WWa performed the data analysis and drafted the manuscript. All authors read and approved the final manuscript.

Availability of data and materials

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

Ethics approval and consent to participate

Informed consent was obtained from all patients and ethical approval was granted by The Ethics Committee of the Qi-Lu Hospital of Shandong University.

Patient consent for publication

Informed consent was obtained from all patients.

Competing interests

The authors declare that they have no competing interests.

References

1 

Gharavi NM, Alva JA, Mouillesseaux KP, Lai C, Yeh M, Yeung W, Johnson J, Szeto WL, Hong L, Fishbein M, et al: Role of the Jak/STAT pathway in the regulation of interleukin-8 transcription by oxidized phospholipids in vitro and in atherosclerosis in vivo. J Biol Chem. 282:31460–31468. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Schober A, Nazari-Jahantigh M, Wei Y, Bidzhekov K, Gremse F, Grommes J, Megens RT, Heyll K, Noels H, Hristov M, et al: MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1. Nat Med. 20:368–376. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Gorenne I, Kumar S, Gray K, Figg N, Yu H, Mercer J and Bennett M: Vascular smooth muscle cell sirtuin 1 protects against DNA damage and inhibits atherosclerosis. Circulation. 127:386–396. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Zheng Y, Li Y, Liu G, Qi X and Cao X: MicroRNA-24 inhibits the proliferation and migration of endothelial cells in patients with atherosclerosis by targeting importin-α3 and regulating inflammatory responses. Exp Ther Med. 15:338–344. 2018.PubMed/NCBI

5 

Baumer Y, McCurdy S, Alcala M, Mehta N, Lee BH, Ginsberg MH and Boisvert WA: CD98 regulates vascular smooth muscle cell proliferation in atherosclerosis. Atherosclerosis. 256:105–114. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Luc G, Bard JM, Juhan-Vague I, Ferrieres J, Evans A, Amouyel P, Arveiler D, Fruchart JC and Ducimetiere P; PRIME Study Group, : C-reactive protein, interleukin-6, and fibrinogen as predictors of coronary heart disease: The PRIME study. Arterioscler Thromb Vasc Biol. 23:1255–1261. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Brunetti ND, Salvemini G, Cuculo A, Ruggiero A, De Gennaro L, Gaglione A and Di Biase M: Coronary artery ectasia is related to coronary slow flow and inflammatory activation. Atherosclerosis. 233:636–640. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Zernecke A and Weber C: Chemokines in the vascular inflammatory response of atherosclerosis. Cardiovasc Res. 86:192–201. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Barton M: Prevention and endothelial therapy of coronary artery disease. Curr Opin Pharmacol. 13:226–241. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Holdt LM, Stahringer A, Sass K, Pichler G, Kulak NA, Wilfert W, Kohlmaier A, Herbst A, Northoff BH, Nicolaou A, et al: Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat Commun. 7:124292016. View Article : Google Scholar : PubMed/NCBI

11 

Bayoumi AS, Aonuma T, Teoh JP, Tang YL and Kim IM: Circular noncoding RNAs as potential therapies and circulating biomarkers for cardiovascular diseases. Acta Pharmacol Sin. 39:1100–1109. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Li CY, Ma L and Yu B: Circular RNA hsa_circ_0003575 regulates oxLDL induced vascular endothelial cells proliferation and angiogenesis. Biomed Pharmacother. 95:1514–1519. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Zheng C, Niu H, Li M, Zhang H, Yang Z, Tian L, Wu Z, Li D and Chen X: Cyclic RNA has-circ-000595 regulates apoptosis of aortic smooth muscle cells. Mol Med Rep. 12:6656–6662. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Diniz GP and Wang DZ: Regulation of skeletal muscle by microRNAs. Compr Physiol. 6:1279–1294. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Lu C, Xie Z and Peng Q: MiRNA-107 enhances chemosensitivity to paclitaxel by targeting antiapoptotic factor Bcl-w in non small cell lung cancer. Am J Cancer Res. 7:1863–1873. 2017.PubMed/NCBI

16 

Chen Z, Zhao L, Zhao F, Yang G and Wang J: MicroRNA-26b regulates cancer proliferation migration and cell cycle transition by suppressing TRAF5 in esophageal squamous cell carcinoma. Am J Transl Res. 8:1957–1970. 2016.PubMed/NCBI

17 

Undi RB, Kandi R and Gutti RK: MicroRNAs as haematopoiesis regulators. Adv Hematol. 2013:6957542013. View Article : Google Scholar : PubMed/NCBI

18 

Wang Y, Han Z, Fan Y, Zhang J, Chen K, Gao L, Zeng H, Cao J and Wang C: MicroRNA-9 Inhibits NLRP3 inflammasome activation in human atherosclerosis inflammation cell models through the JAK1/STAT signaling pathway. Cell Physiol Biochem. 41:1555–1571. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Jiang ZP and Zhou TB: Role of miR-107 and its signaling pathways in diseases. J Recept Signal Transduct Res. 34:338–341. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Liu F, Liu S, Ai F, Zhang D, Xiao Z, Nie X and Fu Y: miR-107 promotes proliferation and inhibits apoptosis of colon cancer cells by targeting prostate apoptosis response-4 (Par4). Oncol Res. 25:967–974. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Nelson PT and Wang WX: MiR-107 is reduced in Alzheimer's disease brain neocortex: Validation study. J Alzheimers Dis. 21:75–79. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Su SG, Yang M, Zhang MF, Peng QZ, Li MY, Liu LP and Bao SY: miR-107-mediated decrease of HMGCS2 indicates poor outcomes and promotes cell migration in hepatocellular carcinoma. Int J Biochem Cell Biol. 91:53–59. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Foley NH and O'Neill LA: miR-107: A Toll-like receptor-regulated miRNA dysregulated in obesity and type II diabetes. J Leukoc Biol. 92:521–527. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Li F, Liu B, Gao Y, Liu Y, Xu Y, Tong W and Zhang A: Upregulation of MicroRNA-107 induces proliferation in human gastric cancer cells by targeting the transcription factor FOXO1. FEBS Lett. 588:538–544. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Norata GD, Sala F, Catapano AL and Fernandez-Hernando C: MicroRNAs and lipoproteins: A connection beyond atherosclerosis? Atherosclerosis. 227:209–215. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Daimiel-Ruiz L, Klett-Mingo M, Konstantinidou V, Micó V, Aranda JF, García B, Martínez-Botas J, Dávalos A, Fernández-Hernando C and Ordovás JM: Dietary lipids modulate the expression of miR-107, an miRNA that regulates the circadian system. Mol Nutr Food Res. 59:552–565. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK and Kjems J: Natural RNA circles function as efficient microRNA sponges. Nature. 495:384–388. 2013. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Kong X, Moran MS, Zhao Y and Yang Q: Inhibition of metadherin sensitizes breast cancer cells to AZD6244. Cancer Biol Ther. 13:43–49. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Tang CM, Zhang M, Huang L, Hu ZQ, Zhu JN, Xiao Z, Zhang Z, Lin QX, Zheng XL, Yang M, et al: CircRNA_000203 enhances the expression of fibrosis-associated genes by derepressing targets of miR-26b-5p, Col1a2 and CTGF, in cardiac fibroblasts. Sci Rep. 7:403422017. View Article : Google Scholar : PubMed/NCBI

31 

O'Shea JJ and Plenge R: JAK and STAT signaling molecules in immunoregulation and immune-mediated disease. Immunity. 36:542–550. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Maschalidi S, Sepulveda FE, Garrigue A, Fischer A and de Saint Basile G: Therapeutic effect of JAK1/2 blockade on the manifestations of hemophagocytic lymphohistiocytosis in mice. Blood. 128:60–71. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Choi KB, Wong F, Harlan JM, Chaudhary PM, Hood L and Karsan A: Lipopolysaccharide mediates endothelial apoptosis by a FADD-dependent pathway. J Biol Chem. 273:20185–20188. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Jiang D, Yang Y and Li D: Lipopolysaccharide induced vascular smooth muscle cells proliferation: A new potential therapeutic target for proliferative vascular diseases. Cell Prolif. 50:e123322017. View Article : Google Scholar

35 

Liu M, Tao G, Liu Q, Liu K and Yang X: MicroRNA let-7g alleviates atherosclerosis via the targeting of LOX-1 in vitro and in vivo. Int J Mol Med. 40:57–64. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Burd CE, Jeck WR, Liu Y, Sanoff HK, Wang Z and Sharpless NE: Expression of linear and novel circular forms of an INK4/ARF-associated non-coding RNA correlates with atherosclerosis risk. PLoS Genet. 6:e10012332010. View Article : Google Scholar : PubMed/NCBI

37 

Wang R, Zhang Y, Xu L, Lin Y, Yang X, Bai L, Chen Y, Zhao S, Fan J, Cheng X and Liu E: Protein inhibitor of activated STAT3 suppresses oxidized LDL-induced cell responses during atherosclerosis in apolipoprotein E-deficient mice. Sci Rep. 6:367902016. View Article : Google Scholar : PubMed/NCBI

38 

Rane SG and Reddy EP: Janus kinases: Components of multiple signaling pathways. Oncogene. 19:5662–5679. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Verma A, Kambhampati S, Parmar S and Platanias LC: Jak family of kinases in cancer. Cancer Metastasis Rev. 22:423–434. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Ortiz-Muñoz G, Martin-Ventura JL, Hernandez-Vargas P, Mallavia B, Lopez-Parra V, Lopez-Franco O, Muñoz-Garcia B, Fernandez-Vizarra P, Ortega L, Egido J and Gomez-Guerrero C: Suppressors of cytokine signaling modulate JAK/STAT-mediated cell responses during atherosclerosis. Arterioscler Thromb Vasc Biol. 29:525–531. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Hiltunen MO, Tuomisto TT, Niemi M, Bräsen JH, Rissanen TT, Törönen P, Vajanto I and Ylä-Herttuala S: Changes in gene expression in atherosclerotic plaques analyzed using DNA array. Atherosclerosis. 165:23–32. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Bennett MR, Evan GI and Newby AC: Deregulated expression of the c-myc oncogene abolishes inhibition of proliferation of rat vascular smooth muscle cells by serum reduction, interferon-gamma, heparin, and cyclic nucleotide analogues and induces apoptosis. Circ Res. 74:525–536. 1994. View Article : Google Scholar : PubMed/NCBI

43 

Kutuk O and Basaga H: Bcl-2 protein family: Implications in vascular apoptosis and atherosclerosis. Apoptosis. 11:1661–1675. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Marin ML, Gordon RE, Veith FJ, Tulchin N and Panetta TF: Distribution of c-myc oncoprotein in healthy and atherosclerotic human carotid arteries. J Vasc Surg. 18:170–177. 1993. View Article : Google Scholar : PubMed/NCBI

45 

Kockx MM, De Meyer GR, Muhring J, Jacob W, Bult H and Herman AG: Apoptosis and related proteins in different stages of human atherosclerotic plaques. Circulation. 97:2307–2315. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Saxena A, McMeekin JD and Thomson DJ: Expression of Bcl-x, Bcl-2, Bax, and Bak in endarterectomy and atherectomy specimens. J Pathol. 196:335–342. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Shi Y and Tokunaga O: Chlamydia pneumoniae (C. pneumoniae) infection upregulates atherosclerosis-related gene expression in human umbilical vein endothelial cells (HUVECs). Atherosclerosis. 177:245–253. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Anikhovskaia IA, Kubatiev AA and Iakovlev Mlu: Endotoxin theory of atherosclerosis. Fiziol Cheloveka. 41:106–116. 2015.(In Russian). PubMed/NCBI

49 

Wort SJ and Evans TW: The role of the endothelium in modulating vascular control in sepsis and related conditions. Br Med Bull. 55:30–48. 1999. View Article : Google Scholar : PubMed/NCBI

50 

Hennessy EJ, Sheedy FJ, Santamaria D, Barbacid M and O'Neill LA: Toll-like receptor-4 (TLR4) down-regulates microRNA-107, increasing macrophage adhesion via cyclin-dependent kinase 6. J Biol Chem. 286:25531–25539. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Kumar V and Mahato RI: Delivery and targeting of miRNAs for treating liver fibrosis. Pharm Res. 32:341–361. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 19 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
Shen L, Hu Y, Lou J, Yin S, Wang W, Wang Y, Xia Y and Wu W: CircRNA‑0044073 is upregulated in atherosclerosis and increases the proliferation and invasion of cells by targeting miR‑107. Mol Med Rep 19: 3923-3932, 2019
APA
Shen, L., Hu, Y., Lou, J., Yin, S., Wang, W., Wang, Y. ... Wu, W. (2019). CircRNA‑0044073 is upregulated in atherosclerosis and increases the proliferation and invasion of cells by targeting miR‑107. Molecular Medicine Reports, 19, 3923-3932. https://doi.org/10.3892/mmr.2019.10011
MLA
Shen, L., Hu, Y., Lou, J., Yin, S., Wang, W., Wang, Y., Xia, Y., Wu, W."CircRNA‑0044073 is upregulated in atherosclerosis and increases the proliferation and invasion of cells by targeting miR‑107". Molecular Medicine Reports 19.5 (2019): 3923-3932.
Chicago
Shen, L., Hu, Y., Lou, J., Yin, S., Wang, W., Wang, Y., Xia, Y., Wu, W."CircRNA‑0044073 is upregulated in atherosclerosis and increases the proliferation and invasion of cells by targeting miR‑107". Molecular Medicine Reports 19, no. 5 (2019): 3923-3932. https://doi.org/10.3892/mmr.2019.10011