Open Access

Cold‑pressed oil from Citrus aurantifolia inhibits the proliferation of vascular smooth muscle cells via regulation of PI3K/MAPK signaling pathways

  • Authors:
    • Byeong-Wook Song
    • Chang Youn Lee
    • Jun-Hee Park
    • Bomi Kim
    • Seahyoung Lee
    • Soyeon Lim
    • Sang Woo Kim
    • Jung-Won Choi
    • Misun Kang
    • Jung Hwa Kang
    • Sung-Suk Lee
    • Mi-Jin Park
    • Hanbyeol Moon
    • Ki-Chul Hwang
    • Il-Kwon Kim
  • View Affiliations

  • Published online on: November 2, 2021     https://doi.org/10.3892/etm.2021.10943
  • Article Number: 21
  • Copyright: © Song 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

Vascular occlusive disease is a chronic disease with significant morbidity and mortality. Although a variety of therapies and medications have been developed, the likelihood of disease re‑emergence is high and this can be life‑threatening. Based on a previous screening experiment related to vascular obstructive diseases using 34 types of essential oils, cold‑pressed oil (CpO) from Citrus aurantifolia (lime) has been demonstrated to have the best effect for the inhibition of vascular smooth muscle cells (VSMCs) proliferation. The aim of the present study was to evaluate the effect of lime CpO on the pathological changes of VSMCs. To determine this, the effect of lime CpO on VSMC proliferation, a major cause of vascular disease, was investigated. To determine the safe concentration interval for toxicity of CpO during VSMC culture, a dilution of 1x10‑5 was determined using Cell Counting Kit‑8 assay, which was confirmed to be non‑toxic using a lactate dehydrogenase assay. To examine the effect of lime CpO in cellular signaling pathways, changes in phosphorylation of both the PI3K/AKT/mTOR and extracellular signal‑regulated MEK/ERK signaling pathways with serum were investigated. Furthermore, lime CpO with FBS also significantly decreased the expression levels of the cell cycle regulators cyclin D1 and proliferating cell nuclear antigen. Additionally, lime CpO with FBS significantly inhibited the sprouting of VSMCs in an ex vivo culture system. These results suggested that lime CpO inhibited the abnormal proliferation of VSMCs and can be developed as a nature‑based therapeutic agent for obstructive vascular disease.

Introduction

Vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) are the main cell types that constitute the blood vessels, with their function being to control blood vessel pressure and tension (1). VSMCs maintain homeostasis in a contractile form with little proliferation under physiological conditions (2,3). However, when ECs are damaged, inflammatory factors secreted in response to pathological conditions cause abnormal proliferation of VSMCs, initiating their transformation to the synthetic phenotype due to internal and external changes (3-5). This abnormal proliferation of VSMCs is known to cause arteriosclerosis and restenosis (6,7). Classical surgical therapies for treating these vascular diseases, such as coronary artery bypass grafting or stent-grafted angioplasty, are effective for a short period of time. However, these surgical treatments can cause blood flow disorders due to the abnormal proliferation of VSMCs over time (8). To overcome this problem, a drug-coated stent has been developed to treat vascular diseases more effectively (9). However, the potential for restenosis due to VSMC proliferation remains a major long-term threat (8). Therefore, the development of novel therapeutic agents for the treatment of vascular diseases is still required.

It has been reported that natural compounds have a positive effect on cardiovascular diseases and improve some vascular functions (9). One of the promising novel treatments of vascular diseases in various natural products is essential oils (EOs). EOs and their constituents are promising as therapeutic agents as they have been demonstrated to functionally improve cardiovascular disease (10,11).

Lime (Citrus aurantifolia) is widely cultivated worldwide and is an excellent source of vitamins, particularly vitamin C. Lime EO has previously been used as a fragrance, an antimicrobial agent and for aromatherapy (12,13). Lime EO contains limonene, β-pinene, γ-terpinene, citral and linarul, among other compounds. Lime EO is a volatile complex mixture and has been used as a means to prevent, improve and treat diseases (14). Furthermore, the potential for the use of lime EO as chemotherapy in the prevention and treatment of inflammatory diseases (15,16), cancer (17,18) and oxidative stress (19,20), has been reported in various studies. Regarding the applications in cardiovascular disease, it has been demonstrated that daily intake of lemon (Citrus limon), a Citrus species that has a similar nutritional value to lime, lowers systolic blood pressure (21).

To the best of our knowledge, the effect of cold-pressed oil (CpO) from limes on VSMC pathological changes is unclear. It was hypothesized that lime CpO could inhibit the proliferation of VSMC through known vascular signaling pathways (22,23) in cardiovascular disease. The present study investigated whether lime CpO could modulate the MAPK and PI3K signaling pathways to reduce abnormal cell proliferation induced by FBS.

Materials and methods

Lime CpO

Cold pressed lime oil was purchased from Sydney Essential Oil Company. Coupled gas chromatography-mass spectrometry (GC-MS) analysis was performed using a Thermo Scientific Model ISQ LT (Thermo Fisher Scientific, Inc.) equipped with both a flame ionization detector (FID) and a mass spectrometer.

GC-MS analysis

A Durabond-5MS capillary column (60 m x 0.25 mm x 0.25 µm film thickness; Agilent Technologies, Inc.) was used. The carrier gas, helium, had a constant flow rate of 1 ml/min. The injection temperature was 250˚C and 1 µl of the sample was injected with a split ratio of 1:20. The oven temperature was maintained at 50˚C for 5 min, then increased by 10˚C/min to 65˚C and held for 30 min, increased by 5˚C/min to 120˚C and held for 15 min, increased by 1˚C/min to 140˚C and held for 11 min, increased by 10˚C/min to 250˚C and held for 5 min, and finally increased by 20˚C/min to 325˚C and held for 6 min. For the FID, the temperature was set to 300˚C, air flow was set to 350 ml/min, hydrogen flow was set to 35 ml/min and the make-up gas (helium) flow was set to 40 ml/min. The mass interface temperature was 250˚C and the ion source temperature was 250˚C. Mass scan data were acquired in electron ionization (EI) mode at a 0.2 sec scan time rate with a scan range of 35-550 amu. The identification of peaks was performed by comparing the peak average mass spectrum of the peak with an electronic library database (National Institute of Standards and Technology, Environmental Protection Agency and National Institutes of Health Mass Spectral Library; version 2.0 g: https://chemdata.nist.gov/). The identity of the compounds was assigned by comparison of the Kovats retention index, determined in relation to a homologous series of n-alkanes (C7-C30).

Animal care

All animal experiments were approved by the Institutional Animal Care and Use Committee of Catholic Kwandong University, International St. Mary's Hospital (Incheon, South Korea; approval no. CKU 01-2019-008) in cooperation with the Association for Assessment and Accreditation of Laboratory Animal Care and performed in accordance with the Guidelines and Regulations for Animal Care (24). Rats were housed in a room with a stable temperature of 22.5±1.5˚C, 50-60% humidity and 12 h light-dark cycles with ad libitum access to food and water. A total of 16 rats were divided into four groups: control (non-treated), lime CpO, FBS (FBS only treated), and FBS and lime CpO. Rats were intramuscularly anesthetized with Zoletil™50 (tiletamine:zolazepam = 1:1; 20 mg/kg; Virbac) and Rompun 2% (xylazine; 5 mg/kg; Bayer AG), and euthanized via an intraperitoneal overdose of sodium pentobarbital (100-200 mg/kg). Euthanasia was confirmed with lack of heartbeat.

Isolation and culture of rat aortic VSMCs

Rat aortic VSMCs were isolated as previously described (25,26). Thoracic aortas from male Sprague-Dawley rats (Orient Bio; n=3; weight, 200-250 g; age, 6-8 weeks) were removed and transferred to incubate in serum-free DMEM (Invitrogen; Thermo Fisher Scientific, Inc.) containing 100 U/ml penicillin and 100 µg/ml streptomycin. The aortas were separated from the connective tissue and transferred to a petri dish containing 5 ml of an enzyme dissociation mixture, composed of DMEM with 1 mg/ml collagenase type I (Sigma-Aldrich; Merck KGaA) and 0.5 µg/ml elastase (Thermo Fisher Scientific, Inc.), and incubated for 30 min at 37˚C. The aortas were then transferred to DMEM and the adventitia was stripped off each aorta with forceps under an optical microscope. Subsequently, the aortas were transferred to a conical tube containing 5 ml of enzyme dissociation mixture containing DMEM with 1 mg/ml of collagenase type I (Sigma-Aldrich; Merck KGaA) and 0.5 µg/ml of elastase (Thermo Fisher Scientific, Inc.) and incubated for 2 h at 37˚C. The suspension was centrifuged at 320 x g for 10 min at room temperature and the pellet was re-suspended in DMEM with 10% FBS (Atlas Biologicals, Inc) under room temperature. Rat aortic VSMCs were cultured in DMEM supplemented with 10% FBS, 1% penicillin-streptomycin in 75-cm2 flasks in a 37˚C incubator with 5% CO2 (Thermo Fisher Scientific, Inc.). Isolated cells from 3 different animals in passages 5-8 were used in the present study.

CpO treatment

The concentration of CpO on VSMCs culture was determined as 10-5 dilution after Cell Counting Kit-8 (CCK-8) assay. For treatment with CpO, VSMCs were seeded into 96-well plates at a density of 5x103 cells/well. VSMCs were serum-starved in DMEM containing 0.5% FBS for 24 h and treated with or without 5% FBS in DMEM for the following 24 h to detect the effects of lime CpO under in vitro and ex vivo conditions in a 37˚C incubator. In order to check the cell signal transduction, MEK1/2 inhibitor U0126 (10 µM; Cell Signaling Technology, Inc.) and mTOR inhibitor Rapamycin (20 nM; Calbiochem; Merck KGaA) were treated with 5% FBS for 24 h.

Cell proliferation assay

A CCK-8 (DoGenBio Co., Ltd.) assay was used according to the manufacturer's protocol. To each well, 10% (v/v) CCK-8 reagent was added and incubated at 37˚C for 2 h to allow for the formation of water-soluble tetrazolium salts (WST)-8 formazan. The absorbance at 450 nm was measured using a microplate reader (Thermo Fisher Scientific, Inc.).

Cytotoxicity assay

The Cytotoxicity Lactate Dehydrogenase (LDH) Assay Kit-WST (cat. no. MK401; Takara Bio, Inc.) was used according to the manufacturer's protocol. Cell suspension and 50 µl DMEM were sequentially added into a 96-well culture plate (7x103 cells/well) and samples were incubated at 37˚C for 24 h. Samples were mixed with 100 µl working solution at room temperature for 30 min in the dark. The absorbance at 490 nm was measured using a microplate reader (Thermo Fisher Scientific, Inc.).

Western blotting

VSMCs were washed once with PBS and lysed using RIPA lysis buffer (Thermo Fisher Scientific, Inc.). Total protein concentration was quantified using an albumin standard (bioPLUS™; BioWORLD). Proteins were then separated by 10% SDS-PAGE by 15-20 mg/ml of protein loading per lane and transferred to a PVDF membrane (MilliporeSigma). After blocking of the membrane with TBS with 0.1% Tween-20 (TBS-T; BioPLUS Chemicals) and 5% (w/v) BSA (bioPLUS™; BioWORLD) in 0.1% TBS-T for 1 h at room temperature, the membrane was washed twice with TBS-T and incubated with the primary antibodies diluted in blocking buffer a ratio of 1:1,000 to 1:2,000 overnight at 4˚C. The membrane was washed three times with TBS-T for 5 min/wash and incubated for 1 h at room temperature with horseradish peroxidase-conjugated secondary antibodies diluted in blocking buffer a ratio of 1:4,000. The membrane was subsequently washed six times with TBS-T for 5 min/wash and bands were detected with an enhanced chemiluminescence reagent (Abclon, Inc.). Band intensities were semi-quantified using a Davinch-Western Imaging System (Davinch-K Co., Ltd.) and ImageJ version 1.44p software (National Institutes of Health). The following antibodies were used in these experiments: anti-PI3K (cat. no. sc-7189; Santa Cruz Biotechnology, Inc.), anti-phosphorylated (p)-PI3K (cat. no. 4228S; Cell Signaling Technology, Inc.), anti-AKT (cat. no. 9272S; Cell Signaling Technology, Inc.), anti-p-AKT (cat. no. 9271S; Cell Signaling Technology, Inc.), anti-mTOR (cat. no. 2972S; Cell Signaling Technology, Inc.), anti-p-mTOR (cat. no. 2971S; Cell Signaling Technology, Inc.), anti-MEK (cat. no. 9122; Cell Signaling Technology, Inc.), anti-p-MEK (cat. no. 9121S; Cell Signaling Technology, Inc.), anti-ERK (cat. no. 9102S; Cell Signaling Technology, Inc.), anti-p-ERK (cat. no. sc-7383; Santa Cruz Biotechnology, Inc.), anti-cyclin D1 (cat. no. 29785; Cell Signaling Technology, Inc.), anti-proliferating cell nuclear antigen (PCNA; cat. no. sc-56; Santa Cruz Biotechnology, Inc.), anti-β-actin (cat. no. ab8227-50; Abcam), anti-caspase-3 (cat. no. ab13847; Abcam) and anti-cleaved caspase-3 (cat. no. ab49822; Abcam). Secondary antibodies were used in these experiments as follows: Goat anti-mouse IgG (H+L)-HRP (cat. no. SA001-500) and goat anti-rabbit IgG (H+L)-HRP (cat. no. SA002-500) from GenDEPOT, LLC. The amount of phosphorylation was calculated by dividing the amount by the total expression amount. This was clarified using magnification of phosphorylation/expression ratio.

Immunocytochemistry

VSMCs were seeded into four-well plastic cell culture dishes (1x105 cells/well) and were treated with CpO for 24 h in a 37˚C incubator. Subsequently, each well was washed with PBS. Cells were then fixed with 4% paraformaldehyde diluted in PBS for 10 min at room temperature, after which they were washed twice with PBS and permeabilized for 10 min at room temperature with 0.2% Triton X-100 diluted in PBS. After washing with PBS, the cells were blocked in blocking solution (2% BSA and 10% horse serum (Vector Laboratories, Inc.; Maravai LifeSciences) in PBS) for 30 min at room temperature and stained for Ki-67 (Dako; cat. no. M7240; 1:200 dilution) for 1 h at 37˚C. The cells were incubated with a FITC-conjugated anti-mouse (cat. no. 115-095-003; 1:500 dilution; Jackson ImmunoResearch Laboratories, Inc.) secondary antibody at room temperature for 1 h and then stained with a DAPI solution (0.1 µl/ml; Thermo Fisher Scientific, Inc.) for 5 min. Immunofluorescence was detected via confocal microscopy (LSM700; Zeiss GmbH) and analyzed using ZEN 2.5 Blue Edition software (Zeiss GmbH) 1.0 for analysis.

Ex vivo aortic ring assay

Ex vivo sprouting of VSMCs was measured via an aortic ring assay using Matrigel (BD Biosciences). The thoracic aortas from the aforementioned 8-week-old Sprague-Dawley rats were removed and transferred into serum-free DMEM. The endothelial lining was removed using a 2-Fr Fogarty balloon catheter (Baxter Healthcare) to minimize the possibility of EC sprouting during the ring assay. The aorta was washed by gradually passing PBS through the aorta three times. After removing perivascular adipose tissue, the aorta was cut into 1-mm-thick segments of aortic ring and placed in Matrigel. The aortic rings were washed with serum-free DMEM twice and starved in DMEM supplemented with 0.5% FBS for 24 h at 37˚C. For the lime CpO group, the medium was changed to DMEM supplemented with 5% FBS containing 1x10-5 diluted lime CpO. The cells were cultured at 37˚C and the media were changed every 3 days and the aortic rings were monitored daily for up to 7 days for sprouting VSMCs. On the 7th day the results were analyzed.

Statistical analysis

Data are presented as the mean ± SEM of at least three independent experiments. For statistical analysis, one-way analysis of variance with Bonferroni's correction was performed for comparisons among more than two groups. All analyses were performed using Prism software (version 5.0; GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Suppression of VSMC proliferation by lime CpO treatment

To evaluate the effect of lime CpO on VSMC proliferation, cells were treated with lime CpO for 24 h. In the group treated with FBS only, the cell proliferation rate was significantly increased by ~30% compared with that of the FBS-negative and lime CpO-negative control groups. Concentration of 10-5 lime CpO was statistically similar to the control, however concentration of 10-4 and 10-3 lime CpO was significantly lower (Fig. 1A). Based on these results, lime CpO significantly inhibited FBS-stimulated VSMC proliferation at low concentrations (10-5). The effect of lime CpO on cell death was assessed via LDH analysis and it was demonstrated to have no statistically significant effect on cell death compared with the FBS-treated only group (Fig. 1B). Furthermore, caspase-3 and cleaved caspase-3 protein levels were also not affected by lime CpO treatment compared with the FBS-treated or FBS-negative and lime CpO-negative control groups (Fig. 1C). Therefore, the results suggested that lime CpO may inhibit FBS-induced proliferation of VSMCs but does not cause cell death.

Changes in AKT and ERK signaling cascades in VSMC proliferation

The mechanism by which lime CpO regulated proliferation was investigated further. Since phosphorylation of the PI3K/AKT/mTOR signaling cascade is known to regulate VSMC proliferation (27), the phosphorylation of these signaling cascades was analyzed. Phosphorylation of the PI3K/AKT/mTOR (Fig. 2A-D) and MEK/ERK signaling pathways (Fig. 2E-G) was significantly increased by 1.5-6.2-fold in FBS-treated cells compared with the FBS-negative and lime CpO-negative control group. However, the signal intensity of each phosphorylated protein band was significantly reduced in the lime CpO-treated group compared with the FBS-treated cells or the U0126 or Rapamycin-treated cells. These results demonstrated that lime CpO inhibited phosphorylation in a manner similar to that of the MEK and mTOR inhibitors in VSMCs (Fig. 2H).

Regulation of cell cycle regulators in VSMCs

It was subsequently demonstrated that the cell cycle-regulating factors cyclin D1 and PCNA were essential for regulating proliferation in lime CpO-treated cells. The protein expression levels of cyclin D1 and PCNA were significantly increased in FBS-treated cells compared with FBS-negative and lime CpO-negative control cells; however, the lime CpO-treated cells displayed significantly reduced protein expression levels compared with the FBS-treated cells (Fig. 3A-C). Ki-67 was observed in the nucleus during cell division, indicating cell proliferation (Fig. 3D-E). The number of Ki-67-positive cells was significantly increased by ~35% in the FBS-treated group compared with the FBS-negative and lime CpO-negative control group. However, the number of Ki-67-positive cells was significantly reduced in the lime CpO-treated group compared with the FBS-treated group (Fig. 3D and E). This result suggested that lime CpO had the potential to suppress the cell cycle of VSMCs and regulate VSMC proliferation.

Inhibition of VSMC proliferation in endothelium-denuded aortic rings

The effect of lime CpO on the proliferation and migration of VSMCs in tissues was investigated and an ex vivo aortic ring analysis was performed (Fig. 4A). EC-denuded vascular tissue was cultured in DMEM containing FBS to observe the proliferation and migration of VSMCs. Intravascular VSMCs spread to the outside of the aortic rings and significantly proliferated in response to FBS compared with the cells in the FBS-negative and lime CpO-negative control group. However, the degree of spread of the lime CpO and FBS-treated VSMCs was significantly decreased compared with the FBS-treated group, to a similar level to that of the control cells (treated with nether CpO or FBS) (Fig. 4B). These results suggested that lime CpO inhibited abnormal VSMC proliferation in injured vascular tissues and possibly contributed to tissue homeostasis.

Analysis of lime CpO components

In the present study, GC-MS analysis was performed to evaluate the characteristics and components of lime CpO (Table I). Table I displays the results of the component analysis of lime CpO by GC-MS. In total, ~36 types of volatile compounds were detected as main materials, including 23 monoterpenes, 9 sesquiterpenes and 4 other components. Monoterpenes (M) and sesquiterpenes (S) accounted for 94.6 and 4.6% of EOs, respectively, in summed area % of GC-MS spectrometry (Table I). D-limonene (41.40%), γ-terpinene (13.19%), terpinolene (10.41%) and α-terpineol (8.61%) were the major monoterpenes among the detected volatile components, overall accounting for 73.61% of the total volatile components (Table I).

Table I

Chemical Composition of Lime CpO by MS and KI identification in GC-MS spectrometry.

Table I

Chemical Composition of Lime CpO by MS and KI identification in GC-MS spectrometry.

RTConstituentArea, %KIaClassification
24.55α-Pinene0.90927(M)
27.08Camphene0.26943(M)
30.36 Linalool-3,7-oxide0.10963(M)
31.89β-Pinene1.48972(M)
34.15β-Myrcene0.36986(M)
39.221,4-Cineole1.391026(M)
39.52α-Terpinene1.051029(M)
40.61ρ-Cymene4.231039(M)
41.27D-Limonene41.401045(M)
41.50Eucalyptol1.111048(M)
42.77Ocimene quintoxide0.121059(O)
42.98(Z)-β-Ocimene0.321061(M)
44.08γ-Terpinene13.191072(M)
46.19Terpinolene10.411092(M)
46.57ρ-Cymenene0.331095(M)
47.14Linalool0.131101(M)
48.49Fenchyl alcohol0.481121(M)
49.531-Terpineol0.581136(M)
50.48β-Terpineol0.721150(M)
51.79Decanal0.251169(O)
52.22endo-Borneol0.181175(M)
52.77Terpinen-4-ol0.441183(M)
53.24ρ-Cymen-8-ol0.201190(M)
53.92α-Terpineol8.611200(M)
54.26γ-Terpineol1.631204(M)
68.01 trans-Caryophyllene0.471434(S)
68.33α-Bergamotene0.781442(S)
68.89β-Farnesene0.101457(S)
69.39α-Humulene0.131471(S)
69.95α-Selinene0.141486(S)
70.42β-Cadinene0.171498(S)
70.69α-Farnesene0.751507(S)
70.95β-Bisabolene1.521516(S)
71.94β-Maaliene0.261548(S)
89.83Butyl palmitate0.242188(O)
91.85Butyl stearate0.202389(O)

[i] aKI on a DB-5 column in reference to n-alkanes. MS, National Institute of Standards and Technology, Environmental Protection Agency and National Institutes of Health Mass Spectral Library; KI, Kovats retention index; RT, Retention time; M, monoterpenes, S, sesquiterpenes; O, others.

Discussion

The present study investigated the effect of lime CpO, a natural substance, on VSMC proliferation. To examine whether lime CpO was involved in VSMC regulation, cellular signaling pathways were analyzed using western blotting. Lime CpO was demonstrated to modulate cell cycle regulators in VSMCs. Furthermore, in ex vivo conditions, lime CpO negatively regulated VSMC proliferation. Among 36 types of volatile compounds identified in lime CpO, four molecules (D-limonene, γ-terpinene, terpinolene and α-terpineol) were specifically detected as the main components.

In the past decade, natural products have been demonstrated to be mostly non-toxic and have been used successfully as therapeutics for numerous diseases (10,28-30). Among these natural products, EOs are promising pharmaceutical agents as they are clinically applicable and can be properly managed and industrialized for human diseases (31). Depending on the EO extraction method, such as steam distillation, expression (for example cold compression) and solvent extraction, the properties of the active compounds may show different distribution ratio in total ingredients, but the main active substances remain unchanged (32). According to a review by Narang and Jiraungkoorskul (17), lime and its related byproducts have been suggested to have potential therapeutic effects in colon cancer, pancreatic cancer, breast cancer, skin cancer and lymphoma. Lime and its related byproducts have also been demonstrated to inhibit the cell cycle and cell proliferation (17). Patil et al (18) reported that lime inhibited cancer cell proliferation by modulating Bax, Bcl-2, caspase-3 and p53 in pancreatic cancer cells. Furthermore, lime has been reported to improve obesity, the atherogenic index and fatty liver disease by inducing antioxidant capacity and hypolipidemic effects (33). Despite the various previous studies on lime CpO, to the best of our knowledge, there are currently no reports on the inhibition of VSMC proliferation, a major cause of occlusive vascular disease.

The present study demonstrated that lime CpO treatment at 10-5 concentrations inhibited FBS-induced VSMC proliferation. Furthermore, lime CpO reduced FBS-induced phosphorylation of the MAPK/ERK and PI3K/AKT/mTOR signaling pathways. Lime CpO inhibited the MAPK/ERK and PI3K/AKT/mTOR signaling pathways at comparable or higher degree. However, there was no change in the expression or activity of caspase-3 under the experimental conditions of the present study. A possible reason for why these results differed from previous reports is that the lime CpO dose is modifiable for different cytotoxic effects under various cell types. In the present study, preliminary experiments were conducted to determine concentrations that were not cytotoxic (Fig. 1A). Compared with conventional chemical drugs, lime CpO is a natural compound with little to no toxicity and controllable doses that can be used in the body (34). The use of natural products as therapeutics is advantageous as they are traditionally used by human and therefore less likely to cause an adverse reaction when developed as medicines (9). However, in future work, lime CpO toxicity will be tested in other vascular constituent cells besides VSMCs, such as vascular ECs and vascular fibroblasts. Furthermore, whether the same effect is shown in other batches of lime CpO and identification of the active compounds via selection experiments with various combinations of constituents from lime CpO should be investigated in future studies.

A previous report suggested that neroli (Citrus aurantium L.) EO is an endothelium and smooth muscle-dependent vasodilator, which alleviates cardiovascular disease (35). It has been reported that neroli EO modulated intracellular Ca2+ concentrations via the inhibition of cation channel-mediated extracellular Ca2+ influx and store-operated Ca2+ release mediated by the ryanodine receptor signaling pathway (35). Among the proposed constituent compounds of neroli EO, D-limonene and α-terpineol are included in the data from the present study, suggesting that lime CpO may also act as a smooth muscle-dependent vasodilator (35). Furthermore, neroli EO have also reported to possess 100% singlet oxygen scavenging activity as a strong antioxidant (12,36,37). Therefore, future work may examine the endothelium and/or smooth muscle-mediated vasodilator effect of lime CpO in a cardiovascular disease model. In the present study, lime CpO inhibited excess VSMC proliferation, which causes occlusive vascular disease, via the regulation of MAPK and PI3K signaling pathways. These results suggest that lime CpO may be developed as a potential therapeutic or health supplement for the treatment of cardiovascular disease. Future studies may explore the different effects of lime CpO compared with lime EO and validate its pharmacological effects on classified compounds including D-limonene, γ-terpinene, terpinolene and α-terpineol to investigate the proposed molecules of in vivo cardiovascular disease.

Supplementary Material

Gas chromatography-mass spectrometry traces of lime cold-pressed oil.

Acknowledgements

Not applicable.

Funding

The present study was supported by the following grants from the Korea Forest Service (Seoul, Korea): Forest Science Technology Research and Development Project (grant no. 2017026B10-1719-BA01) and the National Institute of Forest Science, Forest Science Technology Research and Development Project (grant no. FP0900-2016-01).

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

BWS, CYL and IKK were involved in the conceptualization, writing, editing and data analysis. JHP, BK, SLe, SLi, SWK, JWC, MK, JHK, SSL, MJP, HM and KCH performed and analyzed the experiments, and edited the manuscript. BWS and IKK confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was performed according to a protocol approved by the Institutional Animal Care and Use Committee of Catholic Kwandong University (approval no. CKU 01-2019-008; Incheon, South Korea).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Rensen SSM, Doevendans PAFM and van Eys GJJM: Regulation and characteristics of vascular smooth muscle cell phenotypic diversity. Neth Heart J. 15:100–108. 2007.PubMed/NCBI View Article : Google Scholar

2 

Hao H, Gabbiani G and Bochaton-Piallat ML: Arterial smooth muscle cell heterogeneity: Implications for atherosclerosis and restenosis development. Arterioscler Thromb Vasc Biol. 23:1510–1520. 2003.PubMed/NCBI View Article : Google Scholar

3 

Morgan JP, Perreault CL and Morgan KG: The cellular basis of contraction and relaxation in cardiac and vascular smooth muscle. Am Heart J. 121:961–968. 1991.PubMed/NCBI View Article : Google Scholar

4 

Sprague AH and Khalil RA: Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem Pharmacol. 78:539–552. 2009.PubMed/NCBI View Article : Google Scholar

5 

Gomez D and Owens GK: Smooth muscle cell phenotypic switching in atherosclerosis. Cardiovasc Res. 95:156–164. 2012.PubMed/NCBI View Article : Google Scholar

6 

Anwar MA, Shalhoub J, Lim CS, Gohel MS and Davies AH: The effect of pressure-induced mechanical stretch on vascular wall differential gene expression. J Vasc Res. 49:463–478. 2012.PubMed/NCBI View Article : Google Scholar

7 

Owens GK, Kumar MS and Wamhoff BR: Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev. 84:767–801. 2004.PubMed/NCBI View Article : Google Scholar

8 

Clowes AW and Reidy MA: Prevention of stenosis after vascular reconstruction: Pharmacologic control of intimal hyperplasia - a review. J Vasc Surg. 13:885–891. 1991.PubMed/NCBI View Article : Google Scholar

9 

Garg S and Serruys PW: Coronary stents: Current status. J Am Coll Cardiol. 56 (Suppl 10):S1–S42. 2010.PubMed/NCBI View Article : Google Scholar

10 

de Andrade TU, Brasil GA, Endringer DC, da Nóbrega FR and de Sousa DP: Cardiovascular activity of the chemical constituents of essential oils. Molecules. 22(E1539)2017.PubMed/NCBI View Article : Google Scholar

11 

Amorati R, Foti MC and Valgimigli L: Antioxidant activity of essential oils. J Agric Food Chem. 61:10835–10847. 2013.PubMed/NCBI View Article : Google Scholar

12 

Dosoky NS and Setzer WN: Biological activities and safety of Citrus spp. essential oils. Int J Mol Sci. 19(1966)2018.PubMed/NCBI View Article : Google Scholar

13 

Reis D and Jones T: Aromatherapy: Using Essential Oils as a Supportive Therapy. Clin J Oncol Nurs. 21:16–19. 2017.PubMed/NCBI View Article : Google Scholar

14 

Spadaro F, Costa R, Circosta C and Occhiuto F: Volatile composition and biological activity of key lime Citrus aurantifolia essential oil. Nat Prod Commun. 7:1523–1526. 2012.PubMed/NCBI

15 

Amorim JL, Simas DLR, Pinheiro MMG, Moreno DSA, Alviano CS, da Silva AJR and Fernandes PD: Anti-inflammatory properties and chemical characterization of the essential oils of four Citrus species. PLoS One. 11(e0153643)2016.PubMed/NCBI View Article : Google Scholar

16 

Kummer R, Fachini-Queiroz FC, Estevão-Silva CF, Grespan R, Silva EL, Bersani-Amado CA and Cuman RKN: Evaluation of anti-inflammatory activity of Citrus latifolia Tanaka essential oil and limonene in experimental mouse models. Evid Based Complement Alternat Med. 2013(859083)2013.PubMed/NCBI View Article : Google Scholar

17 

Narang N and Jiraungkoorskul W: Anticancer Activity of Key Lime, Citrus aurantifolia. Pharmacogn Rev. 10:118–122. 2016.PubMed/NCBI View Article : Google Scholar

18 

Patil JR, Chidambara Murthy KN, Jayaprakasha GK, Chetti MB and Patil BS: Bioactive compounds from Mexican lime (Citrus aurantifolia) juice induce apoptosis in human pancreatic cells. J Agric Food Chem. 57:10933–10942. 2009.PubMed/NCBI View Article : Google Scholar

19 

Tosukhowong P, Yachantha C, Sasivongsbhakdi T, Ratchanon S, Chaisawasdi S, Boonla C and Tungsanga K: Citraturic, alkalinizing and antioxidative effects of limeade-based regimen in nephrolithiasis patients. Urol Res. 36:149–155. 2008.PubMed/NCBI View Article : Google Scholar

20 

Boshtam M, Moshtaghian J, Naderi G, Asgary S and Nayeri H: Antioxidant effects of Citrus aurantifolia (Christm) juice and peel extract on LDL oxidation. J Res Med Sci. 16:951–955. 2011.PubMed/NCBI

21 

Kato Y, Domoto T, Hiramitsu M, Katagiri T, Sato K, Miyake Y, Aoi S, Ishihara K, Ikeda H, Umei N, et al: Effect on blood pressure of daily lemon ingestion and walking. J Nutr Metab. 2014(912684)2014.PubMed/NCBI View Article : Google Scholar

22 

Muslin AJ: MAPK signalling in cardiovascular health and disease: Molecular mechanisms and therapeutic targets. Clin Sci (Lond). 115:203–218. 2008.PubMed/NCBI View Article : Google Scholar

23 

Muto A, Fitzgerald TN, Pimiento JM, Maloney SP, Teso D, Paszkowiak JJ, Westvik TS, Kudo FA, Nishibe T and Dardik A: Smooth muscle cell signal transduction: implications of vascular biology for vascular surgeons. J Vasc Surg. 45 (Suppl A):A15–A24. 2007.PubMed/NCBI View Article : Google Scholar

24 

National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals: Guide for the Care and Use of Laboratory Animals, 8th edition. National Academies Press (US), Washington, DC, 2011.

25 

Chang W, Lim S, Song H, Song BW, Kim HJ, Cha MJ, Sung JM, Kim TW and Hwang KC: Cordycepin inhibits vascular smooth muscle cell proliferation. Eur J Pharmacol. 597:64–69. 2008.PubMed/NCBI View Article : Google Scholar

26 

Lim S, Lee SY, Seo HH, Ham O, Lee C, Park JH, Lee J, Seung M, Yun I, Han SM, et al: Regulation of mitochondrial morphology by positive feedback interaction between PKCδ and Drp1 in vascular smooth muscle cell. J Cell Biochem. 116:648–660. 2015.PubMed/NCBI View Article : Google Scholar

27 

Mathew OP, Ranganna K, Mathew J, Zhu M, Yousefipour Z, Selvam C and Milton SG: Cellular effects of butylate on vascular smooth muscle cells are mediated through disparate actions on dual targets, histone deacetylase (HDAC) activity and PI3K/Akt signaling network. Int J Mol Sci. 20(2902)2019.PubMed/NCBI View Article : Google Scholar

28 

Suen J, Thomas J, Kranz A, Vun S and Miller M: Effect of flavonoids on oxidative stress and inflammation in adults at risk of cardiovascular disease: a systematic review. Healthcare (Basel). 4(69)2016.PubMed/NCBI View Article : Google Scholar

29 

Li D, Wu H, Dou H, Guo L and Huang W: Microcapsule of sweet orange essential oil changes gut microbiota in diet-induced obese rats. Biochem Biophys Res Commun. 505:991–995. 2018.PubMed/NCBI View Article : Google Scholar

30 

Jiang D, Li D and Wu W: Inhibitory effects and mechanisms of luteolin on proliferation and migration of vascular smooth muscle cells. Nutrients. 5:1648–1659. 2013.PubMed/NCBI View Article : Google Scholar

31 

Lv X, Zhao S, Ning Z, Zeng H, Shu Y, Tao O, Xiao C, Lu C and Liu Y: Citrus fruits as a treasure trove of active natural metabolites that potentially provide benefits for human health. Chem Cent J. 9(68)2015.PubMed/NCBI View Article : Google Scholar

32 

Aziz ZAA, Ahmad A, Setapar SHM, Karakucuk A, Azim MM, Lokhat D, Rafatullah M, Ganash M, Kamal MA and Ashraf GM: Essential oils: extraction techniques, pharmaceutical and therapeutic potential - a review. Curr Drug Metab. 19:1100–1110. 2018.PubMed/NCBI View Article : Google Scholar

33 

Lin LY, Chuang CH, Chen HC and Yang KM: Lime (Citrus aurantifolia (Christm.) swingle) essential oils: volatile compounds, antioxidant capacity, and hypolipidemic effect. Foods. 8(E398)2019.PubMed/NCBI View Article : Google Scholar

34 

Adokoh CK, Asante DB, Acheampong DO, Kotsuchibashi Y, Armah FA, Sirikyi IH, Kimura K, Gmakame E and Abdul-Rauf S: Chemical profile and in vivo toxicity evaluation of unripe Citrus aurantifolia essential oil. Toxicol Rep. 6:692–702. 2019.PubMed/NCBI View Article : Google Scholar

35 

Kang P, Ryu K-H, Lee J-M, Kim H-K and Seol GH: Endothelium- and smooth muscle-dependent vasodilator effects of Citrus aurantium L. var. amara: Focus on Ca(2+) modulation. Biomed Pharmacother. 82:467–471. 2016.PubMed/NCBI View Article : Google Scholar

36 

Ammar AH, Bouajila J, Lebrihi A, Mathieu F, Romdhane M and Zagrouba F: Chemical composition and in vitro antimicrobial and antioxidant activities of Citrus aurantium L. flowers essential oil (Neroli oil). Pak J Biol Sci. 15:1034–1040. 2012.PubMed/NCBI View Article : Google Scholar

37 

Ao Y, Satoh K, Shibano K, Kawahito Y and Shioda S: Singlet oxygen scavenging activity and cytotoxicity of essential oils from rutaceae. J Clin Biochem Nutr. 43:6–12. 2008.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

January-2022
Volume 23 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song B, Lee CY, Park J, Kim B, Lee S, Lim S, Kim SW, Choi J, Kang M, Kang JH, Kang JH, et al: Cold‑pressed oil from <em>Citrus aurantifolia</em> inhibits the proliferation of vascular smooth muscle cells via regulation of PI3K/MAPK signaling pathways. Exp Ther Med 23: 21, 2022
APA
Song, B., Lee, C.Y., Park, J., Kim, B., Lee, S., Lim, S. ... Kim, I. (2022). Cold‑pressed oil from <em>Citrus aurantifolia</em> inhibits the proliferation of vascular smooth muscle cells via regulation of PI3K/MAPK signaling pathways. Experimental and Therapeutic Medicine, 23, 21. https://doi.org/10.3892/etm.2021.10943
MLA
Song, B., Lee, C. Y., Park, J., Kim, B., Lee, S., Lim, S., Kim, S. W., Choi, J., Kang, M., Kang, J. H., Lee, S., Park, M., Moon, H., Hwang, K., Kim, I."Cold‑pressed oil from <em>Citrus aurantifolia</em> inhibits the proliferation of vascular smooth muscle cells via regulation of PI3K/MAPK signaling pathways". Experimental and Therapeutic Medicine 23.1 (2022): 21.
Chicago
Song, B., Lee, C. Y., Park, J., Kim, B., Lee, S., Lim, S., Kim, S. W., Choi, J., Kang, M., Kang, J. H., Lee, S., Park, M., Moon, H., Hwang, K., Kim, I."Cold‑pressed oil from <em>Citrus aurantifolia</em> inhibits the proliferation of vascular smooth muscle cells via regulation of PI3K/MAPK signaling pathways". Experimental and Therapeutic Medicine 23, no. 1 (2022): 21. https://doi.org/10.3892/etm.2021.10943