Abstract
Natural products have long been considered as a kind of complementary medicine. In this study, we investigate the apoptotic effect of essential oils of Toona sinensis roots (TSR) on human clear cell renal cell carcinomas (ccRCC). The sesquiterpene content of TSR essential oil was determined via GC/MS analysis. TSR decreased ccRCC cell viabilities, inducing ROS generation and reduction of the mitochondrial membrane potential. Moreover, TSR inhibited Bcl-2 and Hsp90 expression but increased PARP-1 cleavage and cytochrome c release. Akt, mTOR and NF-κB phosphorylation and HIF-α expression were all inhibited, which likely contributed to the anti-proliferative and anti-adhesive effects of TSR.
Keywords: Apoptosis, Essential oil, Renal cell carcinoma, Signalling pathway, Toona sinensis
1. Introduction
Renal cell carcinoma (RCC) occurs in renal tubules and is the most common type of kidney cancer [1], with 76,080 cases and 13,780 deaths predicted in America in 2021 [2]. Since RCC is resistant to traditional chemo- and radiotherapies, the rapid development of targeted drugs is important [1]. Approximately 15% of patients with RCC present with locally advanced or metastatic RCC, for which surgery is noncurative [3]. Clear cell renal cell carcinoma (ccRCC), which accounts for 80% of RCC, is the most common histologic subtype, characterised by mutations or epigenetic silencing of the Von Hippel–Lindau (VHL) gene [3]. This leads to the stabilisation and accumulation of HIF-α, which causes the constitutive activation of several downstream oncogenic pathways, such as phosphatidylinositol 3 kinase (PI3K)/Akt/mTOR, ras/raf/mitogen-activated protein kinase and vascular endothelial growth factor pathways [3, 4].
Toona sinensis (A. Juss) M. Roem is a woody plant belonging to the Meliaceae family and is widely distributed in Asia [5]. It has a distinct flavour, especially the leaves which are very popular in vegetarian cuisine [6, 7]. Moreover, it is used in folk medicine to treat infections, diarrhoea, colds, pain, etc. [8]. Recent pharmacological studies on T. sinensis have demonstrated its anti-microbial, anti-oxidant, anti-inflammatory, anti-diabetic, anti-nociceptive and anti-tumour activities owing to its abundant phytochemical constituents (phenols, flavonoids, limonoids, phytols, etc.)[8–13]. To our knowledge, few reports focus on the effects of TSR extracts on tumour cells. In this study, we investigated whether TSR essential oil induces apoptosis in ccRCC cells by using a primary ccRCC cell line, 786-O and a metastatic line, Caki-1 cell.
2. Methods
2.1. Supercritical fluid extraction of TSR
The roots of T. sinensis were collected in 2016 in Yun-Lin, Taiwan, and the specimens were authenticated by Prof. Hseng-Kuang Hsu (Kaohsiung Medical University). Powdered roots (265 g) were extracted by supercritical carbon dioxide (CO2) fluid after 95% ethanol extraction. Pure CO2 was compressed via a high-pressure diaphragm pump to 299.92 bar at 49°C and allowed to flow through the extraction vessel at a flow rate of 4.2 kg of CO2 per hour. After separation, the supercritical CO2 fluid TSR extract was collected from the valve of the separator, and the regenerated CO2 flowed back into the reservoir. The extraction time was in the range of 60–120 min, and it stopped when the yield reached less than 0.1% of TSR as 40 kg of CO2 passed through the vessel. The recovery rate of supercritical CO2 fluid extract of TSR was 1.88%.
2.2. Gas chromatography and mass spectrometry (GC–MS) analysis
Gas chromatography and mass spectrometry (GC–MS) analysis of TSR essential oil was conducted using a high resolution time-of-flight mass spectrometer (Shimadzu Corp., QP2010, Kyoto, Japan) utilising a DB-5MS UI column (30-m length, 0.25-mm diameter, 0.25-μm film thickness). Helium was used as the carrier gas at a flow rate of 1 mL/min with a pressure of 53.5 kPa. The percentage of each of the constituents was calculated by the integral area under its respectively peak in relation to the total area of all sample constituents. Essential oil (1 μL) was directly injected, and a 1:2 split ratio was applied. The temperature of the oven was set to 50°C for 2 min, followed by 85°C for 4 min and then 200°C for 1 min. Next, the oven was heated to 2.0°C/s until 230°C, where it was kept for 4 min. The injector and transfer line temperatures were both set to 230°C. The mass spectrometer was operated in an electron impact mode with the electron energy set to 1 kV. Mass spectral identification was performed using the Shimadzu FFNSC 1.2 and FFNSC 3 library.
2.3. Cell culture
786-O and Caki-1 human ccRCC cell lines were obtained from the Bioresource Collection and Research Centre (HsinChu, Taiwan) and American Type Culture Collection (Manassas, VA, USA), respectively. The 786-O cells were cultured in RPMI1640 and Caki-1 in McCoy's 5A media supplemented with 10% foetal bovine serum and 100 μg/mL penicillin–streptomycin. The cells were maintained at 37°C in a humidified 5% CO2/95% air incubator. 786-O cell line is defective in VHL expression, as it lacks wild-type pVHL and only expresses HIF-2α [14]. Caki-1 cell line harbors wild-type VHL gene and expresses the detectable level of HIF-1α [15].
2.4. Drug treatment
The cells were treated with serial dilutions of TSR for 24–72 h. TSR solution was prepared 1:1 TSR extract to 99.9% ethanol and then diluted 2,000-fold in a culture medium for treatment. Due to the different cytotoxicities of the cell lines, the total treatment periods for 786-O and Caki-1 cells were 24 and 48 h (except for the MTT assay), respectively. In all experiments, 0.1% ethanol was used as a vehicle control. The cells were seeded and cultured at 80%–90% confluence (96-well plates, 7,500 cells/well; 35 mm dishes, 3 × 105 cells/dish; 60 mm dishes, 4 × 105 cells/dish) overnight.
2.5. Cell morphology and viability assays
After stimulation with TSR at concentrations between 62.5 and 250 ppm for 24–72 h, morphological changes were investigated using an inverted phase-contrast microscope (Leica, USA). Images were also taken using a digital camera (Nikon, Japan). Proliferation of TSR-treated cells was determined with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay (BIO BASIC Inc., Markham, ON, Canada). After plating, 50 μL of MTT dye (0.5 mg/mL) was added for 2.5 h; then, the medium was discarded, and 100 μL of dimethyl sulfoxide (DMSO) was added to extract the dye. Optical density was measured at 570 nm using a microplate reader (Bio Tek Instruments Inc., Winooski, VT, USA). Survival rates (%) were normalised to the vehicle-treated control group.
2.6. Acridine orange (AO) and ethidium bromide (EB) staining
After TSR treatment, cell suspensions were prepared in culture media, 97 μL cell suspension was mixed with 3 μL AO and EB dyes, and then the samples were analysed via fluorescence microscopy (Nikon) using a cooled charge-coupled device (CCD) (NIKON, Japan) at magnifications of 200 × and 400 × . AO freely permeates into the cells and stains the nucleus green, whereas EB stains the nucleus during membrane breakdown in late stage of apoptosis. Cells treated with orange or red nuclei were considered necrotic. Herein, the percentage of apoptotic cell population is the combination of early and late apoptotic cells.
2.7. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) measurement
For fluorescent staining (rhodamine 123, Rh123 for the MMP and H2DCFDA for ROS, the cells were washed with phosphate-buffered saline (PBS) and then stained with Rh123 (10 μg/mL, Molecular probes, Eugene, OR, USA) and H2DCFDA (10 μM, AAT Bioquest Inc., Sunnyvale, CA, USA) for 15–30 min, respectively. The cells were visualised via fluorescence microscopy (Nikon), and images were taken using cooled CCD (Nikon) at magnifications of 200 × and 400 × . The quantification of fluorescent signals (Rh123 and ROS), cells were stained with the Rh123 and ROS, then the cells were analyzed by using flow cytometry (Attune NxT Flow Cytometer; Thermo Fisher Scientific, Waltham, MA, USA).
2.8. Clonogenic assay
The proliferative capacity of cells over a long-term period was measured via clonogenic assays. Dishes (35 mm) were seeded with 8,000 cells, and after 24–48 h of TSR treatment, the media was replaced with plain media and then incubated with 5% CO2 at 37°C for 7 days. At the assay endpoint, cells were stained with 0.5% crystal violet containing 6% glutaraldehyde and photographed using an inverted microscope (Leica, Wetzlar, Germany). A group of ~50 cells was counted as one colony.
2.9. Fibronectin adhesion assay
Prior to adhesion, cells were treated with varying concentrations of TSR essential oil for 24–48 h. Subsequently, adherent cells were trypsinised and allowed to recultivate on fibronectin-coated (20 μg/mL) 24-well plates at a density of 104 cells/mL. After 2 h, non-adherent cells were removed by washing with serum free media twice, and the remaining cells were fixed with 10% formalin and stained with 2.5% Coomassie Brilliant Blue in PBS. After visualisation under an inverted microscope (Leica), the dye was extracted in 100 μL of DMSO. The optic absorbance was read at 595 nm using an automated ELISA reader (BioTek).
2.10. Western blot analysis
RCC cells in 35-mm dishes were incubated with TSR. Cell lysates were collected in lysis buffer containing 0.15% Triton X-100, 2 mM magnesium chloride, 25 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES), 60 mM piperazine-N,N′-bis(2-ethanesulfonic acid) (PIPES), 1 mM ethylenediaminetetraacetic acid (EDTA), 1 mM phenylmethylsulfonyl fluoride (PMSF), 1 mM sodium fluoride, 1 mM sodium orthovanadate, 1 mM β-glycerolphosphate, 2.5 mM sodium pyrophosphate, 1 μg/mL aprotinin, 1 μg/mL pepstatin A and 1 μg/mL leupeptin (pH 6.9) and were sonicated for 20 times with 1-s pulses. Protein concentrations were measured using a Bradford Protein Assay Kit (Bio-Rad Life Sciences, Hercules, CA, USA), and the samples were mixed in reducing sample buffer, boiled and stored at −80°C until further analysis. Protein samples were resolved on 10%–12.5% sodium dodecyl sulphate polyacrylamide gels and transferred to a nitrocellulose membrane, as previously described. The membranes were blocked in 5% non-fat milk or 5% bovine serum albumin in Tris-buffered saline with 0.1% Tween 20 (TBS-T) containing 150 mM NaCl and 50 mM Tris base (pH 8.2) for 60 min at room temperature. Blots were then incubated overnight with blocking solution with the primary antibodies (Table 1) at 4°C. After washing the membranes with TBS-T for several times, blots were incubated with alkaline phosphatase- or horseradish peroxidase-conjugated secondary antibodies (1:2,000), followed by the NBT/BCIP substrate solution (Sigma-Aldrich Inc., St. Louis,MO, USA) and/or enhanced chemiluminescence detection kit (Advansta Inc. San Jose, CA, USA). Immunoreactive bands were quantified via densitometry and compared with the expression of GAPDH or related total protein.
Table 1.
List of antibodies.
Primary antibodies | dilution | Company |
---|---|---|
pAkt1 (Ser473) (ab81283) Akt1 (ab32505), mTOR (ab134903), cleaved PARP-1 (ab32064), NF-κB (ab32536), Hsp90 (ab203126) | 1:1000 | Abcam Plc. (Cambridge, MA, USA) |
pmTOR (ser2448) (sc-293132) | 1:1000 | Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA) |
pNF-κB (ser536) (#3033) | 1:1000 | Cell Signaling Technology Inc. (Danvers, MA, USA) |
HIF-1α (2015-S) | 1:1000 | Epitomics, INC., a brand of Abcam (Burlingame, CA, USA) |
HIF-2α (NB-100-122) | 1:1000 | Novus Biologicals, LLC., a Bio-Techne brand (Centennial, CO, USA) |
Bcl-2 (B3170) | 1:1000 | Sigma |
Cytochrome c (45-6100) | 1:1000 | Thermo Fisher Scientific Inc. (Waltha, MA, USA) |
GAPDH (60004-1-Ig) | 1:2000 | Proteintech Group Inc. (Rosemont, IL, USA) |
2.11. Preparation of mitochondrial and cytosolic fraction
After treatment of TSR, cells were collected and homogenized in buffer containing 0.28 M sucrose, 20 mM 4-(2-hydroxyethyl)-1-piperazineethane sulfonic acid (HEPES), 50 mM sodium chloride, 2 mM ethylenediaminetetraacetic acid (EDTA), 2 mM PMSF, 100 mM sodium fluoride, 2 mM sodium orthovanadate, 20 mM sodium pyrophosphate, 10 μg/mL leupeptin. The pellets were discarded after centrifugation at 500g for 10 min. The supernatants were then collected and centrifuged at 12,000 r.p.m. for 60 min. After centrifugation, the supernatants were cytosolic fraction while the pellets contains mitochondria [16, 17].
2.12. Caspase-3/7 activity assay
786-O and Caki-1 cells were seeded on 35 mm dishes overnight and then treated with various concentration of TSR for 24 and 48 h, respectively. Caspase-3 activity was measured using a caspase-3/7 activity assay kit (#10009135) according to the manufacturer's instructions (Cayman Chemical Company, Ann Arbor, MI, USA). One hundred microliters specific caspase-3/7 substrate N-Ac-DEVD-N′-MC-R110 was added into 90 μL cell based assay buffer and incubated at 37°C for 90 min. Caspase-3/7 activity was analyzed at an absorbance of 485 (excitation) and 535 nm (emission) under Synergy H1 multi-mode microplates reader (BioTek).
2.13. Statistical analyses
All data are expressed as mean ± SD. Differences between the groups were evaluated via one-way analysis of variance, followed by correction for Dunnett's post hoc. The results were considered significant if the P-value was less than 0.05.
3. Results
3.1. Chemical composition of TSR essential oil
As presented in Fig. 1 and Table 2, the GC–MS analysis of TSR led to the identification of five major compounds: spathulenol (area: 37.72%; retention time: 46.606 min), caryophyllene oxide (area: 14.84%; retention time: 46.799 min), pogostol (area: 17.22%; retention time: 47.018 min), viridiflorol (area: 16.39%; retention time: 47.357) and cedr-8 (15)-en-9-ol (area: 13.82% retention time: 47.95 min). In addition, some chemicals were detected using a database containing 39 common essential oil standard references, such as fenchone (0.04681 ppm), camphor (0.19215 ppm), cedrol (1.0714 ppm) and guaiol (0.43624 ppm), detected in the ppm range (Table 3).
Fig. 1.
GC–MS analysis of TSR essential oil. Total ion chromatogram of TSR essential oil.
Table 2.
Identification of chemical composition of essential oil from TSR by GC/MS analysis.
Peak# | R.Time | I.Time | F.Time | Peak Report TIC | ||||||
---|---|---|---|---|---|---|---|---|---|---|
| ||||||||||
Area | Area% | Height | Height% | A/H | Mark | Name | ||||
1 | 46.606 | 46.475 | 46.725 | 467302 | 37.72 | 68626 | 45.70 | 6.81 | V | Spathulenol |
2 | 46.799 | 46.725 | 46.933 | 183788 | 14.84 | 22181 | 14.77 | 8.29 | V | Caryophyllene oxisde |
3 | 47.018 | 46.933 | 47.183 | 213356 | 17.22 | 22199 | 14.78 | 9.61 | V | Pogostol |
4 | 47.357 | 47.183 | 47.458 | 203081 | 16.39 | 23529 | 15.67 | 8.63 | V | Viridiflorol |
5 | 47.950 | 47.717 | 48.125 | 171200 | 13.82 | 13623 | 9.07 | 12.57 | V | 1H-3a, 7-Methanoazulen-5-ol, octahydro-3, 8 |
1238727 | 100.00 | 150158 | 100.00 |
Table 3.
Quantitative results of essential oil by GC/MS using 39 essential oil standard compounds.
Quantitative Result Table | ||||||
---|---|---|---|---|---|---|
| ||||||
ID# | R.Time | m/z | Area | Height | Conc. | Name |
1 | - | 93.00 | — | — | N.D.(Peak) ppm | Pinene <alpha-> |
2 | - | 93.00 | — | — | N.D.(Peak) ppm | Sabinene |
3 | - | 93.00 | — | — | N.D.(Peak) ppm | Pinene <beta-> |
4 | - | 93.00 | — | — | N.D.(Peak) ppm | Phellandrene <alpha-> |
5 | - | 121.00 | — | — | N.D.(Peak) ppm | Terpinene <alpha-> |
6 | - | 119.00 | — | — | N.D.(Peak) ppm | Cymene <para-> |
7 | - | 68.00 | — | — | N.D.(Peak) ppm | Limonene |
8 | - | 93.00 | — | — | N.D.(Peak) ppm | Terpinene <gamma-> |
9 | - | 71.00 | — | — | N.D.(Peak) ppm | Sabinene hydrate <trans-> |
10 | - | 93.00 | — | — | N.D.(Peak) ppm | 2,2-Dimethyl-5-methylene norbornane |
11 | - | 93.00 | — | — | N.D.(Peak) ppm | Myrcene |
12 | - | 93.00 | — | — | N.D.(Peak) ppm | Carene <delta-3-> |
13 | - | 43.00 | — | — | N.D.(Ref) ppm | Cineole <1,4-> |
14 | - | 43.00 | — | — | N.D.(Ref) ppm | Eucalyptol |
15 | - | 93.00 | — | — | N.D.(Peak) ppm | Ocimene <(E)-, beta-> |
16 | - | 93.00 | — | — | N.D.(Peak) ppm | Terpinolene |
17 | 17.37 | 81.00 | 1497 | 260 | 0.04681 ppm | Fenchone |
18 | - | 81.00 | — | — | N.D.(Peak) ppm | Fenchyl alcohol |
19 | - | 71.00 | — | — | N.D.(Peak) ppm | Linalool |
20 | 21.33 | 95.00 | 2597 | 422 | 0.19215 ppm | Camphor |
21 | - | 67.00 | — | — | N.D.(Ref) ppm | Isoisopulegol |
22 | - | 95.00 | — | — | N.D.(Peak) ppm | Isoborneol |
23 | - | 71.00 | — | — | N.D.(Peak) ppm | Menthol |
24 | - | 93.00 | — | — | N.D.(Peak) ppm | Caryophyllene <(E)-> |
25 | - | 161.00 | — | — | N.D.(Peak) ppm | Valencene |
26 | - | 93.00 | — | — | N.D.(Peak) ppm | Selinene <alpha-> |
27 | - | 69.00 | — | — | N.D.(Peak) ppm | Nerolidol <(Z)-> |
28 | - | 93.00 | — | — | N.D.(Peak) ppm | Bisabolene <(Z)-, alpha-> |
29 | - | 112.00 | — | — | N.D.(Peak) ppm | Isomenthone |
30 | - | 95.00 | — | — | N.D.(Peak) ppm | Borneol |
31 | - | 59.00 | — | — | N.D.(Peak) ppm | Terpineol <alpha-> |
32 | - | 121.00 | — | — | N.D.(Peak) ppm | Terpineol <gamma-> |
33 | - | 81.00 | — | — | N.D.(Peak) ppm | Pulegone |
34 | - | 119.00 | — | — | N.D.(Ref) ppm | Cedrene <alpha-> |
35 | - | 93.00 | — | — | N.D.(Ref) ppm | Humulene <alpha-> |
36 | - | 69.00 | — | — | N.D.(Ref) ppm | Nerolidol |
37 | 47.35 | 161.00 | 4598 | 552 | 0.43624 ppm | Guaiol |
38 | 47.91 | 95.00 | 7530 | 781 | 1.07140 ppm | Cedrol |
39 | - | 109.00 | — | — | N.D.(Peak) ppm | Bisabolol <alpha-> |
R. time: retention time. N.D.: non determined.
3.2. Cytotoxic effects of TSR essential oil on ccRCC cells
To test the potency of TSR on the cell viability of ccRCC cells, 786-O and Caki-1 cells were incubated with different TSR concentrations (62.5, 93.75, 125 and 250 ppm) for 24, 48 and 72 h to record the morphological changes and measure the survival rates via MTT assays. At 250 ppm, 786-O cells were retracted after 24 h of treatment (Fig. 2A), whereas Caki-1 cell retraction was observed after 48 h of treatment (Fig. 2B). At 250 ppm of TSR, the cell viability of 786-O was 41.86% of the vehicle control (0 ppm, p < 0.001) after 24 h of treatment (Fig. 2C). In Caki-1 cells, TSR demonstrated cytotoxicity at 93.75, 125 and 250 ppm. The viabilities of TSR-treated Caki-1 cells were 89.04%, 80.52% and 44.73% at 24 h (Fig. 2D). Overall, the cytotoxicity of TSR was more prominent after 48 and 72 h at 93.75–250 ppm of TSR in both cell lines.
Fig. 2.
Effects of TSR essential oil on cell morphology and viabilities. (A and B) Cell morphology and (C and D) cell viability assays. Scale bar = 100 μm. Results are expressed as mean ± SD of five independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 compared with the control group.
3.3. Proapoptotic effects of TSR essential oil on 786-O and Caki-1 cells
To further elucidate the effects of TSR on ccRCC cells, we performed dual AO–EB staining to evaluate apoptosis in TSR-treated cells. ccRCC cell lines were treated with TSR for 24 h (786-O) or 48 h (Caki-1), and at concentrations of 62.5 ppm, there were obvious green dots in both TSR-treated cell lines (Fig. 3). Many yellow to orange dots in TSR-treated cells appeared due to the concentrations of over 93.75 ppm (Fig. 3). Compared with control cells, TSR-treated 786-O and Caki-1 cells exhibited high proportions of late apoptotic (19%–33% at 62.5 ppm, 33%–37% at 93.75 ppm, 33.9%–47% at 125 ppm and 54%–59% at 250 ppm; Fig. 3B and C) but low proportions of necrotic cells (<5%, data not shown) after 24 h of treatment.
Fig. 3.
Effects of TSR essential oil on apoptosis. (A) Dual AO–EB staining on TSR-treated 786-O (24 h) and Caki-1 cells (48 h). Scale bar = 50 μm. Arrow head: early apoptotic cells; arrows: late apoptotic cells. Representative images are from one of three independent experiments. (B and C) Late apoptotic populations are expressed as mean ± SD of three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 compared with the control group.
3.4. TSR essential oil inhibits colony formation and adhesion
Although TSR essential oil demonstrated an effect on apoptosis, we wanted to further explore if TSR-treated cells could still proliferate and adhere to an extracellular matrix. The colony-forming ability of both 786-O and Caki-1 cell lines was eliminated by the increasing concentrations of TSR (Fig. 4A). At concentrations of 62.5 and 93.75 ppm, the inhibitory effect of TSR was more prominent in 786-O cells than in Caki-1 cells (Fig. 4B and C). Compared with controls, TSR-treated 786-O and Caki-1 cells formed fewer colonies (the inhibition rates were 20.5%–44.4% at 62.5 ppm, 63.8%–82.4% at 93.75 ppm, 86.6%–97.4% at 125 ppm and 100% at 250 ppm; Fig. 4B and C). Conversely, both 786-O and Caki-1 cells adhered less to fibronectin after TSR treatment (Fig. 4D). At concentrations of 62.5 and 93.75 ppm, Caki-1 cell adhesion was significantly blocked by TSR (Fig. 4E and F). At higher TSR concentrations (125 and 250 ppm), both ccRCC cells adhered less (Fig. 4E and F).
Fig. 4.
Effects of TSR essential oil on colony formation and cell adhesion. Cells were treated with TSR at concentrations of 0, 62.5, 93.75, 125 and 250 ppm for 24 h (786-O) and 48 h (Caki-1), respectively. (A) Colony-forming assay, (B) quantification of colonies and (C) fibronectin adhesion assay. Scale bar = 50 μm; magnification: 100 ×. Representative images are from three independent experiments. Data were quantified for three independent experiments and expressed as mean ± SD. *p < 0.05 compared with the control group. Scale bar = 100 μm.
3.5. TSR essential oil diminishes MMP and induces ROS generation
Disruption of the mitochondrial membrane is an important step in the mitochondria-dependent apoptotic pathway, which leads to the loss of the mitochondrial transmembrane potential [18]. MMP maintenance in both cell lines was examined via Rh123 staining and flow cytometry analysis. In the vehicle-treated control group, long, tubular-shaped mitochondria can be observed in both cell lines (Fig. 5A). After treatment with TSR, the numbers of mitochondria were diminished and swollen in both cell lines (Fig. 5A). 786-O cells treated with the mitochondrial oxidative phosphorylation uncoupler CCCP were used as the positive control. Rh123 quantification via flow cytometry demonstrated that TSR inhibited Rh123 signal intensities in both 786-O and Caki-1 cells (Fig. 5B and C).
Fig. 5.
Effects of TSR essential oil on MMP. Cells were treated with TSR at concentrations of 0, 62.5, 93.75, 125 and 250 ppm for 24 h (786-O) and 48 h (Caki-1), respectively. CCCP (50 μM) was used as a positive control. (A and B) Rhodamine 123 staining of 786-O and Caki-1 cells. (C and D) Flow cytometry histogram of Rh123 staining. (E) The bar graph shows Rh123 intensities of 786-O and Cak-1 cells examined via flow cytometry. *p < 0.05, **p < 0.01, ***p < 0.001 compared with the control group.
Mitochondria-dependent apoptosis was preceded by ROS overproduction [18]. TSR essential oil significantly induced ROS production in a concentration-dependent manner in both cell lines (Fig. 6A–C). Quantification of ROS fluorescence intensity via flow cytometry demonstrated that TSR essential oil (62.5–250 ppm) significantly triggered Caki-1 ROS overproduction, whereas 786-O required a higher concentration (250 ppm) to induce similar effects (Fig. 6C).
Fig. 6.
Effects of TSR essential oil on ROS production. Cells were treated with TSR at concentrations of 0, 62.5, 93.75, 125 and 250 ppm for 24 h (786-O) and 48 h (Caki-1), respectively. (A and B) Flow cytometry histogram of ROS staining. (C) Bar graph showing the quantification of ROS intensities in 786-O and Cak-1 cells. *p < 0.05, **p < 0.01, ***p < 0.001 compared with the control group.
3.6. TSR essential oil promotes the expression of pro-and anti-apoptotic proteins and caspase-3/7 activities
To further evaluate the mechanism of TSR-induced apoptosis, the levels of intrinsic apoptotic proteins were determined via Western blot analysis and caspase-3/7 activity assays. Accordingly, TSR essential oil increased cytosolic but blocked membranous cytochrome c expression (Fig. 7A–E). Anti-apoptotic proteins, such as Bcl-2 and heat shock protein 90, were decreased with TSR treatment (Fig. 7A–E). In addition, caspase activity assays further supported the apoptotic effects of TSR on ccRCC cells. As expected, TSR increased caspase-3/7 activities > twofold in a concentration-dependent manner in both cell lines (Fig. 7F).
Fig. 7.
Effects of TSR essential oil on cytochrome c, Bcl-2, Hsp90, cleaved PARP-1 and caspase activities. Cells were treated with TSR at concentrations of 0, 62.5, 93.75, 125 and 250 ppm for 24 h (786-O) and 48 h (Caki-1), respectively. (A and B) Protein expression of Bcl-2, hsp90, cleaved PARP-1 and cytochrome c (cytosolic and membranous fractions). GAPDH was used as the internal control. (C–G) Bar graphs showing the densitometric analysis (mean ± SD) for three to five independent experiments. (H) Caspase activity assay. Results (mean ± SD) were from three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 compared with the control group.
3.7. Inhibitory effects of TSR essential oil on Akt, mTOR and NF-κB phosphorylation and HIF-1/2α protein expression
Under normoxic conditions, HIF-α expression can increase with growth factor signalling [19]. To determine whether TSR blocks intracellular signalling in 786-O and Caki-1 cells, Western blot analyses were employed to examine the HIF-α, pNF-κB, pAkt and pmTOR expressions. TSR inhibited NF-κB, Akt and mTOR phosphorylation in both cell lines (Fig. 8A–H). Furthermore, TSR reduced HIF-1α and HIF-2α expressions in 786-O and Caki-1 cells at 24 and 48 h, respectively (Fig. 8A, B, and I).
Fig. 8.
The effects of TSR on oncogenic signalling proteins (pAkt, Akt, pmTOR, mTOR and HIF-1α/2α). Cells were treated with TSR at concentrations of 0, 62.5, 93.75, 125 and 250 ppm for 24 h (786-O) and 48 h (Caki-1), respectively. (A and B) Phosphorylation of Akt, mTOR and NF-κB as well as protein expression of total Akt, total mTOR, total NF-κB and HIF-1/2α. GAPDH was used as the internal control. The figures shown are representative of one experiment. (C–I) Bar graphs showing the densitometric analysis for three to five independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001 compared with the control group.
4. Discussion
T. sinensis has long been considered as a multi-functional plant owing to its nutraceutical and medicinal uses [8]. There are only three scientific reports focusing on the effects of T. sinensis roots. Methyl gallate extracted from the methanol extracts of T. sinensis roots exhibited an anti-oxidant activity against hydrogen peroxide-induced DNA damage in MDCK cells [20]. Water extracts of TSR could help improve the anti-oxidant system in senescence-accelerated mouse prone 8 for clearing β-amyloid plaques [21]. Besides, Yang et al. (2013) demonstrated that ethyl acetate extracts of TSR inhibits the proliferation of MGC-803 and PC3 tumour cells [22]. However, the essential oil of TSR and its anti-tumour effects have never been reported. Our study examined the composition of TSR essential oil and identified five major compounds and some rare ingredients from essential oil standard references. Fifty six terpenoids have been isolated from this plant [8]. Two triterpenoids, betulonic acid and 3-oxours-12-en-28-oic acid are from TSR, exhibiting antitumour activity [22]. Compare to our results, specific proportions and concentrations of sesquiterpenoids (pogostol, viridiflorol, and guaiol), sesquiterpenes (spathulenol, cedr-8 (15)-en-9-ol, cedrol, and caryophyllene oxide) as well as monoterpenoids (fenchone and champhor) have been identified. Among them, spathulenol was the predominant compound (37.2%). Sesquiterpenes and their metabolites exhibit numerous pharmacological activities, including anti-tumour, anti-inflammatory, anti-microbial, cytotoxic, immunosuppressive and other effects [23]. Wang et al. (2014) reported that trace amounts (<0.1%) of spathulenol can be found in the essential oil of T. sinensis leaves, whereas caryophyllene oxide accounts for 0.82% [24]. Interestingly, these two compounds are the major ingredients in the essential oil of TSR. Sesquiterpene oxides, such as caryophyllene oxide, are toxic to ants and inhibits the growth of ant-associated fungi [25]. According to the RIFM fragrance ingredient safety assessment, caryophyllene oxide and cedr-8(15)-en-9-ol demonstrate no observed genotoxicity, phototoxicity or photoallergic effects [26, 27]. The LD50 of caryophyllene oxide and cedr-8(15)-en-9-ol for acute oral toxicity are more than 5 gram/kg in rats [26, 27]. The NOAEL of caryophyllene oxide for fertility was considered to be 21000 ppm (equivalent to 1398 mg/kg/day for males and 1660 mg/kg/day for females) [26]. Briefly, the five major compounds identified in TSR essential oil, only spathulenol had been reported to exhibit cytotoxicity on human cancer cell lines [28, 29]. However, the influence of other trace constituents and their synergetic effects should be considered when used in vivo.
Akt, mTOR and NF-κB phosphorylation has been demonstrated in numerous tumours, indicating poor prognoses for patients [30]. Natural products or compounds modulating multiple signalling pathways to suppress tumour progression is current trends for developing anti-cancer drugs. Akt/mTOR and Akt/mTOR/NF-κB are two main mutated pathways costitutively activated in human cancers [30, 31]. Studies have been shown that Akt/mTOR blocks apoptosis by inhibition of the tumour suppressor protein phosphatase and tensin homolog and/or transactivation of the transcription factor NF-κB [31]. In our data, inhibition of pAkt, pmTOR, and pNF-κB were concurrent with induction of apoptosis. Whether TSR essential oil inhibits Akt, mTOR, and NF-κB concomitantly or blocks them in a series signalling cascade, needs more experiments to clarify.
The Akt/mTOR/NF-κB signalling network regulates cell growth and modulates HIF-α activity, translation, expression and stability in RCC, and the accumulation of HIF-α protein is a key feature of ccRCC due to the loss of the functional VHL protein [32–34]. Deletion of either HIF-1α or HIF-2α is sufficient to restrict the formation of tumour, suggesting the pro-tumourigenic activities of HIF-1α/2α in ccRCC [19]. Due to the lack of HIF-1α in 786-O cells and an undetectable level of HIF-2α in Caki-1 cells, we examined the expressions of HIF-2α and HIF-1α in 786-O and Caki-1 cells, respectively, following TSR treatment. We found that TSR inhibited the phosphorylation of Akt, mTOR and NF-κB and simultaneously decreased the downstream expression of HIF-α. Although the exact mechanism of how TSR inhibits the expression of HIF-α in these cells remains unclear, one possibility may be through the inactivation of Akt, mTOR or NF-κB pathways. Furthermore, whether TSR induces HIFα degradation needs further clarification.
Hsps can inhibit or aid the apoptotic machinery through their chaperone functions by modulating protein assembly and folding, ubiquitin-dependent degradation and protein translocation [35]. The inhibition of hsp90-Akt binding increases the sensitivity of cells to apoptotic signals, indicating a direct interaction between hsp90 and Akt [36]. Hsp90 expression and Akt phosphorylation were abrogated by TSR essential oil in both 786-O and Caki-1 cell lines. Several lines of evidence demonstrated that blocking Hsp90 not only reduces TNFα-triggered NF-κB activity and increases cancerous cell death [37] but also downregulates acidosis-induced HIF-1α/2α expression in glioma cells [38]. As a multi-functional protein, Hsp90 might be another key regulator in TSR-induced effects. Together, these findings indicate that TSR essential oil-inhibited HIF-1α/2α expression might occur with the assistance of Hsp90.
In conclusion, TSR essential oil exhibited various anti-proliferative and cytotoxic effects, including the induction of mitochondrial-dependent apoptosis associated with the downregulation of several oncogenic signalling pathways in ccRCC. Altogether, this suggests that TSR essential oil may be a potential cancer treatment.
Acknowledgements
The authors gratefully thank Dr. Ho-Chian Sung to help extract the essential oil of root of T. sinensis and thank Mr. Hsin-Che Yu for assisting data collection. Authors also acknowledge the use of high resolution TOF-Mass spectrometer belonging to the Instrument Centre of National Cheng Kung University.
Funding Statement
This research was funded by Ministry of Science and Technology (109-2635-B-037-001) and E-Da Cancer Hospital (grant number: EDCHP109008).
Footnotes
Conflict of interest
The authors declare no conflict of interest.
Funding Statement
This research was funded by Ministry of Science and Technology (109-2635-B-037-001) and E-Da Cancer Hospital (grant number: EDCHP109008).
References
- 1. Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, et al. Renal cell carcinoma. Nat Rev Dis Primers. 2017;3:17009. doi: 10.1038/nrdp.2017.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. CA Cancer J Clin. 2021;71:7–33. doi: 10.3322/caac.21654. [DOI] [PubMed] [Google Scholar]
- 3. Nabi S, Kessler ER, Bernard B, Flaig TW, Lam ET. Renal cell carcinoma: a review of biology and pathophysiology. F1000Res. 2018;7:307. doi: 10.12688/f1000research.13179.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Braga EA, Fridman MV, Loginov VI, Dmitriev AA, Morozov SG. Molecular mechanisms in clear cell renal cell carcinoma: Role of miRNAs and hypermethylated miRNA genes in crucial oncogenic pathways and processes. Front Genet. 2019;10:320. doi: 10.3389/fgene.2019.00320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Edmonds JM, Staniforth M. Toona sinensis: Meliaceae. Curtis's Botanical magazine. 1998;15:186–93. [Google Scholar]
- 6. Zhai X, Granvogl M. Key odor-active compounds in raw green and red Toona sinensis (A. Juss.) Roem. and their changes during blanching. J Agric Food Chem. 2020;68:7169–83. doi: 10.1021/acs.jafc.0c02012. [DOI] [PubMed] [Google Scholar]
- 7. Zhai X, Granvogl M. Elucidation of the impact of different drying methods on the key odorants of Toona sinensis (A. Juss.) Roem. using the sensomics approach. J Agric Food Chem. 2020;68:7697–709. doi: 10.1021/acs.jafc.0c02144. [DOI] [PubMed] [Google Scholar]
- 8. Peng W, Liu Y, Hu M, Zhang M, Yang J, Liang F, et al. Toona sinensis: a comprehensive review on its traditional usages, phytochemisty, pharmacology and toxicology. Rev Bras Farmacogn. 2019;29:111–24. doi: 10.1016/j.bjp.2018.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Wu JG, Peng W, Yi J, Wu YB, Chen TQ, Wong KH, et al. Chemical composition, antimicrobial activity against Staphylococcus aureus and a pro-apoptotic effect in SGC-7901 of the essential oil from Toona sinensis (A. Juss.) Roem. leaves. J Ethnopharmacol. 2014;154:198–205. doi: 10.1016/j.jep.2014.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Lim HJ, Park IS, Jie EY, Ahn WS, Kim SJ, Jeong SI, et al. Anti-Inflammatory activities of an extract of in vitro grown adventitious shoots of Toona sinensis in LPS-treated RAW2647 and propionibacterium acnes-treated HaCaT cells. Plants (Basel) 2020:9. doi: 10.3390/plants9121701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Chen YC, Chien LH, Huang BM, Chia YC, Chiu HF. Aqueous extracts of Toona sinensis leaves inhibit renal carcinoma cell growth and migration through JAK2/stat3, Akt, MEK/ERK, and mTOR/HIF-2alpha pathways. Nutr Cancer. 2016;68:654–66. doi: 10.1080/01635581.2016.1158292. [DOI] [PubMed] [Google Scholar]
- 12. Su YF, Yang YC, Hsu HK, Hwang SL, Lee KS, Lieu AS, et al. Toona sinensis leaf extract has antinociceptive effect comparable with non-steroidal anti-inflammatory agents in mouse writhing test. BMC Complement Altern Med. 2015;15:70. doi: 10.1186/s12906-015-0599-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Zhang Y, Dong H, Wang M, Zhang J. Quercetin isolated from Toona sinensis leaves attenuates hyperglycemia and protects hepatocytes in high-carbohydrate/high-fat diet and alloxan induced experimental diabetic mice. J Diabetes Res. 2016;2016:8492780. doi: 10.1155/2016/8492780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Gordan JD, Lal P, Dondeti VR, Letrero R, Parekh KN, Oquendo CE, et al. HIF-alpha effects on c-Myc distinguish two subtypes of sporadic VHL-deficient clear cell renal carcinoma. Cancer Cell. 2008;14:435–46. doi: 10.1016/j.ccr.2008.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Raval RR, Lau KW, Tran MG, Sowter HM, Mandriota SJ, Li JL, et al. Contrasting properties of hypoxia-inducible factor 1 (HIF-1) and HIF-2 in von Hippel-Lindau-associated renal cell carcinoma. Mol Cell Biol. 2005;25:5675–86. doi: 10.1128/MCB.25.13.5675-5686.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Clayton DA, Shadel GS. Isolation of mitochondria from cells and tissues. Cold Spring Harb Protoc. 2014;2014 doi: 10.1101/pdb.top074542. pdb top074542. [DOI] [PubMed] [Google Scholar]
- 17. Frezza C, Cipolat S, Scorrano L. Organelle isolation: functional mitochondria from mouse liver, muscle and cultured fibroblasts. Nat Protoc. 2007;2:287–95. doi: 10.1038/nprot.2006.478. [DOI] [PubMed] [Google Scholar]
- 18. Bock FJ, Tait SWG. Mitochondria as multifaceted regulators of cell death. Nat Rev Mol Cell Biol. 2020;21:85–100. doi: 10.1038/s41580-019-0173-8. [DOI] [PubMed] [Google Scholar]
- 19. Hoefflin R, Harlander S, Schafer S, Metzger P, Kuo F, Schonenberger D, et al. HIF-1alpha and HIF-2alpha differently regulate tumour development and inflammation of clear cell renal cell carcinoma in mice. Nat Commun. 2020;11:4111. doi: 10.1038/s41467-020-17873-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Hsieh TJ, Liu TZ, Chia YC, Chern CL, Lu FJ, Chuang MC, et al. Protective effect of methyl gallate from Toona sinensis (Meliaceae) against hydrogen peroxide-induced oxidative stress and DNA damage in MDCK cells. Food Chem Toxicol. 2004;42:843–50. doi: 10.1016/j.fct.2004.01.008. [DOI] [PubMed] [Google Scholar]
- 21. Liao JW, Hsu CK, Wang MF, Hsu WM, Chan YC. Beneficial effect of Toona sinensis Roemor on improving cognitive performance and brain degeneration in senescence-accelerated mice. Br J Nutr. 2006;96:400–7. doi: 10.1079/bjn20061823. [DOI] [PubMed] [Google Scholar]
- 22. Yang S, Zhao Q, Xiang H, Liu M, Zhang Q, Xue W, et al. Antiproliferative activity and apoptosis-inducing mechanism of constituents from Toona sinensis on human cancer cells. Cancer Cell Int. 2013;13:12. doi: 10.1186/1475-2867-13-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sharma A, Bajpai VK, Shukla S. Sesquiterpenes and Cytotoxicity. In: Ramawat K, Mérillon JM, editors. Natural Products. Berlin: Springer; 2013. pp. 3515–50. [Google Scholar]
- 24. Wang CL, Shi JX, Wu Y. Chemical and Antimicrobial Analyses of Essential Oil of Toona sinensis from China. Asian Journal of Chemistry. 2014;26:2557–60. [Google Scholar]
- 25. Yang D, Michel L, Chaumont JP, Millet-Clerc J. Use of caryophyllene oxide as an antifungal agent in an in vitro experimental model of onychomycosis. Mycopathologia. 1999;148:79–82. doi: 10.1023/a:1007178924408. [DOI] [PubMed] [Google Scholar]
- 26. Api AM, Belmonte F, Belsito D, Biserta S, Botelho D, Bruze M, et al. RIFM fragrance ingredient safety assessment, caryophyllene oxide, CAS Registry Number 1139-30-6. Food Chem Toxicol. 2020;138(Suppl 1):111102. doi: 10.1016/j.fct.2019.111102. [DOI] [PubMed] [Google Scholar]
- 27. Panel RE, Belsito D, Bickers D, Bruze M, Calow P, Greim H, et al. A toxicologic and dermatologic assessment of cyclic and non-cyclic terpene alcohols when used as fragrance ingredients. Food Chem Toxicol. 2008;46(Suppl 11):S1–71. doi: 10.1016/j.fct.2008.06.085. [DOI] [PubMed] [Google Scholar]
- 28. do Nascimento KF, Moreira FMF, Alencar Santos J, Kassuya CAL, Croda JHR, Cardoso CAL, et al. Anti-oxidant, anti-inflammatory, antiproliferative and antimycobacterial activities of the essential oil of Psidium guineense Sw. and spathulenol. J Ethnopharmacol. 2018;210:351–8. doi: 10.1016/j.jep.2017.08.030. [DOI] [PubMed] [Google Scholar]
- 29. Martins A, Hajdu Z, Vasas A, Csupor-Loffler B, Molnar J, Hohmann J. Spathulenol inhibit the human ABCB1 efflux pump. Planta Med. 2010;76:1349. [Google Scholar]
- 30. Ghoneum A, Said N. PI3K-AKT-mTOR and NFkappaB pathways in ovarian cancer: Implications for targeted therapeutics. Cancers (Basel) 2019:11. doi: 10.3390/cancers11070949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Dan HC, Cooper MJ, Cogswell PC, Duncan JA, Ting JP, Baldwin AS. Akt-dependent regulation of NF-{kappa}B is controlled by mTOR and Raptor in association with IKK. Genes Dev. 2008;22:1490–500. doi: 10.1101/gad.1662308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Zhou J, Schmid T, Frank R, Brune B. PI3K/Akt is required for heat shock proteins to protect hypoxia-inducible factor 1alpha from pVHL-independent degradation. J Biol Chem. 2004;279:13506–13. doi: 10.1074/jbc.M310164200. [DOI] [PubMed] [Google Scholar]
- 33. Wilczynski J, Duechler M, Czyz M. Targeting NF-kappaB and HIF-1 pathways for the treatment of cancer: part II. Arch Immunol Ther Exp (Warsz) 2011;59:301–7. doi: 10.1007/s00005-011-0132-3. [DOI] [PubMed] [Google Scholar]
- 34. van Uden P, Kenneth NS, Rocha S. Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem J. 2008;412:477–84. doi: 10.1042/BJ20080476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Calderwood SK, Khaleque MA, Sawyer DB, Ciocca DR. Heat shock proteins in cancer: chaperones of tumorigenesis. Trends Biochem Sci. 2006;31:164–72. doi: 10.1016/j.tibs.2006.01.006. [DOI] [PubMed] [Google Scholar]
- 36. Sato S, Fujita N, Tsuruo T. Modulation of Akt kinase activity by binding to Hsp90. Proc Natl Acad Sci U S A. 2000;97:10832–7. doi: 10.1073/pnas.170276797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Lewis J, Devin A, Miller A, Lin Y, Rodriguez Y, Neckers L, et al. Disruption of hsp90 function results in degradation of the death domain kinase, receptor-interacting protein (RIP), and blockage of tumor necrosis factor-induced nuclear factor-kappaB activation. J Biol Chem. 2000;275:10519–26. doi: 10.1074/jbc.275.14.10519. [DOI] [PubMed] [Google Scholar]
- 38. Filatova A, Seidel S, Bogurcu N, Graf S, Garvalov BK, Acker T. Acidosis acts through HSP90 in a PHD/VHL-independent manner to promote HIF function and stem cell maintenance in glioma. Cancer Res. 2016;76:5845–56. doi: 10.1158/0008-5472.CAN-15-2630. [DOI] [PubMed] [Google Scholar]