Skip to main content
Journal of Cellular and Molecular Medicine logoLink to Journal of Cellular and Molecular Medicine
. 2024 Feb 23;28(6):e18050. doi: 10.1111/jcmm.18050

Calotropis procera extract inhibits prostate cancer through regulation of autophagy

Palak Singh 1, Bodhana Dhole 1, Jaganmoy Choudhury 1, Anannya Tuli 2, Deepak Pandey 1, Thirumurthy Velpandian 2, Surabhi Gupta 1, Pradeep Kumar Chaturvedi 1,
PMCID: PMC10941509  PMID: 38400579

Abstract

Current treatment options available for prostate cancer (PCa) patients have many adverse side effects and hence, new alternative therapies need to be explored. Anticancer potential of various phytochemicals derived from Calotropis procera has been studied in many cancers but no study has investigated the effect of leaf extract of C. procera on PCa cells. Hence, we investigated the effect of C. procera leaf extract (CPE) on cellular properties of androgen‐independent PC‐3 and androgen‐sensitive 22Rv1 cells. A hydroalcoholic extract of C. procera was prepared and MTT assay was performed to study the effect of CPE on viability of PCa cells. The effect of CPE on cell division ability, migration capability and reactive oxygen species (ROS) production was studied using colony formation assay, wound‐healing assay and 2′,7′‐dichlorodihydrofluorescein diacetate assay, respectively. Caspase activity assay and LDH assay were performed to study the involvement of apoptosis and necrosis in CPE‐mediated cell death. Protein levels of cell cycle, antioxidant, autophagy and apoptosis markers were measured by western blot. The composition of CPE was identified using untargeted LC–MS analysis. Results showed that CPE decreased the viability of both the PCa cells, PC‐3 and 22Rv1, in a dose‐ and time‐dependent manner. Also, CPE significantly inhibited the colony‐forming ability, migration and endogenous ROS production in both the cell lines. Furthermore, CPE significantly decreased NF‐κB protein levels and increased the protein levels of the cell cycle inhibitor p27. A significant increase in expression of autophagy markers was observed in CPE‐treated PC‐3 cells while autophagy markers were downregulated in 22Rv1 cells after CPE exposure. Hence, it can be concluded that CPE inhibits PCa cell viability possibly by regulating the autophagy pathway and/or altering the ROS levels. Thus, CPE can be explored as a possible alternative therapeutic agent for PCa.

Keywords: apoptosis, autophagy, Calotropis procera, cell migration, cell proliferation, prostate cancer, reactive oxygen species

1. INTRODUCTION

Prostate cancer (PCa) is in fourth position in terms of new incidences and eighth in terms of cancer‐related deaths in both sexes with a death rate of 3.8% worldwide. Amongst men, it is the second most common cancer with a global death rate of 6.8% as of 2020. 1 Androgen deprivation therapy (ADT), surgery and radiation therapy are the commonly available primary treatments for PCa patients. 2 However, conventional treatments have several adverse side effects including cardiotoxicity, musculoskeletal alterations and neurotoxicity. 3 , 4 To reduce the side effects, several new therapies such as immunotherapy, complementary and alternative medicines (CAM), vaccines and their combinations are being tested for their efficacy. 5

Complementary medicines often include crude plant parts in the form of extracts, powder and tablets. 6 These drugs are frequently being used along with surgery, radiotherapy or chemotherapy, either as a replacement of chemical drugs or act in a synergistic manner with other chemical drugs in order to increase the efficacy of the treatment with minimal side effects. 7 , 8 The bioactive compounds naturally found in these plants act against cancer cells by targeting various pro‐ and anti‐oncogenic molecules and signalling pathways. 9 Therefore, the plant derivatives and their constituent bioactive compounds are being actively tested for their therapeutic potential. 8 , 10

The genus Calotropis belongs to Asclepiadaceae family and consists of only six species. 11 Calotropis procera is also known as milkweed, swallowwort, 11 madar and sodom apple. 12 It is a tropical shrub found in parts of Africa, Asia and other tropics. 13 In India, it is majorly found in wastelands of Assam, West Bengal, Rajasthan and Punjab. 12 Different parts of this plant such as seeds, flower, roots, leaves and milky latex are reported to possess insecticidal, analgesic, anti‐ulcers, cytotoxic, anti‐bacterial, hepatoprotective and cardioprotective properties. 13 Different phytochemicals in C. procera latex have shown anticancer potential in both human and animal models, specifically against breast, liver, glioblastoma, colon and PCa. 14 , 15 Recently the leaf extract of C. procera has been shown to exhibit anticancer potential against lung 16 and breast cancer 17 , 18 but its role in PCa has not been explored till date. Thus, the current study investigated the anticancer activity of the leaf extract of C. procera on PCa cells in vitro and its underlying mechanism.

2. MATERIALS AND METHODS

2.1. Reagents

F‐12 K, RPMI‐1640 and KSFM media, fetal bovine serum (FBS), cell culture plates, MPER reagent, CyQUANT LDH Cytotoxicity Assay kit, BCA (Bicinchoninic acid) kit and EnzChek Caspase‐3 Activity Assay Kit were purchased from Thermo Fisher Scientific (Waltham, MA, USA). 3‐(4, 5‐Dimethylthiazol‐2‐yl)‐2, 5‐diphenyl tetrazolium bromide (MTT), doxorubicin hydrochloride, dimethyl sulfoxide (DMSO) and 2′,7′‐dichlorodihydrofluorescein diacetate (DCFDA) were purchased from Sigma Aldrich (St. Louis, MO, USA). Antibodies were purchased from Abcam (Cambridge, UK), Cell Signaling Technology (Danvers, Massachusetts, USA) and Santa Cruz Biotechnology (Dallas, Texas, USA).

2.2. Preparation of C. procera leaf extract

Plant of C. procera was identified by its flowers, and the leaves were washed and air dried for several days to remove moisture. The leaves were then ground to fine powder in liquid nitrogen. The powdered sample was weighed and methanol (Qualigens, India) was added to it in the ratio of 1:10 weight/volume. The methanolic mixture was shaken for 24 h followed by drying in rotary vacuum evaporator at 50°C. The pellet obtained was dissolved in 80% ethanol at a concentration of 80 mg/mL and stored at 4°C till further use.

2.3. Cell culture

All the cell lines, PC‐3, 22Rv1 and RWPE‐1, were purchased from the American Type Culture Collection (ATCC) and grown in a 37°C incubator with 5% CO2. Androgen‐independent PC‐3 cells were cultured in F‐12K medium while androgen‐sensitive 22Rv1 cell line was cultured in RPMI‐1640 medium, along with 10% FBS for both cell lines. The normal prostate epithelial cell line, RWPE‐1, was cultured in KSFM medium containing 5 ng/mL of recombinant human epidermal growth factor and 0.05 mg/mL of bovine pituitary extract.

2.4. Cell viability assay

MTT assay was used to determine the effect of C. procera leaf extract (CPE) on viability of PC‐3, 22Rv1 and RWPE‐1 cells. 19 5000 cells/0.2 mL/well were seeded in 96‐well culture plates. After 24 h of plating, they were treated with varying doses of CPE (25, 50, 100, 200 and 400 μg/mL) for different time points (24, 48 and 72 h). 0.5 mg/mL MTT solution was added to the wells after removing the culture medium, and cells were incubated for 4 h at 37°C, 5% CO2. The MTT solution was then discarded and formazan crystals formed were solubilized in DMSO for 2 h. Absorbance at 570 nm was measured using TECAN plate reader (Mannedorf, Switzerland). The absorbance at 570 nm is directly proportional to the number of viable cells. The percentage cell viability was then calculated as:

Percentage cell viabilityCV%=Absorbance of treated cells/Absorbance of control×100.

2.5. Colony formation assay

To determine the colony formation ability of PCa cells on CPE treatment, colony formation assay was performed as previously described. 20 Briefly, the cells were seeded in 6‐well plates at different cell densities for vehicle control (100, 250, 500 and 1000 cells/well) and treatment group (1000, 2000, 3000, 4000 and 5000 cells/well). After the cells adhered, they were treated with IC₅₀ concentration of CPE (26.5 μg/mL for PC‐3 and 20 μg/mL for 22Rv1) or vehicle control (complete media with ethanol) for 48 h. After 48 h of treatment, the wells were washed and complete fresh media was added. The media was changed after every 3–4 days. When colonies appeared in CPE‐treated wells (>50 cells/colony), the cells were fixed with formaldehyde and stained with crystal violet. The colonies were then visualized and counted under the microscope and plating efficiency (PE) and surviving fraction were calculated as:

PE=No.of colonies formed in control/No.of cells seeded.
Surviving fraction=No.of colonies formed after treatment/No.of cells seeded×PE.

2.6. Wound‐healing assay

To determine the effect of CPE on the migration of PCa cells, wound‐healing assay was performed. For the wound‐healing assay, 2.5 × 105 cells/well were seeded for PC‐3 and 6 × 105 cells/well were seeded for 22Rv1 cells in 24‐well plates. At 80%–90% cell confluency, a scratch was made passing through the middle region of the well, followed by treatment with either IC₅₀ concentration of CPE or vehicle (complete media with ethanol) taken as control. The width of the scratch was measured at 0 h and 48 h of the treatment. The % wound closure was calculated by the formula:

%Wound closure=Widthat0hWidthat48hWidthat0h×100.

2.7. Measurement of reactive oxygen species

The endogenous reactive oxygen species (ROS) levels on CPE treatment, were measured using DCFDA assay. For this assay, 5000 cells/well were seeded in 96‐well plates. Cells were treated with either IC₅₀ concentration of CPE or 500 μM Hydrogen peroxide (H₂O₂) (taken as positive control) for 48 h. After 48 h of treatment, 10 μM of DCFDA was added to each well for 1 h and fluorescence was measured using Tecan multimode microplate reader with excitation and emission at 488 and 525 nm respectively.

2.8. Measurement of necrotic potential

Necrotic potential of CPE on PCa cells was measured using CyQUANT LDH Cytotoxicity Assay kit as per the manufacturer's instruction. Briefly, 5000 cells/well were seeded in 96‐well plate and incubated for 24 h. After 24 h of incubation, treatment of CPE was given in designated wells and 10 μL of ultrapure water was put in cells for measurement of spontaneous LDH activity. The plate was incubated at 37°C with 5% CO₂ for 48 h. After the designated time period of treatment, media from the wells serving as the maximum LDH activity controls was replaced with fresh complete media containing 10 μL of 10× Lysis Buffer. The plate was then incubated at 37°C with 5% CO₂ for 45 min. 50 μL of medium from each sample (spontaneous LDH activity, maximum LDH activity and treatment LDH activity) was transferred to another 96‐well flat‐bottom plate and 50 μL of Reaction Mixture was then added to each sample well and mixed well. The plate was incubated at room temperature for 30 min protected from light. 50 μL of Stop Solution was then added to each sample well, followed by absorbance measurement at 490 nm and 680 nm using TECAN multimode microplate reader. The % Cytotoxicity was determined using the formula:

%Cytotoxicity=TreatmentLDHactivitySpontaneousLDHactivity/MaximumLDHactivitySpontaneousLDHactivity×100.

2.9. Measurement of apoptotic potential

Caspase‐3 activity‐mediated apoptosis of CPE was measured using the EnzChek Caspase‐3 Activity Assay Kit, as per the manufacturer's instruction. 2 × 105 cells were plated in a 24‐well plate in complete media (media with 10% FBS). After 24 h of seeding for PC‐3 (48 h for 22Rv1 cells), cells were treated with IC₅₀ values of the CPE for 48 h. H₂O₂ at a concentration of 50 μM was used as a positive inducer of caspase 3 activity. Post‐treatment, the cells were washed with 1 mL PBS per well and 60 μL of 1× lysis buffer was added to each well with the cells and kept on ice for 30 min. The lysed cells were then centrifuged for 5 min at 5000 rpm and 50 μL of each sample supernatant was added to 50 μL Z‐DEVD‐AMC substrate solution in a 96‐well black plate. The cells were then incubated at room temperature for 60 min and fluorescence reading was taken at excitation 342 nm and emission 441 nm using TECAN multimode microplate reader. A standard curve was prepared using a series of 7‐amino‐4‐methylcoumarin (AMC) solutions of known concentration ranging from 100 to 0.05 μM. The standard curve was used to calculate the amount of AMC produced in each sample. The caspase 3 activity in nmole of AMC released per min per mL of cell lysate or positive control was calculated based on the formula:

Caspase3activitynmolAMC/min/mL=nmolAMC×d/t×v,

where v = volume of sample in mL; d = dilution factor; t = reaction time in minutes.

2.10. Western blot

The whole cell lysate from cells treated with CPE or vehicle control was isolated using MPER reagent along with inhibitors (1% phosphatase inhibitor, 1% protease inhibitor and 1% PMSF). BCA Assay was performed for measurement of protein concentration in the cell lysates. Equal amount of protein sample from vehicle control and CPE‐treated cells was separated using 12% or 15% SDS‐PAGE and then electrotransferred on to a PVDF membrane. The membranes were incubated (at 4°C) overnight, with primary antibody specific for NF‐κB, oxidative stress markers, LC3B, Beclin‐1, p62, BAD, Bcl‐2 (Abcam), BAX, p27 (Cell Signaling Technology) or GAPDH (Santa Cruz Biotechnology), followed by corresponding HRP conjugated anti‐rabbit IgG antibody (Cell Signaling Technology) or anti‐mouse IgG antibody (Cell Signaling Technology) incubation for 2 h. After that, the membranes were thoroughly washed in TBST (1×). Finally, the PVDF membrane was probed with ECL solution (Thermo Fisher) and observed in ChemiDoc Imaging System (Azure Biosystems) to visualize the protein bands.

2.11. LC–MS analysis

Liquid chromatography–mass spectrometry (LC–MS) analysis of the leaf extract of C. procera was performed using a high‐performance liquid chromatography system (HPLC; Agilent Technologies, Santa Clara, CA, USA) hyphenated to a triple quadrupole tandem mass spectrometer (4000 Q‐Trap, AB Sciex, Foster City, CA, USA). All the parameters of mass spectrometer and HPLC were controlled by Analyst software, version 1.7.1 (AB Sciex) and OpenLAB control panel software (Agilent Technologies), respectively. The crude extract was diluted to 1 mg/mL in pure methanol and further diluted to 10 μg/mL in 50% methanol containing 0.1% formic acid (FA) for LC–MS analysis. The chromatographic separation was carried out on a Purospher® STAR RP‐18 endcapped (3 μm) column using a mobile phase combination of solvent A (18.2 MΩ Milli‐Q water with 0.1% FA) and solvent B (pure methanol with 0.1% FA) pumped at a flow rate of 0.5 mL/min. The gradient profile was set as: 0.0 min 5% B eluent, 1.0 min 5% B eluent, 15.0 min 100% B eluent, 19.0 min 100% B eluent, 20.0 min 5% B eluent and 25.0 min 5% B eluent. The temperature of autosampler tray and the column oven were maintained at 10 ± 1 and 25 ± 1°C, respectively, and the samples were injected at a volume of 20 μL for analysis. In order to achieve maximum coverage of the components, ESI source was operated in dual ionization mode with polarity switching. Independent data acquisition using enhanced mass scan (EMS) mode in the range of 50–800 (m/z) was used for capturing the [M+H+] and [M‐H+] masses. The source‐dependent parameters were set as: curtain gas (CUR) = 25 psi, collision gas (CAD) = 12 psi, ion‐spray voltage = 4.5 kV and 5.5 kV for positive and negative mode respectively, source temperature = 500°C, ion source gas 1 (GS1) = 45 psi, ion source gas 2 (GS2) = 60 psi.

2.12. Identification of intracellular bioactive compounds

In order to analyse the CPE metabolites entering the cells, the PC‐3 and 22Rv1 cells were treated with IC₅₀ concentration of CPE. The cells were trypsinized after 24 h of treatment to give maximum time for the metabolites to enter the cells but not enough time for them to cause cell death. After trypsinization of treated cells, the cell pellet was washed with 1× PBS and centrifuged at 13,000×g for 20 min at 4°C, so as to form a tight pellet. The cell pellet was resuspended in 1 mL of nuclease free water. The cells were then lyophilized. The lyophilized cell lysates were reconstituted in 50% MeOH containing 0.1% FA. The resulting solution was vortexed for 1 min and centrifuged at 10,000 rpm for 10 min at 4°C, and the supernatant was subjected for LC–MS analysis as described above.

2.13. Statistical analysis

Data was analysed using GraphPad Prism, version 8.0 (GraphPad Software Inc). The dose‐response curve and IC₅₀ values for each cell line were calculated by non‐linear regression analysis. Mean ± SEM was calculated for three independent experiments, each with at least three technical replicates. Two‐way anova was used to determine statistical significance for all MTT assays, and unpaired t‐test with Welch's correction was used to determine the statistical significance for the results of all other assays and western blots.

3. RESULTS

3.1. Effect of CPE on cell viability

The effect of CPE on viability of human PCa cells, PC‐3 and 22Rv1, was measured by the MTT assay (Figure 1A,B). To measure the effect of CPE on viability of non‐cancerous cells, normal prostate epithelial cell line (RWPE‐1) was used (Figure 1C). Doxorubicin at a concentration of 100 μM was used as a positive control because it is one of the widely used chemotherapeutic drug for PCa. CPE decreased cell viability of PC‐3 and 22Rv1 cells in a dose‐ and time‐dependent manner. The IC₅₀ values of CPE for all the three cell lines (PC‐3, 22Rv1 and RWPE‐1) are given in Table 1. The optimal cytotoxic effect of CPE was observed at 48 h in both PC‐3 and 22Rv1 cell lines. Therefore, the PCa cells were treated with IC₅₀ concentration of CPE for 48 h in all further experiments.

FIGURE 1.

FIGURE 1

Effect of Calotropis procera leaf extract (CPE) on cell viability: Per cent cell viability at different time points (24, 48, 72 h) after treatment with varying concentrations of CPE (25–400 μg/mL). (A) PC‐3 cell line (B) 22Rv1 cell line (C) RWPE‐1 cell line. Data are presented as mean ± SEM (n = 3).

TABLE 1.

IC₅₀ concentrations of Calotropis procera leaf extract at different time points.

IC₅₀ of CPE (μg/mL)
24 h 48 h 72 h
PC‐3 52.20 26.50 12.20
22Rv1 34.58 19.87 12.98
RWPE‐1 14.31 10.02 9.77

3.2. Effects of CPE on cell division ability of PCa cells

To determine the division ability of a single cell to form a colony after treatment with CPE, clonogenic assay was performed. The size and number of colonies formed in each cell line decreased after treatment with CPE as compared to the control. CPE treatment had a more profound effect on cell division capability of PC‐3 cells compared with 22Rv1 cells (Figure 2A,B).

FIGURE 2.

FIGURE 2

Effect of Calotropis procera leaf extract (CPE) on cell division ability measured via clonogenic assay and western blot of cell cycle markers in prostate cancer cell lines. (A, B) Histogram represents the survival fraction of the colonies formed in (A) PC‐3 and (B) 22Rv1 cells treated with CPE or vehicle control. (C–E) Representative western blot and densitometric analysis of cell cycle markers p27 and NF‐κB in PC‐3 cells. (F–H) Representative western blot and densitometric analysis of cell cycle markers p27 and NF‐κB in 22Rv1 cells. Data are represented as mean ± SEM (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001 compared with vehicle control.

3.3. Effect of CPE on cell cycle markers

Since a significant decrease in clonogenic ability of CPE‐treated cells was observed in both PCa cells, the protein levels of cell cycle markers, p27 and NF‐κB, were checked in both PC‐3 and 22Rv1 cells after treatment with CPE (Figure 2C,F). Densitometric analysis of the western blots showed no change in expression of p27 whereas a significant decrease in protein levels of NF‐κB (p ≤ 0.05) was observed in CPE‐treated PC‐3 cells (Figure 2D,E). On the contrary, an increase in p27 expression (p ≤ 0.05) was observed in CPE‐treated 22Rv1 cells as compared to vehicle control (Figure 2G). Furthermore, NF‐κB expression remained downregulated (p ≤ 0.05), in CPE‐treated 22Rv1 cells as well (Figure 2H).

3.4. Effect of CPE on cell migration

Wound‐healing assay was performed in order to investigate the changes in the cell migration ability of PCa cells in the presence of CPE (Figure 3A,C). The percentage of wound closure in PC‐3 cells treated with CPE was 13.61%, which was significantly less (p < 0.001) than that of vehicle control (99.62%; Figure 3B). In 22Rv1 cells also, the cells treated with IC₅₀ value of CPE had significantly (p < 0.01) lower wound closure of 4.19% compared with that of vehicle control (15.08%) (Figure 3D).

FIGURE 3.

FIGURE 3

Effect of Calotropis procera leaf extract (CPE) on cell migration of prostate cancer cells after 48 h of treatment. Representative images showing the migration of cells in the wound area in (A) PC‐3 and (C) 22Rv1 cells. Percentage of wound closure in (B) PC‐3 and (D) 22Rv1 cells treated with CPE or vehicle control. Data are represented as mean ± SEM (n = 3). **p < 0.01, ***p < 0.001 compared with vehicle control.

3.5. Effect of CPE on endogenous ROS levels

DCFDA assay was performed to analyse the effect of CPE on endogenous ROS levels in PCa cells (Figure 4A,B). In both PC‐3 and 22Rv1, the ROS levels increased after treatment with H₂O₂, which was taken as positive control. Cells treated with vehicle control showed no significant change in ROS levels compared with untreated cells. However, upon treatment with IC₅₀ concentration of CPE, significant decrease in ROS levels was observed in both cell lines compared with untreated cells or compared with cells treated with vehicle control (Figure 4A,B).

FIGURE 4.

FIGURE 4

Levels of endogenous reactive oxygen species (ROS) measured by DCFDA assay and protein levels of antioxidants markers in prostate cancer cells treated with Calotropis procera leaf extract (CPE). Endogenous ROS levels after CPE treatment in (A) PC‐3 cells (B) 22Rv1 cells. H₂O₂ was used as positive control in this assay. (C–F) Representative western blot and densitometric analysis of antioxidant markers catalase, SOD1 and thioredoxin in PC‐3 cells. (G–J) Representative western blot and densitometric analysis of antioxidant markers catalase, SOD1 and thioredoxin in 22Rv1 cells. The data are shown as mean ± SEM (n = 3). ns , no significant difference; *p < 0.05, **p < 0.01, ***p < 0.001 compared with vehicle control or only cells.

3.6. Effect of CPE on antioxidant markers

As ROS levels decreased after treatment with CPE, the effect of CPE on protein levels of different antioxidant molecules including catalase, superoxide dismutase 1 (SOD1) and thioredoxin were analysed (Figure 4C,G). Though a reduction in catalase levels was observed after treatment of PC‐3 cells with CPE, it was not statistically significant (Figure 4D). The levels of SOD1 remained unchanged (Figure 4E) while the levels of thioredoxin decreased significantly (p < 0.01; Figure 4F).

Interestingly, in CPE‐treated 22Rv1 cells, protein levels of catalase and SOD1 were significantly downregulated (p < 0.01), while thioredoxin expression remained unchanged, as compared to vehicle control (Figure 4H–J).

3.7. Effect of CPE on necrosis

Lactate dehydrogenase assay was performed to analyse whether CPE induces necrosis in PCa cells (Figure 5A,B). The percentage cell cytotoxicity was significantly reduced in both PC‐3 (p < 0.01) and 22Rv1 (p < 0.05) cells on treatment with CPE indicating a reduction in necrosis as compared to vehicle control (Figure 5A,B).

FIGURE 5.

FIGURE 5

Effect of Calotropis procera leaf extract (CPE) on necrosis of (A) PC‐3 cells and (B) 22Rv1 cells as analysed by the lactate dehydrogenase release assay. Doxorubicin was used as positive control in this assay. The data are shown as mean ± SEM (n = 3). ns , no significant difference; *p < 0.05, **p < 0.01, ***p < 0.001 in CPE‐treated cells compared with vehicle control cells or doxorubicin‐treated cells.

3.8. Effect of CPE on markers of autophagy

Western blotting for autophagy marker proteins, LC3B, Beclin‐1 and p62, was performed to assess whether autophagy is the mechanism involved in decreasing cell viability of PCa cells after CPE treatment (Figure 6A,E). It was observed that in PC‐3 cells, a significant increase in the ratio of LC3‐II to LC3‐I protein levels (p < 0.05) was observed in cells treated with CPE (Figure 6B) but no change in Beclin‐1 level was observed between vehicle control and treatment group (Figure 6C). The levels of p62 protein in PC‐3 cells were significantly upregulated (p < 0.05) as well (Figure 6D).

FIGURE 6.

FIGURE 6

Effect of Calotropis procera leaf extract (CPE) on molecular markers of autophagy in PC‐3 and 22Rv1 cells. (A–D) Representative western blot and densitometric analysis of autophagy markers in PC‐3 cells. (E–H) Representative western blot and densitometric analysis of autophagy markers in 22Rv1 cells. Data are represented as mean ± SEM (n = 3). *p < 0.05, **p < 0.01 between vehicle control and CPE‐treated groups.

However, in 22Rv1 cells, a significant decrease in the ratio of LC3‐II to LC3‐I and Beclin‐1 (p < 0.01) was observed in cells treated with CPE as compared to vehicle control (Figure 6F,G). Though a decrease in p62 levels was also observed in CPE‐treated 22Rv1 cells, it was not statistically significant (Figure 6H).

3.9. Effect of CPE on induction of apoptosis through extrinsic pathway

Caspase activity assay was performed to analyse whether CPE reduces the cell viability of PCa cells by inducing apoptosis through extrinsic pathway. No significant change in caspase activity was observed in CPE‐treated cells as compared to vehicle control in both PC‐3 and 22Rv1 cells (Figure 7A,B).

FIGURE 7.

FIGURE 7

Effect of Calotropis procera leaf extract (CPE) on induction of apoptosis, analysed by caspase activity assay and western blot of apoptotic markers. Caspase activity after CPE treatment in (A) PC‐3 cells and (B) 22Rv1 cells. (C–F) Representative western blot and densitometric analysis of apoptotic markers BAD, BAX and Bcl‐2 in PC‐3 cells. (G–J) Representative western blot and densitometric analysis of apoptotic markers BAD, BAX and Bcl‐2 in 22Rv1 cells. The data are shown as mean ± SEM (n = 3). ns = no significant difference, *p < 0.05 in CPE‐treated cells compared with vehicle control cells.

3.10. Effect of CPE on apoptotic markers

In order to investigate whether decreasing cell viability by CPE was mediated through intrinsic apoptotic pathway, protein expression of apoptotic markers such as BAD, BAX and Bcl‐2 was measured in cells treated with CPE (Figure 7C,G). No change in protein level of BAD was observed while BAX level was significantly downregulated (p < 0.05) in CPE‐treated PC‐3 cells as compared to vehicle control (Figure 7D,E). Level of the anti‐apoptotic protein, Bcl‐2, also remained unchanged (Figure 7F).

A similar observation as PC‐3 cells was obtained in CPE‐treated 22Rv1 cells where BAD protein levels remained unchanged while a significant decrease in BAX level (p < 0.05) was observed (Figure 7H,I). However, a decrease in Bcl‐2 levels was also observed, though it was not statistically significant (Figure 7J).

3.11. LC–MS analysis of C. procera leaf extract

A total of 123 bioactive compounds were recognized in positive and negative ionization modes through untargeted LC–MS analysis of CPE. A list of all identified primary and secondary metabolites is given in Table 2.

TABLE 2.

Compounds identified in Calotropis procera leaf extract.

S. No. Compound Formula Precursor ion Retention time (min) Compound class Compounds present in cell line
Compounds analysed in positive ion mode (M+H+)
cp001 Safranal C10H14O 151.10 (M+H+) 11.47 Cyclic terpenic aldehyde 22Rv1
cp002 Eucalyptol (cineole) C10H18O 155.14 (M+H+) 13.37 Monoterpenoid PC‐3, 22Rv1
cp003 Terpineol C10H18O 155.14 (M+H+) 13.37 Monoterpenoid 22Rv1
cp004 Dihydroedulan II C13H22O 195.17 (M+H+) 13.50 Organic heterobicyclic compound
cp005 Neryl acetone (cis‐geranylacetone) C13H22O 195.17 (M+H+) 13.50 Monoterpene PC‐3, 22Rv1
cp006 cis‐calamenene C15H22 203.17 (M+H+) 15.00 Sesquiterpene PC‐3, 22Rv1
cp007 Bicyclogermacrene C15H24 205.19 (M+H+) 16.45 Sesquiterpene 22Rv1
cp008 α‐copaene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp009 α‐gurjunene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp010 cis‐thujopsene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp011 α‐humulene (alpha‐caryophyllene) C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp012 β‐himachalene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp013 Germacrene D C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp014 α‐selinene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp015 α‐guaiene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp016 α‐cedrene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp017 Junipene (longifolene) C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp018 Trans‐caryophyllene (β‐caryophyllene) C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp019 Aromadendrene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp020 α‐cadinene (α‐amorphene) C15H24 205.19 (M+H+) 16.45 22Rv1
cp021 α‐cubebene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp022 α‐muurolene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp023 γ‐Cadinene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp024 β‐Muurolene C15H24 205.19 (M+H+) 16.45 PC‐3, 22Rv1
cp025 Diepicedrene‐1‐oxide C15H24O 221.18 (M+H+) 13.27 Sesquiterpene 22Rv1
cp026 Spathulenol C15H24O 221.18 (M+H+) 14.85 22Rv1
cp027 Isoaromadendrene epoxide C15H24O 221.18 (M+H+) 14.85 22Rv1
cp028 Aromadendrene oxide C15H24O 221.18 (M+H+) 14.85 22Rv1
cp029 Calarene epoxide C15H24O 221.18 (M+H+) 14.85 22Rv1
cp030 Cedrenol C15H24O 221.18 (M+H+) 14.85 22Rv1
cp031 1,4‐trans‐1,7‐cis‐acorenone C15H24O 221.18 (M+H+) 14.85 22Rv1
cp032 Cedr‐8‐en‐15‐ol C15H24O 221.18 (M+H+) 14.85 22Rv1
cp033 Cedr‐8‐en‐13‐ol C15H24O 221.18 (M+H+) 14.85 22Rv1
cp034 Caryophyllene oxide C15H24O 221.18 (M+H+) 14.85 22Rv1
cp035 Hexahydrofarnesol C15H32O 229.25 (M+H+) 13.55 Fatty alcohol 22Rv1
cp036 Cedrane‐8,13‐diol C15H26O2 239.19 (M+H+) 13.27 PC‐3, 22Rv1
cp037 Palmitic acid C16H32O2 257.24 (M+H+) 17.65 Long‐chain fatty acids PC‐3, 22Rv1
cp038 Farnesyl acetone C18H30O 263.23 (M+H+) 16.65 Terpene ketone PC‐3, 22Rv1
cp039 9‐eicosyne C20H38 279.30 (M+H+) 15.50 Aliphatic hydrocarbon
cp040 Trans‐geranyl geraniol C20H34O 291.30 (M+H+) 13.90 Diterpenoid PC‐3, 22Rv1
cp041 9‐octadecenoic acid (Z)‐methyl ester C19H36O2 297.27 (M+H+) 16.60 Fatty acid methyl ester
cp042 Phytol C20H40O 297.31 (M+H+) 16.60 Terpenoid alcohol
cp043 Isophytol C20H40O 297.31 (M+H+) 16.60
cp044 Methyl stearate C19H38O2 299.29 (M+H+) 16.90 Fatty acid methyl esters PC‐3, 22Rv1
cp045 Coroglaucigenin C23H34O5 391.24 (M+H+) 11.85 Hydroxy steroid PC‐3, 22Rv1
cp046 Calotropagenin C23H32O6 405.22 (M+H+) 10.01 Cardenolide PC‐3, 22Rv1
cp047 Squalene (spinacene) C30H50 411.39 (M+H+) 21.20 Isoprenoid PC‐3, 22Rv1
cp048 Stigmasterol C29H48O 413.37 (M+H+) 18.45 3beta‐sterol 22Rv1
cp049 Hydroxycalotropagenin C23H32O7 421.21 (M+H+) 8.01 PC‐3, 22Rv1
cp050 Dammaradienol C32H52O2 427.38 (M+H+) 17.80 Fatty alcohols PC‐3, 22Rv1
cp051 α‐amyrin C30H50O 427.39 (M+H+) 17.80 Triterpenoid PC‐3, 22Rv1
cp052 Multiflorenol C30H50O 427.39 (M+H+) 17.85 Triterpenoid PC‐3, 22Rv1
cp053 Lupeol (fagarasterol) C30H50O 427.39 (M+H+) 17.80 PC‐3, 22Rv1
cp054 urs‐19(29)‐en‐3‐β‐ol (pyrethrol) C30H50O 427.39 (M+H+) 17.80 22Rv1
cp055 β‐amyrin C30H50O 427.39 (M+H+) 17.80 22Rv1
cp056 Ethyl iso‐allocholate C26H44O5 437.32 (M+H+) 14.60 PC‐3, 22Rv1
cp057 Kaempferol hexoside C21H20O11 449.10 (M+H+) 9.57 Monosaccharide derivative PC‐3, 22Rv1
cp058 Urs‐12‐en‐24‐oic acid, 3‐oxo‐, methyl ester C31H48O3 469.36 (M+H+) 20.15 PC‐3, 22Rv1
cp059 Urs‐19(29)‐en‐3‐yl acetate (calotropenyl acetate) C32H52O2 469.40 (M+H+) 20.15 Triterpenoid PC‐3, 22Rv1
cp060 Isorhamnetin hexoside C22H22O12 479.11 (M+H+) 11.20 Glycosyloxyflavone 22Rv1
cp061 Uscharidin C29H38O9 531.25 (M+H+) 11.50 Cardenolide PC‐3, 22Rv1
cp062 Calotropin (pecilocerin A) C29H40O9 533.27 (M+H+) 11.73 Cardenolide 22Rv1
cp063 Calactin (pecilocerin B) C29H40O9 533.27 (M+H+) 11.73 22Rv1
cp064 Frugoside C29H44O9 537.30 (M+H+) 9.61 Cardenolide PC‐3, 22Rv1
cp065 Desglucouzarin C29H44O9 537.30 (M+H+) 11.07 PC‐3, 22Rv1
cp066 12 hydroxy calactin C29H40O10 549.26 (M+H+) 11.70 Cardenolide PC‐3, 22Rv1
cp067 15 hydroxy calactin C29H40O10 549.26 (M+H+) 11.70 PC‐3, 22Rv1
cp068 16 hydroxy calactin C29H40O10 549.26 (M+H+) 11.70 PC‐3, 22Rv1
cp069 Calactinic acid C29H40O10 549.26 (M+H+) 11.70 PC‐3, 22Rv1
cp070 Calotoxin C29H40O10 549.26 (M+H+) 9.78 PC‐3, 22Rv1
cp071 Calactinic acid methyl ester I C30H42O10 563.28 (M+H+) 9.64 PC‐3, 22Rv1
cp072 Asclepin C31H42O10 575.28 (M+H+) 11.55 22Rv1
cp073 Voruscharin C31H43NO8S 590.27 (M+H+) 16.70 Cardenolide 22Rv1
cp074 Kaempferol‐3‐O‐rutinoside (nicotiflorin) C27H30O15 595.16 (M+H+) 10.45 Flavonoid glycoside PC‐3
cp075 Kaempferol robinoside C27H30O15 595.16 (M+H+) 10.45 PC‐3
cp076 15 hydroxy uscharin C31H41NO9S 604.25 (M+H+) 11.75 PC‐3, 22Rv1
cp077 2″‐oxovoruscharin C31H41NO9S 604.25 (M+H+) 11.75 Cardenolide PC‐3, 22Rv1
cp078 Quercetin‐3‐O‐rutinoside (Rutin) C27H30O16 611.15 (M+H+) 10.01 Flavonoid glycoside PC‐3, 22Rv1
cp079 Isorhamnetin hexoside pentoside C27H30O16 611.15 (M+H+) 10.01 Flavonoid glycoside PC‐3, 22Rv1
cp080 Labriformine C31H39NO10S 618.23 (M+H+) 8.63 Cardenolide PC‐3, 22Rv1
cp081 Isorhamnetin robinoside C28H32O16 625.17 (M+H+) 10.63 Flavonoid glycoside 22Rv1
cp082 Isorhamnetin‐3‐O‐rutinoside (Narcissin) C28H32O16 625.17 (M+H+) 10.63 22Rv1
cp083 Calotropagenin glycoside III 634.27 (M+H+) 9.63 Calotropin derivative
cp084 Calotropagenin glycoside IV 648.28 (M+H+) 10.67 Calotropin derivative 22Rv1
cp085 Calotropisprocerasaponin I C35H54O13 683.36 (M+H+) 17.13 PC‐3, 22Rv1
cp086 Uzarin C35H54O14 699.35 (M+H+) 17.97 PC‐3, 22Rv1
Compounds analysed in negative ion mode (M‐H+)
cp087 Camphor C10H16O 151.12 (M‐H+) 11.47 Cyclic monoterpene Ketone
cp088 α‐cyclocitral C10H16O 151.12 (M‐H+) 11.47 Organic oxide PC‐3
cp089 Trans‐Pinocarveol C10H16O 151.12 (M‐H+) 11.47 Bicyclic monoterpenoid 22Rv1
cp090 Verbenol (Berbenol) C10H16O 151.12 (M‐H+) 11.47 22Rv1
cp091 1‐dodecene C12H24 167.19 (M‐H+) 16.20 Monoterpenoid PC‐3, 22Rv1
cp092 1,1,6‐trimethyl‐1,2‐dihydronaphthalene C13H16 171.13 (M‐H+) 10.35 13C‐norisoprenoid
cp093 Trans‐chrysanthenol C12H18O2 193.13 (M‐H+) 11.03 PC‐3, 22Rv1
cp094 α‐calacorene C15H20 199.16 (M‐H+) 13.40 Sesquiterpenoid PC‐3, 22Rv1
cp095 Methyl‐α‐ionone C14H22O 205.17 (M‐H+) 14.23 Methyl ketone PC‐3, 22Rv1
cp096 α‐terpinyl propionate C13H22O2 209.16 (M‐H+) 11.40 p‐menthane monoterpenoid
cp097 Humulane‐1,6‐dien‐3‐ol C15H26O 221.20 (M‐H+) 14.77 Sesquiterpene 22Rv1
cp098 Nerolidol (peruviol, penetrol) C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp099 Viridiflorol C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp100 Hinesol (agaruspirol) C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp101 α‐acorenol C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp102 Dihydro‐α‐agarofuran C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp103 Epishyobunol C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp104 Epiglobulol C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp105 Palustrol C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp106 β‐eudesmol (beta‐selinenol) C15H26O 221.20 (M‐H+) 14.77 22Rv1
cp107 α‐cedrol C15H26O 221.20 (M‐H+) 14.77 Sesquiterpenoid and a tertiary alcohol 22Rv1
cp108 Z‐7‐Hexadecenal C16H30O 237.23 (M‐H+) 16.20 Fatty aldehyde 22Rv1
cp109 Methyl palmitate C17H34O2 269.26 (M‐H+) 16.47 Fatty acid methyl Ester 22Rv1
cp110 Kaur‐16‐ene C20H32 271.25 (M‐H+) 16.27 Kaurane diterpenoid 22Rv1
cp111 Bolandiol C18H28O2 275.21 (M‐H+) 13.53 3beta‐hydroxy steroid PC‐3, 22Rv1
cp112 n‐Eicosane C20H42 281.33 (M‐H+) 16.60 Alkane 22Rv1
cp113 Ethyl palmitate C18H36O2 283.27 (M‐H+) 16.53 Fatty acid esters
cp114 Quercetin C15H10O7 301.04 (M‐H+) 15.40 Flavonols 22Rv1
cp115 Ethyl linoleate (mandenol) C20H36O2 307.27 (M‐H+) 16.50 Linoleic acids and derivatives
cp116 Ethyl 9,12‐octadecadienoate C20H36O2 307.27 (M‐H+) 16.50
cp117 Hydroxycoroglaucigenin C23H34O6 405.24 (M‐H+) 9.50 Aminoquinoline PC‐3, 22Rv1
cp118 β‐sitosterol (cupreol) C29H50O 413.39 (M‐H+) 18.45 Stigmastane sterol
cp119 Afroside C29H42O9 533.28 (M‐H+) 12.43 22Rv1
cp120 1‐Heptatriacontanol C37H76O 535.59 (M‐H+) 20.10
cp121 Uscharin C31H41NO8S 586.26 (M‐H+) 13.33 Cardenolides PC‐3, 22Rv1
cp122 Calotropagenin glycoside II 600.24 (M‐H+) 17.80 Calotropin derivative
cp123 Calotropagenin glycoside I 634.25 (M‐H+) 16.75 Calotropin derivative PC‐3, 22Rv1

Abbreviations: NI, negative ion; PI, positive ion; RT, retention time.

LC/MS analysis of PC‐3 and 22Rv1 cells treated with CPE for 24 h was performed to identify the compounds which get internalized. It was observed that from the 123 metabolites identified in the crude leaf extract, 63 metabolites were be detected in PC‐3 cells treated with CPE while they were absent in cell lysates of vehicle control (Table 2). In 22Rv1 cells, 105 metabolites were identified in the CPE‐treated cells which were absent in cell lysate of vehicle control (Table 2).

4. DISCUSSION

Conventional treatment methods for PCa have many side effects and hence, treatment with herbal extracts is considered an important form of alternative therapy. Thus, in this study we investigated the effect of the leaf extract of the herb, C. procera on PCa cell lines in vitro. Results showed that leaf extract of C. procera significantly decreased cell viability as well as cell division and migration capability of both androgen‐independent and androgen‐sensitive PCa cell lines. Interestingly, the androgen‐sensitive 22Rv1 cell line was more susceptible to CPE treatment as compared to androgen‐independent PC‐3 cell line as indicated by the IC50 values.

p27kip1 protein directly mitigates inflammation by inhibiting NF‐κB activation thus playing a key role between cell cycle and inflammation. 21 It is also reported that loss of p27 results in increased aggressiveness of PCa. 22 Hence, our observation that p27 increased in CPE‐treated 22Rv1 cells may explain our results showing decreased cell division and migration capability of these cells after treatment with CPE. Similarly, downregulated expression of NF‐κB in both PCa cell lines after CPE treatment is in line with another study showing that C. procera inhibits breast cancer proliferation by inhibiting NF‐κB activation. 23

Elevated ROS levels maintain tumorigenicity and promote genomic instability in cancer cells. A study using various prostate cells showed that ROS generation is higher in PCa cells compared with normal prostate cells and suggested that reducing ROS production may decrease cell migration while increasing cell death. 24 Interestingly, our data showed that CPE treatment results in significant reduction in ROS levels, which may lead to the reduced cell viability and migration capacity of the PCa cells. Moreover, another study in breast cancer has shown that C. procera can induce cytotoxicity by reducing ROS levels resulting in decreased cell migration and induction of apoptosis. 18

Reduction in ROS levels is usually associated with a concomitant increase in antioxidants. 25 Interestingly, our data showed that levels of antioxidant markers, catalase, SOD1 and thioredoxin, decreased after CPE treatment. Since CPE is known to possess significant radical scavenging activity, 16 it can be inferred that the reduction in ROS levels observed in CPE treated PCa cells was direct and not mediated by modulation of antioxidant enzymes.

Also, SOD1 has been proposed as a novel target for anticancer therapy since it remains overexpressed in a number of cancers along with increased ROS levels in order to prevent any damage caused due to excessive ROS and maintain tumorigenesis. 26 Similarly, another antioxidant protein thioredoxin showed upregulated expression in androgen‐independent PCa and its inhibition led to decrease in cancer growth. 27 Thus, it can be proposed that the downregulated protein levels of SOD1 and thioredoxin could contribute to decreasing cell proliferation of PCa cells.

Cancer cells often utilize autophagy for resource reallocation as a survival strategy. 28 , 29 Studies have shown that in PC‐3 cells, autophagy activation is accompanied by upregulated expression of p62, LC3B and Beclin‐1 at both transcript and protein levels. 30 , 31 , 32 These three proteins play a crucial role in different key steps of autophagy and, hence, were chosen for analysis. In our study, LC3‐II and p62 expression was significantly upregulated indicating autophagy induction in CPE‐treated PC‐3 cells. In contrast, CPE treatment significantly reduced the expression of Beclin‐1 and LC3‐II in 22Rv1 cells, thus showing inhibition of autophagy. There are data suggesting that depending on the cellular features of the PCa cell lines, either induction or inhibition of autophagy can result in cell survival. 28 Studies have shown that activation of autophagy in androgen‐independent PC‐3 cells results in cell cycle arrest in G2/M phase. 33 , 34 However, it has been observed that autophagy acts as pro‐survival mechanism in androgen‐responsive 22Rv1 cells. 29

Some previous studies have shown that C. procera extracts inhibit proliferation of human skin melanoma cells and canine mammary tumour cells via apoptosis induction. 35 , 36 However, our results showed that both extrinsic and intrinsic pathways of apoptosis were not activated in PCa cells by CPE treatment. Decrease in cell viability by CPE also did not involve necrosis as shown by the results of LDH Release assay.

In order to identify the compounds of C. procera hydroalcoholic extract which may be inhibiting cell viability of PCa cells, LC–MS analysis of the hydroalcoholic extract as well as the cells treated with CPE was performed. Majority of compounds internalized by both PCa cell lines belonged to the class of terpene derivatives, cardenolides and flavonoid glycosides. Interestingly, more of the compounds were internalized by the androgen‐responsive 22Rv1 cells with only three compounds, namely nicotiflorin, kaempferol robinoside and α‐cyclotral internalized specifically by androgen‐independent PC‐3 cells.

Terpenes like lupeol have been known to suppress tumour angiogenesis via downregulation of TNFα. 37 Another terpene derivative hinesol has shown anti‐proliferative effect in lung cancer cells. 38 Cardenolides like uscharin, calotropin, calactin, 2′‐oxovoruscharin, 19‐dihydrocalactin, 19‐dihydrocalotoxin, 15‐β hydroxy uscharin, asclepin and calotropagenin have been known to inhibit Na+/K+ATPase and HIF‐1α activity, which are important for tumour metastasis. Thus, these cardenolides show cytotoxic effect against breast, colon, cervical, lung and ovarian cancer. 39 , 40 , 41

Flavonoids possess high antioxidant potential as their phenolic hydroxyl group can stabilize free radicals directly or they can activate antioxidant and suppress pro‐oxidant enzymes indirectly. 42 Quercetin derivatives found in C. procera are known to possess high free radical scavenging activity. 43 Also, the phytoestrogens, kaemferol and its derivatives which have anticancer effect against several androgen‐dependent cancers and thus are suitable candidates for anticancer therapy. 44

In summary, our results show that CPE inhibits proliferation and migration of PCa cells by regulating autophagy and reducing the levels of intracellular ROS. Our results also suggest that androgen‐sensitive and androgen‐independent PCa cells respond differently to herbal formulations depicting their uniqueness which can be utilized as a potential target for PCa treatment. This necessitates the need to test the herbal extracts on different kinds of PCa cells.

The untargeted LC–MS analysis showed the presence of several metabolites in CPE, which are known to possess cytotoxic and anti‐proliferative potential against several different cancers. Many of the metabolites also have radical scavenging potential. Further studies are required to delineate the effect of each metabolite individually or in combination on the cell viability and its associated mechanisms in PCa cells. In conclusion, our findings indicate the possibility of using CPE as an alternate therapeutic agent for PCa. This could be a significant advancement in safer treatment modalities for PCa in particular and cancer in general.

AUTHOR CONTRIBUTIONS

Palak Singh: Conceptualization (lead); data curation (lead); formal analysis (lead); investigation (lead); methodology (lead); software (lead); validation (lead); visualization (lead); writing – original draft (lead); writing – review and editing (equal). Bodhana Dhole: Formal analysis (equal); investigation (equal); project administration (equal); resources (equal); supervision (equal); visualization (equal); writing – review and editing (equal). Jaganmoy Choudhury: Conceptualization (supporting); project administration (supporting); software (equal); supervision (supporting); visualization (supporting). Anannya Tuli: Data curation (supporting); formal analysis (supporting). Deepak Pandey: Conceptualization (equal); methodology (equal); supervision (equal); visualization (equal). Thirumurthy Velpandian: Formal analysis (supporting); supervision (supporting). Surabhi Gupta: Conceptualization (lead); formal analysis (equal); funding acquisition (lead); investigation (equal); methodology (lead); project administration (lead); supervision (lead); validation (lead); visualization (lead); writing – review and editing (equal). Pradeep Kumar Chaturvedi: Conceptualization (lead); funding acquisition (lead); project administration (lead); resources (lead); supervision (lead); writing – review and editing (equal).

CONFLICT OF INTEREST STATEMENT

The authors declare that there are no conflicts of interest.

ACKNOWLEDGEMENTS

The authors would like to thank Mr. Mopungkal Marim, Ms. Usha Chauhan, Mr. Shubham Kurve (Department of Reproductive Biology) and Mr. Maan Singh (Department of Ocular Pharmacology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences) from AIIMS, New Delhi, for providing technical assistance.

Singh P, Dhole B, Choudhury J, et al. Calotropis procera extract inhibits prostate cancer through regulation of autophagy. J Cell Mol Med. 2024;28:e18050. doi: 10.1111/jcmm.18050

DATA AVAILABILITY STATEMENT

Data from this study are available from the corresponding author upon reasonable request.

REFERENCES

  • 1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249. [DOI] [PubMed] [Google Scholar]
  • 2. Bayne CE, Williams SB, Cooperberg MR, et al. Treatment of the primary tumor in metastatic prostate cancer: current concepts and future perspectives. Eur Urol. 2016;69(5):775‐787. [DOI] [PubMed] [Google Scholar]
  • 3. Dueregger A, Heidegger I, Ofer P, et al. The use of dietary supplements to alleviate androgen deprivation therapy side effects during prostate cancer treatment. Nutrients. 2014;6(10):4491‐4519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Karavelioglu E, Gonul Y, Aksit H, et al. Cabazitaxel causes a dose‐dependent central nervous system toxicity in rats. J Neurol Sci. 2016;360:66‐71. [DOI] [PubMed] [Google Scholar]
  • 5. Kim JH, Lee H, Shin EA, Kim DH, Choi JB, Kim SH. Implications of Bcl‐2 and its interplay with other molecules and signaling pathways in prostate cancer progression. Expert Opin Ther Targets. 2017;21(9):911‐920. [DOI] [PubMed] [Google Scholar]
  • 6. Atmaca H, Bozkurt E. Apoptotic and anti‐angiogenic effects of Salvia triloba extract in prostate cancer cell lines. Tumour Biol. 2016;37(3):3639‐3646. [DOI] [PubMed] [Google Scholar]
  • 7. Fu W, Hong Z, You X, et al. Enhancement of anticancer activity of docetaxel by combination with Fuzheng Yiliu decoction in a mouse model of castration‐resistant prostate cancer. Biomed Pharmacother. 2019;118:109374. [DOI] [PubMed] [Google Scholar]
  • 8. Tsai CH, Tzeng SF, Hsieh SC, et al. A standardized herbal extract mitigates tumor inflammation and augments chemotherapy effect of docetaxel in prostate cancer. Sci Rep. 2017;7(1):15624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Lu Z, Wang H, Zhu M, et al. Ophiopogonin D', a natural product from radix Ophiopogonis, induces in vitro and in vivo RIPK1‐dependent and caspase‐independent apoptotic death in androgen‐independent human prostate cancer cells. Front Pharmacol. 2018;9:432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Zheng L, Chen S, Cao Y, et al. Combination of comprehensive two‐dimensional prostate cancer cell membrane chromatographic system and network pharmacology for characterizing membrane binding active components from radix et Rhizoma Rhei and their targets. J Chromatogr A. 2018;1564:145‐154. [DOI] [PubMed] [Google Scholar]
  • 11. Ibrahim SR, Mohamed GA, Shaala LA, Banuls LM, Kiss R, Youssef DT. Calotroposides H‐N, new cytotoxic oxypregnane oligoglycosides from the root bark of Calotropis procera . Steroids. 2015;96:63‐72. [DOI] [PubMed] [Google Scholar]
  • 12. Basak SK, Bhaumik A, Mohanta A, Singhal P. Ocular toxicity by latex of Calotropis procera (Sodom apple). Indian J Ophthalmol. 2009;57(3):232‐234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ibrahim SR, Mohamed G, Shaala L, et al. Proceraside a, a new cardiac glycoside from the root barks of Calotropis procera with in vitro anticancer effects. Nat Prod Res. 2014;28(17):1322‐1327. [DOI] [PubMed] [Google Scholar]
  • 14. Juncker T, Schumacher M, Dicato M, Diederich M. UNBS1450 from Calotropis procera as a regulator of signaling pathways involved in proliferation and cell death. Biochem Pharmacol. 2009;78(1):1‐10. [DOI] [PubMed] [Google Scholar]
  • 15. Al‐Qahtani MAM, Farah MA, Abou‐Tarboush FM, et al. Anticancer effects of Calotropis procera latex extract in mcf‐7 breast cancer cells. Pharmacogn Mag. 2020;16(71):550. [Google Scholar]
  • 16. Olajuyin AM, Olajuyin AK, Wang Z, et al. Anti‐proliferative, antioxidant effects of methanol extract of Calotropis procera leaf on lung cancer cells (H1299) and its ameliorative effect on expression of CD146 on blood cells. Clin Phytoscience. 2021;7(1):1‐12. [Google Scholar]
  • 17. Alzahrani HS, Almalki SA, Rizgallah MR. Study of the cytotoxic effect of Calotropis procera on breast cancer cell line, T47D in vitro: a traditional remedy. Int J Sci Innov. 2019;7(1):107‐112. [Google Scholar]
  • 18. Rabelo ACS, Miglino MA, Arbizu S, et al. Calotropis procera selectively impaired the 4T1 breast cancer cells growth by preferentially blocking Akt/mTOR signaling. Current developments. Nutrition. 2021;5(Supplement_2):278. [Google Scholar]
  • 19. Kumar P, Nagarajan A, Uchil PD. Analysis of Cell Viability by the MTT Assay. Vol 2018. Cold Spring Harb Protoc; 2018. doi: 10.1101/pdb.prot095505 [DOI] [PubMed] [Google Scholar]
  • 20. Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315‐2319. [DOI] [PubMed] [Google Scholar]
  • 21. Antony C. The Tumour Suppressor p27kip1 Interacts with NF‐kB Activator IKK and Plays a Role in Inflammation . 2009.
  • 22. Gupta S, Afaq F, Mukhtar H. Involvement of nuclear factor‐kappa B, Bax and Bcl‐2 in induction of cell cycle arrest and apoptosis by apigenin in human prostate carcinoma cells. Oncogene. 2002;21(23):3727‐3738. [DOI] [PubMed] [Google Scholar]
  • 23. Samy RP, Rajendran P, Li F, et al. Identification of a novel Calotropis procera protein that can suppress tumor growth in breast cancer through the suppression of NF‐κB pathway. PloS One. 2012;7(12):e48514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Kumar B, Koul S, Khandrika L, Meacham RB, Koul HK. Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype. Cancer Res. 2008;68(6):1777‐1785. [DOI] [PubMed] [Google Scholar]
  • 25. Kumar VL, Roy S. Calotropis procera latex extract affords protection against inflammation and oxidative stress in Freund's complete adjuvant‐induced monoarthritis in rats. Mediators Inflamm. 2007;2007:47523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Che M, Wang R, Li X, Wang HY, Zheng XFS. Expanding roles of superoxide dismutases in cell regulation and cancer. Drug Discov Today. 2016;21(1):143‐149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Samaranayake GJ, Troccoli CI, Huynh M, et al. Thioredoxin‐1 protects against androgen receptor‐induced redox vulnerability in castration‐resistant prostate cancer. Nat Commun. 2017;8(1):1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Ziparo E, Petrungaro S, Marini ES, et al. Autophagy in prostate cancer and androgen suppression therapy. Int J Mol Sci. 2013;14(6):12090‐12106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Lin C, Blessing AM, Pulliam TL, et al. Inhibition of CAMKK2 impairs autophagy and castration‐resistant prostate cancer via suppression of AMPK‐ULK1 signaling. Oncogene. 2021;40(9):1690‐1705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Zheng Q, Su H, Ranek MJ, Wang X. Autophagy and p62 in cardiac proteinopathy. Circ Res. 2011;109(3):296‐308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Nam RK, Benatar T, Amemiya Y, Sherman C, Seth A. Mir‐139 regulates autophagy in prostate cancer cells through Beclin‐1 and mTOR signaling proteins. Anticancer Res. 2020;40(12):6649‐6663. [DOI] [PubMed] [Google Scholar]
  • 32. Toepfer N, Childress C, Parikh A, Rukstalis D, Yang W. Atorvastatin induces autophagy in prostate cancer PC3 cells through activation of LC3 transcription. Cancer Biol Ther. 2011;12(8):691‐699. [DOI] [PubMed] [Google Scholar]
  • 33. Chen RJ, Hung CM, Chen YL, Wu MD, Yuan GF, Wang YJ. Monascuspiloin induces apoptosis and autophagic cell death in human prostate cancer cells via the Akt and AMPK signaling pathways. J Agric Food Chem. 2012;60(29):7185‐7193. [DOI] [PubMed] [Google Scholar]
  • 34. Tai S, Sun Y, Liu N, et al. Combination of Rad001 (everolimus) and propachlor synergistically induces apoptosis through enhanced autophagy in prostate cancer cells. Mol Cancer Ther. 2012;11(6):1320‐1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Joshi AL, Roham PH, Mhaske R, et al. Calotropis procera extract induces apoptosis and cell cycle arrest at G2/M phase in human skin melanoma (SK‐MEL‐2) cells. Nat Prod Res. 2015;29(23):2261‐2264. [DOI] [PubMed] [Google Scholar]
  • 36. Vahidi R, Abbasloo E, Safi S, Bolourchian M. Bcl(2)‐dependent antineoplastic effects of Calotropis procera root extract against canine mammary tumor cells. Vet Res Forum. 2021;12(2):197‐202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kangsamaksin T, Chaithongyot S, Wootthichairangsan C, Hanchaina R, Tangshewinsirikul C, Svasti J. Lupeol and stigmasterol suppress tumor angiogenesis and inhibit cholangiocarcinoma growth in mice via downregulation of tumor necrosis factor‐α. PloS One. 2017;12(12):e0189628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Guo W, Liu S, Ju X, et al. The antitumor effect of hinesol, extract from Atractylodes lancea (Thunb.) DC. By proliferation, inhibition, and apoptosis induction via MEK/ERK and NF‐κB pathway in non‐small cell lung cancer cell lines A549 and NCI‐H1299. J Cell Biochem. 2019;120(11):18600‐18607. [DOI] [PubMed] [Google Scholar]
  • 39. Zheng Z, Zhou Z, Zhang Q, et al. Non‐classical cardenolides from Calotropis gigantea exhibit anticancer effect as HIF‐1 inhibitors. Bioorg Chem. 2021;109:104740. [DOI] [PubMed] [Google Scholar]
  • 40. Parhira S, Zhu GY, Chen M, Bai LP, Jiang ZH. Cardenolides from Calotropis gigantea as potent inhibitors of hypoxia‐inducible factor‐1 transcriptional activity. J Ethnopharmacol. 2016;194:930‐936. [DOI] [PubMed] [Google Scholar]
  • 41. Pederson PJ, Cai S, Carver C, et al. Triple‐negative breast cancer cells exhibit differential sensitivity to cardenolides from Calotropis gigantea. J Nat Prod. 2020;83(7):2269‐2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Kopustinskiene DM, Jakstas V, Savickas A, Bernatoniene J. Flavonoids as anticancer agents. Nutrients. 2020;12:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Mohamed MA, Hamed MM, Ahmed WS, Abdou AM. Antioxidant and cytotoxic flavonols from Calotropis procera . Z Naturforsch C J Biosci. 2011;66(11–12):547‐554. [DOI] [PubMed] [Google Scholar]
  • 44. Kim SH, Choi KC. Anti‐cancer effect and underlying mechanism(s) of kaempferol, a phytoestrogen, on the regulation of apoptosis in diverse cancer cell models. Toxicol Res. 2013;29(4):229‐234. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data from this study are available from the corresponding author upon reasonable request.


Articles from Journal of Cellular and Molecular Medicine are provided here courtesy of Blackwell Publishing

RESOURCES