Original Article
Split ViewerCannabigerol Treatment Shows Antiproliferative Activity and Causes Apoptosis of Human Colorectal Cancer Cells
1Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea
2Kangwon Center for System Imaging, Chuncheon, Republic of Korea
3Chuncheon Bioindustry Foundation, Chuncheon, Republic of Korea
4Korean Pharmacopuncture Institute, Seoul, Republic of Korea
Correspondence to: Keun-Cheol Kim
Department of Biological Sciences, College of Natural Sciences, Kangwon National University, 1 Kangwondaehak Rd, Chuncheon 24341, Republic of Korea
Tel: +82-33-250-8532
E-mail: kckim@kangwon.ac.kr
This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
J Pharmacopuncture 2024; 27(4): 332-339
Published December 31, 2024 https://doi.org/10.3831/KPI.2024.27.4.332
Copyright © The Korean Pharmacopuncture Institute.
Abstract
Methods: Anti-proliferative effect of CBG was examined using MTT assay and two colorectal cancer cells (SW480 and LoVo cells). Cell death ratio was analyzed using Annexin V/ PI staining experiment. Cell cycle distribution was analyzed using flow cytometry. We also performed western blot analysis on apoptotic marker proteins.
Results: CBG showed growth inhibitory effect in colorectal cancer cells using MTT assay. IC50 concentration of CBG was 34.89 μM in SW480 cells and 23.51 μM in LoVo cells. Annexin V/PI staining showed that CBG treatment increased apoptotic cells from 4.8% to 31.7% in SW480 cells and from 7.7% to 33.9% in LoVo cells. Flow cytometry confirmed that CBG increased sub G1 population via G1 arrest in both SW480 and LoVo cells. Western blot analysis showed that CBG increased expression levels of cell death-related proteins such as cleaved PARP-1, cleaved caspase 9, p53, and caspase 3.
Conclusion: CBG treatment shows antiproliferative activity and causes apoptosis of colorectal cancer cells, suggesting that CBG is applicable as a promising anticancer drug.
Keywords
INTRODUCTION
Colorectal cancer (CRC), characterized by aberrant proliferation of glandular epithelial cells in the colon or rectum, is the second most fatal cancer in the world [1, 2]. The major treatment modality for CRC is surgery, typically followed by chemotherapy [3]. The 5-year survival rate of CRC patients depends on the stage of CRC progression. If the cancer has metastasized, only about 10% of the patients survive beyond 5 years [4, 5]. 5-Fluorouracil (5-FU), an inhibitor of DNA synthesis, is the most widely used chemotherapeutic drug for CRC [6, 7]. The angiogenesis inhibitors bevacizumab (Avastin) and ramucirumab (Cyramza) also find place in CRC treatment. However, these drugs cause substantial side effects [8]. Currently, a variety of therapeutic strategies including chemotherapy agents are utilized for CRC treatment. In addition, many studies have been performed to identify new compounds from natural plant extracts with anticancer activities.
Cannabigerol (CBG) is a phytocannabinoid extracted from
Given that CBG has no psychoactive effects, its therapeutic application in cancer is an interesting proposition [16]. CBG has been studied for its efficacy in neurological disorders, inflammatory diseases, and infections [17, 18]. CBG remains relatively understudied despite its unique chemical profile and potential therapeutic applications in cancer. CBG can prevent the progression of glioblastoma by activating the proapoptotic pathway [19]. CBG can also inhibit cell growth and colony formation and triggers cell cycle arrest. In addition, it was found to induce apoptosis in HuCC-1 and Mz-ChA-1 cholangiocarcinoma cells [20]. CBG can attenuate the production of colony-stimulating factor-1 (CSF-1) by melanoma cells, indicating that it functions as an anticancer agent by acting in the tumor microenvironment [21]. CBG was also found to reduce tumor progression in a xenograft model by lowering the number of tumor-associated macrophages [21].
Therefore, CBG seems to be a promising compound in cancer treatment. However, additional research is required to establish its effect using various cancer models. We performed a large array of molecular assays including the dimethylthiazole-2’, 5’-diphenyl-2-H-tetrazlium bromide (MTT) assay, Annexin V/propidium iodide (PI) staining, fluorescence-activated cell sorting (FACS) analysis, and western blot analysis to clarify the effect of CBG against CRC cells. We found that CBG treatment reduces the proliferation and induces the apoptosis of human CRC cells.
MATERIALS AND METHODS
1. Cell culture and reagents
Human CRC cells LoVo and SW480 were purchased from Korea Cell Bank (Seoul, Korea). LoVo cells were maintained in RPMI-1640 (Welgene, Korea), and SW480 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Welgene) supplemented with 1X penicillin/streptomycin (LS 202-02; Welgene) and 10% fetal bovine serum (Merck Millipore, Germany). All cells were cultured in an incubator at 37℃ with 5% CO2. CBG was purchased from Cayman Chemical (Ann Arbor, MI, USA) and dissolved in dimethyl sulfoxide (DMSO).
2. MTT assay
Cell viability was measured by the MTT assay. For this assay, MTT was diluted in phosphate-buffered saline (PBS). Cells were seeded into a 96-well plate at a cell density of 6 × 103 cells/well. After incubation overnight, the medium was replaced with DMEM containing various CBG concentrations ranging from 0 to 40 μM. The cells were incubated for up to 96 h. Every 24 h, 1X MTT solution was added to the 96-well plate and incubated for 4 h. The medium was removed, and purple formazan crystals were solubilized with 100 μL DMSO. The absorbance was measured at 570/690 nm wavelength using a microplate reader (Allsheng, China).
3. Annexin V/fluorescein isothiocyanate (FITC) staining
Cell death was analyzed using an Annexin V-FITC apoptosis detection kit (BioVision, Milpitas, CA, USA). SW480 and LoVo cells were seeded in 60 π culture dishes at a density of 6 × 105 cells/dish and incubated overnight. The SW480 and LoVo cells were treated with 30 μM CBG for 24 h to observe the fluorescence intensity. Cells were fixed with paraformaldehyde for 20 min. After fixing, the cells were incubated with 10 μL of PI and 5 μL of Annexin V for 10 min in the dark at room temperature. Annexin V was observed under a confocal microscope (Ts2, Nikon, Tokyo, Japan) at the Kangwon Center for System Imaging. To analyze the apoptotic cell population, the cells were washed twice with PBS and incubated with 10 μL of PI and 5 μL of Annexin V for 30 min in the dark at room temperature. Annexin V-FITC and PI fluorescence intensities were analyzed with a FACSymphonyTM A3 Cell Analyzer (BD Bioscience, Franklin Lakes, NJ, USA).
4. FACS analysis and cell morphology
The concentration of CBG used for treatment was based on its IC50 values against LoVo and SW480 cells. After culturing for up to 48 h, the cells were harvested at 12 and 24-hour intervals and washed twice with PBS. The cells were fixed with 70% ethanol for 24 h. After fixing, the cells were stained with PI (50 μg/mL) for 30 min. Cell cycle distribution was analyzed with a FACSymphonyTM A3 Cell Analyzer (BD Bioscience). Cell morphology was imaged using a confocal microscope (Nikon, Eclipse TS100, Tokyo, Japan).
5. Western blot
Cells were seeded into 100 π dishes at a density of 6 × 106 cells/dish and treated with CBG for up to 48 h. Before lysis, the cells were washed once with PBS. Proteins were extracted using radio-immunoprecipitation assay buffer (25 mM Tris-HCl pH 7.6, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS) supplemented with a protease inhibitor cocktail to extract total proteins. Protein concentrations were determined using Bradford reagent kit (Thermo Fisher Scientific, Waltham, MA, USA). Equal amounts of proteins (40 μg) were electrophoresed on 12% and 8% gels and transferred onto a polyvinylidene fluoride (PVDF) membrane at 100 V for 1 h. The PVDF membrane was blocked using 5% skimmed milk for 30 min at room temperature, followed by incubation with specific primary antibodies at 4℃ overnight. Membranes were blotted with caspase 3, caspase 9, p53, poly (ADP-ribose) polymerase-1 (PARP-1), and β-actin primary antibodies. Afterward, the membrane was washed with 1X TBST buffer and incubated with secondary antibodies for 1 h at room temperature. Finally, the proteins were detected using an electrochemiluminescence kit (ATTO, Japan).
RESULTS
1. CBG inhibits cell growth and changes in cell morphology
We performed an MTT assay to analyze whether CBG could inhibit the growth of SW480 and LoVo cells (Fig. 1A). At concentrations below 10 μM, CBG did not affect cell growth. However, CBG at 20 μM inhibited the growth of both cell lines. No cell growth was observed in the presence of CBG at 40 μM. The IC₅₀ value of CBG was estimated to be 34.89 μM for SW480 cells and 23.51 μM for LoVo cells. Therefore, the concentration of CBG was set as 30 μM for both cancer cells for further experiments. CBG-treated cancer cells demonstrated intracytoplasmic vesicles as a distinct time-dependent morphological change (Fig. 1B). Buoyant dead cancer cells were also noted in the CBG-treated group in a time-dependent manner. These results suggest that CBG can inhibit the growth and cause death of human CRC cells.
-
Figure 1.CBG inhibits cell growth and changes cell morphology. (A) Cell viability was determined using MTT assay for 96 hours. IC50 value of CBG was 34.89 μM for SW480 cells and 23.51 μM for LoVo cells. (B) Cell morphology was observed with a phase contrast microscope after CBG treatment. Dead cells and cytoplasmic vesicles were shown in CBG-treated cancer cells.
2. Annexin V is increased during cell death induced by CBG
Phytocannabinoids including CBD and THC can induce apoptosis in CRC cells. Therefore, we examined whether cell death induced by CBG was associated with an increased amount of Annexin V. We counted the number of dead cells using flow cytometry after Annexin V/PI staining (Fig. 2A, B). The proportion of apoptotic cell population increased after 24 h of CBG treatment. The proportion of early apoptotic cells increased 5.8-fold from 4.3% to 26% in SW480 cells and 7-fold from 3.6% to 25.5% in LoVo cells. The proportion of late apoptotic cells also increased by 5.2% from 0.5% to 5.7% in SW480 cells and increased by 4.3% from 4.1% to 8.4% in LoVo cells. When observed under a fluorescence microscope, the fluorescence intensity of Annexin V was observed near the cell membrane of both cancer cell lines after CBG treatment (Fig. 2C). These results suggest that CBG induces the typical cell death mechanism of phytocannabinoids in human CRC cells.
-
Figure 2.CBG treatment shows increased annexin V expression in colorectal cancer cells. (A) Colorectal cancer cells were treated with 30 μM of CBG for 24 hours. Apoptotic cells were analyzed with annexin V-FITC/PI staining using flow cytometry. (B) Graphs showing increased apoptotic colorectal cancer cells after CBG treatment. (C) Annexin V stained cells were observed with a confocal microscope after CBG treatment.
3. CBG treatment induces cell death via G1 arrest
Time-dependent cell cycle distribution analysis after CBG treatment (Fig. 3) revealed that after 12 h of CBG exposure, the proportion of G1 phase population increased from 55.9% to 75.7% in SW480 cells and from 53.6% to 62.6% in LoVo cells. The proportion of dead cell population increased to 38.4% in SW480 cells after 24 h of CBG exposure and 19.2% in LoVo cells after 48 h of CBG exposure. Therefore, CBG treatment altered cell cycle distribution by increasing cell death via G1 arrest.
-
Figure 3.CBG treatment increases sub-G1 population via G1 arrest. (A) SW480 and LoVo cells were treated with 30 μM CBG for 48 hours. After that, cells were stained with propidium iodide for FACS analysis. (B) Graphs show percentage of each cell cycle distribution.
4. CBG treatment upregulates the expression of apoptotic marker proteins
To examine the mechanism by which CBG induces cell death, we examined the expression of cell death-related proteins by western blot analysis (Fig. 4A). CBG increased the expression of cleaved forms of caspase 9, PARP-1, and caspase 3, which are typical marker proteins for programmed cell death, in a time-dependent manner (Fig. 4B). These results indicate that CBG induces apoptosis of CRC cells.
-
Figure 4.CBG treatment increases cleaved forms of caspase and PARP-1. (A) LoVo and SW480 cells were treated with 30 μM CBG, harvested at every 24 hours, and cultured for 72 hours. Western blot experiment was performed using PARP-1, caspase 9, caspase 3, and p53 antibodies. (B) Graphs showing increased intensities for cleaved forms of PARP-1, caspase 9, and caspase 3.
DISCUSSION
CBG is a non-psychoactive cannabinoid derived from
Its neuroprotective effect is similar to that of CBD. Studies on neural cell lines have shown that 2.5-10 μM CBG protects against hydrogen peroxide-induced neurotoxicity effects by reducing oxidative stress and mitochondrial dysfunction [26]. In the present study, we showed that CBG could inhibit cell growth, with IC50 values of 34.89 μM and 23.51 μM for SW480 and LoVo CRC cells, respectively. CBG was reported to reduce the viability of patient-derived primary glioblastoma cells at an IC50 of 100 µM [19]. In addition, CBG can inhibit cholangiocarcinoma growth at high concentrations of 100-200 μM [20]. Therefore, the IC50 of CBG depends on the cancer cell type. Our current study demonstrated that CBG inhibits the growth of two CRC cell lines at a concentration of 30 μM. Exposure at this concentration increased the proportion of buoyant dead cells, confirming the anticancer effects of CBG. Morphologically, we also observed vesicle formation in CBG-treated CRC cells. These morphological changes have also been observed previously in cancer cells treated with other cannabinoids. We previously demonstrated that CBD regulates intracellular vesicle formation in lung and colorectal cancers [27, 28]. Peroxisome proliferator-activated receptor γ (PPARγ), clathrin, and β-adaptin are known to regulate vesicle formation in lung cancer. In contrast, CBD induces the formation of vesicles in MCF7 breast cancer cells, suggesting a mechanism for cell death through autophagy [29]. CBD-induced vesicle formation might also depend on PINK1-Parkin-dependent mitophagy [30], suggesting that the formation of cytoplasmic vacuolation can occur through various mechanisms after cannabinoid treatment. Therefore, we see that CBG and CBD have different pharmacological effects in terms of morphological and functional changes, such as vesicle formation and cell death.
CBG is known to induce the apoptosis of many types of tumors, including prostate carcinoma, glioblastoma, and cholangiocarcinoma both
Cannabinoids are known to improve prognosis and promote cancer regression in patients with different tumor types. A combination of curcumin, piperine, and cannabinoid variants was found to inhibit cell proliferation and induce apoptosis drastically in different CRC models [32]. Combination treatment of CBG and CBD exhibits the highest cytotoxicity via G-protein-coupled receptor 55 (GPR55) and TRPV1 signaling in glioblastoma stem cells, suggesting that CBG has potential in adjuvant standard-of-care therapy [19]. Exposure of ultraviolet A (UVA)-irradiated melanocytes to CBG significantly decreased the content of phosphatidylcholine, phosphatidylinositol, and sphingomyelin, which was increased by UVA, indicating that CBD and CBG can partially reverse the pro-cancerogenic changes in phospholipid profiles induced by UVA [33]. An optimal combination of CBG and CBD exhibited strong antitumor effects in mice with transgenic adenocarcinoma of the mouse prostate (i.e., TRAMP) by altering mitochondrial bioenergetics via voltage-dependent anion-selective channel 1, pointing to their therapeutical potential in prostate cancer [34]. A combination of THC, CBC, and CBG was demonstrated to exhibit synergistic effects with a PARP-1 inhibitor against ovarian cancer cells via the Wnt signaling pathway [35]. Therefore, cannabinoids may be used in combination to achieve synergistic anticancer effects.
CONCLUSION
Our results highlight the anticancer effects of CBG in two CRC cell lines. Although CBG seems to be a promising drug in preclinical cancer treatment, comprehensive studies are necessary to translate these findings into clinical applications. In addition, ongoing research into its mechanisms of action is crucial for developing effective CBG-based therapies for cancer.
ACKNOWLEDGEMENTS
We thank the helpful assistants of the Korea Basic Science Institute (KBSI) National Research Facilities & Equipment Center (NFEC).
CONFLICTS OF INTEREST
Keun-Cheol Kim is an editorial board member of Journal of Pharmacopuncture but has no role in the decision to publish this article. No other potential conflicts of interest relevant to this aricle were reported.
FUNDING
References
- Sawicki T, Ruszkowska M, Danielewicz A, Niedźwiedzka E, Arłukowicz T, Przybyłowicz KE. A review of colorectal cancer in terms of epidemiology, risk factors, development, symptoms and diagnosis. Cancers (Basel). 2021;13(9):2025.
- Kumar A, Gautam V, Sandhu A, Rawat K, Sharma A, Saha L. Current and emerging therapeutic approaches for colorectal cancer: a comprehensive review. World J Gastrointest Surg. 2023;15(4):495-519.
- Tirendi S, Marengo B, Domenicotti C, Bassi AM, Almonti V, Vernazza S. Colorectal cancer and therapy response: a focus on the main mechanisms involved. Front Oncol. 2023;13:1208140.
- Hossain MS, Karuniawati H, Jairoun AA, Urbi Z, Ooi J, John A, et al. Colorectal cancer: a review of carcinogenesis, global epidemiology, current challenges, risk factors, preventive and treatment strategies. Cancers (Basel). 2022;14(7):1732.
- Zeineddine FA, Zeineddine MA, Yousef A, Gu Y, Chowdhury S, Dasari A, et al. Survival improvement for patients with metastatic colorectal cancer over twenty years. NPJ Precis Oncol. 2023;7(1):16.
- Van Cutsem E, Hoff PM, Harper P, Bukowski RM, Cunningham D, Dufour P, et al. Oral capecitabine vs intravenous 5-fluorouracil and leucovorin: integrated efficacy data and novel analyses from two large, randomised, phase III trials. Br J Cancer. 2004;90(6):1190-7.
- Zhang N, Yin Y, Xu SJ, Chen WS. 5-Fluorouracil: mechanisms of resistance and reversal strategies. Molecules. 2008;13(8):1551-69.
- Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5(1):22.
- Gülck T, Møller BL. Phytocannabinoids: origins and biosynthesis. Trends Plant Sci. 2020;25(10):985-1004.
- Stone NL, Murphy AJ, England TJ, O'Sullivan SE. A systematic review of minor phytocannabinoids with promising neuroprotective potential. Br J Pharmacol. 2020;177(19):4330-52.
- Anderson LL, Heblinski M, Absalom NL, Hawkins NA, Bowen MT, Benson MJ, et al. Cannabigerolic acid, a major biosynthetic precursor molecule in cannabis, exhibits divergent effects on seizures in mouse models of epilepsy. Br J Pharmacol. 2021;178(24):4826-41.
- Tahir MN, Shahbazi F, Rondeau-Gagné S, Trant JF. The biosynthesis of the cannabinoids. J Cannabis Res. 2021;3(1):7.
- Kim AL, Yun YJ, Choi HW, Hong CH, Shim HJ, Lee JH, et al. Profiling cannabinoid contents and expression levels of corresponding biosynthetic genes in commercial
Cannabis (Cannabis sativa L.) cultivars. Plants (Basel). 2022;11(22):3088. - Jastrząb A, Jarocka-Karpowicz I, Skrzydlewska E. The origin and biomedical relevance of cannabigerol. Int J Mol Sci. 2022;23(14):7929.
- Filipiuc LE, Ababei DC, Alexa-Stratulat T, Pricope CV, Bild V, Stefanescu R, et al. Major Phytocannabinoids and their related compounds: should we only search for drugs that act on cannabinoid receptors? Pharmaceutics. 2021;13(11):1823.
- di Giacomo V, Chiavaroli A, Recinella L, Orlando G, Cataldi A, Rapino M, et al. Antioxidant and neuroprotective effects induced by cannabidiol and cannabigerol in rat CTX-TNA2 astrocytes and isolated cortexes. Int J Mol Sci. 2020;21(10):3575.
- Gugliandolo A, Pollastro F, Grassi G, Bramanti P, Mazzon E. In vitro model of neuroinflammation: efficacy of cannabigerol, a non-psychoactive cannabinoid. Int J Mol Sci. 2018;19(7):1992.
- Appendino G, Gibbons S, Giana A, Pagani A, Grassi G, Stavri M, et al. Antibacterial cannabinoids from Cannabis sativa: a structure-activity study. J Nat Prod. 2008;71(8):1427-30.
- Lah TT, Novak M, Pena Almidon MA, Marinelli O, Žvar Baškovič B, Majc B, et al. Cannabigerol is a potential therapeutic agent in a novel combined therapy for glioblastoma. Cells. 2021;10(2):340.
- Viereckl MJ, Krutsinger K, Apawu A, Gu J, Cardona B, Barratt D, et al. Cannabidiol and cannabigerol inhibit cholangiocarcinoma growth in vitro via divergent cell death pathways. Biomolecules. 2022;12(6):854.
- Wyrobnik I, Steinberg M, Gelfand A, Rosenblum R, Eid Mutlak Y, Sulimani L, et al. Decreased melanoma CSF-1 secretion by Cannabigerol treatment reprograms regulatory myeloid cells and reduces tumor progression. Oncoimmunology. 2023;12(1):2219164.
- Schilling S, Melzer R, McCabe PF. Cannabis sativa. Curr Biol. 2020;30(1):R8-9.
- Grunfeld Y, Edery H. Psychopharmacological activity of the active constituents of hashish and some related cannabinoids. Psychopharmacologia. 1969;14(3):200-10.
- Nachnani R, Raup-Konsavage WM, Vrana KE. The pharmacological case for cannabigerol. J Pharmacol Exp Ther. 2021;376(2):204-12.
- Zagožen M, Čerenak A, Kreft S. Cannabigerol and cannabichromene in
Cannabis sativa L. Acta Pharm. 2020;71(3):355-64. - Echeverry C, Prunell G, Narbondo C, de Medina VS, Nadal X, Reyes-Parada M, et al. A comparative in vitro study of the neuroprotective effect induced by cannabidiol, cannabigerol, and their respective acid forms: relevance of the 5-HT1A receptors. Neurotox Res. 2021;39(2):335-48.
- Park YJ, Na HH, Kwon IS, Hwang YN, Park HJ, Kwon TH, et al. Cannabidiol regulates PPARγ-dependent vesicle formation as well as cell death in A549 human lung cancer cells. Pharmaceuticals (Basel). 2022;15(7):836.
- Kwon IS, Hwang YN, Park JH, Na HH, Kwon TH, Park JS, et al. Metallothionein family proteins as regulators of zinc ions synergistically enhance the anticancer effect of cannabidiol in human colorectal cancer cells. Int J Mol Sci. 2023;24(23):16621.
- Sharma K, Le N, Alotaibi M, Gewirtz DA. Cytotoxic autophagy in cancer therapy. Int J Mol Sci. 2014;15(6):10034-51.
- Ramirez A, Old W, Selwood DL, Liu X. Cannabidiol activates PINK1-Parkin-dependent mitophagy and mitochondrial-derived vesicles. Eur J Cell Biol. 2022;101(1):151185.
- De Petrocellis L, Ligresti A, Schiano Moriello A, Iappelli M, Verde R, Stott CG, et al. Non-THC cannabinoids inhibit prostate carcinoma growth in vitro and in vivo: pro-apoptotic effects and underlying mechanisms. Br J Pharmacol. 2013;168(1):79-102.
- Yüksel B, Hızlı Deniz AA, Şahin F, Sahin K, Türkel N. Cannabinoid compounds in combination with curcumin and piperine display an anti-tumorigenic effect against colon cancer cells. Front Pharmacol. 2023;14:1145666.
- Łuczaj W, Dobrzyńska I, Skrzydlewska E. Differences in the phospholipid profile of melanocytes and melanoma cells irradiated with UVA and treated with cannabigerol and cannabidiol. Sci Rep. 2023;13(1):16121.
- Mahmoud AM, Kostrzewa M, Marolda V, Cerasuolo M, Maccarinelli F, Coltrini D, et al. Cannabidiol alters mitochondrial bioenergetics via VDAC1 and triggers cell death in hormone-refractory prostate cancer. Pharmacol Res. 2023;189:106683.
- Shalev N, Kendall M, Anil SM, Tiwari S, Peeri H, Kumar N, et al. Phytocannabinoid compositions from cannabis act synergistically with PARP1 inhibitor against ovarian cancer cells in vitro and affect the Wnt signaling pathway. Molecules. 2022;27(21):7523.
Related articles in JoP
Article
Original Article
J Pharmacopuncture 2024; 27(4): 332-339
Published online December 31, 2024 https://doi.org/10.3831/KPI.2024.27.4.332
Copyright © The Korean Pharmacopuncture Institute.
Cannabigerol Treatment Shows Antiproliferative Activity and Causes Apoptosis of Human Colorectal Cancer Cells
Ju-Hee Park1 , Yu-Na Hwang1 , Han-Heom Na1,2 , Do-Yeon Kim1 , Hyo-Jun Lee1 , Tae-Hyung Kwon3 , Jin-Sung Park4 , Keun-Cheol Kim1,2*
1Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea
2Kangwon Center for System Imaging, Chuncheon, Republic of Korea
3Chuncheon Bioindustry Foundation, Chuncheon, Republic of Korea
4Korean Pharmacopuncture Institute, Seoul, Republic of Korea
Correspondence to:Keun-Cheol Kim
Department of Biological Sciences, College of Natural Sciences, Kangwon National University, 1 Kangwondaehak Rd, Chuncheon 24341, Republic of Korea
Tel: +82-33-250-8532
E-mail: kckim@kangwon.ac.kr
This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted noncommercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Abstract
Objectives: To determine growth inhibitory and anti-cancer effects of Cannabigerol (CBG) in human colorectal cancer cells.
Methods: Anti-proliferative effect of CBG was examined using MTT assay and two colorectal cancer cells (SW480 and LoVo cells). Cell death ratio was analyzed using Annexin V/ PI staining experiment. Cell cycle distribution was analyzed using flow cytometry. We also performed western blot analysis on apoptotic marker proteins.
Results: CBG showed growth inhibitory effect in colorectal cancer cells using MTT assay. IC50 concentration of CBG was 34.89 μM in SW480 cells and 23.51 μM in LoVo cells. Annexin V/PI staining showed that CBG treatment increased apoptotic cells from 4.8% to 31.7% in SW480 cells and from 7.7% to 33.9% in LoVo cells. Flow cytometry confirmed that CBG increased sub G1 population via G1 arrest in both SW480 and LoVo cells. Western blot analysis showed that CBG increased expression levels of cell death-related proteins such as cleaved PARP-1, cleaved caspase 9, p53, and caspase 3.
Conclusion: CBG treatment shows antiproliferative activity and causes apoptosis of colorectal cancer cells, suggesting that CBG is applicable as a promising anticancer drug.
Keywords: phytocannabinoid, cannabigerol, IC50, colorectal cancer, apoptosis
INTRODUCTION
Colorectal cancer (CRC), characterized by aberrant proliferation of glandular epithelial cells in the colon or rectum, is the second most fatal cancer in the world [1, 2]. The major treatment modality for CRC is surgery, typically followed by chemotherapy [3]. The 5-year survival rate of CRC patients depends on the stage of CRC progression. If the cancer has metastasized, only about 10% of the patients survive beyond 5 years [4, 5]. 5-Fluorouracil (5-FU), an inhibitor of DNA synthesis, is the most widely used chemotherapeutic drug for CRC [6, 7]. The angiogenesis inhibitors bevacizumab (Avastin) and ramucirumab (Cyramza) also find place in CRC treatment. However, these drugs cause substantial side effects [8]. Currently, a variety of therapeutic strategies including chemotherapy agents are utilized for CRC treatment. In addition, many studies have been performed to identify new compounds from natural plant extracts with anticancer activities.
Cannabigerol (CBG) is a phytocannabinoid extracted from
Given that CBG has no psychoactive effects, its therapeutic application in cancer is an interesting proposition [16]. CBG has been studied for its efficacy in neurological disorders, inflammatory diseases, and infections [17, 18]. CBG remains relatively understudied despite its unique chemical profile and potential therapeutic applications in cancer. CBG can prevent the progression of glioblastoma by activating the proapoptotic pathway [19]. CBG can also inhibit cell growth and colony formation and triggers cell cycle arrest. In addition, it was found to induce apoptosis in HuCC-1 and Mz-ChA-1 cholangiocarcinoma cells [20]. CBG can attenuate the production of colony-stimulating factor-1 (CSF-1) by melanoma cells, indicating that it functions as an anticancer agent by acting in the tumor microenvironment [21]. CBG was also found to reduce tumor progression in a xenograft model by lowering the number of tumor-associated macrophages [21].
Therefore, CBG seems to be a promising compound in cancer treatment. However, additional research is required to establish its effect using various cancer models. We performed a large array of molecular assays including the dimethylthiazole-2’, 5’-diphenyl-2-H-tetrazlium bromide (MTT) assay, Annexin V/propidium iodide (PI) staining, fluorescence-activated cell sorting (FACS) analysis, and western blot analysis to clarify the effect of CBG against CRC cells. We found that CBG treatment reduces the proliferation and induces the apoptosis of human CRC cells.
MATERIALS AND METHODS
1. Cell culture and reagents
Human CRC cells LoVo and SW480 were purchased from Korea Cell Bank (Seoul, Korea). LoVo cells were maintained in RPMI-1640 (Welgene, Korea), and SW480 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Welgene) supplemented with 1X penicillin/streptomycin (LS 202-02; Welgene) and 10% fetal bovine serum (Merck Millipore, Germany). All cells were cultured in an incubator at 37℃ with 5% CO2. CBG was purchased from Cayman Chemical (Ann Arbor, MI, USA) and dissolved in dimethyl sulfoxide (DMSO).
2. MTT assay
Cell viability was measured by the MTT assay. For this assay, MTT was diluted in phosphate-buffered saline (PBS). Cells were seeded into a 96-well plate at a cell density of 6 × 103 cells/well. After incubation overnight, the medium was replaced with DMEM containing various CBG concentrations ranging from 0 to 40 μM. The cells were incubated for up to 96 h. Every 24 h, 1X MTT solution was added to the 96-well plate and incubated for 4 h. The medium was removed, and purple formazan crystals were solubilized with 100 μL DMSO. The absorbance was measured at 570/690 nm wavelength using a microplate reader (Allsheng, China).
3. Annexin V/fluorescein isothiocyanate (FITC) staining
Cell death was analyzed using an Annexin V-FITC apoptosis detection kit (BioVision, Milpitas, CA, USA). SW480 and LoVo cells were seeded in 60 π culture dishes at a density of 6 × 105 cells/dish and incubated overnight. The SW480 and LoVo cells were treated with 30 μM CBG for 24 h to observe the fluorescence intensity. Cells were fixed with paraformaldehyde for 20 min. After fixing, the cells were incubated with 10 μL of PI and 5 μL of Annexin V for 10 min in the dark at room temperature. Annexin V was observed under a confocal microscope (Ts2, Nikon, Tokyo, Japan) at the Kangwon Center for System Imaging. To analyze the apoptotic cell population, the cells were washed twice with PBS and incubated with 10 μL of PI and 5 μL of Annexin V for 30 min in the dark at room temperature. Annexin V-FITC and PI fluorescence intensities were analyzed with a FACSymphonyTM A3 Cell Analyzer (BD Bioscience, Franklin Lakes, NJ, USA).
4. FACS analysis and cell morphology
The concentration of CBG used for treatment was based on its IC50 values against LoVo and SW480 cells. After culturing for up to 48 h, the cells were harvested at 12 and 24-hour intervals and washed twice with PBS. The cells were fixed with 70% ethanol for 24 h. After fixing, the cells were stained with PI (50 μg/mL) for 30 min. Cell cycle distribution was analyzed with a FACSymphonyTM A3 Cell Analyzer (BD Bioscience). Cell morphology was imaged using a confocal microscope (Nikon, Eclipse TS100, Tokyo, Japan).
5. Western blot
Cells were seeded into 100 π dishes at a density of 6 × 106 cells/dish and treated with CBG for up to 48 h. Before lysis, the cells were washed once with PBS. Proteins were extracted using radio-immunoprecipitation assay buffer (25 mM Tris-HCl pH 7.6, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS) supplemented with a protease inhibitor cocktail to extract total proteins. Protein concentrations were determined using Bradford reagent kit (Thermo Fisher Scientific, Waltham, MA, USA). Equal amounts of proteins (40 μg) were electrophoresed on 12% and 8% gels and transferred onto a polyvinylidene fluoride (PVDF) membrane at 100 V for 1 h. The PVDF membrane was blocked using 5% skimmed milk for 30 min at room temperature, followed by incubation with specific primary antibodies at 4℃ overnight. Membranes were blotted with caspase 3, caspase 9, p53, poly (ADP-ribose) polymerase-1 (PARP-1), and β-actin primary antibodies. Afterward, the membrane was washed with 1X TBST buffer and incubated with secondary antibodies for 1 h at room temperature. Finally, the proteins were detected using an electrochemiluminescence kit (ATTO, Japan).
RESULTS
1. CBG inhibits cell growth and changes in cell morphology
We performed an MTT assay to analyze whether CBG could inhibit the growth of SW480 and LoVo cells (Fig. 1A). At concentrations below 10 μM, CBG did not affect cell growth. However, CBG at 20 μM inhibited the growth of both cell lines. No cell growth was observed in the presence of CBG at 40 μM. The IC₅₀ value of CBG was estimated to be 34.89 μM for SW480 cells and 23.51 μM for LoVo cells. Therefore, the concentration of CBG was set as 30 μM for both cancer cells for further experiments. CBG-treated cancer cells demonstrated intracytoplasmic vesicles as a distinct time-dependent morphological change (Fig. 1B). Buoyant dead cancer cells were also noted in the CBG-treated group in a time-dependent manner. These results suggest that CBG can inhibit the growth and cause death of human CRC cells.
-
Figure 1. CBG inhibits cell growth and changes cell morphology. (A) Cell viability was determined using MTT assay for 96 hours. IC50 value of CBG was 34.89 μM for SW480 cells and 23.51 μM for LoVo cells. (B) Cell morphology was observed with a phase contrast microscope after CBG treatment. Dead cells and cytoplasmic vesicles were shown in CBG-treated cancer cells.
2. Annexin V is increased during cell death induced by CBG
Phytocannabinoids including CBD and THC can induce apoptosis in CRC cells. Therefore, we examined whether cell death induced by CBG was associated with an increased amount of Annexin V. We counted the number of dead cells using flow cytometry after Annexin V/PI staining (Fig. 2A, B). The proportion of apoptotic cell population increased after 24 h of CBG treatment. The proportion of early apoptotic cells increased 5.8-fold from 4.3% to 26% in SW480 cells and 7-fold from 3.6% to 25.5% in LoVo cells. The proportion of late apoptotic cells also increased by 5.2% from 0.5% to 5.7% in SW480 cells and increased by 4.3% from 4.1% to 8.4% in LoVo cells. When observed under a fluorescence microscope, the fluorescence intensity of Annexin V was observed near the cell membrane of both cancer cell lines after CBG treatment (Fig. 2C). These results suggest that CBG induces the typical cell death mechanism of phytocannabinoids in human CRC cells.
-
Figure 2. CBG treatment shows increased annexin V expression in colorectal cancer cells. (A) Colorectal cancer cells were treated with 30 μM of CBG for 24 hours. Apoptotic cells were analyzed with annexin V-FITC/PI staining using flow cytometry. (B) Graphs showing increased apoptotic colorectal cancer cells after CBG treatment. (C) Annexin V stained cells were observed with a confocal microscope after CBG treatment.
3. CBG treatment induces cell death via G1 arrest
Time-dependent cell cycle distribution analysis after CBG treatment (Fig. 3) revealed that after 12 h of CBG exposure, the proportion of G1 phase population increased from 55.9% to 75.7% in SW480 cells and from 53.6% to 62.6% in LoVo cells. The proportion of dead cell population increased to 38.4% in SW480 cells after 24 h of CBG exposure and 19.2% in LoVo cells after 48 h of CBG exposure. Therefore, CBG treatment altered cell cycle distribution by increasing cell death via G1 arrest.
-
Figure 3. CBG treatment increases sub-G1 population via G1 arrest. (A) SW480 and LoVo cells were treated with 30 μM CBG for 48 hours. After that, cells were stained with propidium iodide for FACS analysis. (B) Graphs show percentage of each cell cycle distribution.
4. CBG treatment upregulates the expression of apoptotic marker proteins
To examine the mechanism by which CBG induces cell death, we examined the expression of cell death-related proteins by western blot analysis (Fig. 4A). CBG increased the expression of cleaved forms of caspase 9, PARP-1, and caspase 3, which are typical marker proteins for programmed cell death, in a time-dependent manner (Fig. 4B). These results indicate that CBG induces apoptosis of CRC cells.
-
Figure 4. CBG treatment increases cleaved forms of caspase and PARP-1. (A) LoVo and SW480 cells were treated with 30 μM CBG, harvested at every 24 hours, and cultured for 72 hours. Western blot experiment was performed using PARP-1, caspase 9, caspase 3, and p53 antibodies. (B) Graphs showing increased intensities for cleaved forms of PARP-1, caspase 9, and caspase 3.
DISCUSSION
CBG is a non-psychoactive cannabinoid derived from
Its neuroprotective effect is similar to that of CBD. Studies on neural cell lines have shown that 2.5-10 μM CBG protects against hydrogen peroxide-induced neurotoxicity effects by reducing oxidative stress and mitochondrial dysfunction [26]. In the present study, we showed that CBG could inhibit cell growth, with IC50 values of 34.89 μM and 23.51 μM for SW480 and LoVo CRC cells, respectively. CBG was reported to reduce the viability of patient-derived primary glioblastoma cells at an IC50 of 100 µM [19]. In addition, CBG can inhibit cholangiocarcinoma growth at high concentrations of 100-200 μM [20]. Therefore, the IC50 of CBG depends on the cancer cell type. Our current study demonstrated that CBG inhibits the growth of two CRC cell lines at a concentration of 30 μM. Exposure at this concentration increased the proportion of buoyant dead cells, confirming the anticancer effects of CBG. Morphologically, we also observed vesicle formation in CBG-treated CRC cells. These morphological changes have also been observed previously in cancer cells treated with other cannabinoids. We previously demonstrated that CBD regulates intracellular vesicle formation in lung and colorectal cancers [27, 28]. Peroxisome proliferator-activated receptor γ (PPARγ), clathrin, and β-adaptin are known to regulate vesicle formation in lung cancer. In contrast, CBD induces the formation of vesicles in MCF7 breast cancer cells, suggesting a mechanism for cell death through autophagy [29]. CBD-induced vesicle formation might also depend on PINK1-Parkin-dependent mitophagy [30], suggesting that the formation of cytoplasmic vacuolation can occur through various mechanisms after cannabinoid treatment. Therefore, we see that CBG and CBD have different pharmacological effects in terms of morphological and functional changes, such as vesicle formation and cell death.
CBG is known to induce the apoptosis of many types of tumors, including prostate carcinoma, glioblastoma, and cholangiocarcinoma both
Cannabinoids are known to improve prognosis and promote cancer regression in patients with different tumor types. A combination of curcumin, piperine, and cannabinoid variants was found to inhibit cell proliferation and induce apoptosis drastically in different CRC models [32]. Combination treatment of CBG and CBD exhibits the highest cytotoxicity via G-protein-coupled receptor 55 (GPR55) and TRPV1 signaling in glioblastoma stem cells, suggesting that CBG has potential in adjuvant standard-of-care therapy [19]. Exposure of ultraviolet A (UVA)-irradiated melanocytes to CBG significantly decreased the content of phosphatidylcholine, phosphatidylinositol, and sphingomyelin, which was increased by UVA, indicating that CBD and CBG can partially reverse the pro-cancerogenic changes in phospholipid profiles induced by UVA [33]. An optimal combination of CBG and CBD exhibited strong antitumor effects in mice with transgenic adenocarcinoma of the mouse prostate (i.e., TRAMP) by altering mitochondrial bioenergetics via voltage-dependent anion-selective channel 1, pointing to their therapeutical potential in prostate cancer [34]. A combination of THC, CBC, and CBG was demonstrated to exhibit synergistic effects with a PARP-1 inhibitor against ovarian cancer cells via the Wnt signaling pathway [35]. Therefore, cannabinoids may be used in combination to achieve synergistic anticancer effects.
CONCLUSION
Our results highlight the anticancer effects of CBG in two CRC cell lines. Although CBG seems to be a promising drug in preclinical cancer treatment, comprehensive studies are necessary to translate these findings into clinical applications. In addition, ongoing research into its mechanisms of action is crucial for developing effective CBG-based therapies for cancer.
ACKNOWLEDGEMENTS
We thank the helpful assistants of the Korea Basic Science Institute (KBSI) National Research Facilities & Equipment Center (NFEC).
CONFLICTS OF INTEREST
Keun-Cheol Kim is an editorial board member of Journal of Pharmacopuncture but has no role in the decision to publish this article. No other potential conflicts of interest relevant to this aricle were reported.
FUNDING
Fig 1.
Fig 2.
Fig 3.
Fig 4.
References
- Sawicki T, Ruszkowska M, Danielewicz A, Niedźwiedzka E, Arłukowicz T, Przybyłowicz KE. A review of colorectal cancer in terms of epidemiology, risk factors, development, symptoms and diagnosis. Cancers (Basel). 2021;13(9):2025.
- Kumar A, Gautam V, Sandhu A, Rawat K, Sharma A, Saha L. Current and emerging therapeutic approaches for colorectal cancer: a comprehensive review. World J Gastrointest Surg. 2023;15(4):495-519.
- Tirendi S, Marengo B, Domenicotti C, Bassi AM, Almonti V, Vernazza S. Colorectal cancer and therapy response: a focus on the main mechanisms involved. Front Oncol. 2023;13:1208140.
- Hossain MS, Karuniawati H, Jairoun AA, Urbi Z, Ooi J, John A, et al. Colorectal cancer: a review of carcinogenesis, global epidemiology, current challenges, risk factors, preventive and treatment strategies. Cancers (Basel). 2022;14(7):1732.
- Zeineddine FA, Zeineddine MA, Yousef A, Gu Y, Chowdhury S, Dasari A, et al. Survival improvement for patients with metastatic colorectal cancer over twenty years. NPJ Precis Oncol. 2023;7(1):16.
- Van Cutsem E, Hoff PM, Harper P, Bukowski RM, Cunningham D, Dufour P, et al. Oral capecitabine vs intravenous 5-fluorouracil and leucovorin: integrated efficacy data and novel analyses from two large, randomised, phase III trials. Br J Cancer. 2004;90(6):1190-7.
- Zhang N, Yin Y, Xu SJ, Chen WS. 5-Fluorouracil: mechanisms of resistance and reversal strategies. Molecules. 2008;13(8):1551-69.
- Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5(1):22.
- Gülck T, Møller BL. Phytocannabinoids: origins and biosynthesis. Trends Plant Sci. 2020;25(10):985-1004.
- Stone NL, Murphy AJ, England TJ, O'Sullivan SE. A systematic review of minor phytocannabinoids with promising neuroprotective potential. Br J Pharmacol. 2020;177(19):4330-52.
- Anderson LL, Heblinski M, Absalom NL, Hawkins NA, Bowen MT, Benson MJ, et al. Cannabigerolic acid, a major biosynthetic precursor molecule in cannabis, exhibits divergent effects on seizures in mouse models of epilepsy. Br J Pharmacol. 2021;178(24):4826-41.
- Tahir MN, Shahbazi F, Rondeau-Gagné S, Trant JF. The biosynthesis of the cannabinoids. J Cannabis Res. 2021;3(1):7.
- Kim AL, Yun YJ, Choi HW, Hong CH, Shim HJ, Lee JH, et al. Profiling cannabinoid contents and expression levels of corresponding biosynthetic genes in commercial
Cannabis (Cannabis sativa L.) cultivars. Plants (Basel). 2022;11(22):3088. - Jastrząb A, Jarocka-Karpowicz I, Skrzydlewska E. The origin and biomedical relevance of cannabigerol. Int J Mol Sci. 2022;23(14):7929.
- Filipiuc LE, Ababei DC, Alexa-Stratulat T, Pricope CV, Bild V, Stefanescu R, et al. Major Phytocannabinoids and their related compounds: should we only search for drugs that act on cannabinoid receptors? Pharmaceutics. 2021;13(11):1823.
- di Giacomo V, Chiavaroli A, Recinella L, Orlando G, Cataldi A, Rapino M, et al. Antioxidant and neuroprotective effects induced by cannabidiol and cannabigerol in rat CTX-TNA2 astrocytes and isolated cortexes. Int J Mol Sci. 2020;21(10):3575.
- Gugliandolo A, Pollastro F, Grassi G, Bramanti P, Mazzon E. In vitro model of neuroinflammation: efficacy of cannabigerol, a non-psychoactive cannabinoid. Int J Mol Sci. 2018;19(7):1992.
- Appendino G, Gibbons S, Giana A, Pagani A, Grassi G, Stavri M, et al. Antibacterial cannabinoids from Cannabis sativa: a structure-activity study. J Nat Prod. 2008;71(8):1427-30.
- Lah TT, Novak M, Pena Almidon MA, Marinelli O, Žvar Baškovič B, Majc B, et al. Cannabigerol is a potential therapeutic agent in a novel combined therapy for glioblastoma. Cells. 2021;10(2):340.
- Viereckl MJ, Krutsinger K, Apawu A, Gu J, Cardona B, Barratt D, et al. Cannabidiol and cannabigerol inhibit cholangiocarcinoma growth in vitro via divergent cell death pathways. Biomolecules. 2022;12(6):854.
- Wyrobnik I, Steinberg M, Gelfand A, Rosenblum R, Eid Mutlak Y, Sulimani L, et al. Decreased melanoma CSF-1 secretion by Cannabigerol treatment reprograms regulatory myeloid cells and reduces tumor progression. Oncoimmunology. 2023;12(1):2219164.
- Schilling S, Melzer R, McCabe PF. Cannabis sativa. Curr Biol. 2020;30(1):R8-9.
- Grunfeld Y, Edery H. Psychopharmacological activity of the active constituents of hashish and some related cannabinoids. Psychopharmacologia. 1969;14(3):200-10.
- Nachnani R, Raup-Konsavage WM, Vrana KE. The pharmacological case for cannabigerol. J Pharmacol Exp Ther. 2021;376(2):204-12.
- Zagožen M, Čerenak A, Kreft S. Cannabigerol and cannabichromene in
Cannabis sativa L. Acta Pharm. 2020;71(3):355-64. - Echeverry C, Prunell G, Narbondo C, de Medina VS, Nadal X, Reyes-Parada M, et al. A comparative in vitro study of the neuroprotective effect induced by cannabidiol, cannabigerol, and their respective acid forms: relevance of the 5-HT1A receptors. Neurotox Res. 2021;39(2):335-48.
- Park YJ, Na HH, Kwon IS, Hwang YN, Park HJ, Kwon TH, et al. Cannabidiol regulates PPARγ-dependent vesicle formation as well as cell death in A549 human lung cancer cells. Pharmaceuticals (Basel). 2022;15(7):836.
- Kwon IS, Hwang YN, Park JH, Na HH, Kwon TH, Park JS, et al. Metallothionein family proteins as regulators of zinc ions synergistically enhance the anticancer effect of cannabidiol in human colorectal cancer cells. Int J Mol Sci. 2023;24(23):16621.
- Sharma K, Le N, Alotaibi M, Gewirtz DA. Cytotoxic autophagy in cancer therapy. Int J Mol Sci. 2014;15(6):10034-51.
- Ramirez A, Old W, Selwood DL, Liu X. Cannabidiol activates PINK1-Parkin-dependent mitophagy and mitochondrial-derived vesicles. Eur J Cell Biol. 2022;101(1):151185.
- De Petrocellis L, Ligresti A, Schiano Moriello A, Iappelli M, Verde R, Stott CG, et al. Non-THC cannabinoids inhibit prostate carcinoma growth in vitro and in vivo: pro-apoptotic effects and underlying mechanisms. Br J Pharmacol. 2013;168(1):79-102.
- Yüksel B, Hızlı Deniz AA, Şahin F, Sahin K, Türkel N. Cannabinoid compounds in combination with curcumin and piperine display an anti-tumorigenic effect against colon cancer cells. Front Pharmacol. 2023;14:1145666.
- Łuczaj W, Dobrzyńska I, Skrzydlewska E. Differences in the phospholipid profile of melanocytes and melanoma cells irradiated with UVA and treated with cannabigerol and cannabidiol. Sci Rep. 2023;13(1):16121.
- Mahmoud AM, Kostrzewa M, Marolda V, Cerasuolo M, Maccarinelli F, Coltrini D, et al. Cannabidiol alters mitochondrial bioenergetics via VDAC1 and triggers cell death in hormone-refractory prostate cancer. Pharmacol Res. 2023;189:106683.
- Shalev N, Kendall M, Anil SM, Tiwari S, Peeri H, Kumar N, et al. Phytocannabinoid compositions from cannabis act synergistically with PARP1 inhibitor against ovarian cancer cells in vitro and affect the Wnt signaling pathway. Molecules. 2022;27(21):7523.