Article

Novel mechanism of cannabidiol-induced apoptosis in breast cancer cell lines

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... CBD has shown growth inhibitory effects on glioblastoma [14], leukemia [15], lung cancer [16], breast cancer [17], cervical cancer [18], prostate cancer [19], and melanoma [20]. Various breast cancer cell lines, including estrogen-receptor (ER)-positive or negative or triple-negative breast cancer (TNBC) cells, have displayed a dose-dependent response to CBD (mostly with low IC50 values [17,[21][22][23][24][25][26]). As the second major component, CBD (Figure 1a) is not associated with psychoactivity [9,11] and has therefore been applied in the treatment of a wide range of neurodegenerative diseases [12,13] and tumors. ...
... As the second major component, CBD (Figure 1a) is not associated with psychoactivity [9,11] and has therefore been applied in the treatment of a wide range of neurodegenerative diseases [12,13] and tumors. CBD has shown growth inhibitory effects on glioblastoma [14], leukemia [15], lung cancer [16], breast cancer [17], cervical cancer [18], prostate cancer [19], and melanoma [20]. Various breast cancer cell lines, including estrogen-receptor (ER)-positive or negative or triple-negative breast cancer (TNBC) cells, have displayed a dose-dependent response to CBD (mostly with low IC50 values [17,[21][22][23][24][25][26]). ...
... CBD has shown growth inhibitory effects on glioblastoma [14], leukemia [15], lung cancer [16], breast cancer [17], cervical cancer [18], prostate cancer [19], and melanoma [20]. Various breast cancer cell lines, including estrogen-receptor (ER)-positive or negative or triple-negative breast cancer (TNBC) cells, have displayed a dose-dependent response to CBD (mostly with low IC50 values [17,[21][22][23][24][25][26]). ...
Article
Full-text available
Cannabidiol (CBD), a nonpsychoactive major component derived from Cannabis sativa, widely used in neurodegenerative diseases, has now been proven to have growth inhibitory effects on many tumor cell lines, including breast tumors. Meanwhile CBD can effectively alleviate cancer-associated pain, anxiety, and depression, especially tumor cachexia, thus it is very promising as an anti-tumor drug with unique advantages. 20(S)-Protopanaxadiol (PPD) derived from the best-known tonic Chinese herbal medicine Ginseng was designed to be co-loaded with CBD into liposomes to examine their synergistic tumor-inhibitory effect. The CBD-PPD co-loading liposomes (CP-liposomes) presented a mean particle size of 138.8 nm. Further glycosyl-modified CP-liposomes (GMCP-liposomes) were prepared by the incorporation of n-Dodecyl β-D-maltoside (Mal) into the liposomal bilayer with glucose residue anchored on the surface to act as a ligand targeting the GLUT1 receptor highly expressed on tumor cells. In vivo studies on murine breast tumor (4T1 cells)-bearing BALB/c mice demonstrated good dose dependent anti-tumor efficacy of CP-liposomes. A high tumor inhibition rate (TIR) of 82.2% was achieved with good tolerance. However, glycosylation modification failed to significantly enhance TIR of CP-liposomes. In summary, combined therapy with PPD proved to be a promising strategy for CBD to be developed into a novel antitumor drug, with characteristics of effectiveness, good tolerance, and the potential to overcome tumor cachexia.
... Breast cancer is the most common cancer type in women worldwide, with the highest regional incidence reported in Australia and New Zealand [32,33]. CBD was found to inhibit oestrogen-receptor-positive (ER+), -negative (ER−) or -triple-negative breast cancer cells (TNBC) in a dose-dependent manner, with relatively low IC50 values [6,[34][35][36][37][38][39]. This suggests that breast cancer cells are sensitive to CBD, while non-transformed epithelial breast cells (MCF10A) are not significantly affected [10,28,39,40]. ...
... Interestingly, the administration of CBD in several clinical trials, as a single dose of 150-900 mg [41][42][43][44][45][46][47][48][49] or 50-1000 mg/day for up to 13 weeks [50][51][52][53], showed overall good tolerability and safety profile, with mild side effects [54]. A variety of mechanisms for CBD's antiproliferative effects have been reported, including autophagy, cell cycle arrest at the G1/S checkpoint, and apoptosis in MCF7, MDA-MB-231, and 4T1 breast cancer cells [34,35,39,55]. Recent studies have found that the antiproliferative effect of CBD in breast cancer cells was independent of endocannabinoid receptors (to some extent), mediated primarily through ROS-induced cell death [34,35,39,56,57]. ...
... A variety of mechanisms for CBD's antiproliferative effects have been reported, including autophagy, cell cycle arrest at the G1/S checkpoint, and apoptosis in MCF7, MDA-MB-231, and 4T1 breast cancer cells [34,35,39,55]. Recent studies have found that the antiproliferative effect of CBD in breast cancer cells was independent of endocannabinoid receptors (to some extent), mediated primarily through ROS-induced cell death [34,35,39,56,57]. CBD's pro-apoptotic actions were attributed to the downregulation of mTOR, AKT, 4EBP1, and cyclin D, as well as the overexpression of PPAR-γ [35,39], where CBD coordinated apoptosis and autophagy via Beclin-1 translocation and cleavage [39]. ...
Article
Full-text available
Cannabidiol (CBD), a nonpsychoactive phytocannabinoid, has recently emerged as a potential cytotoxic agent in addition to its ameliorative activity in chemotherapy-associated side effects. In this work, the potential interactions of CBD with docetaxel (DOC), doxorubicin (DOX), paclitaxel (PTX), vinorelbine (VIN), and 7-ethyl-10-hydroxycamptothecin (SN−38) were explored in MCF7 breast adenocarcinoma cells using different synergy quantification models. The apoptotic profiles of MCF7 cells after the treatments were assessed via flow cytometry. The molecular mechanisms of CBD and the most promising combinations were investigated via label-free quantification proteomics. A strong synergy was observed across all synergy models at different molar ratios of CBD in combination with SN−38 and VIN. Intriguingly, synergy was observed for CBD with all chemotherapeutic drugs at a molar ratio of 636:1 in almost all synergy models. However, discording synergy trends warranted the validation of the selected combinations against different models. Enhanced apoptosis was observed for all synergistic CBD combinations compared to monotherapies or negative controls. A shotgun proteomics study highlighted 121 dysregulated proteins in CBD-treated MCF7 cells compared to the negative controls. We reported the inhibition of topoisomerase II β and α, cullin 1, V-type proton ATPase, and CDK-6 in CBD-treated MCF7 cells for the first time as additional cytotoxic mechanisms of CBD, alongside sabotaged energy production and reduced mitochondrial translation. We observed 91 significantly dysregulated proteins in MCF7 cells treated with the synergistic combination of CBD with SN−38 (CSN−38), compared to the monotherapies. Regulation of telomerase, cell cycle, topoisomerase I, EGFR1, protein metabolism, TP53 regulation of DNA repair, death receptor signalling, and RHO GTPase signalling pathways contributed to the proteome-wide synergistic molecular mechanisms of CSN−38. In conclusion, we identified significant synergistic interactions between CBD and the five important chemotherapeutic drugs and the key molecular pathways of the combination of CBD and CSN−38 in MCF7 cells. Further in vivo and clinical studies are warranted to evaluate the implementation of CBD-based synergistic adjuvant therapies for breast cancer.
... In preclinical studies, CB1 and CB2 agonists (CBD, THC, and synthetic) have been shown to inhibit the proliferation of estrogen receptors positive breast cancer cell lines [14]. Accordingly, a cytotoxic effect of CBD was observed in several cell lines including estrogenreceptor (ER)-positive cells (MCF-7, ZR-75-1, T47D), ER-negative cells (MDA-MB-231, MDA-MB-468, and SK-BR3), and triple-negative breast cancer cells (SUM159, 4T1up, MVT-1, and SCP2) [115][116][117][118][119]. ...
... In addition, CBD treatment induces an enhancement in the level of ROS in breast cancer cells [70]. Furthermore, CBD induces apoptosis and blocks cell viability through the inhibition of the AKT/mTOR axis and cyclin D together with the enhancement of ROS generation [115,116]. Similarly, THC showed anti-proliferative effects on several breast cancer cell lines [116,122]. Elbaz et al. observed that CBD specifically inhibits the epidermal growth factor-induced proliferation, suggesting it as a novel potential therapeutic option for breast cancer [115]. ...
... Furthermore, CBD induces apoptosis and blocks cell viability through the inhibition of the AKT/mTOR axis and cyclin D together with the enhancement of ROS generation [115,116]. Similarly, THC showed anti-proliferative effects on several breast cancer cell lines [116,122]. Elbaz et al. observed that CBD specifically inhibits the epidermal growth factor-induced proliferation, suggesting it as a novel potential therapeutic option for breast cancer [115]. ...
Article
Full-text available
Cannabis sativa L. is a source of over 150 active compounds known as phytocannabinoids that are receiving renewed interest due to their diverse pharmacologic activities. Indeed, phyto-cannabinoids mimic the endogenous bioactive endocannabinoids effects through activation of CB1 and CB2 receptors widely described in the central nervous system and peripheral tissues. All phyto-cannabinoids have been studied for their protective actions towards different biological mechanisms, including inflammation, immune response, oxidative stress that, altogether, result in an inhibitory activity against the carcinogenesis. The role of the endocannabinoid system is not yet completely clear in cancer, but several studies indicate that cannabinoid receptors and endogenous ligands are overexpressed in different tumor tissues. Recently, in vitro and in vivo evidence support the effectiveness of phytocannabinoids against various cancer types, in terms of proliferation, metastasis, and angiogenesis, actions partially due to their ability to regulate signaling pathways critical for cell growth and survival. The aim of this review was to report the current knowledge about the action of phytocannabinoids from Cannabis sativa L. against cancer initiation and progression with a specific regard to brain, breast, colorectal, and lung cancer as well as their possible use in the therapies. We will also report the known molecular mechanisms responsible for such positive effects. Finally, we will describe the actual therapeutic options for Cannabis sativa L. and the ongoing clinical trials.
... Similarly, in a rat model of induced schizophrenia, mTOR signalling is reduced (Renard et al., 2016), whereas CBD treatment increased mTOR activity and reversed symptoms. In contrast, several preclinical studies focused on tuberous sclerosis complex (TSC)-associated epilepsy and breast cancer, showing pathology-associated elevated mTORC1 activity and showing that CBD treatment reduced pathway activity (Serra et al., 2019;Sultan, Marie, & Sheweita, 2018). Despite numerous suggested mechanisms for CBD on the G protein-coupled receptor 55 (GPR55) (Kaplan, Stella, Catterall, & Westenbroek, 2017), adenosine signalling (Liou et al., 2008), transient receptor potential vanilloid type 1 (TRPV1) (Costa, Giagnoni, Franke, Trovato, & Colleoni, 2004) and the folate one-carbon cycle (Perry, Finch, Muller-Taubenberger, Leung, & Williams, 2019), these mechanisms do not provide direct insight to the effect of these phytocannabinoids on mTOR signalling. ...
... Phytocannabinoids have demonstrated efficacy in preclinical models of MS (Feliu et al., 2015;Giacoppo et al., 2017) and in clinical studies relating to spasticity associated with MS (Etges et al., 2016;Novotna et al., 2011). In addition, CBD has been shown to be effective in preclinical models of psychosis (Renard et al., 2016;Renard, Norris, Rushlow, & Laviolette, 2017), breast cancer (Sultan et al., 2018) and tuberous sclerosis complex (Serra et al., 2019). One suggested mechanism for CBD treatment of epilepsy and cancer is through the downregulation of hyperactivated mTORC1 found in the disease state (Serra et al., 2019;Sultan et al., 2018). ...
... In addition, CBD has been shown to be effective in preclinical models of psychosis (Renard et al., 2016;Renard, Norris, Rushlow, & Laviolette, 2017), breast cancer (Sultan et al., 2018) and tuberous sclerosis complex (Serra et al., 2019). One suggested mechanism for CBD treatment of epilepsy and cancer is through the downregulation of hyperactivated mTORC1 found in the disease state (Serra et al., 2019;Sultan et al., 2018). In contrast, in the spinal cord neurons of a mouse model of MS, the mTORC1 pathway has been suggested to be downregulated after induction of the disease state (Giacoppo et al., 2017), where CBD elevates mTORC1 signalling and this could improve the progression of the disease due to the arrest of myelin loss. ...
Article
Full-text available
BACKGROUND AND PURPOSE: Cannabidiol (CBD) has been shown to differentially regulate the mechanistic target of rapamycin complex 1 (mTORC1) in preclinical models of disease, where it reduces activity in models of epilepsies and cancer and increases it in models of multiple sclerosis (MS) and psychosis. Here, we investigate the effects of phytocannabinoids on mTORC1 and define a molecular mechanism. EXPERIMENTAL APPROACH: A novel mechanism for phytocannabinoids was identified using the tractable model system, Dictyostelium discoideum. Using mouse embryonic fibroblasts, we further validate this new mechanism of action. We demonstrate clinical relevance using cells derived from healthy individuals and from people with MS (pwMS). KEY RESULTS: Both CBD and the more abundant cannabigerol (CBG) enhance mTORC1 activity in D. discoideum. We identify a mechanism for this effect involving inositol polyphosphate multikinase (IPMK), where elevated IPMK expression reverses the response to phytocannabinoids, decreasing mTORC1 activity upon treatment, providing new insight on phytocannabinoids' actions. We further validated this mechanism using mouse embryonic fibroblasts. Clinical relevance of this effect was shown in primary human peripheral blood mononuclear cells, where CBD and CBG treatment increased mTORC1 activity in cells derived from healthy individuals and decreased mTORC1 activity in cells derived from pwMS. CONCLUSION AND IMPLICATIONS: Our findings suggest that both CBD and the abundant CBG differentially regulate mTORC1 signalling through a mechanism dependent on the activity of the upstream IPMK signalling pathway, with potential relevance to the treatment of mTOR-related disorders, including MS.
... Various breast cancer cell lines have been used to demonstrate a dose-dependent response to CBD, including estrogen-receptor (ER)-positive cells, ER-negative cells, and triple-negative breast cancer (TNBC) cells (93)(94)(95)(96)(97)(98)(99). ...
... CBD exerts its anti-proliferative effects on breast cancer cells through a variety of mechanisms, including apoptosis, autophagy, and cell cycle arrest (93,94,98). More recent studies have found the anti-proliferative effects of CBD on breast cancer cells to be independent of the endocannabinoid receptors (98). ...
... In fact, this mechanism was already identified in other models, suggesting that this is a general mechanism of action [91]. The induction of apoptosis by CBD in MDA-MB-231 cells was also recently verified by Sultan et al., where CBD inhibited cell survival and induced apoptosis, favored by an interplay among PPAR, mTOR and cyclin D1 ( Figure 3A) [112]. In contrast, regarding the MDA-MB-231 cell line, a study showed that THC promotes the proliferation of these cells [113]. ...
... On the other hand, THC induced a similar effect on EVSA-T cells, though this effect was mediated by the CB2 receptor through CDK1 inhibition, and activation of the transcription factor JunD [129,130] (Figure 4B). However, there are studies on MCF-7 cells showing that THC is also able to promote cell proliferation, an effect that might involve Act1 and Erb2 [113,[131][132][133]. Regarding CBD, it was demonstrated in the T-47D cell line that this phytocannabinoid inhibits cell survival and promotes apoptosis due to an interplay among PPAR, mTOR and cyclin D1 [112] (Figure 3B), probably through a mechanism similar to the one observed in TNBC cases. Recently, it was also verified that CBD is able to induce endoplasmic reticulum stress, which leads to cell death, in MCF-7 cells through the activation or TRPV1 and the consequent increase in Ca 2+ and ROS levels ( Figure 3B) [134]. ...
Article
Full-text available
Although cannabinoids have been used for centuries for diverse pathological conditions, recently, their clinical interest and application have emerged due to their diverse pharmacological properties. Indeed, it is well established that cannabinoids exert important actions on multiple sclerosis, epilepsy and pain relief. Regarding cancer, cannabinoids were first introduced to manage chemotherapy-related side effects, though several studies demonstrated that they could modulate the proliferation and death of different cancer cells, as well as angiogenesis, making them attractive agents for cancer treatment. In relation to breast cancer, it has been suggested that estrogen receptor-negative (ER−) cells are more sensitive to cannabinoids than estrogen receptor-positive (ER+) cells. In fact, most of the studies regarding their effects on breast tumors have been conducted on triple-negative breast cancer (TNBC). Nonetheless, the number of studies on human epidermal growth factor receptor 2-positive (HER2+) and ER+ breast tumors has been rising in recent years. However, besides the optimistic results obtained thus far, there is still a long way to go to fully understand the role of these molecules. This review intends to help clarify the clinical potential of cannabinoids for each breast cancer subtype.
... CBD is a phytocannabinoid derived from the Cannabis sativa plant with well-documented analgesic, anti-inflammatory, and anxiolytic effects [12][13][14]. Unlike other cannabinoids, CBD does not typically bind to cannabinoid receptors 1 (CB1) and 2 (CB2) [15][16][17][18][19]. Rather, CBD targets receptors such as transient receptor potential cation channel subfamily V receptor 1 (TRPV1), G-protein coupled receptor 55 (GPR55), and peroxisome proliferator-activated receptor gamma (PPARγ). ...
... Unlike other cannabinoids, CBD does not typically bind to cannabinoid receptors 1 (CB1) and 2 (CB2) [15][16][17][18][19]. Rather, CBD targets receptors such as transient receptor potential cation channel subfamily V receptor 1 (TRPV1), G-protein coupled receptor 55 (GPR55), and peroxisome proliferator-activated receptor gamma (PPARγ). In human oncology studies, CBD has been shown to induce tumor cell apoptosis through these non-canonical receptors, among others [15][16][17][18][19]. Furthermore, CBD has been shown to induce apoptosis through an autophagy-apoptosis crosstalk pathway orchestrated by Beclin1. ...
Article
Full-text available
Background Canine urothelial carcinoma is the most common form of canine bladder cancer. Treatment with chemotherapy has variable response rates leading to most dogs succumbing to their disease within a year. Cannabidiol is an emerging treatment within the field of oncology. In reported in vivo studies, cannabidiol has induced apoptosis, reduced cell migration, and acted as a chemotherapy sensitizer in various human tumor types. The aim of this study was to characterize the effects of cannabidiol on canine urothelial carcinoma cell viability and apoptosis as both a single agent and in combination with chemotherapy in vitro . Results Cannabidiol reduced cell viability and induced apoptosis in canine urothelial cells as determined by crystal violet viability assay and annexin V/propidium iodide flow cytometry. Furthermore, combinations of cannabidiol with mitoxantrone and vinblastine chemotherapy yielded significantly reduced cell viability and increased apoptosis compared to single agent treatment alone. The drug interactions were deemed synergistic based on combination index calculations. Conversely, the combination of cannabidiol and carboplatin did not result in decreased cell viability and increased apoptosis compared to single agent treatment. Combination index calculations suggested an antagonistic interaction between these drugs. Finally, the combination of the non-steroidal anti-inflammatory drug piroxicam with cannabidiol did not significantly affect cell viability, although, some cell lines demonstrated decreased cell viability when mitoxantrone was combined with piroxicam. Conclusions Cannabidiol showed promising results as a single agent or in combination with mitoxantrone and vinblastine for treatment of canine urothelial carcinoma cells. Further studies are justified to investigate whether these results are translatable in vivo .
... Indeed, comparison with cisplatin treatment has shown that CBD exposure can repress melanoma tumor growth to a similar extent (Blázquez et al., 2006;Armstrong et al., 2015;Simmerman et al., 2018). In breast cancer, CBD can trigger cell apoptosis by promoting interactions with PPARγ, mTOR, and cyclin D1 (Sultan et al., 2018;Jeong et al., 2019c). Furthermore, the increased activities of the antioxidant enzymes SOD and GPX in CT26 colon cancer cells, accompanied by growth inhibition, may indicate that CBD operates via a novel mechanism (Honarman et al., 2019). ...
... Previous studies have demonstrated that CBD treatment could effectively induce cell apoptosis in tumor cells (Sultan et al., 2018;Alharris et al., 2019;Jeong et al., 2019c;Kis et al., 2019), but whether CBD could induce pyroptosis in tumor cells remains unreported. One type of programmed cell death (PCD), pyroptosis, may provide a possible beneficial effect on tumor cell therapy (Shi et al., 2015;Wang et al., 2017). ...
Article
Full-text available
Cannabidiol (CBD), a phytochemical derived from Cannabis sativa L., has been demonstrated to exhibit promising anti-tumor properties in multiple cancer types. However, the effects of CBD on hepatocellular carcinoma (HCC) cells remain unknown. We have shown that CBD effectively suppresses HCC cell growth in vivo and in vitro, and induced HCC cell pyroptosis in a caspase-3/GSDME-dependent manner. We further demonstrated that accumulation of integrative stress response (ISR) and mitochondrial stress may contribute to the initiation of pyroptotic signaling by CBD. Simultaneously, CBD can repress aerobic glycolysis through modulation of the ATF4–IGFBP1–Akt axis, due to the depletion of ATP and crucial intermediate metabolites. Collectively, these observations indicate that CBD could be considered as a potential compound for HCC therapy.
... In breast cancer, CBD-induced apoptosis is accompanied by down-regulation of the mammalian target of rapamycin (mTOR), which regulates cell proliferation and apoptosis, cell cycle, and localization of PPARγ in the nuclei and cytoplasmic of the tested cells [154]. ...
Article
Full-text available
Cannabidiol (CBD) is the primary non-psychoactive chemical from Cannabis Sativa, a plant used for centuries for both recreational and medicinal purposes. CBD lacks the psychotropic effects of Δ9-tetrahydrocannabinol (Δ9-THC) and has shown great therapeutic potential. CBD exerts a wide spectrum of effects at a molecular, cellular, and organ level, affecting inflammation, oxidative damage, cell survival, pain, vasodilation, and excitability, among others, modifying many physiological and pathophysiological processes. There is evidence that CBD may be effective in treating several human disorders, like anxiety, chronic pain, psychiatric pathologies, cardiovascular diseases, and even cancer. Multiple cellular and pre-clinical studies using animal models of disease and several human trials have shown that CBD has an overall safe profile. In this review article, we summarize the pharmacokinetics data, the putative mechanisms of action of CBD, and the physiological effects reported in pre-clinical studies to give a comprehensive list of the findings and major effects attributed to this compound.
... These compounds have been reported for their potent anticancer activities. Cannabidiol has been reported for its ability to inhibit proliferation [19], down-regulate Id-1 gene expression, mTOR and cyclin D1, upregulate PPARg protein expression [39,40], induce apoptosis and autophagy [41], and arrest cell cycle [42] in breast cancer. The antiproliferative effect of cinnamic acid on breast cancer has been attributed to their ability to inhibit 17β-hydroxysteroid dehydrogenase types1 and 5 activities [43,44], inhibit cell growth [45] and induce apoptosis [46]. ...
Article
Full-text available
The present study investigated the ability of Cannabis sativa leaves infusion (CSI) to modulate major metabolisms implicated in cancer cells survival, as well as to induce cell death in human breast cancer (MCF-7) cells. MCF-7 cell lines were treated with CSI for 48 h, doxorubicin served as the standard anticancer drug, while untreated MCF-7 cells served as the control. CSI caused 21.2% inhibition of cell growth at the highest dose. Liquid chromatography–mass spectroscopy (LC-MS) profiling of the control cells revealed the presence of carbohydrate, vitamins, oxidative, lipids, nucleotides, and amino acids metabolites. Treatment with CSI caused a 91% depletion of these metabolites, while concomitantly generating selenomethionine, l-cystine, deoxyadenosine triphosphate, cyclic AMP, selenocystathionine, inosine triphosphate, adenosine phosphosulfate, 5'-methylthioadenosine, uric acid, malonic semialdehyde, 2-methylguanosine, ganglioside GD2 and malonic acid. Metabolomics analysis via pathway enrichment of the metabolites revealed the activation of key metabolic pathways relevant to glucose, lipid, amino acid, vitamin, and nucleotide metabolisms. CSI caused a total inactivation of glucose, vitamin, and nucleotide metabolisms, while inactivating key lipid and amino acid metabolic pathways linked to cancer cell survival. Flow cytometry analysis revealed an induction of apoptosis and necrosis in MCF-7 cells treated with CSI. High-performance liquid chromatography (HPLC) analysis of CSI revealed the presence of cannabidiol, rutin, cinnamic acid, and ferulic. These results portray the antiproliferative potentials of CSI as an alternative therapy for the treatment and management of breast cancer as depicted by its modulation of glucose, lipid, amino acid, vitamin, and nucleotide metabolisms, while concomitantly inducing cell death in MCF-7 cells.
... These results suggest that the cytotoxic effect of cannabis oils is mainly due to the presence of CBD, flavonoids, and likely other phenolic compounds, which act together via the entourage effect. Previous studies have shown that CBD induces cell death in breast [66][67][68][69] and lung [70] cancer cell lines, whereas its parent molecule, cannabidiolic acid CBD-A, inhibits breast cancer cell migration via a mechanism that inhibits protein kinase [71]. It has also been demonstrated that some common flavonoids, such as quercetin and luteolin, reduce the cell proliferation of MCF-7 [59,72], and other specific flavonoids of C. sativa, cannflavins, show anticancer activities as well [62]. ...
Article
Full-text available
Currently, much effort is being placed into obtaining extracts and/or essential oils from Cannabis sativa L. for specific therapeutic purposes or pharmacological compositions. These potential applications depend mainly on the phytochemical composition of the oils, which in turn are determined by the type of C. sativa and the extraction method used to obtain the oils. In this work, we have evaluated the contents of secondary metabolites, delta-9-tetrahydrocannabinol (THC), and cannabidiol (CBD), in addition to the total phenolic, flavonoids, and anthraquinone content in oils obtained using solid–liquid extraction (SLE) and supercritical fluid extraction (SCF). Different varieties of C. sativa were chosen by using the ratio of THC to CBD concentrations. Additionally, antioxidant, antifungal and anticancer activities on different cancer cell lines were evaluated in vitro. The results indicate that oils extracted by SLE, with high contents of CBD, flavonoids, and phenolic compounds, exhibit a high antioxidant capacity and induce a high decrease in the cell viability of the tested breast cancer cell line (MCF-7). The observed biological activities are attributed to the entourage effect, in which CBD, phenols and flavonoids play a key role. Therefore, it is concluded that the right selection of C. sativa variety and the solvent for SLE extraction method could be used to obtain the optimal oil composition to develop a natural anticancer agent.
... 13,36,[131][132][133][134] Mechanistically, CBD regulates key signaling pathways implicated in cell survival, proliferation, invasion, angiogenesis, stemness, immune evasion, and metastasis. 13,117,[133][134][135][136] The majority of CBD's anticancer properties are explained by its capacity to control key cancer-driving genes and signaling pathways (Fig. 1). 118 CBD modulates ER stress, 137 intracellular ROS levels, 138 AKT/ mammalian target of rapamycin (mTOR) pathway, 20 vascular endothelial growth factor signaling, 134 prometastatic genes, 132 as well as the tumor microenvironment (e.g., regulation of mesenchymal and immune cell activity). ...
Article
Full-text available
Introduction: Cannabidiol (CBD) is the second most abundant Phytocannabinoid in Cannabis extracts. CBD has a binding affinity for several cannabinoid and cannabinoid-associated receptors. Epidiolex (oral CBD solution) has been lately licensed by the Food and Drug Administration (FDA) for the treatment of pediatric epileptic seizures. Methods: In this review, we discussed the most promising applications of CBD for chronic inflammatory conditions, namely CBD's anti-inflammatory effects during inflammatory bowel disease, coronavirus disease (antiviral effect), brain pathologies (neuroprotective and anti-inflammatory properties), as well as CBD immunomodulatory and antitumoral activities in the tumor microenvironment. Special focus was shed on the main therapeutic mechanisms of action of CBD, particularly in the control of the immune system and the endocannabinoid system. Results: Findings suggest that CBD is a potent immunomodulatory drug as it has manifested immunosuppressive properties in the context of sterile inflammation (e.g., inflammatory bowel disease, rheumatoid arthritis, and neurodegenerative diseases), and immunoprotective effects during viral infections (e.g. COVID-19) Similarly, CBD has exhibited a selective response toward cancer types by engaging different targets and signaling pathways. These results are in favor of the primary function of the endocannabinoid system which is homeostatic maintenance. Conclusion: The presented evidence suggests that the endocannabinoid system is a prominent target for the treatment of inflammatory and autoimmune diseases, rheumatoid diseases, viral infections, neurological and psychological pathologies, and cancer. Moreover, the antitumoral activities of CBD have been suggested to be potentially used in combination with chemo- or immunotherapy during cancer. However, clinical results are still lacking, which raises a challenge to apply translational cannabis research to the human immune system.
... Breast cancer (BC), one of the most common tumor types, is the second leading cause of cancer-related mortality among females (1)(2)(3). In China, BC cases account for 12.2% of all new BC diagnoses worldwide and 9.6% of all BC-related deaths (4). ...
Article
Full-text available
Heat shock protein (HSP) 20 belongs to the small HSP family and exhibits diverse functions, including tumor suppression, in addition to being a molecular chaperon, which is the classical property of HSPs. The present study aimed to examine the association between HSP20 expression and breast cancer (BC) progression in patients, and to explore the possible role of HSP20 in malignant phenotypes of BC cells. A series of experiments, including reverse transcription-quantitative PCR, western blotting, Cell Counting Kit-8 and flow cytometry, were performed. Data from Gene Expression Omnibus and Kaplan-Meier Plotter revealed that HSP20 expression was significantly downregulated in BC tissues, and patients with BC with lower HSP20 expression exhibited poorer recurrence-free survival. The data revealed that HSP20 was closely associated with the pathological tumor stage (P=0.015) and pathological tumor node metastasis (P=0.031) of patients with BC. Additionally, HSP20 expression was markedly decreased in BC cell lines. Exogenous overexpression of HSP20 inhibited proliferation and accelerated apoptosis of BC cells. These cells exhibited decreased migration and invasion when HSP20 was overexpressed. Furthermore, HSP20 overexpression suppressed the MAPK and AKT signaling pathways, as evidenced by the reduced phosphorylation levels of AKT, ERK, JNK and p38. Knockdown of HSP20 exerted the opposite effects. Notably, the AKT agonist, SC79, and the ERK agonist, LM22B-10, reversed the decrease in cell proliferation and migration induced by HSP20 overexpression. Overall, the data suggest that the decreased expression of HSP20 in BC tissues may be associated with disease progression. HSP20 also attenuated the malignant phenotype of BC cells and the inhibition of MAPK and AKT signaling may be associated with this effect. Therefore, HSP20 may be a potential prognostic marker or a candidate therapeutic target for BC.
... CBD could also be used as a candidate drug for the treatment of pain (Xiong et al., 2012;De Gregorio et al., 2019). In addition, the pharmacology values of CBD in substance abuse (Lee et al., 2017), anti-tumor (Sultan, 2018), and multiple sclerosis (Mecha et al., 2013) have been investigated in recent years. Another deeply researched topic is the molecular mechanism of CBD in these disorders, which may be related to the endocannabinoid system (ECS), serotonin system, transient receptor potential vanilloid (TRPV) channels, G protein-coupled receptor 55 (GPR55), peroxisome proliferator-activated receptor-gamma (PPARγ), and so on (Campos et al., 2012). ...
Article
Full-text available
Cannabidiol, a non-psychoactive component extracted from the plant cannabis sativa, has gained growing focus in recent years since its extensive pharmacology effects have been founded. The purpose of this study intends to reveal the hot spots and frontiers of cannabidiol research using bibliometrics and data visualization methods. A total of 3,555 publications with 106,793 citations from 2004 to 2021 related to cannabidiol were retrieved in the Web of Science database, and the co-authorships, research categories, keyword burst, and reference citations in the cannabidiol field were analyzed and visualized by VOSviewer and Citespace software. Great importance has been attached to the pharmacology or pharmacy values of cannabidiol, especially in the treatment of neuropsychiatric disorders, such as epilepsy, anxiety, and schizophrenia. The mechanisms or targets of the cannabidiol have attracted the extreme interest of the researchers, a variety of receptors including cannabinoids type 1, cannabinoids type 2, 5-hydroxytriptamine1A, and G protein-coupled receptor 55 were involved in the pharmacology effects of cannabidiol. Moreover, the latest developed topic has focused on the positive effects of cannabidiol on substance use disorders. In conclusion, this study reveals the development and transformation of knowledge structures and research hotspots in the cannabidiol field from a bibliometrics perspective, exploring the possible directions of future research.
... Researchers have focused on its role in treating cancers (Pisanti et al. 2017;Jo et al. 2021). Studies have found that CBD inhibits CRC progression by inducing cancer cell apoptosis (Sultan et al. 2018;Zhang et al. 2019). Some studies found that CBD inhibits cancer cell proliferation and migration in non-small cell lung cancer, prostate cancer, canine urothelial carcinoma cells, and bladder cancer (Milian et al. 2020;Pietrovito et al. 2020;Inkol et al. 2021;Chen et al. 2021). ...
Article
Full-text available
Colorectal cancer (CRC) is the leading cause of cancer deaths worldwide, wherein distant metastasis is the main reason for death. The non-psychoactive phytocannabinoid cannabidiol (CBD) effectively induces the apoptosis of CRC cells. We investigated the role of CBD in the migration and metastasis of CRC cells. CBD significantly inhibited proliferation, migration, and invasion of colon cancer cells in a dose- or time-dependent manner. CBD could also inhibit epithelial–mesenchymal transition (EMT) by upregulating epithelial markers such as E-cadherin and downregulating mesenchymal markers such as N-cadherin, Snail, Vimentin, and HIF-1α. CBD could suppress the activation of the Wnt/β-catenin signaling pathway, inhibit the expression of β-catenin target genes such as APC and CK1, and increase the expression of Axin1. Compared to the control group, the volume and weight of orthotopic xenograft tumors significantly decreased after the CBD treatment. The results demonstrated that CBD inhibits invasion and metastasis in CRC cells. This was the first study elucidating the underlying molecular mechanism of CBD in inhibiting EMT and metastasis via the Wnt/β-catenin signaling pathway in CRC cells. The molecular mechanism by which CBD inhibits EMT and metastasis of CRC cells was shown to be through the Wnt/β-catenin signaling pathway for the first time.
... High concentrations of CBD treatment may possibly show a greater inhibition of A549 cell growth [33]. However, CBD can also induce morphological changes in many types of cancer cells [34,35]. CBD-caused vesicle formation has also been seen in in vitro experimentation and has been suggested to be a mechanism of cell death via autophagy in MCF7 cells [36]. ...
Article
Full-text available
Extracts of phytocannabinoids from Cannabis sativa have been studied for therapeutic purposes. Although nonpsychoactive CBD has been studied as a promising anticancer drug because it induces apoptosis in many cancer cells, it is also known to induce several physiological changes. In this study, we clarify the functional role it plays in the morphological characteristics of intracellular vesicle formation as well as apoptosis in A549 human lung cancer cells. CBD treatment shows growth inhibition at concentrations above 20 μM, but FACS analysis shows low efficacy in terms of cell death. Microscopic observations suggest that multiple vesicles were detected in the cytoplasmic region of CBD-treated A549 cells. CBD treatment upregulates apoptosis-related proteins, such as p53, PARP, RIP1, RIP3, Atg12, and Beclin, indicating that CBD regulates several types of cell death. CBD treatment also induced E-cadherin, PPARγ, clathrin, β-adaptin, and Tsg101, also known to be cellular-differentiation inducers or vesicle-formation components. Treatment combining CBD with GW9662, a PPARγ inhibitor, reduced CBD-induced cytoplasmic vesicle formation. This indicates that PPARγ regulates the vesicle-formation mechanism. However, CBD-treated E-cad KO clones did not show this regulatory mechanism. These results elucidate the pharmacological and molecular networks associated with CBD in PPARγ-dependent vesicle formation and the induction of apoptosis.
... In some cells, such as cardiomyocytes and neurons, CBD and AEA protect against apoptosis [50][51][52]. However, in both immune and neoplastic cells, they induce apoptosis [34,53,54]. As was observed for chorionic cells (BeWo), AEA intensifies apoptosis by activating both initiator caspase 9 and executive caspases 3 and 7 [55]. ...
Article
Full-text available
Inflammation and apoptosis are regulated by similar factors, including ultraviolet B (UVB) radiation and cannabinoids, which are metabolized by cyclooxygenase-2 (COX-2) into pro-apoptotic prostaglandin derivatives. Thus, the aim of this study was to evaluate the impact of cyclooxygenase-2 inhibition by celecoxib on the apoptosis of keratinocytes modulated by UVB, anandamide (AEA) and cannabidiol (CBD). For this purpose, keratinocytes were non-treated/treated with celecoxib and/or with UVB and CBD and AEA. Apoptosis was evaluated using microscopy, gene expressions using quantitate reverse-transcriptase polymerase chain reaction; prostaglandins using liquid chromatography tandem mass spectrometry and cyclooxygenase activity using spectrophotometry. UVB enhances the percentage of apoptotic keratinocytes, which can be caused by the increased prostaglandin generation by cyclooxygenase-2, or/and induced cannabinoid receptor 1/2 (CB1/2) expression. AEA used alone intensifies apoptosis by affecting caspase expression, and in UVB-irradiated keratinocytes, cyclooxygenase-2 activity is increased, while CBD acts as a cytoprotective when used with or without UVB. After COX-2 inhibition, UVB-induced changes are partially ameliorated, when anandamide becomes an anti-apoptotic agent. It can be caused by observed reduced generation of anandamide pro-apoptotic derivative prostaglandin-ethanolamide by COX. Therefore, products of cyclooxygenase-dependent lipid metabolism seem to play an important role in the modulation of UVB-induced apoptosis by cannabinoids, which is particularly significant in case of AEA as inhibition of cyclooxygenase reduces the generation of pro-apoptotic lipid mediators and thus prevents apoptosis.
... Other synthetic CBR agonists also inhibit tumor growth and metastasis of BC. In one study, CBD was the most cytotoxic of all cannabinoids tested in ERþ and ER-cell lines [53][54][55][56]. Most recent studies have focused on the combination of THC and CBD, as these two compounds target different receptors and may act synergistically [57][58][59]. ...
Article
Full-text available
Breast cancer is the most common malignancy in women worldwide. Sixty-five percent of breast cancers are estrogen and/or progesterone receptor positive. Estrogen receptor expression is a prognostic and predictive biomarker of response to endocrine therapy, which consists of the selective estrogen receptor modulator tamoxifen, aromatase inhibitors, and the selective estrogen receptor degrader fulvestrant. Cannabidiol is a phytocannabinoid that is emerging as a potential therapeutic agent. The aim of this study was to investigate the effect of cannabidiol on estrogen receptor-positive and estrogen receptor-negative representative breast cancer cell lines in combination with standard therapeutic agents used in clinical practice. To compare the effects of cannabidiol on breast cancer cell viability, cancer cell lines were exposed to increasing concentrations of cannabidiol. The effects of cannabidiol in combination with the endocrine therapeutics tamoxifen, fulvestrant, and the cyclin-dependent kinase inhibitor palbociclib on breast cancer cell viability were examined. We demonstrated that cannabidiol dose-dependently decreased the viability of all breast cancer cell lines independent of estrogen expression. The addition of cannabidiol to tamoxifen had an additive negative effect on cell viability in ER + T-47D. Cannabidiol did not attenuate the effect of standard treatment of hormone receptor-positive breast cancer with fulvestrant and palbociclib. In addition, cannabidiol did not attenuate the effect of standard treatment of triple-negative breast cancer and HER2-positive breast cancer cell lines with trastuzumab and cisplatin.
... In 2018, Zhang et al. (2018a) reported that 20(S)-PPD could downregulate the protein expression of PI3K/AKT/mTOR in MCF-7 cells. In 2018, Sultan et al. (2018) reported that cannabidiol suppressed cell survival and prompted apoptosis in a dose-dependent manner, accompanied by downregulation of mTOR and upregulation and localization of PPARγ protein in the nuclei and cytoplasm. In 2018, another report by Hu et al. (2018) indicated that curcumin inhibited the phosphorylation of AKT and mTOR, decreased Bcl-2 and increased Bax, and cleaved caspase-3, subsequently inducing apoptosis in MCF-7 and MDA-MB-231 cells. ...
Article
Full-text available
Breast cancer is one of the top-ranked malignant carcinomas associated with morbidity and mortality in women worldwide. Chemotherapy is one of the main approaches to breast cancer treatment. Breast cancer initially responds to traditional first- and second-line drugs (aromatase inhibitor, tamoxifen, and carboplatin), but eventually acquires resistance, and certain patients relapse within 5 years. Chemotherapeutic drugs also have obvious toxic effects. In recent years, natural products have been widely used in breast cancer research because of their low side effects, low toxicity, and good efficacy based on their multitarget therapy. Apoptosis, a programmed cell death, occurs as a normal and controlled process that promotes cell growth and death. Inducing apoptosis is an important strategy to control excessive breast cancer cell proliferation. Accumulating evidence has revealed that natural products become increasingly important in breast cancer treatment by suppressing cell apoptosis. In this study, we reviewed current studies on natural product–induced breast cancer cell apoptosis and summarized the proapoptosis mechanisms including mitochondrial, FasL/Fas, PI3K/AKT, reactive oxygen species, and mitogen-activated protein kinase–mediated pathway. We hope that our review can provide direction in the search for candidate drugs derived from natural products to treat breast cancer by promoting cell apoptosis.
... Oxymatrine, which is extracted from conventional Chinese medicine Sophora flavescens, is an alkaloid with a variety of pharmacological effects. Studies have shown that oxymatrine has anti-inflammatory, anti-bacterial and anti-oxidant effects [14,15]. In addition, it has also revealed that oxymatrine can restrain the proliferation and induce apoptosis of various tumor cells, reverse multidrug resistance, and inhibit tumor angiogenesis, with the increase of drug concentration [18,19]. ...
Article
Objective: Oxymatrine has shown strong anti-cancer ability, but its mechanism is not well-studied. Methods: The inhibitory rates of oxymatrine with various concentrations (0, 1, 2, 4, 6, 8, 16, 32 mg/ml) on MCF-7 cells were detected by CCK-8. The effects of oxymatrine on the expression of miRNA-140-5P in MCF-7 cells were detected by real-time fluorescent quantitative PCR (RT-PCR). miRNA-140-5P mimics or NC mimics were transfected into cells using Lipofectamine 2000. Eventually, the cells were divided into control-group, drug-group, miRNA-140-5P mimics group, NC mimics group, and miRNA-140-5P mimics + drug group. Cell viability was detected by CCK-8 assay and apoptosis rate of each group were measured by using Flow cytometry. Western blot was carried out to detect the protein expression of TGFBR1 and FGF9. Results: Oxymatrine at various concentrations had conspicuous inhibitory effect on the proliferation of MCF-7 cells (P<0.05), and the inhibitory effect of oxymatrine on MCF-7 cells showed both dose- and time-dependent manners. The relative expression of miRNA-140-5P in MCF cells was remarkably lower than that in MCF-10A. Oxymatrine could effectively promote the expression of miRNA-140-5P in MCF-7 cells, and the relative expression of miRNA-140-5P increased significantly with the increased dose of oxymatrine (P<0.05). Both transfection of miRNA-140-5P mimics and oxymatrine treatment could reduce the proliferation of MCF-7 cells (P<0.05), and the proliferation of cells in miRNA-140-5P mimics + drug-group was significantly lower than that of other groups (P<0.05). Compared with the control-group, the protein expressions of TGFbR1 and FGF9 in low-dose, medium-dose and high-dose groups were dramatically decreased (P<0.05), in a dose-dependent manner (P<0.05). Conclusion: Oxymatrine inhibits proliferation and promotes cell apoptosis of breast cancer MCF-7 cells. The mechanism may contribute to the regulation of miRNA-140-5p and its target genes.
... Apoptosis linked to ER-stress was also reported in breast, prostate and in GBM [132][133][134]. Interestingly, in lung and breast cancers CBD stimulated apoptosis by direct and indirect regulation of PPAR-γ [135,136]. In breast cancer, CDB induced a crosstalk between apoptosis and autophagy modulating cancer cell death [137]. ...
Article
Full-text available
The tumour microenvironment (TME) is now recognised as a hallmark of cancer, since tumour:stroma crosstalk supports the key steps of tumour growth and progression. The dynamic co-evolution of the tumour and stromal compartments may alter the surrounding microenvironment, including the composition in metabolites and signalling mediators. A growing number of evidence reports the involvement of the endocannabinoid system (ECS) in cancer. ECS is composed by a complex network of ligands, receptors, and enzymes, which act in synergy and contribute to several physiological but also pathological processes. Several in vitro and in vivo evidence show that ECS deregulation in cancer cells affects proliferation, migration, invasion, apoptosis, and metastatic potential. Although it is still an evolving research, recent experimental evidence also suggests that ECS can modulate the functional behaviour of several components of the TME, above all the immune cells, endothelial cells and stromal components. However, the role of ECS in the tumour:stroma interplay remains unclear and research in this area is particularly intriguing. This review aims to shed light on the latest relevant findings of the tumour response to ECS modulation, encouraging a more in-depth analysis in this field. Novel discoveries could be promising for novel anti-tumour approaches, targeting the microenvironmental components and the supportive tumour:stroma crosstalk, thereby hindering tumour development.
... Several studies have shown that CBD induces apoptosis in cancer cell lines [17,29,[35][36][37]. Here, we show that CBD also induced apoptosis in the lung cancer spheres, enriched in cancer stem cells. ...
Article
Full-text available
Currently, there is no effective therapy against lung cancer due to the development of resistance. Resistance contributes to disease progression, recurrence, and mortality. The presence of so-called cancer stem cells could explain the ineffectiveness of conventional treatment, and the development of successful cancer treatment depends on the targeting also of cancer stem cells. Cannabidiol (CBD) is a cannabinoid with anti-tumor properties. However, the effects on cancer stem cells are not well understood. The effects of CBD were evaluated in spheres enriched in lung cancer stem cells and adherent lung cancer cells. We found that CBD decreased viability and induced cell death in both cell populations. Furthermore, we found that CBD activated the effector caspases 3/7, increased the expression of pro-apoptotic proteins, increased the levels of reactive oxygen species, as well as a leading to a loss of mitochondrial membrane potential in both populations. We also found that CBD decreased self-renewal, a hallmark of cancer stem cells. Overall, our results suggest that CBD is effective against the otherwise treatment-resistant cancer stem cells and joins a growing list of compounds effective against cancer stem cells. The effects and mechanisms of CBD in cancer stem cells should be further explored to find their Achilles heel.
... Therapeutic potential of cannabinoids (Δ9-THC, Δ8-THC and CBD) is well demonstrated in lung cancer, glioma, leukemia, neuroblastoma, skin, uterus, breast, gastric, colorectal, pancreatic, and prostate carcinomas 42 . CBD, a phytocannabinoid from Hemp (Cannabis sativa) induces autophagy, apoptosis 111 , cell cycle arrest and inhibits the migration, invasion, epithelial to mesenchymal transition (EMT) and angiogenesis of various tumors by acting on CB1 and CB2 receptors, and TRPV1 ion channels and regulating ERK, MAPK, PI3K, p38, AKT and ceramide signaling pathways 112,113 . CBD interacts with G-protein-coupled receptors (Gi/o) and regulates various cellular signaling pathways by inhibition of adenylyl cyclase 114 . ...
Article
Extracellular Vesicles (EVs) were isolated from human umbilical cord mesenchymal stem cells (hUCMSCs) and were further encapsulated with cannabidiol (CBD) through sonication method (CBD EVs). CBD EVs displayed an average particle size of 114.1±1.02 nm, zeta potential of -30.26±0.12 mV, entrapment efficiency of 92.3±2.21% and stability for several months at 4 °C. CBD release from the EVs was observed as 50.74±2.44% and 53.99±1.4% at pH 6.8 and pH 7.4, respectively after 48 h. Our in-vitro studies demonstrated that CBD either alone or in EVs form significantly sensitized MDA-MB-231 cells to doxorubicin (DOX) (*P<0.05). Flow cytometry and migration studies revealed that CBD EVs either alone or in combination with DOX induced G1 phase cell cycle arrest and decreased migration of MDA-MB-231 cells, respectively. CBD EVs and DOX combination significantly reduced tumor burden (***P<0.001) in MDA-MB-231 xenograft tumor model. Western blotting and immunocytochemical analysis demonstrated that CBD EVs and DOX combination decreased the expression of proteins involved in inflammation, metastasis and increased the expression of proteins involved in apoptosis. CBD EVs and DOX combination will have profound clinical significance in not only decreasing the side effects but also increasing the therapeutic efficacy of DOX in TNBC.
... VGCCs inhibition CBD inhibits human T-Type and L-type Ca 2+ channels in rat myocytes (40,41) man breast cancer cells by inhibiting the expression of cyclin D1 and mTOR pathway, and by increasing the peroxisome proliferator-activated (PPARγ) receptor expression (14). CBD is a PPARγ receptor agonist which inhibits the expression of pro-inflammatory genes and inflammatory signaling (15). ...
... Cannabidiol has an antiproliferative effect against aggressive subtype of BC cells, and also inhibits tumor growth in murine model by interfering in the recruitment of macrophages [83]. The anti-cancer effects of CBD on BC cells are related to regulatory effects on the biogenesis of exosomes and microvesicles released by cells and involved in intercellular communication [87] and by inducing apoptosis with down-regulation of mammalian target of rapamycin (mTOR) and cyclin D1 and up-regulation of peroxisome proliferator-activated receptor gamma (PPARγ) protein expression [88]. CBD also blocks and reverts the effect of IL-1β involved in the change to a malignant phenotype through epithelialmesenchymal transition [89]. ...
Chapter
Full-text available
The importance of a new anticancer drug for breast cancer is well established. Natural compounds that can prevent this disease or be used as an adjuvant treatment associated with conventional drugs could be the solution for this. This chapter is an overview of agents extracted from plants with outstand results in the last six years. Green tea, berberine, thymoquinone and cannabidiol are compounds isolated from medicinal plants. These agents showed action through induction of apoptosis, down regulation of inflammation, epigenetics, hormonal modulation, among other. In vitro effect against cancer cells, in vivo experiments mainly with murine model and clinical trials reassured their efficacy against breast cancer. A protective effect against recurrence cases and chemosensitization to standard drugs was also successful. The use of nanotechnology provided a optimize delivery of these therapeutical molecules. Taken together this information led us to acknowledgement that we do probably have the natural agents for a future adjuvant treatment against breast cancer.
... This, combined with CBD induced decrease in lymphocyte proliferation, also reduces the proinflammatory response in organisms [13][14][15]. For these reasons, CBD has been widely considered as a potential therapeutic compound for many disease states, including skin diseases such as psoriasis [16], as well as various types of cancer [17,18]. However, to use CBD safely and effectively, a comprehensive understanding of its effects on cell metabolism is necessary. ...
Article
Full-text available
Human epidermal keratinocytes are constantly exposed to UV radiation. As a result, there is a significant need for safe and effective compounds to protect skin cells against this environmental damage. This study aimed to analyze the effect of phytocannabinoid-cannabinoid (CBD)-on the proteome of UVA/B irradiated keratinocytes. The keratinocytes were cultured in a three-dimensional (3D) system, designed to mimic epidermal conditions closely. The obtained results indicate that CBD protected against the harmful effects of UVA/B radiation. CBD decreased the expression of proinflammatory proteins, including TNFα/NFκB and IκBKB complex and decreased the expression of proteins involved in de novo protein biosynthesis, which are increased in UVA/B-irradiated cells. Additionally, CBD enhanced the UV-induced expression of 20S proteasome subunits. CBD also protected protein structures from 4-hydroxynonenal (HNE)-binding induced by UV radiation, which primarily affects antioxidant enzymes. CBD-through its antioxidant/anti-inflammatory activity and regulation of protein biosynthesis and degradation-protects skin cells against UVA/B-induced changes. In the future, its long-term use in epidermal cells should be investigated.
... SK-BR3 cells were coated in a six-well plate at about a density of 1 Â 10 6 cells/well and were kept for incubation at a temperature of about 37 C in a humidified 5% CO 2 incubator for about 24 h [24]. The cells/wells were rinsed gently with sterile PBS and treated with the extracted compound sample at the concentration of 24.59 lg/mL in serum-free McCoy's 5A medium to induce apoptosis and incubated for 24 h at 37 C in a humidified 5% CO 2 incubator. ...
Article
Full-text available
The present research aimed to screen and evaluate the anticancer effect of Naringi crenulata phytocompounds on HER2+ human breast cancer cells. The cytotoxicity assay was performed to select effective solvent extraction. Extracted compounds showed lower toxicity on normal breast cells and significant cytotoxicity on human HER2+ breast cancer cells (SK-BR3) with an IC50 value of 24.59 μg/mL. The results indicate that the Naringi crenulata extract (NCE) shows anticancer potential via stimulating the cellular death of cancerous cells. The findings of the present study thus suggest that NCE supplementation might help in the treatment of HER2+ breast cancer.
Article
Full-text available
In this review article, we embark on a thorough exploration of cannabinoids, compounds that have garnered considerable attention for their potential therapeutic applications. Initially, this article delves into the fundamental background of cannabinoids, emphasizing the role of endogenous cannabinoids in the human body and outlining their significance in studying neurodegenerative diseases and cancer. Building on this foundation, this article categorizes cannabinoids into three main types: phytocannabinoids (plant-derived cannabinoids), endocannabinoids (naturally occurring in the body), and synthetic cannabinoids (laboratory-produced cannabinoids). The intricate mechanisms through which these compounds interact with cannabinoid receptors and signaling pathways are elucidated. A comprehensive overview of cannabinoid pharmacology follows, highlighting their absorption, distribution, metabolism, and excretion, as well as their pharmacokinetic and pharmacodynamic properties. Special emphasis is placed on the role of cannabinoids in neurodegenerative diseases, showcasing their potential benefits in conditions such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and multiple sclerosis. The potential antitumor properties of cannabinoids are also investigated, exploring their potential therapeutic applications in cancer treatment and the mechanisms underlying their anticancer effects. Clinical aspects are thoroughly discussed, from the viability of cannabinoids as therapeutic agents to current clinical trials, safety considerations, and the adverse effects observed. This review culminates in a discussion of promising future research avenues and the broader implications for cannabinoid-based therapies, concluding with a reflection on the immense potential of cannabinoids in modern medicine.
Article
Full-text available
According to World Health Organisation data, cancer is one of the most influential diseases that goes head to head with heart diseases in the ranking of causes of death, causing approximately 10 million deaths annually and accounting for 20% of all deaths. Lung cancer is a leading cause of cancer-related deaths globally for both men and women. In some pre-clinical studies in cancer cell line in vitro experiments, some results have been obtained that Aloe Vera Extract (AVE) obtained from Aloe vera plant and Cannabindiol (CBD) obtained from Cannabis sativa may have anticancer effect against cancer, but more analysis is required for the reliability of these results. In this study, the cytotoxic and apoptotic effects of AVE and CBD in human lung cancer (A549 and BEAS) cells were studied in a multifaceted manner. Diagnosis of lung cancer disease is approximately 15% in the early stage and 85% in the late stage or metastatic stage. Therefore, despite the use of targeted drugs today, the 5-year survival rate of patients can only be 5-10%. In this study, the cytotoxic and apoptotic effects of AVE and CBD on human lung cancer cells A549 and healthy normal BEAS-2B cells were studied in a versatile and comparative manner. Cytotoxic effect of AVE and CBD was evaluated by xCELLigence RTCA System and AnnexinV-FITC/PI Apoptosis Assay by Flow Cytometry. In the current investigation, we discovered that AVE and CBD might incite a relative higher ratio of cell death in lung cancer cells (A549) than in non-cancer human epithelial lung cell line BEAS-2B, which may be achieved through regulating mitochondrial metabolism. In summary, our research is under Patent submission and shows that CBD and AVE have the potential to develop into a potent new anti-lung cancer drug.
Chapter
The International Agency for Research on Cancer report on the global burden of cancer estimated 18.1 million new cancer cases (17.0 million excluding nonmelanoma skin cancers) and 9.6 million cancer deaths (9.5 million excluding nonmelanoma skin cancers) in 2018. In both sexes combined, lung cancer is the most commonly diagnosed cancer (11.6% of the total cases) and the leading cause of cancer death (18.4% of the total cancer deaths), closely followed by female breast cancer (11.6%), prostate cancer (7.1%), and colorectal cancer (6.1%) for incidence and colorectal cancer (9.2%), stomach cancer (8.2%), and liver cancer (8.2%) for mortality [33].
Article
Full-text available
It is known that gynecological cancers remain a worldwide problem and as shown by the statistics, there is a need for new gynecological cancer treatments. Cannabinoids, the pharmacologically active compounds of the Cannabis sativa plant, have been used for many centuries by individuals as a symptomatic treatment to alleviate pain, nausea, vomiting, and to help stimulate appetite. Research has revealed that cannabinoids also exert anti-cancer activity such as anti-proliferative and pro-apoptotic effects through a variety of mechanisms. There is significant value in the development of these compounds as anti-cancer therapies in clinical practice as they do not produce the typical toxic side effects that exist with conventional therapies and recent clinical trials have shown their great tolerability by patients at high doses. Cannabinoids can induce psychoactive effects that could limit their progression. Therefore, non-psychoactive cannabinoids are attracting pharmacological interest due to their inability to produce psychological effects. Recent studies have focussed on non-psychoactive cannabinoids in ovarian cancer and have revealed promising pre-clinical results that indicate that these compounds may have potential benefits in the treatment of these cancers. However, there are still unanswered questions and research gaps that need to be addressed. This review summarizes the current understanding of this topic and identifies the current gaps in knowledge that provide a useful direction for future work.
Article
Cannabidiol (CBD) is the non-psychoactive component of the plant Cannabis sativa (L.) that has great anti-inflammatory benefits and wound healing effects. However, its high lipophilicity, chemical instability, and extensive metabolism impair its bioavailability and clinical use. Here, we report on the preparation of a human cornea substitute in vitro and validate this substitute for the evaluation of drug penetration. CBD nanoemulsion was developed and evaluated for stability and biological activity. The physicochemical properties of CBD nanoemulsion were maintained during storage for 90 days under room conditions. In the scratch assay, nanoformulation showed significantly ameliorated wound closure rates compared to the control and pure CBD. Due to the lower cytotoxicity of nanoformulated CBD, a higher anti-inflammatory activity was demonstrated. Neither nanoemulsion nor pure CBD can penetrate the cornea after the four-hour apical treatment. For nanoemulsion, 94 % of the initial amount of CBD remained in the apical compartment while only 54% of the original amount of pure CBD was detected in the apical medium, and 7 % in the cornea, the rest was most likely metabolized. In summary, the nanoemulsion developed in this study enhanced the stability and biological activity of CBD.
Article
Background The growing usage of synthetic cannabinoids (SCs) has become a critical public health issue. Aside from skin toxicity and hepatotoxicity, SCs has been linked to several life-threatening adverse effects, including cardiovascular, gastrointestinal, neurological, renal, metabolic, ophthalmologic, and pulmonary consequences. This study aimed to evaluate the effects of SCs usage on oxidative stress, cell proliferation, and apoptosis in a group of SCs users by measuring the plasma levels of 4-hydroxy-2-nonenal (4-HNE), malondialdehyde (MDA), and glutathione peroxidase (GPX), as well as the expression of phosphoinositol-3-kinase (PI3K)/AKT and Caspase-9. Also, to find any association between the duration and frequency of SCs usage and the measured parameters. This study comprised 30 SCs users (aged 18–38 years) from Kasr Alainy Psychiatry and Addiction outpatient clinics, Cairo University, and 30 age-matched healthy males as a control group. Results The mean values of 4-HNE, MDA, and Caspase-9 were significantly higher (P ˂ 0.001) in the SCs users’ group than in the control group, while the mean value of GPX was significantly lower (P ˂ 0.001). The mean relative expression values of PI3K and AKT were significantly lower (P ˂ 0.001) in the SCs users’ group than in the control group. Significant correlations were detected between the duration and frequency of SCs use and oxidative stress, cell proliferation, and apoptosis biomarkers. Conclusion SCs can cause an increase in oxidative stress and a decrease in the expression of the proliferative pathways, consequently promoting apoptosis.
Article
Cannabinoids have been utilized for recreational and therapeutic purposes for over 4,000 years. As the primary ingredient in exogenous cannabinoids, Cannabidiol (CBD) has drawn a lot of interest from researchers due to its negligible psychotropic side effects and potential tumor-suppressing properties. However, the obscure mechanisms that underlie them remain a mystery. Complex biological mechanisms are involved in the progression of cancer, and malignancies have a variety of acquired biological capabilities, including sustained proliferation, death evasion, neovascularization, tissue invasion and metastasis, immune escape, metabolic reprogramming, induction of tumor-associated inflammation, cancerous stemness and genomic instability. Nowadays, the role of CBD hidden in these hallmarks is gradually revealed. Nevertheless, flaws or inconsistencies in the recent studies addressing the anti-cancer effects of CBD still exist. The purpose of this review is to evaluate the potential mechanisms underlying the role of CBD in a range of tumor-acquired biological capabilities. We propose potential drugs that may have a synergistic effect with CBD and provide optional directions for future research.
Article
Triple negative breast cancer (TNBC) represents an aggressive subtype of breast cancer, which is deficient in estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Thus, TNBC cells are unable to respond to the conventional hormonal therapies, making chemotherapy the only therapeutic choice. Patients with TNBC develop metastasis and recurrence over time and have reduced survival compared to patients with other subtypes of breast cancer. Therefore, there is a need for innovative therapies. Data emerged from pre-clinical studies, highlighted various antitumor activities of plant-derived Cannabis sativa and synthetic cannabinoids (CBs), including delta-9-tetrahydrocannabinol (THC) and non-psychoactive cannabidiol (CBD). On the contrary, some studies indicated that CBs might also promote tumor progression. At present, clinical studies on the effects of CBs from Cannabis sativa in cancer patients are few. In the present study, we reviewed known and possible interactions between cannabinoids and TNBC therapies.
Article
The significant resistance to currently available chemotherapeutics makes treatment for TNBC a key clinical concern. Herein, we studied the anti-cancer potentials of synthetic cannabidiol (CBD) and Tetrahydrocannabivarin (THCV) when used alone or in combination with doxorubicin (DOX) against MDA-MB-231 resistant cells. Pre-treatment with CBD and THCV significantly increased the cytotoxicity of DOX in MDA-MB-231 2D and 3D cultures that were DOX-resistant. Transcriptomics and Proteomics studies revealed that CBD and THCV, by downregulating PD-L1, TGF-β, sp1, NLRP3, P38-MAPK, and upregulating AMPK induced apoptosis leading to improved DOX's chemosensitivity against DOX resistant MDA-MB-231 tumors in BALB/c nude mice. CBD/THCV in combination with DOX significantly inhibited H3k4 methylation and H2K5 acetylation as demonstrated by western blotting and RT-PCR. Based on these findings, CBD and THCV appear to counteract histone modifications and their subsequent effects on DOX, resulting in chemo-sensitization against MDA-MB-231 resistant cancers.
Article
Abnormal energy metabolism, as one of the important hallmarks of cancer, was induced by multiple carcinogenic factors and tumor-specific microenvironments. It comprises aerobic glycolysis, de novo lipid biosynthesis, and glutamine-dependent anaplerosis. Considering that metabolic reprogramming provides various nutrients for tumor survival and development, it has been considered a potential target for cancer therapy. Cannabinoids have been shown to exhibit a variety of anticancer activities by unclear mechanisms. This paper first reviews the recent progress of related signaling pathways (reactive oxygen species (ROS), AMP-activated protein kinase (AMPK), mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K), hypoxia-inducible factor-1alpha (HIF-1α), and p53) mediating the reprogramming of cancer metabolism (including glucose metabolism, lipid metabolism, and amino acid metabolism). Then we comprehensively explore the latest discoveries and possible mechanisms of the anticancer effects of cannabinoids through the regulation of the above-mentioned related signaling pathways, to provide new targets and insights for cancer prevention and treatment.
Article
Full-text available
Cannabidiol (CBD) is a non-psychoactive phytocannabinoid that has been discussed for its safety and efficacy in cancer treatments. For this reason, we have inquired into its use on triple-negative human breast cancer. Analyzing the biological effects of CBD on MDA-MB-231, we have demonstrated that both CBD dosage and serum concentrations in the culture medium influence its outcomes; furthermore, light scattering studies demonstrated that serum impacts the CBD aggrega-tion state by acting as a surfactant agent. Pharmacological studies on CBD in combination with chemotherapeutic agents reveal that CBD possesses a protective action against the cytotoxic effect exerted by cisplatin on MDA-MB-231 grown in standard conditions. Furthermore, in a low serum condition (0.5%), starting from a threshold concentration (5 μM), CBD forms aggregates, exerts cy-tostatic antiproliferative outcomes, and promotes cell cycle arrest activating autophagy. At doses above the threshold, CBD exerts a highly cytotoxic effect inducing bubbling cell death. Finally, IGF-1 and EGF antagonize the antiproliferative effect of CBD protecting cells from harmful consequences of CBD aggregates. In conclusion, CBD effect is strongly associated with the physical state and concentration that reaches the treated cells, parameters not taken into account in most of the research papers.
Article
This systematic review examine the biological effects of CBD, a major component of therapeutic Cannabis, on human pathological and cancer cell populations of integumentary, gastro-intestinal, genital and breast, respiratory, nervous, haematopoietic and skeletal districts in terms of cell viability, proliferation, migration, apoptosis, inflammation, metastasis, and CBD receptor expression. The included studies were in English, on human cell lines and primary culture from non-healthy donors with CBD exposure as variable and no CBD exposure as control. Quality assessment was based on ToxRtool with a reliability score ranging from 15 to 18. Following the PRISMA statement 4 independent reviewers performed an electronic search using MEDLINE via PubMed, Scopus and Web of Science. From 3974 articles, 83 studies have been selected. Data showed conflicting results due to different concentration exposure, administrations and time points. CBD inhibited cell viability and proliferation in most cellular districts except the integumentary apparatus. Also a significant inhibition of migration was observed in all cell types, while an increase in apoptosis at both high and low doses (greater and less than 10 μM respectively). Considering inflammation, CBD caused an anti-inflammatory effect on nervous cells at low doses and on gastro-intestinal cells at high doses, while metastatic power was reduced even at low doses, but in a skeletal cell line there was an increased angiogenesis. CB1 receptor has been related to viability effects, CB2 to apoptosis and TRPV1 to inflammation and invasiveness. A detailed insight into these aspects would allow therapeutic use of this substance without possible side effects.
Article
Full-text available
Cannabidiol (CBD), a major non-psychotropic component of cannabis, is receiving growing attention as a potential anticancer agent. CBD suppresses the development of cancer in both in vitro (cancer cell culture) and in vivo (xenografts in immunodeficient mice) models. For critical evaluation of the advances of CBD on its path from laboratory research to practical application, in this review, we wish to call the attention of scientists and clinicians to the following issues: (a) the biological effects of CBD in cancer and healthy cells; (b) the anticancer effects of CBD in animal models and clinical case reports; (c) CBD’s interaction with conventional anticancer drugs; (d) CBD’s potential in palliative care for cancer patients; (e) CBD’s tolerability and reported side effects; (f) CBD delivery for anticancer treatment.
Article
To date, promising therapy for triple negative breast cancer (TNBC) remains a serious concern clinically because of poor prognosis, resistance, and recurrence. Herein, anti-cancer potential of synthetic cannabidiol (CBD; Purisys, GA; GMP grade) was explored either alone or as a chemosensitizer followed by post-treatment with doxorubicin (DOX) in TNBC (i.e., MDA-MB-231 and MDA-MB-468) cells. In comparison to 2D cultures, CBD showed greater IC50 values in 3D (LDP2 hydrogel based) cultures of MDA-MB-231 (6.26-fold higher) and MDA-MB-468 (10.22-fold higher) cells. Next-generation RNA sequencing revealed GADD45A, GADD45G, FASN, LOX, and integrin (i.e., -α5, -β5) genes to be novelly altered by CBD in MDA-MB-231 cells. CIM-16 plate-based migration assay and western blotting disclosed that CBD induces anti-migratory effects in TNBC cells by decreasing fibronectin, vimentin, and integrins-α5, -β5, and -β1. Western blotting, RT-qPCR, and immunocytochemistry revealed that CBD inhibited autophagy (decreased Beclin1, and ATG-5, -7, and -16) of TNBC cells. CBD pre-treatment increased DOX sensitivity in TNBC cells. CBD pre-treatment accompanied by DOX treatment decreased LOX and integrin-α5, and increased caspase 9 protein respectively in MDA-MB-468 cells.
Chapter
The endocannabinoid system plays a role in tumorigenesis and metastasis. The publication of reports on the regulation of bone remodeling by the classical cannabinoid receptor type 1 in 2005 and type 2 in 2006 kicked start research into the role of cannabinoid system in bone metabolism in health and disease. This chapter focuses on the complex role that cannabinoid ligands and receptors play in the regulation of bone–cancer cell cross talk, and reviews studies that tested the effects of endogenous, plant-derived, and synthetic cannabinoid ligands on bone metastasis, skeletal tumor burden, osteolysis, and bone pain in preclinical models of primary bone and metastatic cancers. Lastly, the chapter highlights the limitations of these studies and attempts to speculate on the potential medicinal and harmful effects associated with therapeutic targeting of the skeletal endocannabinoid system for the treatment of cancer-induced bone disease.
Article
Cannabinoids are the major chemical constituents of the plant Cannabis sativa L. and are known to exhibit a wide range of pharmacological effects viz., psychotropic, analgesic, anticancer, antiinflammatory, antidiabetic, anticonvulsive, antibacterial and antifungal etc. The use of cannabis, cannabinoids and their products is restricted in several countries due to the high risk of misuse. Recently, cannabinoids have regained the interest of the researchers due to their therapeutic applications. Ever since the discovery of the cannabinoids, most of the studies carried out on the evaluation of their biological activities were limited to only preclinical levels. The quality of the preclinical data still remains only low to moderate, thus, leaving behind an uncertainty in their use for therapeutic applications. Problems associated with the solubility, stability and bioavailability of the cannabinoid drugs are also a major concern in the quality of the study. Nanoparticle based drug delivery system could be a potential method to increase the reliability of the data. While considering the immense pharmacological properties of the cannabinoids, there is an urgency to perform intensive clinical trials and to know their mechanism of action in various disease conditions, evaluate their efficacy and safety, and register them as drug candidates. This review highlights the chemistry, types and biological activities of the cannabinoids such as THC, CBD and CBN in focus with their anticancer activity, neuroprotective effect and nanoformulating the cannabinoid drugs.
Preprint
Full-text available
Multidrug-resistant (MDR) and extensively drug-resistant (XDR) bacteria are a major worldwide public health problem. In the last decades, resistance to last-resort antibiotics such as polymyxin B (PB) have been increasingly observed among these superbugs, compromising the effectiveness of antimicrobial therapy. The present study aimed (i) to assess the ultrapure Cannabidiol (CBD) antibacterial activity against a broad diversity of Gram-negative (GN) and Gram-positive (GP) bacteria (44 different species, 95 strains), comprising standard strains and clinical isolates, and (ii) to investigate the antibacterial activity of the combination CBD + PB against GN bacteria, including chromosomal- and plasmid-acquired PB-resistant and intrinsically PB-resistant GNB. We evaluated CBD in vitro antibacterial activity using the standard broth microdilution method, and the antibacterial activity of the combination CBD + PB was screened using the standard broth microdilution and confirmed by checkerboard assay. CBD exhibited antibacterial activity against different GP bacterial species, lipooligosaccharide (LOS)-expressing GN diplococcus (GND) ( Neisseria gonorrhoeae , Neisseria meningitidis , and Moraxella catarrhalis ), and Mycobacterium tuberculosis . The combination CBD + PB exhibited antibacterial activity against PB-resistant GNB (e.g., Klebsiella pneumoniae ) as well as additive and/or synergistic effect against LOS-expressing GND. The antibacterial activity of the combination CBD + PB against Pseudomonas aeruginosa and plasmid-mediated colistin-resistant (MCR-1) E. coli strains could be only demonstrated in the presence of phenylalanine-arginine-β-naphthylamide (PAβN). In conclusion, our results show promising translational potential of the combination CBD + PB against MDR and XDR GNB, including PB-resistant K. pneumoniae , highlighting its potential as a rescue treatment for life-threatening infections caused by these superbugs. One Sentence Summary Promising combination of cannabidiol (CBD) + polymyxin B (PB) against superbugs (e.g., PB-resistant Gram-negative bacilli): Repurposing CBD
Thesis
Full-text available
In Colombia, breast cancer has been the second most diagnosed &the fifth with the highest mortality. Cisplatin is one of the most recommended drugs for its treatment and, although a wide of its biology is currently known &there are equipment for its early detection &treatment available, the emergence of chemoresistance is a problem that is taking increasingly importance, making necessary to implement a model to achieve a standardized protocol that allows evaluating alternative treatments with promising bioactive molecules such as CBD, in chemoresistant tumors. In the present work, obtaining a cisplatin resistant MCF-7 cell line was standardized using 2 different methods, its viability was compared with that of the parental line, against exposure to cisplatin &CBD by means of the MTT assay. Morphological changes in cells adapted to cisplatin were found: increase in cell size, increased amount of cytoplasmic granules &irregularities in the cell membrane. There was a significant difference in the proliferation of the parental line &the resistant line obtained here after exposure to cisplatin, as well as a differential response after 48 hours of exposure to 100 µM of CBD. In conclusion, the procedure here reported is effective in obtaining a line with resistance characteristics. This requires maintaining the culture with a confluence greater than 90%, limiting the maximum exposure to 3 µg / ml cisplatin. https://ciencia.lasalle.edu.co/biologia/83/
Article
Introduction Iris taochia is an endemic plant in Turkey. Iris species has many biological effects such as antibacterial, antiinflammatory, antioxidant and anticancer properties. Apoptosis is a programmed cell death and this mechanism regulates the death of cancer cells. Purpose The aim of our work is to investigate how the Iris taochia extracts affect the apoptotic activity in the MCF7 cells. Methods Cytotoxic dose and cell viability is determined by the MTT assay. Bad, Bax, Bcl-2, Bcl-W, Bid, Bim, Caspase 3, Caspase 8, CD40, CD40L, cIAP-2, CytoC, DR6, Fas, FasL, HSP27, HSP60, HSP70, HTRA, IGF-I, IGF-II, IGFBP-1, IGFBP-2, IGFBP-3, IGFBP-4, IGFBP-5, IGFBP-6, IGF-1sR, Livin, p21, p27, p53, SMAC, Survivin, sTNF-R1, sTNF-R2, TNF-α, TNF-β, TRAILR-1, TRAILR-2, TRAILR-3, TRAILR-4 and XIAP proteins were measured by the membrane array kit. Results Iris taochia extracts exhibited significant cytotoxic effects on MCF7 cells and IC50 values ranging from 1.56 to 100 μg/mL. Our results indicate that MeOH extract of Iris taochia in MCF7 cells may be a regulator of cell death proteins, cell cycle and growth factors. DCM and EtOH extracts of Iris taochia have a limited effect on MCF7 cells. Especially, HSPs, which play a significant role in chemoresistance downregulating DCM and EtOH extracts of Iris taochia, whereas ligands and receptors of extrinsic apoptotic pathway are upregulated by these extracts. Conclusion This is the first study to investigate the cytotoxic and apoptotic effect of Iris taochia extracts on MCF7 cells. Results also showed that Iris taochia reduced cell viability and induced apoptotic pathways as a potential regulator of cancer cell death.
Article
Breast cancer is the leading cause of cancer-related death in women worldwide. In the last years, cannabinoids have gained attention in the clinical setting and clinical trials with cannabinoid-based preparations are underway. However, contradictory anti-tumour properties have also been reported. Thus, the elucidation of the molecular mechanisms behind their anti-tumour efficacy is crucial to better understand its therapeutic potential. Considering this, our work aims to clarify the molecular mechanisms underlying the anti-cancer properties of the endocannabinoid anandamide (AEA) and of the phytocannabinoids, cannabidiol (CBD) and Δ9-tetrahydrocannabinol (THC), in estrogen receptor-positive (ER+) breast cancer cells that overexpress aromatase (MCF-7aro). Their in vitro effects on cell proliferation, cell death and activity/expression of aromatase, ERα, ERβ and AR were investigated. Our results demonstrated that cannabinoids disrupted MCF-7aro cell cycle progression. Unlike AEA and THC that induced apoptosis, CBD triggered autophagy to promote apoptotic cell death. Interestingly, all cannabinoids reduced aromatase and ERα expression levels in cells. On the other hand, AEA and CBD not only exhibited high anti-aromatase activity but also induced up-regulation of ERβ. Therefore, all cannabinoids, albeit by different actions, target aromatase and ERs, impairing, in that way, the growth of ER+ breast cancer cells, which is dependent on estrogen signalling. As aromatase and ERs are key targets for ER+ breast cancer treatment, cannabinoids can be considered as potential and attractive therapeutic compounds for this type of cancer, being CBD the most promising one. Thus, from an in vitro perspective, this work may contribute to the growing mass of evidence of cannabinoids and cannabinoids-based medicines as potential anti-cancer drugs.
Article
Full-text available
Background: A gradual decline in the incidence of breast cancer is documented in developed countries especially over last two decades, while in developing countries the incidence continues to rise. We conducted this study to examine trends in incidence of breast cancer in a developing country, Sri Lanka. Methods: A retrospective cohort evaluation of patients with breast cancer during 2001-2010 was performed using population based data from the Sri Lanka National Cancer Registry. Trends in incidence were analysed using Joinpoint regression analysis. Results: The age standardized incidence of female breast cancer in Sri Lanka appears to have increased from 17.3 per 100,000 in 2001 (95% confidence interval [95% CI] 16.5-18.2) to 24.7 per 100,000 in 2010 (95% CI 23.7-25.7); a 1.4-fold increase (p < 0.05) with an estimated annual percentage change (EAPC) of 4.4 (95% CI 3.3-5.5). Highest incidence of breast cancer was seen among women of 60 to 64-year age group which has increased from 68.1 to 100.2 per 100,000 over this period (EAPC 4.6%, 95% CI 3.9-5.2, p < 0.001 for trend). A substantially greater increase was observed among women older than 50 years (from 50.4 to 76.9 per 100,000; EAPC 5.5, 95% CI 4.1-7.0, p < 0.05) compared with women younger than 50 years (from 32.0 to 39.6 per 100,000; EAPC 2.3, 95% CI 1.1-3.5, p < 0.05). Conclusions: A gradual but a significant increase in the incidence of female breast cancer is observed in Sri Lanka. A rapid rise in the breast cancer incidence among post-menopausal women appears to be the major contributor towards this increase. Improving cancer data collection appears to have been a contributor to the observed increase. However, an inherent increase is also likely as differential rates of increase were observed by age groups. Further research is needed to identify the reasons for the observed increase which may help with future cancer control efforts in Sri Lanka.
Article
Full-text available
Abstract Background Breast cancer is a heterogeneous disease with several morphological and molecular subtypes. Widely accepted molecular classification system uses assessment of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and proliferation marker Ki67. Few studies have been conducted on the incidence and molecular types of breast cancer in Sub-Saharan Africa. Previous studies mainly from Western and Central Africa, showed breast cancer to occur at younger ages and to present with aggressive features, such as high-grade, advanced stage and triple-negative phenotype (negative for ER, PR and HER2). Limited data from East Africa including Ethiopia however shows hormone receptor negative tumors to account for a lower proportion of all breast cancers than has been reported from elsewhere in Africa. Methods In this study from Tikur Anbessa Specialized Hospital, 114 breast cancer patients diagnosed between 2012 and 2015 were enrolled. ER, PR, Ki67 and HER2 receptor status were assessed using immunohistochemistry from tissue microarrays. FISH was used for assessment of gene amplification in all equivocal tumor samples and for confirmation in HER2-enriched cases. Results The distribution of molecular subtypes was: Luminal A: 40%; Luminal B: 26%; HER2-enriched: 10%; TNBC: 23%. ER were positive in 65% of all tumors and 43% the cases were positive for PR. There was statistically significant difference in median age at diagnosis between the molecular subtypes (P
Article
Full-text available
Breast cancer is the leading cause of cancer-related deaths in women worldwide. GLOBOCAN estimated about 1.7 million new cases of breast cancer diagnoses worldwide and about 522,000 deaths in 2012. The burden of breast cancer mortality lies in the developing low-income and middle-income countries, where about 70% of such deaths occur. The incidence of breast cancer is also rising in low-income and middle-income countries in Africa as trend towards urbanization, and adoption of Western lifestyles increases. In general, the triple-negative breast cancer (TNBC) subtype tends to be frequent in women of African ancestry. What are the factors contributing to this prevalence? Are there genetic predispositions to TNBC in African women? This review addresses these questions and provides an update on the incidence, survival, and mortality of breast cancer in Africans, with a focus on sub-Saharan Africans. We have also addressed factors that could account for ethical disparities in incidence and mortality. Further, we have highlighted challenges associated with access to essential drug and to healthcare treatment in some African countries and outlined alternative/herbal treatment methods that are increasingly implemented in Africa and other developing nations.
Article
Full-text available
Targeting mTORC1 has been thoroughly explored in cancer therapy. Following encouraging preclinical studies, mTORC1 inhibitors however failed to provide substantial benefits in cancer patients. Several resistance mechanisms have been identified including mutations of mTOR and activation of alternate proliferation pathways. Moreover, emerging evidence discloses intratumoral heterogeneity of mTORC1 activity that further contributes to a reduced anticancer efficacy of mTORC1 inhibitors. Genetic heterogeneity as well as heterogeneous conditions of the tumor environment such as hypoxia profoundly modifies mTORC1 activity in tumors and hence influences the response of tumors to mTORC1 inhibitors. Intriguingly, the heterogeneity of mTORC1 activity also occurs towards its substrates at the single cell level, as mutually exclusive pattern of activation of mTORC1 downstream effectors has been reported in tumors. After briefly describing mTORC1 biology and the use of mTORC1 inhibitors in patients, this review will give an overview on concepts of resistance to mTORC1 inhibition in cancer with a particular focus on intratumoral heterogeneity of mTORC1 activity.
Article
Full-text available
"Cannabinoid" is the collective term for a group of chemical compounds that either are derived from the Cannabis plant, are synthetic analogues, or occur endogenously. Although cannabinoids interact mostly at the level of the currently recognized cannabinoid receptors, they might have cross reactivity, such as at opioid receptors. Patients with malignant disease represent a cohort within health care that have some of the greatest unmet needs despite the availability of a plethora of guideline-driven disease-modulating treatments and pain and symptom management options. Cannabinoid therapies are varied and versatile, and can be offered as pharmaceuticals (nabilone, dronabinol, and nabiximols), dried botanical material, and edible organic oils infused with cannabis extracts. Cannabinoid therapy regimens can be creative, involving combinations of all of the aforementioned modalities. Patients with malignant disease, at all points of their disease trajectory, could be candidates for cannabinoid therapies whether as monotherapies or as adjuvants. The most studied and established roles for cannabinoid therapies include pain, chemotherapy-induced nausea and vomiting, and anorexia. Moreover, given their breadth of activity, cannabinoids could be used to concurrently optimize the management of multiple symptoms, thereby reducing overall polypharmacy. The use of cannabinoid therapies could be effective in improving quality of life and possibly modifying malignancy by virtue of direct effects and in improving compliance or adherence with disease-modulating treatments such as chemotherapy and radiation therapy.
Article
Full-text available
Approximately 70% of patients have breast cancers that are oestrogen receptor alpha positive (ER+) and are therefore candidates for endocrine treatment. Many of these patients relapse in the years during or following completion of adjuvant endocrine therapy. Thus, many ER+ cancers have primary resistance or develop resistance to endocrine therapy during treatment. Recent improvements in our understanding of how tumours evolve during treatment with endocrine agents have identified both changes in gene expression and mutational profiles, in the primary cancer as well as in circulating tumour cells. Analysing these changes has the potential to improve the prediction of which specific patients will respond to endocrine treatment. Serially profiled biopsies during treatment in the neoadjuvant setting offer promise for accurate and early prediction of response to both current and novel drugs and allow investigation of mechanisms of resistance. In addition, recent advances in monitoring tumour evolution through non-invasive (liquid) sampling of circulating tumour cells and cell-free tumour DNA may provide a method to detect resistant clones and allow implementation of personalized treatments for metastatic breast cancer patients. This review summarises current and future biomarkers and signatures for predicting response to endocrine treatment, and discusses the potential for using approved drugs and novel agents to improve outcomes. Increased prediction accuracy is likely to require sequential sampling, utilising preoperative or neoadjuvant treatment and/or liquid biopsies and an improved understanding of both the dynamics and heterogeneity of breast cancer.
Article
Full-text available
Tumor heterogeneity of triple-negative breast cancer (TNBC) has been the main barrier in conquering breast cancer. To dissect the molecular diversity of TNBC and discover therapeutic targets for TNBC, the molecular classification of TNBC is a prioritized issue in research area. Accordingly, recent studies have been successful in classifying TNBC into several distinct subtypes with specific biologic pathways. Despite the different methodologies used and varied number of final subtypes, these studies identically suggested that TNBC consists of four major subtypes: basal like, mesenchymal, lumina! androgen receptor, and immune-enriched. By reviewing these methods of classifications of TNBC, we highlight the unmet need to develop a molecular classifier suited for TNBC.
Article
Full-text available
In addition to the well-known palliative effects of cannabinoids on some cancer-associated symptoms, a large body of evidence shows that these molecules can decrease tumour growth in animal models of cancer. They do so by modulating key cell signalling pathways involved in the control of cancer cell proliferation and survival. In addition, cannabinoids inhibit angiogenesis and decrease metastasis in various tumour types in laboratory animals. In this review, we discuss the current understanding of cannabinoids as antitumour agents, focusing on recent discoveries about their molecular mechanisms of action, including resistance mechanisms and opportunities for their use in combination therapy. Those observations have already contributed to the foundation for the development of the first clinical studies that will analyze the safety and potential clinical benefit of cannabinoids as anticancer agents.
Article
Full-text available
Breast cancer is composed of multiple subtypes with distinct morphologies and clinical implications. The advent of microarrays has led to a new paradigm in deciphering breast cancer heterogeneity, based on which the intrinsic subtyping system using prognostic multigene classifiers was developed. Subtypes identified using different gene panels, though overlap to a great extent, do not completely converge, and the avail of new information and perspectives has led to the emergence of novel subtypes, which complicate our understanding towards breast tumor heterogeneity. This review explores and summarizes the existing intrinsic subtypes, patient clinical features and management, commercial signature panels, as well as various information used for tumor classification. Two trends are pointed out in the end on breast cancer subtyping, i.e., either diverging to more refined groups or converging to the major subtypes. This review improves our understandings towards breast cancer intrinsic classification, current status on clinical application, and future trends.
Article
Full-text available
Rapamycin is an effective anti-angiogenic drug. However, the mode of its action remains unclear. Therefore, in this study, we aimed to elucidate the antitumor mechanism of rapamycin, hypothetically via apoptotic promotion, using MCF-7 breast cancer cells. MCF-7 cells were plated at a density of 15105 cells/well in 6-well plates. After 24h, cells were treated with a series of concentrations of rapamycin while only adding DMEM medium with PEG for the control regiment and grown at 37oC, 5% CO2 and 95% air for 72h. Trypan blue was used to determine the cell viability and proliferation. Untreated and rapamycin-treated MCF-7 cells were also examined for morphological changes with an inverted-phase contrast microscope. Alteration in cell morphology was ascertained, along with a stage in the cell cycle and proliferation. In addition, cytotoxicity testing was performed using normal mouse breast mammary pads. Our results clearly showed that rapamycin exhibited inhibitory activity on MCF-7 cell lines. The IC50 value of rapamycin on the MCF-7 cells was determined as 0.4μg/ml (p<0.05). Direct observation by inverted microscopy demonstrated that the MCF-7 cells treated with rapamycin showed characteristic features of apoptosis including cell shrinkage, vascularization and autophagy. Cells underwent early apoptosis up to 24% after 72h. Analysis of the cell cycle showed an increase in the G0G1 phase cell population and a corresponding decrease in the S and G2M phase populations, from 81.5% to 91.3% and 17.3% to 7.9%, respectively. This study demonstrated that rapamycin may potentially act as an anti-cancer agent via the inhibition of growth with some morphological changes of the MCF-7 cancer cells, arrest cell cycle progression at G0/G1 phase and induction of apoptosis in late stage of apoptosis. Further studies are needed to further characterize the mode of action of rapamycin as an anti-cancer agent.
Article
Full-text available
Genes encoding components of the PI3K–AKT–mTOR signaling axis are frequently mutated in cancer, but few mutations have been characterized in MTOR, the gene encoding the mTOR kinase. Using publicly available tumor genome sequencing data, we generated a comprehensive catalog of mTOR pathway mutations in cancer, identifying 33 MTOR mutations that confer pathway hyperactivation. The mutations cluster in six distinct regions in the C-terminal half of mTOR and occur in multiple cancer types, with one cluster particularly prominent in kidney cancer. The activating mutations do not affect mTOR complex assembly, but a subset reduces binding to the mTOR inhibitor DEPTOR. mTOR complex 1 (mTORC1) signaling in cells expressing various activating mutations remains sensitive to pharmacologic mTOR inhibition, but is partially resistant to nutrient deprivation. Finally, cancer cell lines with hyperactivating MTOR mutations display heightened sensitivity to rapamycin both in culture and in vivo xenografts, suggesting that such mutations confer mTOR pathway dependency. Significance: We report that a diverse set of cancer-associated MTOR mutations confer increased mTORC1/2 pathway activity and that cells harboring these mutations are highly sensitive to rapamycin in culture and in vivo. These findings are clinically relevant as the MTOR mutations characterized herein may serve as biomarkers for predicting tumor responses to mTOR inhibitors.
Article
Full-text available
The aim of our study was to investigate HER-2 and TOP2A gene status and their correlation with Bcl-2, p53, Ki67, ssDNA, and clinicopathological parameters in four molecular subtypes of breast cancer. Seventy-four paraffin-embedded samples are immunohistochemically studied for the expression of estrogen receptor (ER), progesterone receptor (PR), HER-2, p53, Bcl-2, ssDNA, and Ki67, while HER-2 and TOP2A gene status by fluorescence in situ hybridization was investigated in 60 samples. Luminal A and B subtypes were characterized with small tumor size, intermediate histological grade, negative lymph node, and metastatic status, while triple negative and HER-2 positive subtypes were associated with larger tumor size, poorly differentiated tumors, and positive lymph node status. p53, Ki67, and ssDNA expression was higher in triple negative and HER-2 positive than in luminal subtypes, while ER, PR, and Bcl-2 dominated in luminal subtypes. HER-2 gene status was higher in luminal B and HER-2 positive than in luminal A and triple negative subtypes, while TOP2A gene status was similar. HER-2 gene status positively correlated with TOP2A gene status, HER-2 receptor, and histological grade, while negative correlation characterized relationship between HER-2 gene status and ER, PR, and Bcl-2. The shortened overall survival period characterized patients from triple negative breast cancer subtype (18.7 months). HER-2 and TOP2A gene amplification showed a tendency to be associated with larger tumor size, positive lymph node status, high level of apoptotic and proliferative indexes, and low level of p53 and Bcl-2 expression, which all together indicate group of patients with similar outcome during the progression of the disease.
Article
Full-text available
Induction of differentiation and apoptosis in cancer cells through ligands of nuclear hormone receptors (NHRs) is a novel and promising approach to cancer therapy. All-trans-retinoic acid (ATRA), an RA receptor-specific NHR ligand, is now used for selective cancers. The NHR, peroxisome proliferator-activated receptor γ (PPARγ) is expressed in breast cancer cells. Activation of PPARγ through a synthetic ligand, troglitazone (TGZ), and other PPARγ-activators cause inhibition of proliferation and lipid accumulation in cultured breast cancer cells. TGZ (10−5 M, 4 days) reversibly inhibits clonal growth of MCF7 breast cancer cells and the combination of TGZ (10−5 M) and ATRA (10−6 M, 4 days) synergistically and irreversibly inhibits growth and induces apoptosis of MCF7 cells, associated with a dramatic decrease of their bcl-2 protein levels. Similar effects are noted with in vitro cultured breast cancer tissues from patients, but not with normal breast epithelial cells. The observed apoptosis mediated by TGZ and ATRA may be related to the striking down-regulation of bcl-2, because forced over-expression of bcl-2 in MCF7 cells cultured with TGZ and ATRA blocks their cell death. TGZ significantly inhibits MCF7 tumor growth in triple immunodeficient mice. Combined administration of TGZ and ATRA causes prominent apoptosis and fibrosis of these tumors without toxic effects on the mice. Taken together, this combination may provide a novel, nontoxic and selective therapy for human breast cancers.
Article
Full-text available
mTOR, the mammalian target of rapamycin, has been widely implicated in signals that promote cell cycle progression and survival in cancer cells. Rapamycin, which inhibits mTOR with high specificity, has consequently attracted much attention as an anti-cancer therapeutic. Rapamycin suppresses phosphorylation of S6 kinase at nano-molar concentrations, however at higher micro-molar doses, rapamycin induces apoptosis in several human cancer cell lines. While much is known about the effect of low dose rapamycin treatment, the mechanistic basis for the apoptotic effects of high-dose rapamycin treatment is not understood. We report here that the apoptotic effects of high-dose rapamycin treatment correlate with suppressing phosphorylation of the mTOR complex 1 substrate, eukaryotic initiation factor 4E (eIF4E) binding protein-1 (4E-BP1). Consistent with this observation, ablation of eIF4E also resulted in apoptorsis in MDA-MB 231 breast cancer cells. We also provide evidence that the differential dose effects of rapamycin are correlated with partial and complete dissociation of Raptor from mTORC1 at low and high doses, respectively. In contrast with MDA-MB-231 cells, MCF-7 breast cancer cells survived rapamycin-induced suppression of 4E-BP1 phosphorylation. We show that survival correlated with a hyper-phosphorylation of Akt at S473 at high rapamycin doses, the suppression of which conferred rapamycin sensitivity. This study reveals that the apoptotic effect of rapamycin requires doses that completely dissociate Raptor from mTORC1 and suppress that phosphorylation of 4E-BP1 and inhibit eIF4E.
Article
Full-text available
Cannabidiol (CBD), a major nonpsychoactive constituent of cannabis, is considered an antineoplastic agent on the basis of its in vitro and in vivo activity against tumor cells. However, the exact molecular mechanism through which CBD mediates this activity is yet to be elucidated. Here, we have shown CBD-induced cell death of breast cancer cells, independent of cannabinoid and vallinoid receptor activation. Electron microscopy revealed morphologies consistent with the coexistence of autophagy and apoptosis. Western blot analysis confirmed these findings. We showed that CBD induces endoplasmic reticulum stress and, subsequently, inhibits AKT and mTOR signaling as shown by decreased levels of phosphorylated mTOR and 4EBP1, and cyclin D1. Analyzing further the cross-talk between the autophagic and apoptotic signaling pathways, we found that beclin1 plays a central role in the induction of CBD-mediated apoptosis in MDA-MB-231 breast cancer cells. Although CBD enhances the interaction between beclin1 and Vps34, it inhibits the association between beclin1 and Bcl-2. In addition, we showed that CBD reduces mitochondrial membrane potential, triggers the translocation of BID to the mitochondria, the release of cytochrome c to the cytosol, and, ultimately, the activation of the intrinsic apoptotic pathway in breast cancer cells. CBD increased the generation of reactive oxygen species (ROS), and ROS inhibition blocked the induction of apoptosis and autophagy. Our study revealed an intricate interplay between apoptosis and autophagy in CBD-treated breast cancer cells and highlighted the value of continued investigation into the potential use of CBD as an antineoplastic agent.
Article
Full-text available
Invasion and metastasis of aggressive breast cancer cells are the final and fatal steps during cancer progression. Clinically, there are still limited therapeutic interventions for aggressive and metastatic breast cancers available. Therefore, effective, targeted, and non-toxic therapies are urgently required. Id-1, an inhibitor of basic helix-loop-helix transcription factors, has recently been shown to be a key regulator of the metastatic potential of breast and additional cancers. We previously reported that cannabidiol (CBD), a cannabinoid with a low toxicity profile, down-regulated Id-1 gene expression in aggressive human breast cancer cells in culture. Using cell proliferation and invasion assays, cell flow cytometry to examine cell cycle and the formation of reactive oxygen species, and Western analysis, we determined pathways leading to the down-regulation of Id-1 expression by CBD and consequently to the inhibition of the proliferative and invasive phenotype of human breast cancer cells. Then, using the mouse 4T1 mammary tumor cell line and the ranksum test, two different syngeneic models of tumor metastasis to the lungs were chosen to determine whether treatment with CBD would reduce metastasis in vivo. We show that CBD inhibits human breast cancer cell proliferation and invasion through differential modulation of the extracellular signal-regulated kinase (ERK) and reactive oxygen species (ROS) pathways, and that both pathways lead to down-regulation of Id-1 expression. Moreover, we demonstrate that CBD up-regulates the pro-differentiation factor, Id-2. Using immune competent mice, we then show that treatment with CBD significantly reduces primary tumor mass as well as the size and number of lung metastatic foci in two models of metastasis. Our data demonstrate the efficacy of CBD in pre-clinical models of breast cancer. The results have the potential to lead to the development of novel non-toxic compounds for the treatment of breast cancer metastasis, and the information gained from these experiments broaden our knowledge of both Id-1 and cannabinoid biology as it pertains to cancer progression.
Article
Full-text available
Mantle cell lymphoma (MCL) is characterized by the uncontrolled overexpression of cyclin D1. Styryl sulfonyl compounds have shown potent antitumor activity against MCL by inducing cell-cycle arrest and apoptosis. However, the exact molecular mechanism by which these compounds function is yet to be elucidated. Here, we show that the prototypical styryl sulfonyl compound ON 01910.Na decreased cyclin D1 and c-Myc protein levels in MCL cells, whereas mRNA levels of cyclin D1 were minimally affected. Notably, ON 01910.Na suppressed eukaryotic translation initiation factor 4E (eIF4E)-mediated cyclin D1 mRNA translation, decreased levels of phosphorylated Akt, mammalian target of Rapamycin (mTOR) and eIF4E-binding protein (eIF4E-BP), lowered the cap site binding activity of eIF4E and directly inhibited activity of phosphatidylinositol-3 kinase (PI-3K). Analysis of apoptotic signaling pathways revealed that ON 01910.Na induced the release of cytochrome c from mitochondria, altered expression of Bcl-2 family of proteins and stimulated activation of caspases. Taken together, styryl sulfonyls can cause a rapid decrease of cyclin D1 by blocking cyclin D1 mRNA translation through inhibition of the PI-3K/Akt/mTOR/eIF4E-BP signaling pathway and triggering a cytochrome c-dependent apoptotic pathway in MCL cells.
Article
Full-text available
The cyclin D1 gene is overexpressed in human breast cancers and is required for oncogene-induced tumorigenesis. Peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear receptor selectively activated by ligands of the thiazolidinedione class. PPARγ induces hepatic steatosis, and liganded PPARγ promotes adipocyte differentiation. Herein, cyclin D1 inhibited ligand-induced PPARγ function, transactivation, expression, and promoter activity. PPARγ transactivation induced by the ligand BRL49653 was inhibited by cyclin D1 through a pRB- and cdk-independent mechanism, requiring a region predicted to form an helix-loop-helix (HLH) structure. The cyclin D1 HLH region was also required for repression of the PPARγ ligand-binding domain linked to a heterologous DNA binding domain. Adipocyte differentiation by PPARγ-specific ligands (BRL49653, troglitazone) was enhanced in cyclin D1−/− fibroblasts and reversed by retroviral expression of cyclin D1. Homozygous deletion of the cyclin D1 gene, enhanced expression by PPARγ ligands of PPARγ and PPARγ-responsive genes, and cyclin D1−/− mice exhibit hepatic steatosis. Finally, reduction of cyclin D1 abundance in vivo using ponasterone-inducible cyclin D1 antisense transgenic mice, increased expression of PPARγ in vivo. The inhibition of PPARγ function by cyclin D1 is a new mechanism of signal transduction cross talk between PPARγ ligands and mitogenic signals that induce cyclin D1.
Article
Full-text available
The mammalian target of rapamycin (mTOR) integrates nutrient and mitogen signals to regulate cell growth (increased cell mass and cell size) and cell division. The immunosuppressive drug rapamycin inhibits cell cycle progression via inhibition of mTOR; however, the signaling pathways by which mTOR regulates cell cycle progression have remained poorly defined. Here we demonstrate that restoration of mTOR signaling (by using a rapamycin-resistant mutant of mTOR) rescues rapamycin-inhibited G1-phase progression, and restoration of signaling along the mTOR-dependent S6K1 or 4E-BP1/eukaryotic translation initiation factor 4E (eIF4E) pathways provides partial rescue. Furthermore, interfering RNA-mediated reduction of S6K1 expression or overexpression of mTOR-insensitive 4E-BP1 isoforms that block eIF4E activity inhibit G1-phase progression individually and additively. Thus, the activities of both the S6K1 and 4E-BP1/eIF4E pathways are required for and independently mediate mTOR-dependent G1-phase progression. In addition, overexpression of constitutively active mutants of S6K1 or wild-type eIF4E accelerates serum-stimulated G1-phase progression, and stable expression of wild-type S6K1 confers a proliferative advantage in low-serum-containing media, suggesting that the activity of each of these pathways is limiting for cell proliferation. These data demonstrate that, as for the regulation of cell growth and cell size, the S6K1 and 4E-BP1/eIF4E pathways each represent critical mediators of mTOR-dependent cell cycle control.
Article
Full-text available
To evaluate the presence of histones and nucleosomes in cell lysates of freshly isolated peripheral blood mononuclear cells (PBMC), fully activated lymphoblasts, or lymphoblasts after induction of apoptosis. Each histone class (H1, H2A, H2B, H3, and H4) was detected by western blot analysis with specific antibodies. Annexin V/propidium iodide (PI) staining was performed for each treatment to distinguish viable, early, and late apoptotic, and necrotic cells. DNA degradation was evaluated by PI staining in a hypotonic buffer. All five histones were detected in cell lysates of activated lymphoblasts in higher concentrations than in lysates of PBMC. An additional significant increase of H2A, H2B, H3, H4, and complete nucleosomes in cell lysates of lymphoblasts was found during interleukin (IL)2 deprivation for 8 or 24 hours. The content of these histones or nucleosomes in cell lysates decreased after delayed IL2 readdition. H1 content in cell lysates was not affected by IL2 deprivation or addition. Quantities of H2A, H2B, H3, and H4 in cell lysates correlated significantly with signs of early apoptosis. UV-B light exposure or etoposide treatment of lymphoblasts resulted in a distinct increase for each histone class in cell lysates compared with standard curves. No bands for post-translationally modified histones were detected in cell lysates in contrast with signals in nuclear preparations. Extranuclear accumulation of histones and nucleosomes is an early event of apoptosis in human lymphoblasts. Dysregulation of early apoptosis might support the induction of autoimmunity against nuclear components.
Article
Full-text available
The cyclin D1 gene encodes the labile serum-inducible regulatory subunit of a holoenzyme that phosphorylates and inactivates the retinoblastoma protein. Overexpression of cyclin D1 promotes cellular proliferation and normal physiological levels of cyclin D1 function to inhibit adipocyte differentiation in vivo. We have previously shown that cyclin D1 inhibits peroxisome proliferator-activated receptor (PPAR)gamma-dependent activity through a cyclin-dependent kinase- and retinoblastoma protein-binding-independent mechanism. In this study, we determined the molecular mechanism by which cyclin D1 regulated PPARgamma function. Herein, murine embryonic fibroblast (MEF) differentiation by PPARgamma ligand was associated with a reduction in histone deacetylase (HDAC1) activity. Cyclin D1-/- MEFs showed an increased propensity to undergo differentiation into adipocytes. Genetic deletion of cyclin D1 reduced HDAC1 activity. Reconstitution of cyclin D1 into the cyclin D1-/- MEFs increased HDAC1 activity and blocked PPARgamma-mediated adipogenesis. PPARgamma activity was enhanced in cyclin D1-/- cells. Reintroduction of cyclin D1 inhibited basal and ligand-induced PPARgamma activity and enhanced HDAC repression of PPARgamma activity. Cyclin D1 bound HDAC in vivo and preferentially physically associated with HDAC1, HDAC2, HDAC3, and HDAC5. Chromatin immunoprecipitation assay demonstrated that cyclin D1 enhanced recruitment of HDAC1 and HDAC3 and histone methyltransferase SUV39H1 to the PPAR response element of the lipoprotein lipase promoter and decreased acetylation of total histone H3 and histone H3 lysine 9. Collectively, these studies suggest an important role of cyclin D1 in regulation of PPARgamma-mediated adipocyte differentiation through recruitment of HDACs to regulate PPAR response element local chromatin structure and PPARgamma function.
Article
Full-text available
Delta(9)-Tetrahydrocannabinol (THC) exhibits antitumor effects on various cancer cell types, but its use in chemotherapy is limited by its psychotropic activity. We investigated the antitumor activities of other plant cannabinoids, i.e., cannabidiol, cannabigerol, cannabichromene, cannabidiol acid and THC acid, and assessed whether there is any advantage in using Cannabis extracts (enriched in either cannabidiol or THC) over pure cannabinoids. Results obtained in a panel of tumor cell lines clearly indicate that, of the five natural compounds tested, cannabidiol is the most potent inhibitor of cancer cell growth (IC(50) between 6.0 and 10.6 microM), with significantly lower potency in noncancer cells. The cannabidiol-rich extract was equipotent to cannabidiol, whereas cannabigerol and cannabichromene followed in the rank of potency. Both cannabidiol and the cannabidiol-rich extract inhibited the growth of xenograft tumors obtained by s.c. injection into athymic mice of human MDA-MB-231 breast carcinoma or rat v-K-ras-transformed thyroid epithelial cells and reduced lung metastases deriving from intrapaw injection of MDA-MB-231 cells. Judging from several experiments on its possible cellular and molecular mechanisms of action, we propose that cannabidiol lacks a unique mode of action in the cell lines investigated. At least for MDA-MB-231 cells, however, our experiments indicate that cannabidiol effect is due to its capability of inducing apoptosis via: direct or indirect activation of cannabinoid CB(2) and vanilloid transient receptor potential vanilloid type-1 receptors and cannabinoid/vanilloid receptor-independent elevation of intracellular Ca(2+) and reactive oxygen species. Our data support the further testing of cannabidiol and cannabidiol-rich extracts for the potential treatment of cancer.
Article
Full-text available
There is currently substantial interest in the regulation of cell function by mammalian target of rapamycin (mTOR), especially effects linked to the rapamycin-sensitive mTOR complex 1 (mTORC1). Rapamycin induces G(1) arrest and blocks proliferation of many tumor cells, suggesting that the inhibition of mTORC1 signaling may be useful in cancer therapy. In MCF7 breast adenocarcinoma cells, rapamycin decreases levels of cyclin D1, without affecting cytoplasmic levels of its mRNA. In some cell-types, rapamycin does not affect cyclin D1 levels, whereas the starvation for leucine (which impairs mTORC1 signaling more profoundly than rapamycin) does. This pattern correlates with the behavior of eukaryotic initiation factor 4E-binding protein 1 (4E-BP1, an mTORC1 target that regulates translation initiation). siRNA-mediated knock-down of 4E-BP1 abrogates the effect of rapamycin on cyclin D1 expression and increases the polysomal association of the cyclin D1 mRNA. Our data identify 4E-BP1 as a key regulator of cyclin D1 expression, indicate that this effect is not mediated through the changes in cytoplasmic levels of cyclin D1 mRNA and suggest that, in some cell types, interfering with the amino acid input to mTORC1, rather than using rapamycin, may inhibit proliferation.
Article
Full-text available
Cyclin D1 is a key regulator of cell proliferation. It also controls other aspects of the cell fate, such as cellular senescence, apoptosis and tumourigenesis. We used B-lymphoid cell lines producing cyclin D1 to investigate the role of this protein in B-cell lymphomas and leukaemias. Constitutive low levels of cyclin D1 had no effect per se on cell proliferation, but conferred resistance to various apoptotic stimuli in B cells. Activation of the pro-apoptotic protein, Bax, was reduced and mitochondrial permeabilization and phosphatidylserine exposure following cytokine withdrawal were delayed in cyclin D1-producing cells. Proteomic analysis showed that the presence of cyclin D1 led to intracellular accumulation of various molecular chaperones. The chaperone, heat shock protein (Hsp)70, bound to both Bax and the mitochondrial apoptosis inducing factor following cytokine withdrawal, and impeded inhibitors of kappaB (IkappaB)-mediated inhibition of nuclear factor-kappaB anti-apoptotic signalling. Impairment of Hsp70 activity--using a pharmacological Hsp inhibitor or transfecting cells with an Hsp70-blocking antibody--restored the cellular response to mitochondrial apoptosis triggering. Thus, constitutive de-novo cyclin D1 production in B cells delays commitment to apoptosis by inducing Hsp70 chaperoning activity on pre- and post-mitochondrial pro-apoptotic factors.
Article
Full-text available
In 1997, the first published reports highlighted PPARγ as a novel cancer therapeutic target regulating differentiation of cancer cells. A subsequent flurry of papers described these activities more widely and fuelled further enthusiasm for differentiation therapy, as the ligands for the PPARγ were seen as well tolerated and in several cases well-established in other therapeutic contexts. This initial enthusiasm and promise was somewhat tempered by contradictory findings in several murine cancer models and equivocal trial findings. As more understanding has emerged in recent years, a renaissance has occurred in targeting PPARγ within the context of either chemoprevention or chemotherapy. This clarity has arisen in part through a clearer understanding of PPARγ biology, how the receptor interacts with other proteins and signaling events, and the mechanisms that modulate its transcriptional actions. Equally greater translational understanding of this target has arisen from a clearer understanding of in vivo murine cancer models. Clinical exploitation will most likely require precise and quantifiable description of PPARγ actions, and resolution of which targets are the most beneficial to target combined with an understanding of the mechanisms that limits its anticancer effectiveness.
Article
The mTOR pathway was discovered in the late 1970s after the compound and natural inhibitor of mTOR, rapamycin was isolated from the bacterium Streptomyces hygroscopicus. mTOR is serine/threonine kinase belonging to the phosphoinositide 3-kinase related kinase (PIKK) family. It forms two distinct complexes; mTORC1 and mTORC2. mTORC1 has a key role in regulating protein synthesis and autophagy whilst mTORC2 is involved in regulating kinases of the AGC family. mTOR signaling is often over active in multiple cancer types including breast cancer. This can involve mutations in mTOR itself but more commonly, in breast cancer, this is related to an increase in activity of ErbB family receptors or alterations and mutations of PI3K signaling. Rapamycin and its analogues (rapalogues) bind to the intercellular receptor FKBP12, and then predominantly inhibit mTORC1 signaling via an allosteric mechanism. Research has shown that inhibition of mTOR is a useful strategy in tackling cancers, with it acting to slow tumor growth and limit the spread of a cancer. Rapalogues have now made their way into the clinic with the rapalogue everolimus (RAD-001/Afinitor) approved for use in conjunction with exemestane, in post-menopausal breast cancer patients with advanced disease who are HER-2 negative (normal expression), hormone receptor positive and whose prior treatment with non-steroidal aromatase inhibitors has failed. Testing across multiple trials has proven that everolimus and other rapalogues are a viable way of treating certain types of cancer. However, rapalogues have shown some drawbacks both in research and clinically, with their use often activating feedback pathways that counter their usefulness. As such, new types of inhibitors are being explored that work via different mechanisms, including inhibitors that are ATP competitive with mTOR and which act to perturb signaling from both mTOR complexes.
Article
Over the past years, several lines of evidence support a therapeutic potential of Cannabis derivatives and in particular phytocannabinoids. Δ(9)-THC and cannabidiol (CBD) are the most abundant phytocannabinoids in Cannabis plants and therapeutic application for both compounds have been suggested. However, CBD is recently emerging as a therapeutic agent in numerous pathological conditions since devoid of the psychoactive side effects exhibited instead by Δ(9)-THC. In this review, we highlight the pharmacological activities of CBD, its cannabinoid receptor-dependent and -independent action, its biological effects focusing on immunomodulation, angiogenetic properties, and modulation of neuronal and cardiovascular function. Furthermore, the therapeutic potential of cannabidiol is also highlighted, in particular in nuerological diseases and cancer.
Article
The mammalian target of rapamycin (mTOR) is a crucial signaling node that integrates environmental cues to regulate cell survival, proliferation and metabolism, and is often deregulated in human cancer. mTOR kinase acts in two functionally distinct complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), whose activities and substrate specificities are regulated by complex co-factors. Deregulation of this centralized signaling pathway has been associated with a variety of human diseases including diabetes, neurodegeneration and cancer. Although mTORC1 signaling has been extensively studied in cancer, recent discoveries indicate a subset of human cancers harboring amplifications in mTORC2-specific genes as the only actionable genomic alterations, suggesting a distinct role for mTORC2 in cancer as well. This review will summarize recent advances in dissecting the relative contributions of mTORC1 versus mTORC2 in cancer, their role in tumor-associated blood vessels and tumor immunity, and provide an update on mTOR inhibitors.Oncogene advance online publication, 17 October 2016; doi:10.1038/onc.2016.363.
Article
The mechanistic/mammalian target of rapamycin (mTOR) is a conserved protein kinase that controls several anabolic processes required for cell growth and proliferation. As such, mTOR has been implicated in an increasing number of pathological conditions, including cancer, obesity, type 2 diabetes and neurodegeneration. As part of the mTOR complex 1 (mTORC1), mTOR regulates cell growth by promoting the biosynthesis of proteins, lipids and nucleic acids. Several mTORC1 substrates have been shown to regulate protein synthesis, including the eukaryotic initiation factor 4E (eIF4E)-binding proteins (4E-BPs) and the ribosomal S6 kinases (S6Ks) 1 and 2. In this work, we focus on the signalling pathways that lie both upstream and downstream of mTORC1, as well as their relevance to human pathologies. We further discuss pharmacological approaches that target mTOR and their applications for the treatment of cancer. © The Author 2015. Published by Oxford University Press on behalf of the UK Environmental Mutagen Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.