1
|
Amaryllidaceae Alkaloids Decrease the Proliferation, Invasion, and Secretion of Clinically Relevant Cytokines by Cultured Human Colon Cancer Cells. Biomolecules 2022; 12:biom12091267. [PMID: 36139106 PMCID: PMC9496155 DOI: 10.3390/biom12091267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 12/30/2022] Open
Abstract
Alkaloids isolated from members of the Amaryllidaceae plant family are promising anticancer agents. The purpose of the current study was to determine if the isocarbostyrils narciclasine, pancratistatin, lycorane, lycorine, crinane, and haemanthamine inhibit phenomena related to cancer progression in vitro. To achieve this, we examined the proliferation, adhesion, and invasion of cultured human colon cancer cells via MTT assay and Matrigel-coated Boyden chambers. In addition, Luminex assays were used to quantify the secretion of matrix metalloproteinases (MMP) and cytokines associated with poor clinical outcomes. We found that all alkaloids decreased cell proliferation regardless of TP53 status, with narciclasine exhibiting the greatest potency. The effects on cell proliferation also appear to be specific to cancer cells. Narciclasine, lycorine, and haemanthamine decrease both adhesion and invasion but with various potencies depending on the cell line. In addition, narciclasine, lycorine, and haemanthamine decreased the secretion of MMP-1, -2, and -7, as well as the secretion of the cytokines pentraxin 3 and vascular endothelial growth factor. In conclusion, the present study shows that Amaryllidaceae alkaloids decrease phenomena and cytokines associated with colorectal cancer progression, supporting future investigations regarding their potential as multifaceted drug candidates.
Collapse
|
2
|
Chemoprevention and therapeutic role of essential oils and phenolic compounds: Modeling tumor microenvironment in glioblastoma. Pharmacol Res 2021; 169:105638. [PMID: 33933637 DOI: 10.1016/j.phrs.2021.105638] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/28/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022]
Abstract
Glioblastoma (GBM) is the most common primary tumor of the central nervous system. Current treatments available for GBM entails surgical resection followed by temozolomide chemotherapy and/or radiotherapy, which are associated with multidrug resistance and severe side effects. While this treatment could yield good results, in almost all cases, patients suffer from relapse, which leads to reduced survival rates. Thus, therapeutic approaches with improved efficiency and reduced off-target risks are needed to overcome these problems. Regarding this, natural products appear as a safe and attractive strategy as chemotherapeutic agents or adjuvants in the treatment of GBM. Besides the increasing role of natural compounds for chemoprevention of GBM, it has been proposed to prevent carcinogenesis and metastasis of GBM. Numerous investigations showed that natural products are able to inhibit proliferation and angiogenesis, to induce apoptosis, and to target GBM stem cells, which are associated with tumor development and recurrence. This review gives a timely and comprehensive overview of the current literature regarding chemoprevention and therapy of GBM by natural products with a focus on essential oils and phenolic compounds and their molecular mechanisms.
Collapse
|
3
|
Sala L, Cirillo G, Riva G, Romano G, Giussani C, Cialdella A, Todisco A, Virtuoso A, Cerrito MG, Bentivegna A, Grassilli E, Ardizzoia A, Bonoldi E, Giovannoni R, Papa M, Lavitrano M. Specific Expression of a New Bruton Tyrosine Kinase Isoform (p65BTK) in the Glioblastoma Gemistocytic Histotype. Front Mol Neurosci 2019; 12:2. [PMID: 30733667 PMCID: PMC6353843 DOI: 10.3389/fnmol.2019.00002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
Bruton's tyrosine-kinase (BTK) is a non-receptor tyrosine kinase recently associated with glioma tumorigenesis and a novel prognostic marker for poor survival in patients with glioma. The p65BTK is a novel BTK isoform involved in different pathways of drug resistance of solid tumors, thus we aimed to investigate the expression and the putative role of p65BTK in tumors of the central nervous system (CNS). We selected a large cohort of patients with glial tumors (n = 71) and analyzed the expression of p65BTK in different histotypes and correlation with clinical parameters. Sections were stained with glial fibrillary acidic protein (GFAP), p53, epidermal growth factor receptor (EGFR), S100, vimentin, and epithelial membrane antigen (EMA) antibodies. Glioma stem cell (GSC) lines, isolated from glioblastoma multiforme (GBM), were treated with different concentrations of ibrutinib, a specific inhibitor of BTK, in order to evaluate their metabolic activity, mitotic index and mortality. Moreover, an orthotopic xenotransplant of GSC from human GBM was used to evaluate the expression of p65BTK in the brain of immunodeficient mice. p65BTK was expressed in GSC and in gemistocytes in human gliomas at different histological grade. We found a significant correlation between BTK expression and low-grade (LG) tumors (p ≤ 0.05) and overall survival (OS) of patients with grade III gliomas (p ≤ 0.05), suggestive of worst prognosis. Interestingly, the expression of p65BTK remained restricted exclusively to gemistocytic cells in the xenograft mouse model. Ibrutinib administration significantly reduced metabolic activity and mitotic index and increased mortality in GSC, highlighting the specific role of p65BTK in cell proliferation and survival. In conclusion, our data demonstrated that p65BTK is expressed in glioma tumors, restricted to gemistocytic cells, has a key role in GSC and has a bad prognostic value, thus highlighting the importance of future research for targeted therapy of human gliomas.
Collapse
Affiliation(s)
- Luca Sala
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Giovanni Cirillo
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gabriele Riva
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Gabriele Romano
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Carlo Giussani
- Unit of Neurosurgery, School of Medicine and Surgery, Neuroscience Center, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | | | - Antonio Todisco
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Assunta Virtuoso
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Angela Bentivegna
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Emanuela Grassilli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | | | - Roberto Giovannoni
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Michele Papa
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | |
Collapse
|
4
|
Marsico G, Pignataro BA, Masi M, Evidente A, Casella F, Zonno MC, Tak JH, Bloomquist JR, Superchi S, Scafato P. Asymmetric synthesis and structure-activity studies of the fungal metabolites colletorin A, colletochlorin A and their halogenates analogues. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.05.077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
5
|
Lefranc F, Tabanca N, Kiss R. Assessing the anticancer effects associated with food products and/or nutraceuticals using in vitro and in vivo preclinical development-related pharmacological tests. Semin Cancer Biol 2017; 46:14-32. [PMID: 28602819 DOI: 10.1016/j.semcancer.2017.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/02/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
This review is part of a special issue entitled "Role of dietary pattern, foods, nutrients and nutraceuticals in supporting cancer prevention and treatment" and describes a pharmacological strategy to determine the potential contribution of food-related components as anticancer agents against established cancer. Therefore, this review does not relate to chemoprevention, which is analysed in several other reviews in the current special issue, but rather focuses on the following: i) the biological events that currently represent barriers against the treatment of certain types of cancers, primarily metastatic cancers; ii) the in vitro and in vivo pharmacological pre-clinical tests that can be used to analyse the potential anticancer effects of food-related components; and iii) several examples of food-related components with anticancer effects. This review does not represent a catalogue-based listing of food-related components with more or less anticancer activity. By contrast, this review proposes an original pharmacological strategy that researchers can use to analyse the potential anticancer activity of any food-related component-e.g., by considering the crucial characteristics of cancer biological aggressiveness. This review also highlights that cancer patients undergoing chemotherapy should restrict the use of "food complements" without supervision by a medical nutritionist. By contrast, an equilibrated diet that includes the food-related components listed herein would be beneficial for cancer patients who are not undergoing chemotherapy.
Collapse
Affiliation(s)
- Florence Lefranc
- Service de Neurochirurgie, Hôpital Erasme, Université Libre de Bruxelles, 808 route de Lennik, 1070 Brussels, Belgium.
| | - Nurhayat Tabanca
- U.S Department of Agriculture-Agricultural Research Service, Subtropical Horticulture Research Station,13601 Old Cutler Rd., Miami, FL 33158, USA.
| | - Robert Kiss
- Retired-formerly at the Fonds National de la Recherche Scientifique (FRS-FNRS, Brussels, Belgium), 5 rue d'Egmont, 1000 Brussels, Belgium.
| |
Collapse
|
6
|
Mirzaei H, Naseri G, Rezaee R, Mohammadi M, Banikazemi Z, Mirzaei HR, Salehi H, Peyvandi M, Pawelek JM, Sahebkar A. Curcumin: A new candidate for melanoma therapy? Int J Cancer 2016; 139:1683-95. [PMID: 27280688 DOI: 10.1002/ijc.30224] [Citation(s) in RCA: 197] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 05/25/2016] [Indexed: 02/06/2023]
Abstract
Melanoma remains among the most lethal cancers and, in spite of great attempts that have been made to increase the life span of patients with metastatic disease, durable and complete remissions are rare. Plants and plant extracts have long been used to treat a variety of human conditions; however, in many cases, effective doses of herbal remedies are associated with serious adverse effects. Curcumin is a natural polyphenol that shows a variety of pharmacological activities including anti-cancer effects, and only minimal adverse effects have been reported for this phytochemical. The anti-cancer effects of curcumin are the result of its anti-angiogenic, pro-apoptotic and immunomodulatory properties. At the molecular and cellular level, curcumin can blunt epithelial-to-mesenchymal transition and affect many targets that are involved in melanoma initiation and progression (e.g., BCl2, MAPKS, p21 and some microRNAs). However, curcumin has a low oral bioavailability that may limit its maximal benefits. The emergence of tailored formulations of curcumin and new delivery systems such as nanoparticles, liposomes, micelles and phospholipid complexes has led to the enhancement of curcumin bioavailability. Although in vitro and in vivo studies have demonstrated that curcumin and its analogues can be used as novel therapeutic agents in melanoma, curcumin has not yet been tested against melanoma in clinical practice. In this review, we summarized reported anti-melanoma effects of curcumin as well as studies on new curcumin formulations and delivery systems that show increased bioavailability. Such tailored delivery systems could pave the way for enhancement of the anti-melanoma effects of curcumin.
Collapse
Affiliation(s)
- Hamed Mirzaei
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Naseri
- Department of Anatomical Sciences, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ramin Rezaee
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohsen Mohammadi
- Razi Herbal Medicines Research Center and Department of pharmaceutical biotechnology, Faculty of pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Zarrin Banikazemi
- Biochemistry of Nutrition Research Center, School of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Hamid Reza Mirzaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mostafa Peyvandi
- Department of Anatomical Sciences, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Anatomical Sciences, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - John M Pawelek
- Department of Dermatology and the Yale Cancer Center, Yale University School of Medicine, New Haven, CT
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Thida M, Kim DW, Tran TTT, Pham MQ, Lee H, Kim I, Lee JW. Gambogic acid induces apoptotic cell death in T98G glioma cells. Bioorg Med Chem Lett 2016; 26:1097-1101. [DOI: 10.1016/j.bmcl.2015.11.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/10/2015] [Accepted: 11/14/2015] [Indexed: 10/22/2022]
|
8
|
Erratum. J Cell Mol Med 2015. [PMID: 26223272 PMCID: PMC4549055 DOI: 10.1111/jcmm.12640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
9
|
Fusicoccin a, a phytotoxic carbotricyclic diterpene glucoside of fungal origin, reduces proliferation and invasion of glioblastoma cells by targeting multiple tyrosine kinases. Transl Oncol 2013; 6:112-23. [PMID: 23544164 DOI: 10.1593/tlo.12409] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 01/27/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a deadly cancer that possesses an intrinsic resistance to pro-apoptotic insults, such as conventional chemotherapy and radiotherapy, and diffusely invades the brain parenchyma, which renders it elusive to total surgical resection. We found that fusicoccin A, a fungal metabolite from Fusicoccum amygdali, decreased the proliferation and migration of human GBM cell lines in vitro, including several cell lines that exhibit varying degrees of resistance to pro-apoptotic stimuli. The data demonstrate that fusicoccin A inhibits GBM cell proliferation by decreasing growth rates and increasing the duration of cell division and also decreases two-dimensional (measured by quantitative video microscopy) and three-dimensional (measured by Boyden chamber assays) migration. These effects of fusicoccin A treatment translated into structural changes in actin cytoskeletal organization and a loss of GBM cell adhesion. Therefore, fusicoccin A exerts cytostatic effects but low cytotoxic effects (as demonstrated by flow cytometry). These cytostatic effects can partly be explained by the fact that fusicoccin inhibits the activities of a dozen kinases, including focal adhesion kinase (FAK), that have been implicated in cell proliferation and migration. Overexpression of FAK, a nonreceptor protein tyrosine kinase, directly correlates with the invasive phenotype of aggressive human gliomas because FAK promotes cell proliferation and migration. Fusicoccin A led to the down-regulation of FAK tyrosine phosphorylation, which occurred in both normoxic and hypoxic GBM cell culture conditions. In conclusion, the current study identifies a novel compound that could be used as a chemical template for generating cytostatic compounds designed to combat GBM.
Collapse
|
10
|
Novel phenolic inhibitors of small/intermediate-conductance Ca²⁺-activated K⁺ channels, KCa3.1 and KCa2.3. PLoS One 2013; 8:e58614. [PMID: 23516517 PMCID: PMC3597730 DOI: 10.1371/journal.pone.0058614] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/05/2013] [Indexed: 01/25/2023] Open
Abstract
Background KCa3.1 channels are calcium/calmodulin-regulated voltage-independent K+ channels that produce membrane hyperpolarization and shape Ca2+-signaling and thereby physiological functions in epithelia, blood vessels, and white and red blood cells. Up-regulation of KCa3.1 is evident in fibrotic and inflamed tissues and some tumors rendering the channel a potential drug target. In the present study, we searched for novel potent small molecule inhibitors of KCa3.1 by testing a series of 20 selected natural and synthetic (poly)phenols, synthetic benzoic acids, and non-steroidal anti-inflammatory drugs (NSAIDs), with known cytoprotective, anti-inflammatory, and/or cytostatic activities. Methodology/Principal Findings In electrophysiological experiments, we identified the natural phenols, caffeic acid (EC50 1.3 µM) and resveratrol (EC50 10 µM) as KCa3.1 inhibitors with moderate potency. The phenols, vanillic acid, gallic acid, and hydroxytyrosol had weak or no blocking effects. Out of the NSAIDs, flufenamic acid was moderately potent (EC50 1.6 µM), followed by mesalamine (EC50≥10 µM). The synthetic fluoro-trivanillic ester, 13b ([3,5-bis[(3-fluoro-4-hydroxy-benzoyl)oxymethyl]phenyl]methyl 3-fluoro-4-hydroxy-benzoate), was identified as a potent mixed KCa2/3 channel inhibitor with an EC50 of 19 nM for KCa3.1 and 360 pM for KCa2.3, which affected KCa1.1 and Kv channels only at micromolar concentrations. The KCa3.1/KCa2-activator SKA-31 antagonized the 13b-blockade. In proliferation assays, 13b was not cytotoxic and reduced proliferation of 3T3 fibroblasts as well as caffeic acid. In isometric vessel myography, 13b increased contractions of porcine coronary arteries to serotonin and antagonized endothelium-derived hyperpolarization-mediated vasorelaxation to pharmacological KCa3.1/KCa2.3 activation. Conclusions/Significance We identified the natural phenols, caffeic acid and resveratrol, the NSAID, flufenamic acid, and the polyphenol 13b as novel KCa3.1 inhibitors. The high potency of 13b with pan-activity on KCa3.1/KCa2 channels makes 13b a new pharmacological tool to manipulate inflammation and cancer growth through KCa3.1/KCa2 blockade and a promising template for new drug design.
Collapse
|
11
|
FTIR spectroscopy: a new valuable tool to classify the effects of polyphenolic compounds on cancer cells. Biochim Biophys Acta Mol Basis Dis 2012; 1832:46-56. [PMID: 23085492 DOI: 10.1016/j.bbadis.2012.10.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 09/11/2012] [Accepted: 10/15/2012] [Indexed: 12/31/2022]
Abstract
Polyphenolic compounds are an important part of human diet and regular consumption of fruits, vegetables and tea is associated with reduced risk of cancer. Dietary polyphenols display a vast array of cellular effects but the large number of data published in the literature makes it difficult to determine the main mechanisms of action associated and to identify molecules with original mechanisms. Therefore, there is an increasing demand for more systemic approaches in order to obtain a global insight of the biochemical processes mediated by polyphenols. Here, we used Fourier transform infrared (FTIR) spectroscopy to analyze cancer cells exposed in vitro to 6 polyphenols: 3 natural well documented polyphenols (curcumin, epigallocatechin gallate (EGCG) and quercetin) and 3 synthetic molecules with a very closely related chemical structure. Statistical analyses on FTIR spectra allowed the comparison of global effects of the 6 compounds and evidenced some common or different features in the cell perturbations among natural and synthetic molecules. Interestingly, marked metabolic changes induced by polyphenols closely related from a chemical point of view were identified. Furthermore, many metabolic changes could be detected as early as after 2h incubation with the drugs.
Collapse
|
12
|
Frédérick R, Bruyère C, Vancraeynest C, Reniers J, Meinguet C, Pochet L, Backlund A, Masereel B, Kiss R, Wouters J. Novel trisubstituted harmine derivatives with original in vitro anticancer activity. J Med Chem 2012; 55:6489-501. [PMID: 22770529 DOI: 10.1021/jm300542e] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To overcome the intrinsic resistance of cancer cells to apoptotic stimuli, we designed and synthesized approximately 50 novel β-carbolines structurally related to harmine. Harmine is known for its anticancer properties and is a DYRK1A inhibitor. Of the synthesized compounds, the most active in terms of growth inhibition of five cancer cell lines are cytostatic and approximately 100 times more potent than harmine but demonstrated no DYRK1A inhibitory activity. These novel β-carbolines display similar growth inhibitory activity in cancer cells that are sensitive and resistant to apoptotic stimuli. Using ChemGPS-NP, we found that the more active β-carbolines are all more lipophilic and larger than the less active compounds. Lastly, on the basis of the NCI human tumor cell line anticancer drug screen and the NCI COMPARE algorithm, it appears that some of these compounds, including 5a and 5k, seem to act as protein synthesis inhibitors.
Collapse
Affiliation(s)
- Raphaël Frédérick
- Namur Medicine and Drug Innovation Center (NAMEDIC-NARILIS), University of Namur (FUNDP), 61, Rue de Bruxelles, 5000 Namur, Belgium.
| | | | | | | | | | | | | | | | | | | |
Collapse
|