1
|
Stompor-Gorący M, Włoch A, Sengupta P, Nasulewicz-Goldeman A, Wietrzyk J. Synergistic Proliferation Effects of Xanthohumol and Niflumic Acid on Merkel and Glioblastoma Cancer Cells: Role of Cell Membrane Interactions. Int J Mol Sci 2024; 25:11015. [PMID: 39456799 PMCID: PMC11508127 DOI: 10.3390/ijms252011015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The objective of our research was to determine the effects of xanthohumol (XN), a flavonoid isolated from hops (Humulus lupulus), and the anti-inflammatory drug niflumic acid (NA), separately and in combination with each other, on the proliferation of human cancer cells. Additionally, so as to understand the mechanism underlying the anticancer properties of the tested compounds, their effects on the biophysical parameters of a model membrane were assessed. The cells were incubated with XN and NA at various concentrations, either individually or in combination with each other. Cell proliferation was quantified using the sulforodamine B (SRB) assay. In addition, the IC50 values for niflumic acid and xanthohumol applied separately were determined by cell proliferation tests for the following human cancer cell lines: 5637 (urinary bladder carcinoma), A-431 (epidermoid carcinoma), UM-SCC-17A (head and neck squamous carcinoma), SK-MEL-3 (melanoma), MCC13 (Merkel cell cancer), and A172 (glioblastoma), in comparison with the mouse normal fibroblasts (BALB/3T3 clone A31). The results show that the two-compound combinations of XN and NA significantly decreased the proliferation of cancer cells in a dose-dependent manner, and the effects were stronger than the additive responses to XN and NA individually. The membrane studies revealed a synergistic effect on the membrane rigidity when using the mixture of XN and NA, which may explain the observed increase in anticancer activity for the combined XN and NA. Our results suggest that NSAIDs, such as niflumic acid, may be a promising strategy for co-application with xanthohumol as anticancer drugs.
Collapse
Affiliation(s)
- Monika Stompor-Gorący
- Department of Pathophysiology, Institute of Medical Sciences, University of Rzeszów, Warzywna 1a, 35-310 Rzeszów, Poland
| | - Aleksandra Włoch
- Department of Physics and Biophysics, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland; (A.W.); (P.S.)
| | - Priti Sengupta
- Department of Physics and Biophysics, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland; (A.W.); (P.S.)
| | - Anna Nasulewicz-Goldeman
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland; (A.N.-G.); (J.W.)
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114 Wrocław, Poland; (A.N.-G.); (J.W.)
| |
Collapse
|
2
|
Song J, Zhang S, Zhang B, Ma J. The anti-breast cancer therapeutic potential of 1,2,3-triazole-containing hybrids. Arch Pharm (Weinheim) 2024; 357:e2300641. [PMID: 38110853 DOI: 10.1002/ardp.202300641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/20/2023]
Abstract
Breast cancer, as one of the most common invasive malignancies and the leading cause of cancer-related deaths in women globally, poses a significant challenge in the world health system. Substantial advances in diagnosis and treatment have significantly improved the survival rate of breast cancer patients, but the number of incidences and deaths of breast cancer are projected to increase by 40% and 50%, respectively, by 2040. Chemotherapy is one of the principal treatments for breast cancer therapy, but multidrug resistance and severe side effects remain the major obstacles to the success of treatment. Hence, there is a vital need to develop novel chemotherapeutic agents to combat this deadly disease. 1,2,3-Triazole, which can be effectively constructed by click chemistry, not only can serve as a linker to connect different anti-breast cancer pharmacophores but also is a valuable pharmacophore with anti-breast cancer potential and favorable properties such as hydrogen bonding, moderate dipole moment, and enhanced water solubility. Particularly, 1,2,3-triazole-containing hybrids have demonstrated promising in vitro and in vivo anti-breast cancer potential against both drug-sensitive and drug-resistant forms and possessed excellent selectivity by targeting different biological pathways associated with breast cancer, representing privileged scaffolds for the discovery of novel anti-breast cancer candidates. This review concentrates on the latest advancements of 1,2,3-triazole-containing hybrids with anti-breast cancer potential, including work published between 2020 and the present. The structure-activity relationships (SARs) and mechanisms of action are also reviewed to shed light on the development of more effective and multitargeted candidates.
Collapse
Affiliation(s)
- Juntao Song
- Department of Oncology and Hematology, Zibo 148 Hospital, Zibo, China
| | - Shuai Zhang
- Department of General Surgery, People's Hospital of Zhoucun District, Zibo, China
| | - Bo Zhang
- Emergency Department, People's Hospital of Zhoucun District, Zibo, China
| | - Junwei Ma
- Department of General Surgery, Zibo 148 Hospital, Zibo, China
| |
Collapse
|
3
|
Nowak D, Huczyński A, Bachorz RA, Hoffmann M. Machine Learning Application for Medicinal Chemistry: Colchicine Case, New Structures, and Anticancer Activity Prediction. Pharmaceuticals (Basel) 2024; 17:173. [PMID: 38399388 PMCID: PMC10892630 DOI: 10.3390/ph17020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/02/2024] [Accepted: 01/12/2024] [Indexed: 02/25/2024] Open
Abstract
In the contemporary era, the exploration of machine learning (ML) has gained widespread attention and is being leveraged to augment traditional methodologies in quantitative structure-activity relationship (QSAR) investigations. The principal objective of this research was to assess the anticancer potential of colchicine-based compounds across five distinct cell lines. This research endeavor ultimately sought to construct ML models proficient in forecasting anticancer activity as quantified by the IC50 value, while concurrently generating innovative colchicine-derived compounds. The resistance index (RI) is computed to evaluate the drug resistance exhibited by LoVo/DX cells relative to LoVo cancer cell lines. Meanwhile, the selectivity index (SI) is computed to determine the potential of a compound to demonstrate superior efficacy against tumor cells compared to its toxicity against normal cells, such as BALB/3T3. We introduce a novel ML system adept at recommending novel chemical structures predicated on known anticancer activity. Our investigation entailed the assessment of inhibitory capabilities across five cell lines, employing predictive models utilizing various algorithms, including random forest, decision tree, support vector machines, k-nearest neighbors, and multiple linear regression. The most proficient model, as determined by quality metrics, was employed to predict the anticancer activity of novel colchicine-based compounds. This methodological approach yielded the establishment of a library encompassing new colchicine-based compounds, each assigned an IC50 value. Additionally, this study resulted in the development of a validated predictive model, capable of reasonably estimating IC50 values based on molecular structure input.
Collapse
Affiliation(s)
- Damian Nowak
- Department of Quantum Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznan, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznan, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland;
| | - Rafał Adam Bachorz
- Institute of Medical Biology of Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland;
- Institute of Computing Science, Faculty of Computing, Poznań University of Technology, Piotrowo 2, 60-965 Poznań, Poland
| | - Marcin Hoffmann
- Department of Quantum Chemistry, Faculty of Chemistry, Adam Mickiewicz University in Poznan, Uniwersytetu Poznanskiego 8, 61-614 Poznan, Poland
| |
Collapse
|
4
|
Rastogi SK, Ciliberto VC, Trevino MZ, Campbell BA, Brittain WJ. Green Approach Toward Triazole Forming Reactions for Developing Anticancer Drugs. Curr Org Synth 2024; 21:380-420. [PMID: 37157212 DOI: 10.2174/1570179420666230508125144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 05/10/2023]
Abstract
Compounds containing triazole have many significant applications in the dye and ink industry, corrosion inhibitors, polymers, and pharmaceutical industries. These compounds possess many antimicrobial, antioxidant, anticancer, antiviral, anti-HIV, antitubercular, and anticancer activities. Several synthetic methods have been reported for reducing time, minimizing synthetic steps, and utilizing less hazardous and toxic solvents and reagents to improve the yield of triazoles and their analogues synthesis. Among the improvement in methods, green approaches towards triazole forming biologically active compounds, especially anticancer compounds, would be very important for pharmaceutical industries as well as global research community. In this article, we have reviewed the last five years of green chemistry approaches on click reaction between alkyl azide and alkynes to install 1,2,3-triazole moiety in natural products and synthetic drug-like molecules, such as in colchicine, flavanone cardanol, bisphosphonates, thiabendazoles, piperazine, prostanoid, flavonoid, quinoxalines, C-azanucleoside, dibenzylamine, and aryl-azotriazole. The cytotoxicity of triazole hybrid analogues was evaluated against a panel of cancer cell lines, including multidrug-resistant cell lines.
Collapse
Affiliation(s)
- Shiva K Rastogi
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Veronica C Ciliberto
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Monica Z Trevino
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Brooke A Campbell
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - William J Brittain
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| |
Collapse
|
5
|
Qiang X, Peng Y, Wang Z, Fu W, Li W, Zhao Q, He D. Synthesis of glycyrrhizin analogues as HMGB1 inhibitors and their activity against sepsis in acute kidney injury. Eur J Med Chem 2023; 259:115696. [PMID: 37542990 DOI: 10.1016/j.ejmech.2023.115696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023]
Abstract
Glycyrrhizin (GL) is one of the antagonists of highly conserved nuclear protein (HMGB1). The researches have shown that the glycosyl of GL is an important pharmacophore for GL binding to HMGB1, and it is the determinant factor for mechanism of action. To get the HMGB1 inhibitors with higher activity and good pharmacokinetic properties, two classes of GL analogues containing C-N glycoside bond were synthesized, and their anti-inflammatory, anti-oxidative stress and anti-septic kidney injury were evaluated. The results are as follows. First, in the anti-inflammatory assay, all the compounds inhibited NO release in some degree; among them, compound 6 displayed the strongest NO inhibitory effect with IC50 value of 15.9 μM, and compound 15 with IC50 of 20.2 μM. The two compounds not only decreased IL-1β and TNF-α levels in RAW264.7 cells and HK-2 cells, but also downregulated the levels of NLRP3, P-NF-κB p65 and HMGB1 in activated HK-2 cells in a dose-dependent manner. Second, in the renal protection assay with H2O2-stimulated HK-2 cell line, they reduced MDA level and increased SOD in HK-2 cells; additionally, they also inhibited the HK-2 cell apoptosis and downregulated the Caspase-1 p20 level. Third, in the in vivo activity tests of the septic mouse, they also showed good activities just like in vitro, decreasing the IL-1β, TNF-α, MDA, blood creatinine (Scr) and urea nitrogen (BUN) in serum, and increasing SOD levels in a dose-dependent manner. The immunoblotting results showed the two compounds downregulated the levels of HMGB1, P-NF-κB p65, NLRP3 and Caspase-1 p20 protein. All in all, the two compounds improved the renal injury of septic mice, and alleviated the tube wall structure damage and renal tubular dilation in kidney, which further proved by H&E staining. This suggests the two compounds have septic acute kidney injury activity, and they will be potential therapeutic drugs for septic acute kidney injury.
Collapse
Affiliation(s)
- Xin Qiang
- Materia Medica Development Group, Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou, 730000, China
| | - Yijie Peng
- Materia Medica Development Group, Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou, 730000, China
| | - Zongyuan Wang
- Materia Medica Development Group, Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou, 730000, China
| | - Wenjie Fu
- Materia Medica Development Group, Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou, 730000, China
| | - Wei Li
- Materia Medica Development Group, Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou, 730000, China
| | - Quanyi Zhao
- Materia Medica Development Group, Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou, 730000, China.
| | - Dian He
- Materia Medica Development Group, Institute of Medicinal Chemistry, School of Pharmacy of Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
6
|
Benramdane S, De Loose J, Filippi N, Espadinha M, Beyens O, Rymenant YV, Dirkx L, Bozdag M, Feijens PB, Augustyns K, Caljon G, De Winter H, De Meester I, Van der Veken P. Highly Selective Inhibitors of Dipeptidyl Peptidase 9 (DPP9) Derived from the Clinically Used DPP4-Inhibitor Vildagliptin. J Med Chem 2023; 66:12717-12738. [PMID: 37721854 DOI: 10.1021/acs.jmedchem.3c00609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Dipeptidyl peptidase 9 (DPP9) is a proline-selective serine protease that plays a key role in NLRP1- and CARD8-mediated inflammatory cell death (pyroptosis). No selective inhibitors have hitherto been reported for the enzyme: all published molecules have grossly comparable affinities for DPP8 and 9 because of the highly similar architecture of these enzymes' active sites. Selective DPP9 inhibitors would be highly instrumental to address unanswered research questions on the enzyme's role in pyroptosis, and they could also be investigated as therapeutics for acute myeloid leukemias. Compounds presented in this manuscript (42 and 47) combine low nanomolar DPP9 affinities with unprecedented DPP9-to-DPP8 selectivity indices up to 175 and selectivity indices >1000 toward all other proline-selective proteases. To rationalize experimentally obtained data, a molecular dynamics study was performed. We also provide in vivo pharmacokinetics data for compound 42.
Collapse
Affiliation(s)
- Siham Benramdane
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Joni De Loose
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Nicolò Filippi
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Margarida Espadinha
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Olivier Beyens
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Yentl Van Rymenant
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Murat Bozdag
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Pim-Bart Feijens
- Laboratory of Microbiology, Parasitology and Hygiene, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Pieter Van der Veken
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
7
|
Göktürk T, Sakallı Çetin E, Hökelek T, Pekel H, Şensoy Ö, Aksu EN, Güp R. Synthesis, Structural Investigations, DNA/BSA Interactions, Molecular Docking Studies, and Anticancer Activity of a New 1,4-Disubstituted 1,2,3-Triazole Derivative. ACS OMEGA 2023; 8:31839-31856. [PMID: 37692230 PMCID: PMC10483525 DOI: 10.1021/acsomega.3c03355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023]
Abstract
We report herein a new 1,2,3-triazole derivative, namely, 4-((1-(3,4-dichlorophenyl)-1H-1,2,3-triazol-4-yl)methoxy)-2-hydroxybenzaldehyde, which was synthesized by copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC). The structure of the compound was analyzed using Fourier transform infrared spectroscopy (FTIR), 1H NMR, 13C NMR, UV-vis, and elemental analyses. Moreover, X-ray crystallography studies demonstrated that the compound adapted a monoclinic crystal system with the P21/c space group. The dominant interactions formed in the crystal packing were found to be hydrogen bonding and van der Waals interactions according to Hirshfeld surface (HS) analysis. The volume of the crystal voids and the percentage of free spaces in the unit cell were calculated as 152.10 Å3 and 9.80%, respectively. The evaluation of energy frameworks showed that stabilization of the compound was dominated by dispersion energy contributions. Both in vitro and in silico investigations on the DNA/bovine serum albumin (BSA) binding activity of the compound showed that the CT-DNA binding activity of the compound was mediated via intercalation and BSA binding activity was mediated via both polar and hydrophobic interactions. The anticancer activity of the compound was also tested by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay using human cell lines including MDA-MB-231, LNCaP, Caco-2, and HEK-293. The compound exhibited more cytotoxic activity than cisplatin and etoposide on Caco-2 cancer cell lines with an IC50 value of 16.63 ± 0.27 μM after 48 h. Annexin V suggests the induction of cell death by apoptosis. Compound 3 significantly increased the loss of mitochondrial membrane potential (MMP) levels in Caco-2 cells, and the reactive oxygen species (ROS) assay proved that compound 3 could induce apoptosis by ROS generation.
Collapse
Affiliation(s)
- Tolga Göktürk
- Department
of Chemistry, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Esin Sakallı Çetin
- Department
of Medical Biology, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Tuncer Hökelek
- Department
of Physics, Hacettepe University, 06800 Ankara, Türkiye
| | - Hanife Pekel
- Department
of Pharmacy Services, Vocational School of Health Services, Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Özge Şensoy
- Department
of Computer Engineering, Istanbul Medipol
University, 34000 Istanbul, Türkiye
| | - Ebru Nur Aksu
- Department
of Medical Biology, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Ramazan Güp
- Department
of Chemistry, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| |
Collapse
|
8
|
Alexeev AA, Nurieva EV, Elisseev IA, Milaeva ER, Lyssenko KA, Zefirova ON. Bicyclic isothioureas for conjugation with tubulin targeted anticancer agents. MENDELEEV COMMUNICATIONS 2022. [DOI: 10.1016/j.mencom.2022.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
9
|
Synthesis of 1,2,3-Triazolyl-Substituted Derivatives of the Alkaloids Sinomenine and Tetrahydrothebaine on Ring A and Their Analgesic Activity. Chem Nat Compd 2022. [DOI: 10.1007/s10600-022-03824-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
Goel B, Dey B, Chatterjee E, Tripathi N, Bhardwaj N, Kumar S, Guru SK, Jain SK. Antiproliferative Potential of Gloriosine: A Lead for Anticancer Drug Development. ACS OMEGA 2022; 7:28994-29001. [PMID: 36033689 PMCID: PMC9404168 DOI: 10.1021/acsomega.2c02688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
Gloriosine, a colchicine-like natural product, is widely obtained from Gloriosa superba roots. Despite having remarkable anticancer potential, colchicine could not pave its way to the clinic, while gloriosine is yet to be investigated for its pharmacological effects. In the present work, 14 compounds, including gloriosine, were isolated from the G. superba roots and were characterized by NMR spectroscopy. Gloriosine (11) was evaluated for its antiproliferative activity against a panel of 15 human cancer cell lines of different tissues and normal breast cells. Gloroisine (11) displayed significant antiproliferative activity against various cancer cell lines selectively, with IC50 values ranging from 32.61 to 100.28 nM. Further, gloriosine (11) was investigated for its apoptosis-inducing ability and found to form apoptotic bodies. It also inhibited A549 cell migration in the wound healing assay. Finally, molecular docking studies were performed to explore the possible binding modes of gloriosine with the colchicine-binding site of tubulin protein. Our findings suggested that gloriosine might be a potential lead for anticancer drug discovery.
Collapse
Affiliation(s)
- Bharat Goel
- Department
of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Biswajit Dey
- Department
of Biological Sciences, National Institute
of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Essha Chatterjee
- Department
of Biological Sciences, National Institute
of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Nancy Tripathi
- Department
of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Nivedita Bhardwaj
- Department
of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Sanjay Kumar
- Department
of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| | - Santosh Kumar Guru
- Department
of Biological Sciences, National Institute
of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Shreyans K. Jain
- Department
of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|