Small molecule 6''-O-Malonylglycitin inhibits ERK2 activity and promotes cytotoxicity in colon adenocarcinoma cells paving for the colon cancer treatment
Abstract
Background: Extracellular signal-regulated kinase 2 (ERK2), an important component of the MAPK signalling pathway, plays an critical role in the proliferation & survival of colon adenocarcinoma cells. Inhibition of ERK2 can interrupt these oncogenic pathways, offering a capable therapeutic strategy. Natural product-derived small molecules offer substantial potential in this context due to their favourable bioavailability & typically low toxicity.
Methods: This study examines the inhibitory effects of 6''-O-Malonylglycitin on ERK2 kinase activity & its cytotoxic effects on HCT-15 colon adenocarcinoma cells. The crystal structure of ERK2 (PDB ID: 2OJJ) was prepared for molecular docking studies, and 188 isoflavone natural compounds were screened using AutoDock Vina. Among these, 6''-O-Malonylglycitin exhibited the highest binding affinity (-9.3 kcal/mol) with ERK2, forming multiple hydrogen bonds & van der Waals interactions.
Results: The compound was evaluated for cytotoxicity by using an MTT assay, showing a dose-dependent reduction in cell viability with an IC50 of 15.33 µM. Time-dependent studies presented demonstrated significant decreases in cell viability up to 48 hours. Quantitative PCR indicated a significant reduction in ERK2 mRNA expression following treatment with 6''-O-Malonylglycitin. Kinase inhibition assay confirmed a concentration-dependent decrease in ERK2 activity.
Conclusion: The findings indicate that 6''-O-malonylglycitin effectively inhibits ERK2 kinase activity and exhibits potent cytotoxic effects against HCT-15 colon adenocarcinoma cells, suggesting its potential as a therapeutic agent for colon cancer.
Key words: Colon adenocarcinoma; ERK2; HCT-15 cells; small molecule inhibitors; 6''-O-Malonylglycitin
Full Text:
PDFReferences
Parascandolo A, Benincasa G, Corcione F, Laukkanen MO. ERK2 Is a Promoter of Cancer Cell Growth and Migration in Colon Adenocarcinoma. Antioxidants, (2024); 13(1): 119.
Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell research, (2002); 12(1): 9-18.
Roux PP, & Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiology and molecular biology reviews: MMBR, (2004); 68(2): 320-344.
Sugiura R, Satoh R, & Takasaki T. ERK: A Double-Edged Sword in Cancer. ERK-Dependent Apoptosis as a Potential Therapeutic Strategy for Cancer. Cells, (2021); 10(10): 2509.
Cicenas J, Zalyte E, Rimkus A, Dapkus D, Noreika R, & Urbonavicius S. JNK, p38, ERK, and SGK1 Inhibitors in Cancer. Cancers, (2017); 10(1): 1.
Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y, Hu LL. ERK/MAPK signalling pathway and tumorigenesis. Experimental and therapeutic medicine, (2020); 19(3): 1997-2007.
Gupta J, del Barco Barrantes I, Igea A, Sakellariou S, Pateras IS, Gorgoulis VG, Nebreda AR. Dual function of p38α MAPK in colon cancer: suppression of colitis-associated tumor initiation but requirement for cancer cell survival. Cancer cell, (2014); 25(4): 484-500.
Pranteda A, Piastra V, Stramucci L, Fratantonio D, Bossi G. The p38 MAPK Signaling Activation in Colorectal Cancer upon Therapeutic Treatments. International journal of molecular sciences, (2020); 21(8): 2773.
Qi XM, Chen G. p38γ MAPK Inflammatory and Metabolic Signaling in Physiology and Disease. Cells, (2023); 12(13): 1674.
Martínez-Limón A, Joaquin M, Caballero M, Posas F, de Nadal E. The p38 Pathway: From Biology to Cancer Therapy. International journal of molecular sciences, (2020); 21(6): 1913.
Raucci A, Laplantine E, Mansukhani A, Basilico C. Activation of the ERK1/2 and p38 mitogen-activated protein kinase pathways mediates fibroblast growth factor-induced growth arrest of chondrocytes. The Journal of biological chemistry, (2024); 279(3): 1747-1756.
Shraim BA, Moursi MO, Benter IF, Habib AM, Akhtar S. The Role of Epidermal Growth Factor Receptor Family of Receptor Tyrosine Kinases in Mediating Diabetes-Induced Cardiovascular Complications. Frontiers in pharmacology, (2021); 12: 701390.
Kong T, Liu M, Ji B, Bai B, Cheng B, & Wang C. Role of the Extracellular Signal-Regulated Kinase 1/2 Signaling Pathway in Ischemia-Reperfusion Injury. Frontiers in physiology, (2019); 10, 1038.
Lake D, Corrêa SA, Müller J. Negative feedback regulation of the ERK1/2 MAPK pathway. Cellular and molecular life sciences: CMLS, (2016); 73(23): 4397-4413.
Lee MW, Bach JH, Lee HJ, Lee DY, Joo WS, Kim YS, et al. The activation of ERK1/2 via a tyrosine kinase pathway attenuates trail-induced apoptosis in HeLa cells. Cancer investigation, (2005); 23(7): 586-592.
Guégan JP, Frémin C, Baffet G. The MAPK MEK1/2-ERK1/2 Pathway and Its Implication in Hepatocyte Cell Cycle Control. International journal of hepatology, (2012); 2012: 328372.
Marampon F, Ciccarelli C, Zani BM. Biological Rationale for Targeting MEK/ERK Pathways in Anti-Cancer Therapy and to Potentiate Tumour Responses to Radiation. International journal of molecular sciences, (2019); 20(10): 2530.
Martin-Vega A, & Cobb MH. Navigating the ERK1/2 MAPK Cascade. Biomolecules, (2023); 13(10): 1555.
Roskoski R, Jr. ERK1/2 MAP kinases: structure, function, and regulation. Pharmacological research, (2012); 66(2): 105-143.
Sipieter F, Cappe B, Leray A, De Schutter E, Bridelance J, Hulpiau P, et al. Characteristic ERK1/2 signaling dynamics distinguishes necroptosis from apoptosis. iScience, (2021); 24(9): 103074.
Sugiura R, Satoh R, Takasaki T. ERK: A Double-Edged Sword in Cancer. ERK-Dependent Apoptosis as a Potential Therapeutic Strategy for Cancer. Cells, (2021); 10(10): 2509.
Zou J, Lei T, Guo P, Yu J, Xu Q, Luo Y, et al. Mechanisms shaping the role of ERK1/2 in cellular senescence (Review). Molecular medicine reports, (2019); 19(2): 759-770.
McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Montalto G, Cervello M, et al. Mutations and deregulation of Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascades which alter therapy response. Oncotarget, (2012); 3(9): 954-987.
Van Emburgh BO, Sartore-Bianchi A, Di Nicolantonio F, Siena S, Bardelli A. Acquired resistance to EGFR-targeted therapies in colorectal cancer. Molecular oncology, (2014); 8(6): 1084-1094.
Zhou J, Ji Q, Li Q. Resistance to anti-EGFR therapies in metastatic colorectal cancer: underlying mechanisms and reversal strategies. Journal of experimental & clinical cancer research: CR, (2021); 40(1): 328.
Kohno M, Pouyssegur J. Targeting the ERK signaling pathway in cancer therapy. Annals of medicine, (2006); 38(3): 200-211.
Grogan L, Shapiro P. Progress in the development of ERK1/2 inhibitors for treating cancer and other diseases. Advances in pharmacology (San Diego, Calif.), (2024); 100: 181-207.
Timofeev O, Giron P, Lawo S, Pichler M, Noeparast M. ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework. NPJ precision oncology, (2024); 8(1): 70.
Aronov AM, Baker C, Bemis GW, Cao J, Chen G, Ford PJ, et al. Flipped out: structure-guided design of selective pyrazolylpyrrole ERK inhibitors. Journal of medicinal chemistry, (2007); 50(6): 1280-1287.
Salmaso V, Moro S. Bridging Molecular Docking to Molecular Dynamics in Exploring Ligand-Protein Recognition Process: An Overview. Frontiers in pharmacology, (2018); 9: 923.
Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of computational chemistry, (2010); 31(2): 455-461.
Wang P, Gong Y, Guo T, Li M, Fang L, Yin S, et al. Activation of Aurora A kinase increases YAP stability via blockage of autophagy. Cell death & disease, (2019); 10(6), 432.
Asma ST, Acaroz U, Imre K, Morar A, Shah SRA, Hussain SZ, et al. Natural Products/Bioactive Compounds as a Source of Anticancer Drugs. Cancers, (2022); 14(24): 6203.
Irfan Dar M, Qureshi MI, Zahiruddin S, Abass S, Jan B, Sultan A, et al. In Silico Analysis of PTP1B Inhibitors and TLC-MS Bioautography-Based Identification of Free Radical Scavenging and α-Amylase Inhibitory Compounds from Heartwood Extract of Pterocarpus marsupium. ACS omega, (2022); 7(50): 46156-46173.
Wu J, Li Y, He Q, Yang X. Exploration of the Use of Natural Compounds in Combination with Chemotherapy Drugs for Tumor Treatment. Molecules (Basel, Switzerland), (2023); 28(3): 1022.
Chen X, Xue B, Wahab S, Sultan A, Khalid M, Yang S. Structure-based molecular docking and molecular dynamics simulations study for the identification of dipeptidyl peptidase 4 inhibitors in type 2 diabetes. Journal of biomolecular structure & dynamics, (2023); 1-14.
Sultan A, Ali R, Sultan T, Ali S, Khan NJ, Parganiha A. Circadian clock modulating small molecules repurposing as inhibitors of SARS-CoV-2 Mpro for pharmacological interventions in COVID-19 pandemic. Chronobiology international, (2021); 38(7): 971-985.
Ahmad M, Das P, Ali R, Husain SA, Sultan A. Molecular docking, MD simulation and MM/GBSA predicted natural triterpene compound Ursolic Acid as a potential inhibitor of mitotic arrest deficient 2 like 1 (MAD2L1) for the therapeutics of celiac disease. Biochemical and Cellular Archives, (2024); 1557-1570.
Bhat SA, Siddiqui ZI, Parray ZA, Sultan A, Afroz M, Azam SA, et al. Naturally occurring HMGB1 inhibitor delineating the anti-hepatitis B virus mechanism of glycyrrhizin via in vitro and in silico studies. Journal of Molecular Liquids, (2022); 356: 119029.
Sultan A, Ali R, Ishrat R, Ali S. Anti-HIV and anti-HCV small molecule protease inhibitors in-silico repurposing against SARS-CoV-2 Mpro for the treatment of COVID-19. Journal of biomolecular structure & dynamics, (2022); 40(23): 12848-12862.
Daina A, Michielin O, & Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific reports, (2017); 7: 42717.
Nada H, Choi Y, Kim S, Jeong KS, Meanwell NA, Lee K. New insights into protein-protein interaction modulators in drug discovery and therapeutic advance. Signal transduction and targeted therapy, (2024); 9(1): 341.
Adan A, Kiraz Y, Baran Y. Cell Proliferation and Cytotoxicity Assays. Current pharmaceutical biotechnology, (2016); 17(14): 1213-1221.
Ghasemi M, Turnbull T, Sebastian S, Kempson I. The MTT Assay: Utility, Limitations, Pitfalls, and Interpretation in Bulk and Single-Cell Analysis. International journal of molecular sciences, (2021); 22(23): 12827.
Barba-Ostria C, Carrera-Pacheco SE, Gonzalez-Pastor R, Heredia-Moya J, Mayorga-Ramos A, Rodríguez-Pólit C, et al. Evaluation of Biological Activity of Natural Compounds: Current Trends and Methods. Molecules, (2022); 27(14), 4490.
Issa NT, Wathieu H, Ojo A, Byers SW, Dakshanamurthy S. Drug Metabolism in Preclinical Drug Development: A Survey of the Discovery Process, Toxicology, and Computational Tools. Current drug metabolism, (2017); 18(6): 556-565.
Qin S, Tang X, Chen Y, Chen K, Fan N, Xiao W, et al. mRNA-based therapeutics: powerful and versatile tools to combat diseases. Signal transduction and targeted therapy, (2022); 7(1): 166.
Bai JP, Alekseyenko AV, Statnikov A, Wang IM, Wong PH. Strategic applications of gene expression: from drug discovery/development to bedside. The AAPS journal, (2013); 15(2): 427-437.
Khorkova O, Stahl J, Joji A, Volmar CH, Wahlestedt C. Amplifying gene expression with RNA-targeted therapeutics. Nature reviews. Drug discovery, (2023); 22(7): 539-561.
Fang JY, Richardson BC. The MAPK signalling pathways and colorectal cancer. The Lancet. Oncology, (2005); 6(5): 322-327.
Chaikuad A, Tacconi EM, Zimmer J, Liang Y, Gray NS, Tarsounas M, et al. A unique inhibitor binding site in ERK1/2 is associated with slow binding kinetics. Nature chemical biology, (2014); 10(10): 853-860.
Germann UA, Furey BF, Markland W, Hoover RR, Aronov AM, Roix JJ, Hale M, et al. Targeting the MAPK Signaling Pathway in Cancer: Promising Preclinical Activity with the Novel Selective ERK1/2 Inhibitor BVD-523 (Ulixertinib). Molecular cancer therapeutics, (2017); 16(11): 2351-2363.
Alshehri MM, Sharifi-Rad J, Herrera-Bravo J, Jara EL, Salazar LA, Kregiel D, et al. Therapeutic Potential of Isoflavones with an Emphasis on Daidzein. Oxidative medicine and cellular longevity, (2021); 2021: 6331630.
Magee PJ, Allsopp P, Samaletdin A, Rowland IR. Daidzein, R-(+) equol and S-(-) equol inhibit the invasion of MDA-MB-231 breast cancer cells potentially via the down-regulation of matrix metalloproteinase-2. European journal of nutrition, (2014); 53(1): 345-350.
Pavese JM, Farmer RL, Bergan RC. Inhibition of cancer cell invasion and metastasis by genistein. Cancer metastasis reviews, (2010); 29(3): 465-482.
Tuli HS, Tuorkey MJ, Thakral F, Sak K, Kumar M, Sharma AK, et al. Molecular Mechanisms of Action of Genistein in Cancer: Recent Advances. Frontiers in pharmacology, (2019); 10, 1336.
DOI: http://dx.doi.org/10.62940/als.v12i4.3765
Refbacks
- There are currently no refbacks.