Generic placeholder image

Clinical Cancer Drugs

Editor-in-Chief

ISSN (Print): 2212-697X
ISSN (Online): 2212-6988

Research Article

A Marine-based Meriolin (3-Pyrimidinylazaindole) Derivative (4ab) Targets PI3K/AKT /mTOR Pathway Inducing Cell Cycle Arrest and Apoptosis in Molt-4 Cells

Author(s): Gousia Chashoo*, Umed Singh, Parvinder P. Singh, Dilip M. Mondhe and Ram A. Vishwakarma

Volume 6, Issue 1, 2019

Page: [33 - 40] Pages: 8

DOI: 10.2174/2212697X06666190509094514

Abstract

Background: Cyclin-dependent kinases play a central role in the control of cell division and therefore it is not surprising that cancer exhibits some features that disturb the normal controls over the cell cycle. Previous studies related to the development of 3-Pyrimidinylazaindole (Meriolin) derivatives as novel Cyclin dependent kinase inhibitors highlighted 4ab as the most potent inhibitor.

Objective: The main objective of the current study was to understand the mode of cell death and the effect of 4ab on major cellular networking pathways in cancer.

Methods: Preliminary apoptotic studies were carried out using flowcytometer and electron microscope. The effect on cellular signalling was studied via western blotting.

Results: 4ab was found to inhibit the enzymatic activity of CDK2. The inhibition of CDK2 activity was found to be associated with the down-regulation of P-cdc-25 and arrest of cells in G0-G1 phase of the cell cycle in lymphoblastic leukemia cells. Further, 4ab was found to affect AKT-mToR pathway by down-regulating the expression of major proteins including P-m-TOR (2448), P110α, P-AKT (S473) and P-p-70S6K.

Conclusion: Current study shows that the potent anticancer potential of 4ab is mediated via cellular apoptosis, dysregulation of mitochondrial membrane potential and arrest of G1 phase in Molt-4 cells. Further, target-based studies showed the effect of 4ab on one of the major cellular signalling pathways deregulated in cancer.

Keywords: CDK, meriolin, PI3k/AKT/mTOR signalling, cell cycle, apoptosis, Molt-4.

Graphical Abstract
[1]
Asghar U, Witkiewicz AK, Turner NC, Knudsen ES. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov 2015; 14: 130-46.
[2]
Heptinstall AB, Adiyasa IWS, Cano C, Hardcastle IR. Recent advances in CDK inhibitors for cancer therapy. Future Med Chem 2018; 10: 1369-88.
[3]
Newman DJ, Cragg GM. Natural products as sources of new drugs from 1981 to 2014. J Nat Prod 2016; 79: 629-61.
[4]
Bharate SB, Sawant SD, Singh PP, Vishwakarma RA. Kinase inhibitors of marine origin. Chem Rev 2013; 113: 6761-815.
[5]
Singh U, Chashoo G, Khan SU, et al. Design of novel 3-Pyrimidinylazaindole CDK2/9 inhibitors with potent in vitro and in vivo antitumor efficacy in a triple-negative breast cancer model. J Med Chem 2017; 60: 9470-89.
[6]
Meerloo VJ, Kaspers GJL, Cloos J. Cell Sensitivity assays: The MTT assay In: Cree I, Ed. Cancer Cell Culture Methods in Molecular Biology (Methods and Protocols). 2000; p. 731.
[7]
Pozarowski P, Darzynkiewicz Z. Analysis of cell cycle by flow cytometry methods. Mol Biol 2004; 281: 301-11.
[8]
Chashoo G, Singh SK, Sharma PR, et al. A propionyloxy derivative of 11-keto-β-boswellic acid induces apoptosis in HL-60 cells mediated through topoisomerase I & II inhibition. Chem Biol Interact 2011; 189: 60-71.
[9]
Kumar A, Qayum A, Sharma PR, Singh SK, Shah BA. Synthesis of b-boswellic acid derivatives as cytotoxic and apoptotic agents. Bioorg Med Chem Lett 2016; 26: 76-81.
[10]
Majeed R, Hamid A, Sangwan PL, et al. Inhibition of phosphotidylinositol-3 kinase pathway by a novel naphthol derivative of betulinic acid induces cell cycle arrest and apoptosis in cancer cells of different origin. Cell Death Dis 2014; 5e1459
[11]
Pozzi S, Cirstea D, Santo L, et al. CYC065, A potent derivative of seliciclib is active in multiple myeloma in preclinical studies. Blood 2010; 116: 2999.
[12]
Ingrid AM, Arteaga CL. The Pi3k/Akt Pathway As A Target For Cancer Treatment ingrid A. Annu Rev Med 2016; 67: 11-28.
[13]
Foster KG, Fingar DC. Mammalian Target of Rapamycin (mTOR): conducting the cellular signaling symphony. J Biol Chem 2010; 285: 14071-7.
[14]
Wang Q, Chen X, Hay N. Akt as a target for cancer therapy: More is not always better (lessons from studies in mice). Br J Cancer 2017; 17: 159-63.
[15]
Liang J, Zubovitz J, Petrocelli T, et al. PKB/Akt phosphorylates p27, impairs nuclear import of p27 and opposes p27-mediated G1 arrest. Nat Med 2002; 8: 1153-60.
[16]
Maddika S, Panigrahi S, Wiechec E, Wesselborg S, Fischer U, Schulze-Osthoff K. Los Marek. Unscheduled Akt-triggered activation of cyclin-dependent kinase 2 as a key effector mechanism of apoptin’s anticancer toxicity. Mol Cell Biol 2009; 29: 1235-48.
[17]
Saxton RA, Sabatini DM. mtor signaling in growth, metabolism, and disease. Cell 2017; 169: 361-71.
[18]
Ilagan E, Manning DB. Emerging role of mTOR in the response to cancer therapeutics. Trends Cancer 2016; 2: 241-51.

© 2024 Bentham Science Publishers | Privacy Policy