1
|
Janković N, Ristovski J, Žižak Ž, Radan M, Cvijić S, Nikolić K, Ignjatović NL. Designing and the anticancer activity of chitosan and chitosan oligosaccharide lactate nanobeads loaded with Biginelli hybrid. RSC Adv 2024; 14:31526-31534. [PMID: 39372042 PMCID: PMC11450446 DOI: 10.1039/d4ra05783j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024] Open
Abstract
This study focuses on the designing and characterization, and anticancer evaluation of chitosan-based nanoparticles (NPs) loaded (enriched) with a Biginelli hybrid compound (BH). NPs based on chitosan (CH) or chitosan oligosaccharide lactate (CHOL), are carefully designed to encapsulate a tetrahydropyrimidine derivative (BH) with already proven anticancer properties. The formulations were evaluated for their physicochemical properties, including particle size distribution and morphology, using techniques such as infrared spectroscopy, scanning electron microscopy, and X-ray diffraction. The cytotoxicity profiles were assessed on different cancer cell lines, showing a higher selectivity towards HeLa and A549 cells related to BH. BH-CH showed better cytotoxic profile related to BH-CHOL NPs. A cell cycle analysis revealed an accumulation of cells in the G2/M phase after a treatment with these NPs, indicating the ability to induce mitotic arrest in cancer cells. In summary, the results underscore the promising application of CH-based natural nanocarriers for the targeted delivery of Biginelli hybrids, showcasing a significant potential for further in vivo testing.
Collapse
Affiliation(s)
- Nenad Janković
- University of Kragujevac, Department of Science, Institute for Information Technologies Kragujevac Jovana Cvijića bb Kragujevac 34000 Serbia
| | - Jovana Ristovski
- University of Novi Sad, Faculty of Medicine Hajduk Veljkova 3 Novi Sad 21000 Serbia
| | - Željko Žižak
- Institute of Oncology and Radiology of Serbia Pasterova 14 Belgrade 11000 Serbia
| | - Milica Radan
- The Institute for the Study of Medicinal Herbs "Dr Josif Pančić" Tadeuša Košćuška 1 Belgrade 11000 Serbia
| | - Sandra Cvijić
- University of Belgrade, Faculty of Pharmacy, Department of Pharmaceutical Technology and Cosmetology Vojvode Stepe 450 Belgrade 11221 Serbia
| | - Katarina Nikolić
- University of Belgrade, Faculty of Pharmacy, Department of Pharmaceutical Chemistry Vojvode Stepe 450 Belgrade 11221 Serbia
| | - Nenad L Ignjatović
- Institute of Technical Sciences of the Serbian Academy of Sciences and Arts Knez Mihailova 35/IV, P.O. Box 377 Belgrade Serbia
| |
Collapse
|
2
|
Shahabipour S, Shamkhali AN, Razzaghi-Asl N. Cytotoxic monastrol derivatives as adjective inhibitors of drug-resistant Eg5: a molecular dynamics perspective. J Biomol Struct Dyn 2024:1-14. [PMID: 38450658 DOI: 10.1080/07391102.2024.2326195] [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: 11/06/2023] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
The mitotic kinesin Eg5 is a motor protein involved in the formation of bipolar spindle and cell division. Eg5 is overexpressed in various cancer cells and Eg5 targeting agents are promising candidates for cancer therapy. Subsequent to the discovery of monastrol as a small-molecule Eg5 modulator, numerous inhibitors/modulators have been reported from which a few entered clinical trials. Mutagenic investigations specified declined sensitivity of Eg5 allosteric site to monastrol due to the occurrence of drug-resistant mutations in some cell cultures. Accordingly, identification of tight binders to the mutant Eg5 allosteric site is an invaluable strategy to devise more efficient Eg5 modulators. We have previously synthesized a few dihydropyrimidinethione (DHPMT)-based 5-carboxamide monastrol derivatives (1-5) with higher cytotoxicities against AGS (IC50 9.90-98.48 µM) and MCF-7 (IC50 15.20-149.13 µM) cancer cell lines than monastrol. Within a current study, a structural insight was offered into the binding mechanism of intended derivatives inside the mutant Eg5 loop5/α2/α3 allosteric pocket. Molecular docking of the DHPMT R and S-enantiomers unraveled top-scored Eg5 complexes. Molecular dynamics (MD) simulations were carried out on 5 superior complexes as (R)-2/D130V-Eg5, (R)-4/D130V-Eg5, (R)-5/D130V-Eg5, (R)-5/L214I-Eg5, (R)-5/R119L-Eg5, and the control groups monastrol/D130V-Eg5, monastrol/L214I-Eg5, monastrol/R119L-Eg5. Free energy calculations were conducted through conformational sampling of MD-driven binding trajectories. Our results provided structural details on probable interaction mechanism of the cytotoxic DHPMTs that are difficult to address experimentally. The outputs of the current study propose new monastrol derivatives as probable resistance-overwhelming Eg5 modulators.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- S Shahabipour
- Department of Applied Chemistry, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - A N Shamkhali
- Department of Applied Chemistry, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - N Razzaghi-Asl
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
3
|
Sebastian J, Raghav D, Rathinasamy K. MD simulation-based screening approach identified tolvaptan as a potential inhibitor of Eg5. Mol Divers 2022:10.1007/s11030-022-10482-w. [DOI: 10.1007/s11030-022-10482-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 06/10/2022] [Indexed: 11/28/2022]
|
4
|
Hong FU, Castro M, Linse K. Tumor-specific lytic path “hyperploid progression mediated death”: Resolving side effects through targeting retinoblastoma or p53 mutant. World J Clin Oncol 2020; 11:854-867. [PMID: 33312882 PMCID: PMC7701912 DOI: 10.5306/wjco.v11.i11.854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/08/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
A major advance was made to reduce the side effects of cancer therapy via the elucidation of the tumor-specific lytic path “hyperploid progression-mediated death” targeting retinoblastoma (Rb) or p53-mutants defective in G1 DNA damage checkpoint. The genetic basis of human cancers was uncovered through the cloning of the tumor suppressor Rb gene. It encodes a nuclear DNA-binding protein whose self-interaction is regulated by cyclin-dependent kinases. A 3D-structure of Rb dimer is shown, confirming its multimeric status. Rb assumes a central role in cell cycle regulation and the “Rb pathway” is universally inactivated in human cancers. Hyperploidy refers to a state in which cells contain one or more extra chromosomes. Hyperploid progression occurs due to continued cell-cycling without cytokinesis in G1 checkpoint-defective cancer cells. The evidence for the triggering of hyperploid progression-mediated death in RB-mutant human retinoblastoma cells is shown. Hence, the very genetic mutation that predisposes to cancer can be exploited to induce lethality. The discovery helped to establish the principle of targeted cytotoxic cancer therapy at the mechanistic level. By triggering the lytic path, targeted therapy with tumor specificity at the genetic level can be developed. It sets the stage for systematically eliminating side effects for cytotoxic cancer therapy.
Collapse
Affiliation(s)
- Frank-Un Hong
- Department of Research and Development, Bio-Synthesis, Lewisville, TX 75057, United States
| | - Miguel Castro
- Department of Research and Development, Bio-Synthesis, Lewisville, TX 75057, United States
| | - Klaus Linse
- Department of Research and Development, Bio-Synthesis, Lewisville, TX 75057, United States
| |
Collapse
|
5
|
Makala H, Ulaganathan V, Sivasubramanian A, Rajendran N, Subramanian S. Evaluating Phenyl Propanoids Isolated from Citrus medica as Potential Inhibitors for Mitotic kinesin Eg5. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817999200630125449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Human mitotic kinesins play an essential role in mitotic cell division. Targeting
the spindle separation phase of mitosis has gained much attention in cancer chemotherapy.
Spindle segregation is carried out mainly by the kinesin, Eg5. Many Eg5 inhibitors are in different
phases of clinical trials as cancer drugs. This enzyme has two allosteric binding sites to which the
inhibitors can bind. The first site is formed by loop L5, helix α2 and helix α3 and all the current drug
candidates bind un-competitively to this site with ATP/ADP. The second site, formed by helix α4
and helix α6, which has gained attention recently, has not been explored well. Some inhibitors that
bind to this site are competitive, while others are uncompetitive to ATP/ADP. Phenylpropanoids are
pharmacologically active secondary metabolites.
Methods:
In this study, we have evaluated fourteen phenyl propanoids extracted from Citrus medica
for inhibitory activity against human mitotic kinesin Eg5 in vitro steady-state ATPase assay. Ther
interactions and stability using molecular docking and molecular dynamics simulations.
Results and Discussions:
Of the fourteen compounds tested, naringin and quercetin showed good
activity with IC50 values in the micromolar range. Molecular docking studies of these complexes
showed that both the molecules interact with the key residues of the active site predominantly thorough
hydrophobic & aromatic π–π interactions consistent with the known inhibitors. Besides, these
molecules also form hydrogen bonding interactions stabilizing the complexes. Molecular dynamics
simulations of these complexes confirm the stability of these interactions.
Conclusion:
These results can be used as a strong basis for further modification of these compounds
to design new inhibitors with higher potency using structure-based drug design.
Collapse
Affiliation(s)
- Himesh Makala
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu-613401, India
| | - Venkatasubramanian Ulaganathan
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu-613401, India
| | - Aravind Sivasubramanian
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu-613401, India
| | - Narendran Rajendran
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu-613401, India
| | - Shankar Subramanian
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu-613401, India
| |
Collapse
|
6
|
Zeng X, Xu WK, Lok TM, Ma HT, Poon RYC. Imbalance of the spindle-assembly checkpoint promotes spindle poison-mediated cytotoxicity with distinct kinetics. Cell Death Dis 2019; 10:314. [PMID: 30952840 PMCID: PMC6450912 DOI: 10.1038/s41419-019-1539-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/24/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022]
Abstract
Disrupting microtubule dynamics with spindle poisons activates the spindle-assembly checkpoint (SAC) and induces mitotic cell death. However, mitotic exit can occur prematurely without proper chromosomal segregation or cytokinesis by a process termed mitotic slippage. It remains controversial whether mitotic slippage increases the cytotoxicity of spindle poisons or the converse. Altering the SAC induces either mitotic cell death or mitotic slippage. While knockout of MAD2-binding protein p31comet strengthened the SAC and promoted mitotic cell death, knockout of TRIP13 had the opposite effect of triggering mitotic slippage. We demonstrated that mitotic slippage prevented mitotic cell death caused by spindle poisons, but reduced subsequent long-term survival. Weakening of the SAC also reduced cell survival in response to spindle perturbation insufficient for triggering mitotic slippage, of which mitotic exit was characterized by displaced chromosomes during metaphase. In either mitotic slippage or mitotic exit with missegregated chromosomes, cell death occurred only after one cell cycle following mitotic exit and increased progressively during subsequent cell cycles. Consistent with these results, transient inhibition of the SAC using an MPS1 inhibitor acted synergistically with spindle perturbation in inducing chromosome missegregation and cytotoxicity. The specific temporal patterns of cell death after mitotic exit with weakened SAC may reconcile the contradictory results from many previous studies.
Collapse
Affiliation(s)
- Xiaofang Zeng
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.,Department of Oncology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wendy Kaichun Xu
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.,Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
| | - Tsun Ming Lok
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hoi Tang Ma
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y C Poon
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong.
| |
Collapse
|
7
|
Lang PY, Gershon TR. A New Way to Treat Brain Tumors: Targeting Proteins Coded by Microcephaly Genes?: Brain tumors and microcephaly arise from opposing derangements regulating progenitor growth. Drivers of microcephaly could be attractive brain tumor targets. Bioessays 2018; 40:e1700243. [PMID: 29577351 PMCID: PMC5910257 DOI: 10.1002/bies.201700243] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/12/2018] [Indexed: 02/06/2023]
Abstract
New targets for brain tumor therapies may be identified by mutations that cause hereditary microcephaly. Brain growth depends on the repeated proliferation of stem and progenitor cells. Microcephaly syndromes result from mutations that specifically impair the ability of brain progenitor or stem cells to proliferate, by inducing either premature differentiation or apoptosis. Brain tumors that derive from brain progenitor or stem cells may share many of the specific requirements of their cells of origin. These tumors may therefore be susceptible to disruptions of the protein products of genes that are mutated in microcephaly. The potential for the products of microcephaly genes to be therapeutic targets in brain tumors are highlighted hereby reviewing research on EG5, KIF14, ASPM, CDK6, and ATR. Treatments that disrupt these proteins may open new avenues for brain tumor therapy that have increased efficacy and decreased toxicity.
Collapse
Affiliation(s)
- Patrick Y. Lang
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Department of Neurology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Timothy R. Gershon
- Department of Neurology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
- Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
8
|
Matos LHS, Masson FT, Simeoni LA, Homem-de-Mello M. Biological activity of dihydropyrimidinone (DHPM) derivatives: A systematic review. Eur J Med Chem 2017; 143:1779-1789. [PMID: 29133039 DOI: 10.1016/j.ejmech.2017.10.073] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/23/2017] [Accepted: 10/27/2017] [Indexed: 12/21/2022]
Abstract
Dihydropyrimidinones are heterocycles with a pyrimidine moiety in the ring nucleus, which, in recent decades, have aroused interest in medicinal chemistry due to alleged versatile biological activity. In this systematic review, we describe the currently published activities of dihydropyrimidinone derivatives. Between 1990 and December 31st, 2016, 115 articles outlined biological activities or toxicity of DHPM derivatives, 12 of those involved in vivo experiments. The main activities associated with this class of compounds are antitumoral (43 articles), anti-inflammatory (12 articles), antibacterial (20 articles) and calcium channel antagonism/inhibition (14 articles). Antitumoral activity is the main biological property evaluated, since the main representative compound of this class (monastrol) is a known Eg5 kinesin inhibitor. This review depicts a variety of other pharmacological activities associated with DHPM derivatives, but the main findings are essentially in vitro characteristics of the substances. This review presents the current state of the art of DHPM biological activities and demonstrates that there is still a need for further in vivo studies to better delineate the pharmacological potential of this class of substances.
Collapse
Affiliation(s)
| | - Flávia Teixeira Masson
- Department of Pharmaceutical Sciences, Health Sciences School, University of Brasilia, Brazil
| | - Luiz Alberto Simeoni
- Department of Pharmaceutical Sciences, Health Sciences School, University of Brasilia, Brazil
| | - Mauricio Homem-de-Mello
- Department of Pharmaceutical Sciences, Health Sciences School, University of Brasilia, Brazil.
| |
Collapse
|
9
|
Antiproliferative activity of monastrol in human adenocarcinoma (MCF-7) and non-tumor (HB4a) breast cells. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:1279-1288. [PMID: 27592117 DOI: 10.1007/s00210-016-1292-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022]
Abstract
Monastrol is an allosteric inhibitor of the mitotic kinesin Eg5 that exhibits an antiproliferative effect against several cell lines. We investigated the antiproliferative effect of monastrol on human breast adenocarcinoma cells (MCF-7) and mammary epithelial cells (HB4a, non-tumoral). Monastrol treatment decreased cell viability only in MCF-7 tumor cells. Real-time cell growth kinetic analysis showed a decrease in the proliferation of MCF-7 cells exposed to monastrol, while in the HB4a cells, only a concentration of 100 μM was able to induce this effect. In a cell cycle analysis, exposure of MCF-7 cells to monastrol led to an increased population of cells in both the G1 and G2/M phases. In HB4a cells, the proportion of cells in the G2/M phase was increased. Monastrol led to an increased mitotic index in both cell lines. Monastrol was not able to induce cell death by apoptosis in any of the cell lines studied. Gene expression analysis was performed to measure the mRNA levels of cell cycle genes, DNA damage indicator gene, and apoptotic related genes. Treatment with monastrol induced in MCF-7 cells a 5-fold increase in the mRNA levels of the CDKN1A gene, an inhibitor of CDKs related with cell cycle arrest in response a stress stimulus, and a 2-fold decrease in CDKN1C mRNA levels in HB4a cells. These results provide evidence that monastrol has a greater antiproliferative effect on MCF-7 tumor cells compared with non-tumor HB4a cells; however, no selective is observed.
Collapse
|
10
|
Lenhart R, Kirov S, Desilva H, Cao J, Lei M, Johnston K, Peterson R, Schweizer L, Purandare A, Ross-Macdonald P, Fairchild C, Wong T, Wee S. Sensitivity of Small Cell Lung Cancer to BET Inhibition Is Mediated by Regulation of ASCL1 Gene Expression. Mol Cancer Ther 2015; 14:2167-74. [PMID: 26253517 DOI: 10.1158/1535-7163.mct-15-0037] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 08/03/2015] [Indexed: 11/16/2022]
Abstract
The BET (bromodomain and extra-terminal) proteins bind acetylated histones and recruit protein complexes to promote transcription elongation. In hematologic cancers, BET proteins have been shown to regulate expression of MYC and other genes that are important to disease pathology. Pharmacologic inhibition of BET protein binding has been shown to inhibit tumor growth in MYC-dependent cancers, such as multiple myeloma. In this study, we demonstrate that small cell lung cancer (SCLC) cells are exquisitely sensitive to growth inhibition by the BET inhibitor JQ1. JQ1 treatment has no impact on MYC protein expression, but results in downregulation of the lineage-specific transcription factor ASCL1. SCLC cells that are sensitive to JQ1 are also sensitive to ASCL1 depletion by RNAi. Chromatin immunoprecipitation studies confirmed the binding of the BET protein BRD4 to the ASCL1 enhancer, and the ability of JQ1 to disrupt the interaction. The importance of ASCL1 as a potential driver oncogene in SCLC is further underscored by the observation that ASCL1 is overexpressed in >50% of SCLC specimens, an extent greater than that observed for other putative oncogenes (MYC, MYCN, and SOX2) previously implicated in SCLC. Our studies have provided a mechanistic basis for the sensitivity of SCLC to BET inhibition and a rationale for the clinical development of BET inhibitors in this disease with high unmet medical need.
Collapse
Affiliation(s)
- Ryan Lenhart
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Stefan Kirov
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | | | - Jian Cao
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Ming Lei
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | | | | | | | | | | | | | - Tai Wong
- Bristol-Myers Squibb Company, Princeton, New Jersey
| | - Susan Wee
- Bristol-Myers Squibb Company, Princeton, New Jersey.
| |
Collapse
|
11
|
Haschka MD, Soratroi C, Kirschnek S, Häcker G, Hilbe R, Geley S, Villunger A, Fava LL. The NOXA-MCL1-BIM axis defines lifespan on extended mitotic arrest. Nat Commun 2015; 6:6891. [PMID: 25922916 PMCID: PMC4423218 DOI: 10.1038/ncomms7891] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 03/09/2015] [Indexed: 11/09/2022] Open
Abstract
Cell death on extended mitotic arrest is considered arguably most critical for the efficacy of microtubule-targeting agents (MTAs) in anticancer therapy. While the molecular machinery controlling mitotic arrest on MTA treatment, the spindle assembly checkpoint (SAC), appears well defined, the molecular components executing cell death, as well as factors connecting both networks remain poorly understood. Here we conduct a mini screen exploring systematically the contribution of individual BCL2 family proteins at single cell resolution to death on extended mitotic arrest, and demonstrate that the mitotic phosphorylation of BCL2 and BCLX represent a priming event for apoptosis that is ultimately triggered by NOXA-dependent MCL1 degradation, enabling BIM-dependent cell death. Our findings provide a comprehensive model for the initiation of apoptosis in cells stalled in mitosis and provide a molecular basis for the increased efficacy of combinatorial treatment of cancer cells using MTAs and BH3 mimetics. Cells experiencing extended mitotic arrest often undergo cell death as a result of steadily declining levels of the apoptotic inhibitor MCL1, but the mechanism controlling this process is poorly understood. Here, Haschka et al. show that the BH3-only protein NOXA promotes the degradation of MCL1, enabling BIM-dependent cell death.
Collapse
Affiliation(s)
- Manuel D Haschka
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Claudia Soratroi
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Susanne Kirschnek
- Institute for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Georg Häcker
- Institute for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Richard Hilbe
- Division of Molecular Pathophysiology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Luca L Fava
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
12
|
Choi M, Kim W, Cheon MG, Lee CW, Kim JE. Polo-like kinase 1 inhibitor BI2536 causes mitotic catastrophe following activation of the spindle assembly checkpoint in non-small cell lung cancer cells. Cancer Lett 2015; 357:591-601. [PMID: 25524551 DOI: 10.1016/j.canlet.2014.12.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/05/2014] [Accepted: 12/09/2014] [Indexed: 01/01/2023]
Abstract
Polo-like kinase 1 (PLK1), a critical kinase that regulates multiple steps in mitosis, is overexpressed in diverse human cancers; thus many PLK1 inhibitors have been developed as potential cancer therapeutic agents. One of these compounds, the PLK1-specific inhibitor BI2536, has been investigated as a cytotoxic drug in several cancers, including lung cancer; however, the detailed mechanism by which BI2536 induces defects in cell proliferation of non-small cell lung cancer (NSCLC) has not yet been determined. We found that BI2536 treatment resulted in mitotic arrest due to improper formation of the mitotic spindles and mitotic centrosomes. The unattached kinetochores in BI2536-treated NSCLC cells activated the spindle assembly checkpoint (SAC). The prolonged activation of the SAC led to a type of apoptotic cell death referred to as mitotic catastrophe. Finally, BI2536-treated NSCLC cells show a defect in cell proliferation. Overall, these data indicate that PLK1 inhibition via mitotic disruption represents a potential approach for the treatment of NSCLC.
Collapse
Affiliation(s)
- Minji Choi
- Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 130-701, Korea; Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 130-701, Korea
| | - Wootae Kim
- Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 130-701, Korea; Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 130-701, Korea
| | - Min Gyeong Cheon
- Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 130-701, Korea; Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 130-701, Korea
| | - Chang Woo Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi 440-746, Korea
| | - Ja Eun Kim
- Department of Pharmacology, School of Medicine, Kyung Hee University, Seoul 130-701, Korea; Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 130-701, Korea.
| |
Collapse
|
13
|
Polyakov VR, Moorcroft ND, Drawid A. Enrichment Analysis for Discovering Biological Associations in Phenotypic Screens. J Chem Inf Model 2014; 54:377-86. [DOI: 10.1021/ci400245c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Valery R. Polyakov
- Sanofi, 55 Corporate Drive, Bridgewater, New Jersey 08807, United States
| | - Neil D. Moorcroft
- Sanofi, 55 Corporate Drive, Bridgewater, New Jersey 08807, United States
| | - Amar Drawid
- Sanofi, 55 Corporate Drive, Bridgewater, New Jersey 08807, United States
| |
Collapse
|
14
|
Salmela AL, Kallio MJ. Mitosis as an anti-cancer drug target. Chromosoma 2013; 122:431-49. [PMID: 23775312 DOI: 10.1007/s00412-013-0419-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 05/23/2013] [Accepted: 05/27/2013] [Indexed: 12/15/2022]
Abstract
Suppression of cell proliferation by targeting mitosis is one potential cancer intervention. A number of existing chemotherapy drugs disrupt mitosis by targeting microtubule dynamics. While efficacious, these drugs have limitations, i.e. neuropathy, unpredictability and development of resistance. In order to overcome these issues, a great deal of effort has been spent exploring novel mitotic targets including Polo-like kinase 1, Aurora kinases, Mps1, Cenp-E and KSP/Eg5. Here we summarize the latest developments in the discovery and clinical evaluation of new mitotic drug targets.
Collapse
Affiliation(s)
- Anna-Leena Salmela
- VTT Biotechnology for Health and Wellbeing, VTT Technical Research Centre of Finland, Itäinen Pitkäkatu 4C, Pharmacity Bldg, 4th Floor, P.O. Box 106, 20521, Turku, Finland
| | | |
Collapse
|
15
|
Song H, Zhou S, Wang R, Li S. Kinesin spindle protein (KSP) inhibitors in combination with chemotherapeutic agents for cancer therapy. ChemMedChem 2013; 8:1736-49. [PMID: 23964020 DOI: 10.1002/cmdc.201300228] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/20/2013] [Indexed: 12/20/2022]
Abstract
A diverse group of proteins, the activities of which are precisely orchestrated during mitosis, have emerged as targets for cancer therapeutics; these include the Aurora kinases (AKs), Polo-like kinases (PLKs), and the kinesin spindle protein (KSP). KSP is essential for the proper separation of spindle poles during mitosis. Agents that target KSP selectively act on cells undergoing cell division, which means that KSP inhibitors are mitosis-specific drugs, and have demonstrated remarkable activities in vitro. However, a significant obstacle to the success of KSP inhibitors is that these compounds, with tremendous efficacy in vitro, have demonstrated little or even no antitumor activity in vivo. Accumulated data suggest that a combination of KSP inhibitors with various cytostatic drugs will result in a more powerful tumor-killing effect than monotherapy. Combination therapies might predominate and represent the next frontier in the discovery research of KSP inhibitors as potential anticancer drugs. Few published studies have reviewed combination therapy using KSP inhibitors. Herein we provide a comprehensive review of the literature on KSP inhibitor monotherapy and therapeutic combinations. The current state and problems are also discussed.
Collapse
Affiliation(s)
- Hualong Song
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai (PR China)
| | | | | | | |
Collapse
|
16
|
Wissing MD, van Diest PJ, van der Wall E, Gelderblom H. Antimitotic agents for the treatment of patients with metastatic castrate-resistant prostate cancer. Expert Opin Investig Drugs 2013; 22:635-61. [PMID: 23586879 DOI: 10.1517/13543784.2013.789858] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Metastatic castrate-resistant prostate cancer (mCRPC) is the second deadliest cancer in men. The group of taxanes, which target microtubules of mitotic cells, is currently the only chemotherapy which has proven to increase overall survival in mCRPC patients. Other mitotic inhibitors are being explored for their clinical potential in mCRPC treatment. AREAS COVERED In this review, we summarize recent developments in the application of mitotic inhibitors for mCRPC from a clinical perspective. The four main groups of mitotic inhibitors currently being tested in clinical trials are microtubule-inhibitors, polo-like kinase 1 inhibitors, aurora kinase inhibitors and kinesin-spindle protein inhibitors. Compounds of these groups of inhibitors that are in clinical development for mCRPC are discussed. For this extensive overview, relevant literature was searched in PubMed and retrieved from clinicaltrials.gov and presentations at ASCO/AACR meetings. EXPERT OPINION In general, mitotic inhibitors are clinically well tolerated but exert limited antitumor activity compared to preclinical study results. However, efficacy of mitotic inhibitors is improving, either by personalizing treatment, by introducing more active compounds, by decreasing resistance of cancer cells against mitotic inhibitors or by using mitotic inhibitors in combination therapies.
Collapse
Affiliation(s)
- Michel D Wissing
- Department of Clinical Oncology, Leiden University Medical Center, Albinusdreef 2 K1-62, 2333ZA Leiden, The Netherlands
| | | | | | | |
Collapse
|
17
|
Jahn SC, Corsino PE, Davis BJ, Law ME, Nørgaard P, Law BK. Constitutive Cdk2 activity promotes aneuploidy while altering the spindle assembly and tetraploidy checkpoints. J Cell Sci 2013; 126:1207-17. [PMID: 23321641 DOI: 10.1242/jcs.117382] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The cell has many mechanisms for protecting the integrity of its genome. These mechanisms are often weakened or absent in many cancers, leading to high rates of chromosomal instability in tumors. Control of the cell cycle is crucial for the function of these checkpoints, and is frequently lost in cancers as well. Overexpression of Cyclin D1 in a large number of breast cancers causes overactivation of the cyclin-dependent kinases, including Cdk2. Constitutive Cdk2 activation through Cyclin D1 generates tumors in mice that are aneuploid and have many characteristics indicative of chromosomal instability. Expression of these complexes in the MCF10A cell line leads to retinoblastoma protein (Rb) hyperphosphorylation, a subsequent increase in proliferation rate, and increased expression of the spindle assembly checkpoint protein Mad2. This results in a strengthening of the spindle assembly checkpoint and renders cells more sensitive to the spindle poison paclitaxel. Constitutive Rb phosphorylation also causes a weakening of the p53-dependent tetraploidy checkpoint. Cells with overactive Cdk2 fail to arrest after mitotic slippage in the presence of paclitaxel or cytokinesis failure during treatment with cytochalasin-B, generating 8N populations. This additional increase in DNA content appears to further intensify the tetraploidy checkpoint in a step-wise manner. These polyploid cells are not viable long-term, either failing to undergo division or creating daughter cells that are unable to undergo subsequent division. This study raises intriguing questions about the treatment of tumors with overactive Cdk2.
Collapse
Affiliation(s)
- Stephan C Jahn
- Department of Pharmacology and Therapeutics and the Shands Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | |
Collapse
|
18
|
Kim N, Song K. KIFC1 is essential for bipolar spindle formation and genomic stability in the primary human fibroblast IMR-90 cell. Cell Struct Funct 2013; 38:21-30. [PMID: 23318213 DOI: 10.1247/csf.12014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Kinesin family member C1 (KIFC1) is the only member of the minus-end-directed kinesin-14 family in human cells. In cancer cells, KIFC1 plays an essential role in bipolar spindle formation by clustering the multiple poles during mitosis. However, it has not been clearly demonstrated whether KIFC1 also functions to mediate bipolar spindle formation and to maintain genomic stability in normal cells. In this study, by using human primary lung fibroblast IMR-90 cells, we showed that KIFC1 knock-down with lentiviral KIFC1 shRNA induced 17% of cells with multiple microtubule organizing centers (MTOCs) and delayed cyclin A degradation for more than 2 hr in early mitosis. However, these cells eventually carried out mitosis, resulting in 24% of cells with lagging chromosomes and 9% of cells with micronuclei after mitosis. Karyotyping of KIFC1-depleted IMR-90 cells demonstrated that cells with various abnormal numbers of chromosomes are produced. When IMR-90 cells treated with KIFC1 or the control shRNA for 60 hr were compared, 20% less cells were observed in KIFC1-depleted cells without an obvious immediate cell death. As reported for Mad2 depletion in IMR-90 cells, KIFC1-depleted IMR-90 cells showed typical features of senescence, like senescence-associated (SA) β-galactosidase expression, when incubated 6 days or more. However, IMR-90 cells knocked down with both KIFC1 and Mad2 underwent apoptosis, suggesting that KIFC1 and Mad2 likely function in different pathways during mitosis. Taken together, we suggest that KIFC1 plays an essential role for bipolar MTOC formation and maintaining chromosomal stability in the mitosis of human primary fibroblast IMR-90.
Collapse
Affiliation(s)
- Namil Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | | |
Collapse
|
19
|
Kaur J, Dutta S, Chang KP, Singh N. A member of the Ras oncogene family, RAP1A, mediates antileishmanial activity of monastrol. J Antimicrob Chemother 2013; 68:1071-80. [PMID: 23292345 PMCID: PMC3625431 DOI: 10.1093/jac/dks507] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Objectives To investigate the mode of action of monastrol in intracellular Leishmania. Methods Microarray experiments were conducted on an Affymetrix GeneChip® Human Genome U133 Plus 2.0 Array, to determine the genes that encode proteins related to pathological alterations of cell signalling pathways in intracellular Leishmania amastigotes in response to monastrol treatment. Results Monastrol induced unprenylated Rap1A in intracellular Leishmania when exposed to this anticancer drug at the IC50 (10 μM). Monastrol, known to cause mitotic arrest in cancer cells, inhibited Rap1A prenylation (geranylgeranylation) in intracellular Leishmania, which resulted in blockade at the G1 phase of the cell cycle. Growth inhibition, rather than apoptosis, was found to be the mechanism by which monastrol displays antileishmanial activity. Conclusions Prenylation inhibitors (unprenylation) of cell signalling pathways can be exploited in Leishmania parasites as novel therapeutic tools.
Collapse
Affiliation(s)
- Jaspreet Kaur
- Drug Target Discovery & Development Division, Central Drug Research Institute (CSIR), Chattar Manzil Palace, Lucknow, India
| | | | | | | |
Collapse
|
20
|
Silió V, Redondo-Muñoz J, Carrera AC. Phosphoinositide 3-kinase β regulates chromosome segregation in mitosis. Mol Biol Cell 2012; 23:4526-42. [PMID: 23051731 PMCID: PMC3510015 DOI: 10.1091/mbc.e12-05-0371] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The phosphoinositide 3-kinase (PI3K) pathway is mutated in approximately half of tumors; it is therefore important to define its functions. This study shows that PI3Kα activity regulates mitotic entry and spindle orientation; in contrast, PI3Kβ controls dynein/dynactin and Aurora B activation at kinetochores and, in turn, chromosome segregation. Class IA phosphoinositide 3-kinases (PI3K) are enzymes composed of a p85 regulatory and a p110 catalytic subunit that control formation of 3-poly-phosphoinositides (PIP3). The PI3K pathway regulates cell survival, migration, and division, and is mutated in approximately half of human tumors. For this reason, it is important to define the function of the ubiquitous PI3K subunits, p110α and p110β. Whereas p110α is activated at G1-phase entry and promotes protein synthesis and gene expression, p110β activity peaks in S phase and regulates DNA synthesis. PI3K activity also increases at the onset of mitosis, but the isoform activated is unknown; we have examined p110α and p110β function in mitosis. p110α was activated at mitosis entry and regulated early mitotic events, such as PIP3 generation, prometaphase progression, and spindle orientation. In contrast, p110β was activated near metaphase and controlled dynein/dynactin and Aurora B activities in kinetochores, chromosome segregation, and optimal function of the spindle checkpoint. These results reveal a p110β function in preserving genomic stability during mitosis.
Collapse
Affiliation(s)
- Virginia Silió
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Campus Universidad Autónoma de Madrid, Cantoblanco, Madrid E-28049, Spain
| | | | | |
Collapse
|
21
|
Ghosh S, Adhikary A, Chakraborty S, Nandi P, Mohanty S, Chakraborty S, Bhattacharjee P, Mukherjee S, Putatunda S, Chakraborty S, Chakraborty A, Sa G, Das T, Sen PC. Nifetepimine, a dihydropyrimidone, ensures CD4+ T cell survival in a tumor microenvironment by maneuvering sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA). J Biol Chem 2012; 287:32881-96. [PMID: 22851172 DOI: 10.1074/jbc.m112.357889] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple mechanisms have been proposed by which tumors induce T cell apoptosis to circumvent tumor immune-surveillance. Although sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) have long been known to regulate intracellular Ca(2+) homeostasis, few studies have examined the role of SERCA in processes of T lymphocyte survival and activation. In this context it remains largely unexplored as to how tumors jeopardize SERCA function to disable T cell-mediated anti-tumor immunity. Here, we show that human CD4(+) T cells in the presence of tumor conditions manifested an up-regulation of SERCA3 expression that resulted in development of endoplasmic reticulum stress leading to CD4(+) T cell apoptosis. Prostaglandin E(2) produced by the tumor cell plays a critical role in up-regulating SERCA3 by enhancing the binding of its transcription factor Sp1. Gene manipulation and pharmacological approaches further established that an increase in SERCA expression also resulted in subsequent inhibition of PKCα and -θ and retention of NFκB in the cytosol; however, down-modulation of SERCA3 expression by a dihydropyrimidone derivative, ethyl-4-(3-nitro)-phenyl-6-methyl-2-oxo-1,2,3,4-tetrahydropyrimidine-5 carboxylate (nifetepimine), protected the CD4(+) T cells from tumor-induced apoptosis. In fact, nifetepimine-mediated restoration of PKC activity resulted in nuclear translocation of p65NFκB, thereby ensuring its survival. Studies further undertaken in a tumor-bearing mice model revalidated the immunoprotective role of nifetepimine. Our present study thus strongly suggests that imbalance in cellular calcium homeostasis is an important factor leading to CD4(+) T cell death during cancer and holds promise that nifetepimine may have the potential to be used as an immunorestoring agent in cancer bearers.
Collapse
Affiliation(s)
- Swatilekha Ghosh
- Division of Molecular Medicine, Bose Institute, P1/12 Calcutta Improvement Trust Scheme VIIM Kolkata 700054, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Marquis L, Tran M, Choi W, Lee IL, Huszar D, Siefker-Radtke A, Dinney C, McConkey DJ. p63 expression correlates with sensitivity to the Eg5 inhibitor ZD4877 in bladder cancer cells. Cancer Biol Ther 2012; 13:477-86. [PMID: 22361733 DOI: 10.4161/cbt.19590] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Antimitotics such as taxanes are being considered as alternatives to conventional cisplatin-based chemotherapy in patients with bladder cancer, but the molecular determinants of sensitivity or resistance to these agents in bladder cancer cells have not been defined. Here we examined the cytotoxic effects of a novel antimitotic, the Eg5 inhibitor AZD4877, in a molecularly diverse panel of human bladder cancer cell lines. The cells displayed heterogeneous responses to the drug that correlated closely with sensitivity to docetaxel but not with sensitivity to cisplatin. Global gene expression profiling identified p63 as the top gene that was differentially expressed between sensitive and resistant cell lines. Stable knockdown of p63 inhibited cell death induced by either AZD4877 or docetaxel and was associated with decreased proliferation and decreased expression of c-myc. Furthermore, c-myc knockdown also rendered cells resistant to AZD4877 or docetaxel. Together, our results implicate p63 and its downstream target c-myc as determinants of sensitivity to anti-mitotics in bladder cancer cells. Our data also suggest that anti-mitotics and cisplatin target different subsets of bladder cancer cells, a conclusion that may have important implications for the therapy of muscle-invasive bladder cancers.
Collapse
Affiliation(s)
- Lauren Marquis
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Theoclitou ME, Aquila B, Block MH, Brassil PJ, Castriotta L, Code E, Collins MP, Davies AM, Deegan T, Ezhuthachan J, Filla S, Freed E, Hu H, Huszar D, Jayaraman M, Lawson D, Lewis PM, Nadella MVP, Oza V, Padmanilayam M, Pontz T, Ronco L, Russell D, Whitston D, Zheng X. Discovery of (+)-N-(3-aminopropyl)-N-[1-(5-benzyl-3-methyl-4-oxo-[1,2]thiazolo[5,4-d]pyrimidin-6-yl)-2-methylpropyl]-4-methylbenzamide (AZD4877), a kinesin spindle protein inhibitor and potential anticancer agent. J Med Chem 2011; 54:6734-50. [PMID: 21899292 DOI: 10.1021/jm200629m] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Structure-activity relationship analysis identified (+)-N-(3-aminopropyl)-N-[1-(5-benzyl-3-methyl-4-oxo-[1,2]thiazolo[5,4-d]pyrimidin-6-yl)-2-methylpropyl]-4-methylbenzamide (AZD4877), from a series of novel kinesin spindle protein (KSP) inhibitors, as exhibiting both excellent biochemical potency and pharmaceutical properties suitable for clinical development. The selected compound arrested cells in mitosis leading to the formation of the monopolar spindle phenotype characteristic of KSP inhibition and induction of cellular death. A favorable pharmacokinetic profile and notable in vivo efficacy supported the selection of this compound as a clinical candidate for the treatment of cancer.
Collapse
Affiliation(s)
- Maria-Elena Theoclitou
- Cancer & Infection Research Area, AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bartoli KM, Jakovljevic J, Woolford JL, Saunders WS. Kinesin molecular motor Eg5 functions during polypeptide synthesis. Mol Biol Cell 2011; 22:3420-30. [PMID: 21795388 PMCID: PMC3172266 DOI: 10.1091/mbc.e11-03-0211] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The microtubule motor Eg5 is well known for its functions during mitosis. It is shown that during interphase, Eg5 associates with ribosomes and is required for efficient protein synthesis. The kinesin-related molecular motor Eg5 plays roles in cell division, promoting spindle assembly. We show that during interphase Eg5 is associated with ribosomes and is required for optimal nascent polypeptide synthesis. When Eg5 was inhibited, ribosomes no longer bound to microtubules in vitro, ribosome transit rates slowed, and polysomes accumulated in intact cells, suggesting defects in elongation or termination during polypeptide synthesis. These results demonstrate that the molecular motor Eg5 associates with ribosomes and enhances the efficiency of translation.
Collapse
Affiliation(s)
- Kristen M Bartoli
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | |
Collapse
|
25
|
Braconi C, Swenson E, Kogure T, Huang N, Patel T. Targeting the IL-6 dependent phenotype can identify novel therapies for cholangiocarcinoma. PLoS One 2010; 5:e15195. [PMID: 21179572 PMCID: PMC3002961 DOI: 10.1371/journal.pone.0015195] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 10/29/2010] [Indexed: 02/07/2023] Open
Abstract
Background The need for new therapies for cholangiocarcinoma is highlighted by their poor prognosis and refractoriness to chemotherapy. Increased production of Interleukin-6 promotes cholangiocarcinoma growth and contributes to chemoresistance by activating cell survival mechanisms. We sought to identify biologically active compounds capable of ameliorating the phenotypic effects of IL-6 expression and to explore their potential therapeutic use for cholangiocarcinoma. Methodology A genomic signature associated with Interleukin-6 expression in Mz-ChA-1 human malignant cholangiocytes was derived. Computational bioinformatics analysis was performed to identify compounds that induced inverse gene changes to the signature. The effect of these compounds on cholangiocarcinoma growth was then experimentally verified in vitro and in vivo. Interactions with other therapeutic agents were evaluated using median effects analysis. Principal Findings A group of structurally related compounds, nitrendipine, nifedipine and felodipine was identified. All three compounds were cytotoxic to Mz-ChA-1 cells with an IC50 for felodipine of 26 µM, nitrendipine, 44 µM and nifedipine, 15 µM. Similar results were observed in KMCH-1, CC-LP-1 and TFK-1 cholangiocarcinoma cell lines. At a fractional effect of 0.5, all three agents were synergistic with either camptothecin or gemcitabine in Mz-ChA-1 cells in vitro. Co-administration of felodipine and gemcitabine decreased the growth of Mz-ChA-1 cell xenografts in nude athymic mice. Conclusions Computational bioinformatics analysis of phenotype-based genomic expression can be used to identify therapeutic agents. Using this drug discovery approach based on targeting a defined tumor associated phenotype, we identified compounds with the potential for therapeutic use in cholangiocarcinoma.
Collapse
Affiliation(s)
- Chiara Braconi
- Department of Internal Medicine, College of Medicine, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, Ohio, United States of America
| | | | | | | | | |
Collapse
|
26
|
Ke N, Xi B, Ye P, Xu W, Zheng M, Mao L, Wu MJ, Zhu J, Wu J, Zhang W, Zhang J, Irelan J, Wang X, Xu X, Abassi YA. Screening and identification of small molecule compounds perturbing mitosis using time-dependent cellular response profiles. Anal Chem 2010; 82:6495-503. [PMID: 20617823 DOI: 10.1021/ac1007877] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cellular processes such as cell cycle progression, mitosis, apoptosis, and cell migration are characterized by well-defined events that are modulated as a function of time. Measuring these events in the context of time and its perturbation by small molecule compounds and RNAi can provide mechanistic information about cellular pathways being affected. We have used impedance-based time-dependent cell response profiling (TCRP) to measure and characterize cellular responses to antimitotic compounds or siRNAs. Our findings indicate that small molecule perturbation of mitosis leads to unique TCRP. We have further used this unique TCRP signature to screen 119 595 compound library and identified novel antimitotic compounds based on clustering analysis of the TCRPs. Importantly, 113 of the 117 hit compounds in the TCRP antimitotic cluster were confirmed as antimitotic based on independent assays, thus establishing the robust predictive nature of this profiling approach. In addition, potent and novel agents that induce mitotic arrest either by directly interfering with tubulin polymerization or by other mechanisms were identified. The TCRP approach allows for a practical and unbiased phenotypic profiling and screening tool for small molecule and RNAi perturbation of specific cellular pathways and time resolution of the TCRP approach can serve as a complement for other existing multidimensional profiling approaches.
Collapse
Affiliation(s)
- Ning Ke
- Department of Cell Biology and Assay Development, ACEA Biosciences, San Diego, California 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Peyre M, Commo F, Dantas-Barbosa C, Andreiuolo F, Puget S, Lacroix L, Drusch F, Scott V, Varlet P, Mauguen A, Dessen P, Lazar V, Vassal G, Grill J. Portrait of ependymoma recurrence in children: biomarkers of tumor progression identified by dual-color microarray-based gene expression analysis. PLoS One 2010; 5:e12932. [PMID: 20885975 PMCID: PMC2945762 DOI: 10.1371/journal.pone.0012932] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 07/14/2010] [Indexed: 01/06/2023] Open
Abstract
Background Children with ependymoma may experience a relapse in up to 50% of cases depending on the extent of resection. Key biological events associated with recurrence are unknown. Methodology/Principal Findings To discover the biology behind the recurrence of ependymomas, we performed CGHarray and a dual-color gene expression microarray analysis of 17 tumors at diagnosis co-hybridized with the corresponding 27 first or subsequent relapses from the same patient. As treatment and location had only limited influence on specific gene expression changes at relapse, we established a common signature for relapse. Eighty-seven genes showed an absolute fold change ≥2 in at least 50% of relapses and were defined as the gene expression signature of ependymoma recurrence. The most frequently upregulated genes are involved in the kinetochore (ASPM, KIF11) or in neural development (CD133, Wnt and Notch pathways). Metallothionein (MT) genes were downregulated in up to 80% of the recurrences. Quantitative PCR for ASPM, KIF11 and MT3 plus immunohistochemistry for ASPM and MT3 confirmed the microarray results. Immunohistochemistry on an independent series of 24 tumor pairs at diagnosis and at relapse confirmed the decrease of MT3 expression at recurrence in 17/24 tumor pairs (p = 0.002). Conversely, ASPM expression was more frequently positive at relapse (87.5% vs 37.5%, p = 0.03). Loss or deletion of the MT genes cluster was never observed at relapse. Promoter sequencing after bisulfite treatment of DNA from primary tumors and recurrences as well as treatment of short-term ependymoma cells cultures with a demethylating agent showed that methylation was not involved in MT3 downregulation. However, in vitro treatment with a histone deacetylase inhibitor or zinc restored MT3 expression. Conclusions/Significance The most frequent molecular events associated with ependymoma recurrence were over-expression of kinetochore proteins and down-regulation of metallothioneins. Metallothionein-3 expression is epigenetically controlled and can be restored in vitro by histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Matthieu Peyre
- Université Paris-Sud, CNRS UMR 8203 “Vectorology and Anticancer Treatments”, Gustave Roussy Institute, Villejuif, France
| | - Frédéric Commo
- CNRS FRE 2939, Bioinformatics Group, Gustave Roussy Institute, Villejuif, France
| | - Carmela Dantas-Barbosa
- Université Paris-Sud, CNRS UMR 8203 “Vectorology and Anticancer Treatments”, Gustave Roussy Institute, Villejuif, France
| | - Felipe Andreiuolo
- Université Paris-Sud, CNRS UMR 8203 “Vectorology and Anticancer Treatments”, Gustave Roussy Institute, Villejuif, France
- Translational Research Laboratory, Gustave Roussy Institute, Villejuif, France
| | - Stéphanie Puget
- Université Paris-Sud, CNRS UMR 8203 “Vectorology and Anticancer Treatments”, Gustave Roussy Institute, Villejuif, France
- Department of Neurosurgery, Necker Sick Children's Hospital, Université Paris V Descartes, Paris, France
| | - Ludovic Lacroix
- Translational Research Laboratory, Gustave Roussy Institute, Villejuif, France
| | - Françoise Drusch
- Translational Research Laboratory, Gustave Roussy Institute, Villejuif, France
| | - Véronique Scott
- Université Paris-Sud, CNRS UMR 8203 “Vectorology and Anticancer Treatments”, Gustave Roussy Institute, Villejuif, France
| | - Pascale Varlet
- Department of Neuropathology, Sainte-Anne Hospital, Paris, France
| | - Audrey Mauguen
- Department of Biostatistics, Gustave Roussy Institute, Villejuif, France
| | - Philippe Dessen
- CNRS FRE 2939, Bioinformatics Group, Gustave Roussy Institute, Villejuif, France
| | - Vladimir Lazar
- Functional Genomics Unit, Gustave Roussy Institute, Villejuif, France
| | - Gilles Vassal
- Université Paris-Sud, CNRS UMR 8203 “Vectorology and Anticancer Treatments”, Gustave Roussy Institute, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Institute, Villejuif, France
| | - Jacques Grill
- Université Paris-Sud, CNRS UMR 8203 “Vectorology and Anticancer Treatments”, Gustave Roussy Institute, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Institute, Villejuif, France
- * E-mail:
| |
Collapse
|
28
|
Prokopcová H, Dallinger D, Uray G, Kaan HYK, Ulaganathan V, Kozielski F, Laggner C, Kappe CO. Structure-Activity Relationships and Molecular Docking of Novel Dihydropyrimidine-Based Mitotic Eg5 Inhibitors. ChemMedChem 2010; 5:1760-9. [DOI: 10.1002/cmdc.201000252] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
29
|
Ingemarsdotter C, Keller D, Beard P. The DNA damage response to non-replicating adeno-associated virus: Centriole overduplication and mitotic catastrophe independent of the spindle checkpoint. Virology 2010; 400:271-86. [PMID: 20199789 DOI: 10.1016/j.virol.2010.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 01/25/2010] [Accepted: 02/03/2010] [Indexed: 12/17/2022]
Abstract
Adeno-associated virus (AAV) type 2 or UV-inactivated AAV (UV-AAV2) infection provokes a DNA damage response that leads to cell cycle arrest at the G2/M border. p53-deficient cells cannot sustain the G2 arrest, enter prolonged impaired mitosis, and die. Here, we studied how non-replicating AAV2 kills p53-deficient osteosarcoma cells. We found that the virus uncouples centriole duplication from the cell cycle, inducing centrosome overamplification that is dependent on Chk1, ATR and CDK kinases, and on G2 arrest. Interference with spindle checkpoint components Mad2 and BubR1 revealed unexpectedly that mitotic catastrophe occurs independently of spindle checkpoint function. We conclude that, in the p53-deficient cells, UV-AAV2 triggers mitotic catastrophe associated with a dramatic Chk1-dependent overduplication of centrioles and the consequent formation of multiple spindle poles in mitosis. As AAV2 acts through cellular damage response pathways, the results provide information on the role of Chk1 in mitotic catastrophe after DNA damage signaling in general.
Collapse
Affiliation(s)
- Carin Ingemarsdotter
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Swiss Institute for Experimental Cancer Research (ISREC), 1015 Lausanne, Switzerland.
| | | | | |
Collapse
|
30
|
Abstract
The process of mitosis is a validated point of intervention in cancer therapy and a variety of anti-mitotic drugs are successfully being used in the clinic. To date, all approved antimitotics target the spindle microtubules, thus interfering with spindle dynamics, leading to mitotic arrest and apoptosis. While effective, these drugs are also associated with a variety of side effects, including neurotoxicity. In recent years, mitotic kinesins have attracted significant attention in the search for novel, alternative mitotic drug targets. Due to their specific function in mitosis, targeting these proteins creates an opportunity for the development of more selective antimitotics with an improved side effect profile. In addition, kinesin inhibitors may overcome resistance to microtubule targeting drugs. Drug discovery efforts in this area have initially focused on the plus-end directed kinesin spindle protein (KSP) and a variety of compounds are currently undergoing clinical testing.
Collapse
|
31
|
Nakai R, Iida SI, Takahashi T, Tsujita T, Okamoto S, Takada C, Akasaka K, Ichikawa S, Ishida H, Kusaka H, Akinaga S, Murakata C, Honda S, Nitta M, Saya H, Yamashita Y. K858, a novel inhibitor of mitotic kinesin Eg5 and antitumor agent, induces cell death in cancer cells. Cancer Res 2009; 69:3901-9. [PMID: 19351824 DOI: 10.1158/0008-5472.can-08-4373] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of this study was to investigate the mechanism of inhibition of Eg5 (kinesin spindle protein), a mitotic kinesin that plays an essential role in establishing mitotic spindle bipolarity, by the novel small molecule inhibitor K858. K858 was selected in a phenotype-based forward chemical genetics screen as an antimitotic agent, and subsequently characterized as an inhibitor of Eg5. K858 blocked centrosome separation, activated the spindle checkpoint, and induced mitotic arrest in cells accompanied by the formation of monopolar spindles. Long-term continuous treatment of cancer cells with K858 resulted in antiproliferative effects through the induction of mitotic cell death, and polyploidization followed by senescence. In contrast, treatment of nontransformed cells with K858 resulted in mitotic slippage without cell death, and cell cycle arrest in G(1) phase in a tetraploid state. In contrast to paclitaxel, K858 did not induce the formation of micronuclei in either cancer or nontransformed cells, suggesting that K858 has minimal effects on abnormalities in the number and structure of chromosomes. K858 exhibited potent antitumor activity in xenograft models of cancer, and induced the accumulation of mitotic cells with monopolar spindles in tumor tissues. Importantly, K858, unlike antimicrotubule agents, had no effect on microtubule polymerization in cell-free and cell-based assays, and was not neurotoxic in a motor coordination test in mice. Taken together, the Eg5 inhibitor K858 represents an important compound for further investigation as a novel anticancer therapeutic.
Collapse
Affiliation(s)
- Ryuichiro Nakai
- Drug Discovery Research Laboratories, Kyowa Hakko Kirin Co, Ltd, Shizuoka, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Engstrom JU, Suzuki T, Kmiec EB. Regulation of targeted gene repair by intrinsic cellular processes. Bioessays 2009; 31:159-68. [PMID: 19204988 DOI: 10.1002/bies.200800119] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Targeted gene alteration (TGA) is a strategy for correcting single base mutations in the DNA of human cells that cause inherited disorders. TGA aims to reverse a phenotype by repairing the mutant base within the chromosome itself, avoiding the introduction of exogenous genes. The process of how to accurately repair a genetic mutation is elucidated through the use of single-stranded DNA oligonucleotides (ODNs) that can enter the cell and migrate to the nucleus. These specifically designed ODNs hybridize to the target sequence and act as a beacon for nucleotide exchange. The key to this reaction is the frequency with which the base is corrected; this will determine whether the approach becomes clinically relevant or not. Over the course of the last five years, workers have been uncovering the role played by the cells in regulating the gene repair process. In this essay, we discuss how the impact of the cell on TGA has evolved through the years and illustrate ways that inherent cellular pathways could be used to enhance TGA activity. We also describe the cost to cell metabolism and survival when certain processes are altered to achieve a higher frequency of repair.
Collapse
Affiliation(s)
- Julia U Engstrom
- University of Delaware, Department of Biological Sciences, Newark, DE 19716, USA
| | | | | |
Collapse
|
33
|
Sarli V, Giannis A. Targeting the kinesin spindle protein: basic principles and clinical implications. Clin Cancer Res 2009; 14:7583-7. [PMID: 19047082 DOI: 10.1158/1078-0432.ccr-08-0120] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kinesin spindle protein (KSP), a member of the kinesin superfamily of microtubule-based motors, plays a critical role in mitosis as it mediates centrosome separation and bipolar spindle assembly and maintenance. Inhibition of KSP function leads to cell cycle arrest at mitosis with the formation of monoastral microtubule arrays, and ultimately, to cell death. Several KSP inhibitors are currently being studied in clinical trials and provide new opportunities for the development of novel anticancer therapeutics alternative from the available microtubule targeting drugs.
Collapse
Affiliation(s)
- Vasiliki Sarli
- Institute for Organic Chemistry, University of Leipzig, Leipzig, Germany.
| | | |
Collapse
|
34
|
Orth JD, Tang Y, Shi J, Loy CT, Amendt C, Wilm C, Zenke FT, Mitchison TJ. Quantitative live imaging of cancer and normal cells treated with Kinesin-5 inhibitors indicates significant differences in phenotypic responses and cell fate. Mol Cancer Ther 2008; 7:3480-9. [PMID: 18974392 DOI: 10.1158/1535-7163.mct-08-0684] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Kinesin-5 inhibitors (K5I) are promising antimitotic cancer drug candidates. They cause prolonged mitotic arrest and death of cancer cells, but their full range of phenotypic effects in different cell types has been unclear. Using time-lapse microscopy of cancer and normal cell lines, we find that a novel K5I causes several different cancer and noncancer cell types to undergo prolonged arrest in monopolar mitosis. Subsequent events, however, differed greatly between cell types. Normal diploid cells mostly slipped from mitosis and arrested in tetraploid G(1), with little cell death. Several cancer cell lines died either during mitotic arrest or following slippage. Contrary to prevailing views, mitotic slippage was not required for death, and the duration of mitotic arrest correlated poorly with the probability of death in most cell lines. We also assayed drug reversibility and long-term responses after transient drug exposure in MCF7 breast cancer cells. Although many cells divided after drug washout during mitosis, this treatment resulted in lower survival compared with washout after spontaneous slippage likely due to chromosome segregation errors in the cells that divided. Our analysis shows that K5Is cause cancer-selective cell killing, provides important kinetic information for understanding clinical responses, and elucidates mechanisms of drug sensitivity versus resistance at the level of phenotype.
Collapse
Affiliation(s)
- James D Orth
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Hayashi N, Koller E, Fazli L, Gleave ME. Effects of Eg5 knockdown on human prostate cancer xenograft growth and chemosensitivity. Prostate 2008; 68:1283-95. [PMID: 18512732 DOI: 10.1002/pros.20783] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVES Microtubular inhibitors, including docetaxel, are active cytotoxics in many cancers, including prostate cancer (CaP). The Eg5 gene, a member of the kinesin-5 family, plays critical roles in proper mitotic spindle function, and is a potential microtubule-related target for proliferating cancer cells. To investigate the functional activities of Eg5 in CaP, we used an antisense oligonucleotide (ASO) targeting Eg5 to assess the potency and anti-cancer activity of Eg5 ASO treatment for androgen-independent CaP cells in vitro and in vivo. RESULTS PC3 cells express higher Eg5 protein and mRNA levels compared to LNCaP cells. In both cell lines, Eg5 ASO treatment reduced mRNA and protein levels in a dose-dependent manner and a complete reduction of Eg5 protein levels was observed at 100 nM. Dose-dependent inhibition in cell growth, potent G2/M phase arrest, and increases in apoptotic sub-G1 fraction were also observed using Eg5 ASO. Surprisingly, low dose Eg5 ASO significantly antagonized cytotoxic effects of paclitaxel. In vivo, Eg5 ASO monotherapy significantly reduced both LNCaP and PC-3 tumor growth but combination treatment with paclitaxel did not yield additive benefits. CONCLUSIONS These findings suggest that while Eg5 is a potential target to delay androgen-independent CaP growth, combination treatment with paclitaxel may not be desirable.
Collapse
Affiliation(s)
- Norihiro Hayashi
- The Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
36
|
Shi J, Orth JD, Mitchison T. Cell type variation in responses to antimitotic drugs that target microtubules and kinesin-5. Cancer Res 2008; 68:3269-76. [PMID: 18451153 DOI: 10.1158/0008-5472.can-07-6699] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
To improve cancer chemotherapy, we need to understand the mechanisms that determine drug sensitivity in cancer and normal cells. Here, we investigate this question across a panel of 11 cell lines at a phenotypic and molecular level for three antimitotic drugs: paclitaxel, nocodazole, and an inhibitor of kinesin-5 (also known as KSP, Eg5, Kif11). Using automated microscopy with markers for mitosis and apoptosis (high content screening), we find that the mitotic arrest response shows relatively little variation between cell types, whereas the tendency to undergo apoptosis shows large variation. We found no correlation between levels of mitotic arrest and apoptosis. Apoptosis depended on entry into mitosis and occurred both from within mitosis and after exit. Response to the three drugs strongly correlated, although paclitaxel caused more apoptosis in some cell lines at similar levels of mitotic arrest. Molecular investigations showed that sensitivity to apoptosis correlated with loss of an antiapoptotic protein, XIAP, during the drug response, but not its preresponse levels, and to some extent also correlated with activation of the p38 and c-Jun NH(2) kinase pathways. We conclude that variation in sensitivity to antimitotic drugs in drug-naive cell lines is governed more by differences in apoptotic signaling than by differences in mitotic spindle or spindle assembly checkpoint proteins and that antimitotics with different mechanisms trigger very similar, but not identical, responses.
Collapse
Affiliation(s)
- Jue Shi
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
37
|
Vijapurkar U, Wang W, Herbst R. Potentiation of Kinesin Spindle Protein Inhibitor–Induced Cell Death by Modulation of Mitochondrial and Death Receptor Apoptotic Pathways. Cancer Res 2007; 67:237-45. [PMID: 17210704 DOI: 10.1158/0008-5472.can-06-2406] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Targeting the mitotic motor kinesin kinesin spindle protein (KSP) is a new strategy for cancer therapy. We have examined the molecular events induced by KSP inhibition and explored possible mechanisms of resistance and sensitization of tumor cells to KSP inhibitors. We found that KSP inhibition induced cell death primarily via activation of the mitochondrial death pathway. In HeLa cells, inhibition of KSP by small-molecule inhibitor monastrol resulted in mitotic arrest and rapid caspase activation. BclXL phosphorylation and loss of mitochondrial membrane potential was detected before significant caspase activation, which was required to trigger the subsequent apoptotic pathway. In A549 cells, however, KSP inhibition did not induce mitochondrial damage, significant caspase activity, or cell death. A549 cells aberrantly exited mitosis, following a prolonged drug-induced arrest, and arrested in a G(1)-like state with 4N DNA content in a p53-dependent manner. Overexpression of BclXL provided a protective mechanism, and its depletion rescued the apoptotic response to monastrol. In addition, Fas receptor was up-regulated in A549 cells in response to monastrol. Treatment with Fas receptor agonists sensitized the cells to monastrol-induced cell death, following exit from mitosis. Thus, activation of the death receptor pathway offered another mechanism to enhance KSP inhibitor-induced apoptosis. This study has elucidated cellular responses induced by KSP inhibitors, and the results provide insights for a more effective cancer treatment with these agents.
Collapse
|