1
|
Sawada JI, Matsuno K, Ogo N, Asai A. Identification of antimitotic sulfonamides inhibiting chromosome congression. Biochem Pharmacol 2025; 232:116718. [PMID: 39701545 DOI: 10.1016/j.bcp.2024.116718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/28/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
The discovery of new small-molecule inhibitors is essential to enhancing our understanding of biological events at the molecular level and driving advancements in drug discovery. Mitotic inhibitors have played a crucial role in development of anticancer drugs. Beyond traditional microtubule inhibitors, various inhibitors targeting specific mitotic factors have been developed. This study aimed to develop novel mitotic inhibitors targeting chromosome alignment. We established a cell-based screening method using Cell Division Cycle Associated 5 (CDCA5) and kinesin-5 as markers, designed to efficiently detect mitotic phenotypes characterized by aberrant bipolar spindles with some misaligned chromosomes. Through this screening, we identified CAIS-1, an aryl sulfonamide with unique antimitotic properties. CAIS-1 exhibits dual functionality by inhibiting chromosome congression at low concentrations and spindle microtubule formation at high concentrations, causing a concentration-dependent mitotic arrest, followed by apoptotic cell death. Mechanistic studies revealed that CAIS-1 directly acts on tubulin at high concentrations, thereby inhibiting tubulin polymerization in vitro. In contrast, at low concentrations, CAIS-1 functions through a mechanism distinct from GSK923295, a conventional chromosome congression inhibitor targeting Centromere-associated protein-E (CENP-E), highlighting its unique mode of action. Moreover, CAIS-2, a structural analog of CAIS-1, selectively inhibits chromosome congression without significantly affecting spindle microtubules. This observation suggests that CAIS-1 and CAIS-2 function as antimitotic sulfonamides with distinct targets beyond tubulin, thus offering additional biological potential of sulfonamide compounds. Together, CAIS-1 and CAIS-2 represent promising tools for providing new molecular insights into kinetochore function during mitosis and for exploring new approaches in anticancer drug development.
Collapse
Affiliation(s)
- Jun-Ichi Sawada
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Kenji Matsuno
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Naohisa Ogo
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| | - Akira Asai
- Center for Drug Discovery, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
2
|
Exploiting immune-dependent effects of microtubule-targeting agents to improve efficacy and tolerability of cancer treatment. Cell Death Dis 2020; 11:361. [PMID: 32398657 PMCID: PMC7217828 DOI: 10.1038/s41419-020-2567-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 02/08/2023]
Abstract
Microtubule-targeting agents (MTAs), like taxanes and vinca alkaloids, are tubulin-binding drugs that are very effective in the treatment of various types of cancers. In cell cultures, these drugs appear to affect assembly of the mitotic spindle and to delay progression through mitosis and this correlates with their ability to induce cell death. Their clinical efficacy is, however, limited by resistance and toxicity. For these reasons, other spindle-targeting drugs, affecting proteins such as certain kinesins like Eg5 and CENP-E, or kinases like Plk1, Aurora A and B, have been developed as an alternative to MTAs. However, these attempts have disappointed in the clinic since these drugs show poor anticancer activity and toxicity ahead of positive effects. In addition, whether efficacy of MTAs in cancer treatment is solely due to their ability to delay mitosis progression remains controversial. Here we discuss recent findings indicating that the taxane paclitaxel can promote a proinflammatory response by activation of innate immunity. We further describe how this can help adaptive antitumor immune response and suggest, on this basis and on the recent success of immune checkpoint inhibitors in cancer treatment, that a combination therapy based on low doses of taxanes and immune checkpoint inhibitors may be of high clinical advantage in terms of wide applicability, reduced toxicity, and increased antitumor response.
Collapse
|
3
|
Motuhi SE, Feizbakhsh O, Foll-Josselin B, Baratte B, Delehouzé C, Cousseau A, Fant X, Bulinski JC, Payri CE, Ruchaud S, Mehiri M, Bach S. Neurymenolide A, a Novel Mitotic Spindle Poison from the New Caledonian Rhodophyta Phacelocarpus neurymenioides. Mar Drugs 2019; 17:md17020093. [PMID: 30717235 PMCID: PMC6410418 DOI: 10.3390/md17020093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 12/30/2022] Open
Abstract
The marine α-pyrone macrolide neurymenolide A was previously isolated from the Fijian red macroalga, Neurymenia fraxinifolia, and characterized as an antibacterial agent against antibiotic-resistant strains that also exhibited moderate cytotoxicity in vitro against cancer cell lines. This compound was also shown to exhibit allelopathic effects on Scleractinian corals. However, to date no mechanism of action has been described in the literature. The present study showed, for the first time, the isolation of neurymenolide A from the New Caledonian Rhodophyta, Phacelocarpus neurymenioides. We confirmed the compound’s moderate cytotoxicity in vitro against several human cell lines, including solid and hematological malignancies. Furthermore, we combined fluorescence microscopy and flow cytometry to demonstrate that treatment of U-2 OS osteosarcoma human cells with neurymenolide A could block cell division in prometaphase by inhibiting the correct formation of the mitotic spindle, which induced a mitotic catastrophe that led to necrosis and apoptosis. Absolute configuration of the stereogenic center C-17 of neurymenolide A was deduced by comparison of the experimental and theoretical circular dichroism spectra. Since the total synthesis of this compound has already been described, our findings open new avenues in cancer treatment for this class of marine molecules, including a new source for the natural product.
Collapse
Affiliation(s)
- Sofia-Eléna Motuhi
- UMR ENTROPIE (IRD-Université de La Réunion-CNRS), Laboratoire d'Excellence Labex-CORAIL, Institut de Recherche pour le Développement (IRD), BP A5, 98848 Nouméa CEDEX, New Caledonia, France.
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
- UMR 7272 CNRS, Marine Natural Products Team, Nice Institute of Chemistry (ICN), University Nice Sophia Antipolis, Parc Valrose, 02 F-06108 Nice CEDEX, France.
| | - Omid Feizbakhsh
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
- Sorbonne Université, CNRS, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
| | - Béatrice Foll-Josselin
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
| | - Blandine Baratte
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
| | - Claire Delehouzé
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
| | - Arnaud Cousseau
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
- UMR 7272 CNRS, Marine Natural Products Team, Nice Institute of Chemistry (ICN), University Nice Sophia Antipolis, Parc Valrose, 02 F-06108 Nice CEDEX, France.
| | - Xavier Fant
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
| | - Jeannette Chloë Bulinski
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA.
| | - Claude Elisabeth Payri
- UMR ENTROPIE (IRD-Université de La Réunion-CNRS), Laboratoire d'Excellence Labex-CORAIL, Institut de Recherche pour le Développement (IRD), BP A5, 98848 Nouméa CEDEX, New Caledonia, France.
| | - Sandrine Ruchaud
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
| | - Mohamed Mehiri
- UMR 7272 CNRS, Marine Natural Products Team, Nice Institute of Chemistry (ICN), University Nice Sophia Antipolis, Parc Valrose, 02 F-06108 Nice CEDEX, France.
| | - Stéphane Bach
- Sorbonne Université, CNRS, USR 3151, Protein Phosphorylation & Human Diseases, Station Biologique de Roscoff, CS 90074, 29688 Roscoff CEDEX, France.
| |
Collapse
|
4
|
Paier CRK, Maranhão SS, Carneiro TR, Lima LM, Rocha DD, da Silva Santos R, de Farias KM, de Moraes-Filho MO, Pessoa C. Natural products as new antimitotic compounds for anticancer drug development. Clinics (Sao Paulo) 2018; 73:e813s. [PMID: 30540125 PMCID: PMC6256996 DOI: 10.6061/clinics/2018/e813s] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/09/2018] [Indexed: 12/19/2022] Open
Abstract
Cell cycle control genes are frequently mutated in cancer cells, which usually display higher rates of proliferation than normal cells. Dysregulated mitosis leads to genomic instability, which contributes to tumor progression and aggressiveness. Many drugs that disrupt mitosis have been studied because they induce cell cycle arrest and tumor cell death. These antitumor compounds are referred to as antimitotics. Vinca alkaloids and taxanes are natural products that target microtubules and inhibit mitosis, and their derivatives are among the most commonly used drugs in cancer therapy worldwide. However, severe adverse effects such as neuropathies are frequently observed during treatment with microtubule-targeting agents. Many efforts have been directed at developing improved antimitotics with increased specificity and decreased likelihood of inducing side effects. These new drugs generally target specific components of mitotic regulation that are mainly or exclusively expressed during cell division, such as kinases, motor proteins and multiprotein complexes. Such small molecules are now in preclinical studies and clinical trials, and many are products or derivatives from natural sources. In this review, we focused on the most promising targets for the development of antimitotics and discussed the advantages and disadvantages of these targets. We also highlighted the novel natural antimitotic agents under investigation by our research group, including combretastatins, withanolides and pterocarpans, which show the potential to circumvent the main issues in antimitotic therapy.
Collapse
Affiliation(s)
- Carlos Roberto Koscky Paier
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
- *Corresponding author. E-mail:
| | - Sarah Sant'Anna Maranhão
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Farmacologia, Universidade Federal do Ceara, Fortaleza, CE, BR
| | - Teiliane Rodrigues Carneiro
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Biotecnologia, Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal do Ceara, Fortaleza, CE, BR
- Laboratorio de Avaliacao e Sintese de Substancias Bioativas (LASSBio), Instituto de Ciencia e Tecnologia de Farmacos e Medicamentos (INCT-INOFAR), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | - Lídia Moreira Lima
- Laboratorio de Avaliacao e Sintese de Substancias Bioativas (LASSBio), Instituto de Ciencia e Tecnologia de Farmacos e Medicamentos (INCT-INOFAR), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, BR
| | - Danilo Damasceno Rocha
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
| | - Renan da Silva Santos
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Farmacologia, Universidade Federal do Ceara, Fortaleza, CE, BR
| | - Kaio Moraes de Farias
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Biotecnologia, Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal do Ceara, Fortaleza, CE, BR
| | - Manoel Odorico de Moraes-Filho
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Farmacologia, Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Biotecnologia, Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal do Ceara, Fortaleza, CE, BR
| | - Claudia Pessoa
- Laboratorio de Oncologia Experimental, Nucleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Farmacologia, Universidade Federal do Ceara, Fortaleza, CE, BR
- Programa de Pos graduacao em Biotecnologia, Rede Nordeste de Biotecnologia (RENORBIO), Universidade Federal do Ceara, Fortaleza, CE, BR
| |
Collapse
|
5
|
Huang S, Tang R, Poon RYC. BCL-W is a regulator of microtubule inhibitor-induced mitotic cell death. Oncotarget 2018; 7:38718-38730. [PMID: 27231850 PMCID: PMC5122423 DOI: 10.18632/oncotarget.9586] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/28/2016] [Indexed: 02/06/2023] Open
Abstract
Microtubule inhibitors including taxanes and vinca alkaloids are among the most widely used anticancer agents. Disrupting the microtubules activates the spindle-assembly checkpoint and traps cells in mitosis. Whether cells subsequently undergo mitotic cell death is an important factor for the effectiveness of the anticancer agents. Given that apoptosis accounts for the majority of mitotic cell death induced by microtubule inhibitors, we performed a systematic study to determine which members of the anti-apoptotic BCL-2 family are involved in determining the duration of mitotic block before cell death or slippage. Depletion of several anti-apoptotic BCL-2-like proteins significantly shortened the time before apoptosis. Among these proteins, BCL-W has not been previously characterized to play a role in mitotic cell death. Although the expression of BCL-W remained constant during mitotic block, it varied significantly between different cell lines. Knockdown of BCL-W with siRNA or disruption of the BCL-W gene with CRISPR-Cas9 speeded up mitotic cell death. Conversely, overexpression of BCL-W delayed mitotic cell death, extending the mitotic block to allow mitotic slippage. Taken together, these results showed that BCL-W contributes to the threshold of anti-apoptotic activity during mitosis.
Collapse
Affiliation(s)
- Shan Huang
- 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
| | - Rui Tang
- 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
|
6
|
Ferrari S, Gentili C. Maintaining Genome Stability in Defiance of Mitotic DNA Damage. Front Genet 2016; 7:128. [PMID: 27493659 PMCID: PMC4954828 DOI: 10.3389/fgene.2016.00128] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/06/2016] [Indexed: 01/08/2023] Open
Abstract
The implementation of decisions affecting cell viability and proliferation is based on prompt detection of the issue to be addressed, formulation and transmission of a correct set of instructions and fidelity in the execution of orders. While the first and the last are purely mechanical processes relying on the faithful functioning of single proteins or macromolecular complexes (sensors and effectors), information is the real cue, with signal amplitude, duration, and frequency ultimately determining the type of response. The cellular response to DNA damage is no exception to the rule. In this review article we focus on DNA damage responses in G2 and Mitosis. First, we set the stage describing mitosis and the machineries in charge of assembling the apparatus responsible for chromosome alignment and segregation as well as the inputs that control its function (checkpoints). Next, we examine the type of issues that a cell approaching mitosis might face, presenting the impact of post-translational modifications (PTMs) on the correct and timely functioning of pathways correcting errors or damage before chromosome segregation. We conclude this essay with a perspective on the current status of mitotic signaling pathway inhibitors and their potential use in cancer therapy.
Collapse
Affiliation(s)
- Stefano Ferrari
- Institute of Molecular Cancer Research, University of Zurich Zurich, Switzerland
| | - Christian Gentili
- Institute of Molecular Cancer Research, University of Zurich Zurich, Switzerland
| |
Collapse
|
7
|
Lee HS, Lee NCO, Kouprina N, Kim JH, Kagansky A, Bates S, Trepel JB, Pommier Y, Sackett D, Larionov V. Effects of Anticancer Drugs on Chromosome Instability and New Clinical Implications for Tumor-Suppressing Therapies. Cancer Res 2016; 76:902-11. [PMID: 26837770 PMCID: PMC4827779 DOI: 10.1158/0008-5472.can-15-1617] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/09/2015] [Indexed: 12/21/2022]
Abstract
Whole chromosomal instability (CIN), manifested as unequal chromosome distribution during cell division, is a distinguishing feature of most cancer types. CIN is generally considered to drive tumorigenesis, but a threshold level exists whereby further increases in CIN frequency in fact hinder tumor growth. While this attribute is appealing for therapeutic exploitation, drugs that increase CIN beyond this therapeutic threshold are currently limited. In our previous work, we developed a quantitative assay for measuring CIN based on the use of a nonessential human artificial chromosome (HAC) carrying a constitutively expressed EGFP transgene. Here, we used this assay to rank 62 different anticancer drugs with respect to their effects on chromosome transmission fidelity. Drugs with various mechanisms of action, such as antimicrotubule activity, histone deacetylase inhibition, mitotic checkpoint inhibition, and targeting of DNA replication and damage responses, were included in the analysis. Ranking of the drugs based on their ability to induce HAC loss revealed that paclitaxel, gemcitabine, dactylolide, LMP400, talazoparib, olaparib, peloruside A, GW843682, VX-680, and cisplatin were the top 10 drugs demonstrating HAC loss at a high frequency. Therefore, identification of currently used compounds that greatly increase chromosome mis-segregation rates should expedite the development of new therapeutic strategies to target and leverage the CIN phenotype in cancer cells.
Collapse
Affiliation(s)
- Hee-Sheung Lee
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Nicholas C O Lee
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Natalay Kouprina
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jung-Hyun Kim
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Alex Kagansky
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Susan Bates
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jane B Trepel
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland
| | - Dan Sackett
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland.
| | - Vladimir Larionov
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
8
|
Marugán C, Torres R, Lallena MJ. Phenotypic Screening Approaches to Develop Aurora Kinase Inhibitors: Drug Discovery Perspectives. Front Oncol 2016; 5:299. [PMID: 26779442 PMCID: PMC4703775 DOI: 10.3389/fonc.2015.00299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 12/14/2015] [Indexed: 01/10/2023] Open
Abstract
Targeting mitotic regulators as a strategy to fight cancer implies the development of drugs against key proteins, such as Aurora-A and -B. Current drugs, which target mitosis through a general mechanism of action (stabilization/destabilization of microtubules), have several side effects (neutropenia, alopecia, and emesis). Pharmaceutical companies aim at avoiding these unwanted effects by generating improved and selective drugs that increase the quality of life of the patients. However, the development of these drugs is an ambitious task that involves testing thousands of compounds through biochemical and cell-based assays. In addition, molecules usually target complex biological processes, involving several proteins and different molecular pathways, further emphasizing the need for high-throughput screening techniques and multiplexing technologies in order to identify drugs with the desired phenotype. We will briefly describe two multiplexing technologies [high-content imaging (HCI) and flow cytometry] and two key processes for drug discovery research (assay development and validation) following our own published industry quality standards. We will further focus on HCI as a useful tool for phenotypic screening and will provide a concrete example of HCI assay to detect Aurora-A or -B selective inhibitors discriminating the off-target effects related to the inhibition of other cell cycle or non-cell cycle key regulators. Finally, we will describe other assays that can help to characterize the in vitro pharmacology of the inhibitors.
Collapse
Affiliation(s)
- Carlos Marugán
- Discovery Chemistry Research and Technology, Lilly Research Laboratories, Eli Lilly and Company , Alcobendas , Spain
| | - Raquel Torres
- Discovery Chemistry Research and Technology, Lilly Research Laboratories, Eli Lilly and Company , Alcobendas , Spain
| | - María José Lallena
- Discovery Chemistry Research and Technology, Lilly Research Laboratories, Eli Lilly and Company , Alcobendas , Spain
| |
Collapse
|
9
|
Asteriti IA, De Mattia F, Guarguaglini G. Cross-Talk between AURKA and Plk1 in Mitotic Entry and Spindle Assembly. Front Oncol 2015; 5:283. [PMID: 26779436 PMCID: PMC4688340 DOI: 10.3389/fonc.2015.00283] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
The Aurora kinase A (AURKA) is involved in different aspects of mitotic control, from mitotic entry to cytokinesis. Consistent with its pleiotropic roles, several AURKA interactors are able to modulate its activity, the best characterized being the microtubule-binding protein TPX2, the centrosomal protein Cep192, and Bora. Bora has been described as an essential cofactor of AURKA for phosphorylation-mediated activation of the mitotic kinase polo-like kinase 1 (Plk1) at the G2/M transition. A complex AURKA/Plk1 signaling axis is emerging, with multiple involved actors; recent data suggest that this control network is not restricted to mitotic entry only, but operates throughout mitosis. Here, we integrate available data from the literature to depict the complex interplay between AURKA and Plk1 in G2 and mitosis and how it contributes to their mitotic functions. We will particularly focus on how the activity of specifically localized AURKA/Plk1 pools is modulated in time and space by their reciprocal regulation to ensure the timely and coordinated unfolding of downstream mitotic events.
Collapse
Affiliation(s)
- Italia Anna Asteriti
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| | - Fabiola De Mattia
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| | - Giulia Guarguaglini
- Institute of Molecular Biology and Pathology, National Research Council (CNR), c/o Department of Biology and Biotechnology, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
10
|
Asteriti IA, Di Cesare E, De Mattia F, Hilsenstein V, Neumann B, Cundari E, Lavia P, Guarguaglini G. The Aurora-A inhibitor MLN8237 affects multiple mitotic processes and induces dose-dependent mitotic abnormalities and aneuploidy. Oncotarget 2015; 5:6229-42. [PMID: 25153724 PMCID: PMC4171625 DOI: 10.18632/oncotarget.2190] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Inhibition of Aurora kinase activity by small molecules is being actively investigated as a potential anti-cancer strategy. A successful therapeutic use of Aurora inhibitors relies on a comprehensive understanding of the effects of inactivating Aurora kinases on cell division, a challenging aim given the pleiotropic roles of those kinases during mitosis. Here we have used the Aurora-A inhibitor MLN8237, currently under phase-I/III clinical trials, in dose-response assays in U2OS human cancer cells synchronously proceeding towards mitosis. By following the behaviour and fate of single Aurora-inhibited cells in mitosis by live microscopy, we show that MLN8237 treatment affects multiple processes that are differentially sensitive to the loss of Aurora-A function. A role of Aurora-A in controlling the orientation of cell division emerges. MLN8237 treatment, even in high doses, fails to induce efficient elimination of dividing cells, or of their progeny, while inducing significant aneuploidy in daughter cells. The results of single-cell analyses show a complex cellular response to MLN8237 and evidence that its effects are strongly dose-dependent: these issues deserve consideration in the light of the design of strategies to kill cancer cells via inhibition of Aurora kinases.
Collapse
Affiliation(s)
- Italia Anna Asteriti
- Institute of Biology, Molecular Medicine and Nanobiotechnology (formerly Institute of Molecular Biology and Pathology), CNR National Research Council, Sapienza University of Rome, Rome, Italy
| | - Erica Di Cesare
- Institute of Biology, Molecular Medicine and Nanobiotechnology (formerly Institute of Molecular Biology and Pathology), CNR National Research Council, Sapienza University of Rome, Rome, Italy
| | - Fabiola De Mattia
- Institute of Biology, Molecular Medicine and Nanobiotechnology (formerly Institute of Molecular Biology and Pathology), CNR National Research Council, Sapienza University of Rome, Rome, Italy
| | - Volker Hilsenstein
- Advanced Light Microscopy Facility, EMBL, Meyerhofstraße 1, Heidelberg, Germany
| | - Beate Neumann
- Advanced Light Microscopy Facility, EMBL, Meyerhofstraße 1, Heidelberg, Germany
| | - Enrico Cundari
- Institute of Biology, Molecular Medicine and Nanobiotechnology (formerly Institute of Molecular Biology and Pathology), CNR National Research Council, Sapienza University of Rome, Rome, Italy
| | - Patrizia Lavia
- Institute of Biology, Molecular Medicine and Nanobiotechnology (formerly Institute of Molecular Biology and Pathology), CNR National Research Council, Sapienza University of Rome, Rome, Italy
| | - Giulia Guarguaglini
- Institute of Biology, Molecular Medicine and Nanobiotechnology (formerly Institute of Molecular Biology and Pathology), CNR National Research Council, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
11
|
Targeting the Mitotic Catastrophe Signaling Pathway in Cancer. Mediators Inflamm 2015; 2015:146282. [PMID: 26491220 PMCID: PMC4600505 DOI: 10.1155/2015/146282] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 05/30/2015] [Indexed: 12/14/2022] Open
Abstract
Mitotic catastrophe, as defined in 2012 by the International Nomenclature Committee on Cell Death, is a bona fide intrinsic oncosuppressive mechanism that senses mitotic failure and responds by driving a cell to an irreversible antiproliferative fate of death or senescence. Thus, failed mitotic catastrophe can promote the unrestrained growth of defective cells, thereby representing a major gateway to tumour development. Furthermore, the activation of mitotic catastrophe offers significant therapeutic advantage which has been exploited in the action of conventional and targeted anticancer agents. Yet, despite its importance in tumour prevention and treatment, the molecular mechanism of mitotic catastrophe is not well understood. A better understanding of the signals that determine cell fate following failed or defective mitosis will reveal new opportunities to selectively target and enhance the programme for therapeutic benefit and reveal biomarkers to predict patient response. This review is focused on the molecular mechanism of mitotic catastrophe induction and signalling and highlights current strategies to exploit the process in cancer therapy.
Collapse
|
12
|
AMPK and PFKFB3 mediate glycolysis and survival in response to mitophagy during mitotic arrest. Nat Cell Biol 2015; 17:1304-16. [PMID: 26322680 DOI: 10.1038/ncb3231] [Citation(s) in RCA: 201] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 07/24/2015] [Indexed: 12/20/2022]
Abstract
Blocking mitotic progression has been proposed as an attractive therapeutic strategy to impair proliferation of tumour cells. However, how cells survive during prolonged mitotic arrest is not well understood. We show here that survival during mitotic arrest is affected by the special energetic requirements of mitotic cells. Prolonged mitotic arrest results in mitophagy-dependent loss of mitochondria, accompanied by reduced ATP levels and the activation of AMPK. Oxidative respiration is replaced by glycolysis owing to AMPK-dependent phosphorylation of PFKFB3 and increased production of this protein as a consequence of mitotic-specific translational activation of its mRNA. Induction of autophagy or inhibition of AMPK or PFKFB3 results in enhanced cell death in mitosis and improves the anti-tumoral efficiency of microtubule poisons in breast cancer cells. Thus, survival of mitotic-arrested cells is limited by their metabolic requirements, a feature with potential implications in cancer therapies aimed to impair mitosis or metabolism in tumour cells.
Collapse
|
13
|
Aurora B Overexpression Causes Aneuploidy and p21Cip1 Repression during Tumor Development. Mol Cell Biol 2015; 35:3566-78. [PMID: 26240282 DOI: 10.1128/mcb.01286-14] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 07/29/2015] [Indexed: 12/21/2022] Open
Abstract
Aurora kinase B, one of the three members of the mammalian Aurora kinase family, is the catalytic component of the chromosomal passenger complex, an essential regulator of chromosome segregation in mitosis. Aurora B is overexpressed in human tumors although whether this kinase may function as an oncogene in vivo is not established. Here, we report a new mouse model in which expression of the endogenous Aurkb locus can be induced in vitro and in vivo. Overexpression of Aurora B in cultured cells induces defective chromosome segregation and aneuploidy. Long-term overexpression of Aurora B in vivo results in aneuploidy and the development of multiple spontaneous tumors in adult mice, including a high incidence of lymphomas. Overexpression of Aurora B also results in a reduced DNA damage response and decreased levels of the p53 target p21(Cip1) in vitro and in vivo, in line with an inverse correlation between Aurora B and p21(Cip1) expression in human leukemias. Thus, overexpression of Aurora B may contribute to tumor formation not only by inducing chromosomal instability but also by suppressing the function of the cell cycle inhibitor p21(Cip1).
Collapse
|
14
|
Sistigu A, Manic G, Obrist F, Vitale I. Trial watch - inhibiting PARP enzymes for anticancer therapy. Mol Cell Oncol 2015; 3:e1053594. [PMID: 27308587 DOI: 10.1080/23723556.2015.1053594] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/16/2015] [Accepted: 05/18/2015] [Indexed: 12/25/2022]
Abstract
Poly(ADP-ribose) polymerases (PARPs) are a members of family of enzymes that catalyze poly(ADP-ribosyl)ation (PARylation) and/or mono(ADP-ribosyl)ation (MARylation), two post-translational protein modifications involved in crucial cellular processes including (but not limited to) the DNA damage response (DDR). PARP1, the most abundant family member, is a nuclear protein that is activated upon sensing distinct types of DNA damage and contributes to their resolution by PARylating multiple DDR players. Recent evidence suggests that, along with DDR, activated PARP1 mediates a series of prosurvival and proapoptotic processes aimed at preserving genomic stability. Despite this potential oncosuppressive role, upregulation and/or overactivation of PARP1 or other PARP enzymes has been reported in a variety of human neoplasms. Over the last few decades, several pharmacologic inhibitors of PARP1 and PARP2 have been assessed in preclinical and clinical studies showing potent antineoplastic activity, particularly against homologous recombination (HR)-deficient ovarian and breast cancers. In this Trial Watch, we describe the impact of PARP enzymes and PARylation in cancer, discuss the mechanism of cancer cell killing by PARP1 inactivation, and summarize the results of recent clinical studies aimed at evaluating the safety and therapeutic profile of PARP inhibitors in cancer patients.
Collapse
Affiliation(s)
| | - Gwenola Manic
- Regina Elena National Cancer Institute , Rome, Italy
| | - Florine Obrist
- Université Paris-Sud/Paris XI, Le Kremlin-Bicêtre, France; INSERM, UMRS1138, Paris, France; Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute, Rome, Italy; Department of Biology, University of Rome "TorVergata", Rome, Italy
| |
Collapse
|
15
|
Obrist F, Manic G, Kroemer G, Vitale I, Galluzzi L. Trial Watch: Proteasomal inhibitors for anticancer therapy. Mol Cell Oncol 2015; 2:e974463. [PMID: 27308423 PMCID: PMC4904962 DOI: 10.4161/23723556.2014.974463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/12/2023]
Abstract
The so-called "ubiquitin-proteasome system" (UPS) is a multicomponent molecular apparatus that catalyzes the covalent attachment of several copies of the small protein ubiquitin to other proteins that are generally (but not always) destined to proteasomal degradation. This enzymatic cascade is crucial for the maintenance of intracellular protein homeostasis (both in physiological conditions and in the course of adaptive stress responses), and regulates a wide array of signaling pathways. In line with this notion, defects in the UPS have been associated with aging as well as with several pathological conditions including cardiac, neurodegenerative, and neoplastic disorders. As transformed cells often experience a constant state of stress (as a result of the hyperactivation of oncogenic signaling pathways and/or adverse microenvironmental conditions), their survival and proliferation are highly dependent on the integrity of the UPS. This rationale has driven an intense wave of preclinical and clinical investigation culminating in 2003 with the approval of the proteasomal inhibitor bortezomib by the US Food and Drug Administration for use in multiple myeloma patients. Another proteasomal inhibitor, carfilzomib, is now licensed by international regulatory agencies for use in multiple myeloma patients, and the approved indications for bortezomib have been extended to mantle cell lymphoma. This said, the clinical activity of bortezomib and carfilzomib is often limited by off-target effects, innate/acquired resistance, and the absence of validated predictive biomarkers. Moreover, the antineoplastic activity of proteasome inhibitors against solid tumors is poor. In this Trial Watch we discuss the contribution of the UPS to oncogenesis and tumor progression and summarize the design and/or results of recent clinical studies evaluating the therapeutic profile of proteasome inhibitors in cancer patients.
Collapse
Affiliation(s)
- Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “Tor Vergata”
| | - Lorenzo Galluzzi
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
16
|
Manic G, Obrist F, Kroemer G, Vitale I, Galluzzi L. Chloroquine and hydroxychloroquine for cancer therapy. Mol Cell Oncol 2014; 1:e29911. [PMID: 27308318 PMCID: PMC4905171 DOI: 10.4161/mco.29911] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Macroautophagy (herein referred to as autophagy) is a highly conserved mechanism for the lysosomal degradation of cytoplasmic components. Autophagy is critical for the maintenance of intracellular homeostasis, both in baseline conditions and in the context of adaptive responses to stress. In line with this notion, defects in the autophagic machinery have been etiologically associated with various human disorders including infectious, inflammatory and neoplastic conditions. Once tumors are established, however, autophagy sustains the survival of malignant cells, hence representing an appealing target for the design of novel anticancer regimens. Accordingly, inhibitors of autophagy including chloroquine and hydroxychloroquine have been shown to mediate substantial antineoplastic effects in preclinical models, especially when combined with chemo- or radiotherapeutic interventions. The pharmacological profile of chloroquine and hydroxychloroquine, however, appear to involve mechanisms other than autophagy inhibition. Here, we discuss the dual role of autophagy in oncogenesis and tumor progression, and summarize the results or design of clinical studies recently completed or initiated to evaluate the therapeutic activity of chloroquine derivatives in cancer patients.
Collapse
Affiliation(s)
| | - Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France; INSERM, UMRS1138; Villejuif, France; Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | - Guido Kroemer
- INSERM, UMRS1138; Villejuif, France; Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
| | - Lorenzo Galluzzi
- Regina Elena National Cancer Institute; Rome, Italy; Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
17
|
Cross RA, McAinsh A. Prime movers: the mechanochemistry of mitotic kinesins. Nat Rev Mol Cell Biol 2014; 15:257-71. [PMID: 24651543 DOI: 10.1038/nrm3768] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mitotic spindles are self-organizing protein machines that harness teams of multiple force generators to drive chromosome segregation. Kinesins are key members of these force-generating teams. Different kinesins walk directionally along dynamic microtubules, anchor, crosslink, align and sort microtubules into polarized bundles, and influence microtubule dynamics by interacting with microtubule tips. The mechanochemical mechanisms of these kinesins are specialized to enable each type to make a specific contribution to spindle self-organization and chromosome segregation.
Collapse
Affiliation(s)
- Robert A Cross
- Warwick Medical School, Gibbet Hill, Coventry CV4 7AL, UK
| | - Andrew McAinsh
- Warwick Medical School, Gibbet Hill, Coventry CV4 7AL, UK
| |
Collapse
|
18
|
Eguren M, Álvarez-Fernández M, García F, López-Contreras AJ, Fujimitsu K, Yaguchi H, Luque-García JL, Fernández-Capetillo O, Muñoz J, Yamano H, Malumbres M. A synthetic lethal interaction between APC/C and topoisomerase poisons uncovered by proteomic screens. Cell Rep 2014; 6:670-83. [PMID: 24508461 DOI: 10.1016/j.celrep.2014.01.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/30/2013] [Accepted: 01/14/2014] [Indexed: 11/24/2022] Open
Abstract
The Anaphase-promoting complex/cyclosome (APC/C) cofactor Cdh1 modulates cell proliferation by targeting multiple cell-cycle regulators for ubiquitin-dependent degradation. Lack of Cdh1 results in structural and numerical chromosome aberrations, a hallmark of genomic instability. By using a proteomic approach in Cdh1-null cells and mouse tissues, we have identified kinesin Eg5 and topoisomerase 2α as Cdh1 targets involved in the maintenance of genomic stability. These proteins are ubiquitinated and degraded through specific KEN and D boxes in a Cdh1-dependent manner. Whereas Cdh1-null cells display partial resistance to Eg5 inhibitors such as monastrol, lack of Cdh1 results in a dramatic sensitivity to Top2α poisons as a consequence of increased levels of trapped Top2α-DNA complexes. Chemical inhibition of the APC/C in cancer cells results in increased sensitivity to Top2α poisons. This work identifies in vivo targets of the mammalian APC/C-Cdh1 complex and reveals synthetic lethal interactions of relevance in anticancer treatments.
Collapse
Affiliation(s)
- Manuel Eguren
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Mónica Álvarez-Fernández
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Fernando García
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | | | - Kazuyuki Fujimitsu
- Cell Cycle Control Group, University College London Cancer Institute, London WC1E 6BT, UK
| | - Hiroko Yaguchi
- Cell Cycle Control Group, University College London Cancer Institute, London WC1E 6BT, UK
| | - José Luis Luque-García
- Department of Analytical Chemistry, Complutense University of Madrid, Madrid 28015, Spain
| | | | - Javier Muñoz
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Hiroyuki Yamano
- Cell Cycle Control Group, University College London Cancer Institute, London WC1E 6BT, UK
| | - Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain.
| |
Collapse
|
19
|
Mashaghi A, Dekker C. Systems and synthetic biology approaches to cell division. SYSTEMS AND SYNTHETIC BIOLOGY 2014; 8:173-8. [PMID: 25136378 DOI: 10.1007/s11693-014-9132-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 01/17/2014] [Indexed: 01/08/2023]
Abstract
Cells proliferate by division into similar daughter cells, a process that lies at the heart of cell biology. Extensive research on cell division has led to the identification of the many components and control elements of the molecular machinery underlying cellular division. Here we provide a brief review of prokaryotic and eukaryotic cell division and emphasize how new approaches such as systems and synthetic biology can provide valuable new insight.
Collapse
Affiliation(s)
- Alireza Mashaghi
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Lorentzweg 1, 2628 CJ Delft, The Netherlands
| | - Cees Dekker
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Lorentzweg 1, 2628 CJ Delft, The Netherlands
| |
Collapse
|
20
|
Pérez de Castro I, Aguirre-Portolés C, Fernández-Miranda G, Cañamero M, Cowley DO, Van Dyke T, Malumbres M. Requirements for Aurora-A in tissue regeneration and tumor development in adult mammals. Cancer Res 2014; 73:6804-15. [PMID: 24242071 DOI: 10.1158/0008-5472.can-13-0586] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aurora-A is a kinase involved in the formation and maturation of the mitotic spindle and chromosome segregation. This kinase is frequently overexpressed in human cancer, and its activity may confer resistance to antitumoral drugs such as Taxol. Inhibition of Aurora-A results in mitotic defects, and this kinase is considered as an attractive therapeutic target for cancer. Nevertheless, the specific requirements for this kinase in adult mammalian tissues remain unclear. Conditional genetic ablation of Aurora-A in adult tissues results in polyploid cells that display a DNA-damage-like response characterized by the upregulation of p53 and the cell-cycle inhibitor p21(Cip1). This is accompanied by apoptotic, differentiation, or senescence markers in a tissue-specific manner. Therapeutic elimination of Aurora-A prevents the progression of skin and mammary gland tumors. However, this is not due to significant levels of apoptosis or senescence, but because Aurora-A-deficient tumors accumulate polyploid cells with limited proliferative potential. Thus, Aurora-A is required for tumor formation in vivo, and the differential response observed in various tissues might have relevant implications in current therapeutic strategies aimed at inhibiting this kinase in the treatment of human cancer.
Collapse
Affiliation(s)
- Ignacio Pérez de Castro
- Authors' Affiliations: Cell Division and Cancer Group; Histopathology Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; and Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| | | | | | | | | | | | | |
Collapse
|