1
|
Conway PJ, Dao J, Kovalskyy D, Mahadevan D, Dray E. Polyploidy in Cancer: Causal Mechanisms, Cancer-Specific Consequences, and Emerging Treatments. Mol Cancer Ther 2024; 23:638-647. [PMID: 38315992 PMCID: PMC11174144 DOI: 10.1158/1535-7163.mct-23-0578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/19/2023] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Drug resistance is the major determinant for metastatic disease and fatalities, across all cancers. Depending on the tissue of origin and the therapeutic course, a variety of biological mechanisms can support and sustain drug resistance. Although genetic mutations and gene silencing through epigenetic mechanisms are major culprits in targeted therapy, drug efflux and polyploidization are more global mechanisms that prevail in a broad range of pathologies, in response to a variety of treatments. There is an unmet need to identify patients at risk for polyploidy, understand the mechanisms underlying polyploidization, and to develop strategies to predict, limit, and reverse polyploidy thus enhancing efficacy of standard-of-care therapy that improve better outcomes. This literature review provides an overview of polyploidy in cancer and offers perspective on patient monitoring and actionable therapy.
Collapse
Affiliation(s)
- Patrick J Conway
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Jonathan Dao
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Dmytro Kovalskyy
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eloise Dray
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
2
|
Streeter SA, Williams AG, Evans JR, Wang J, Guarnaccia AD, Florian AC, Al-Tobasei R, Liu Q, Tansey WP, Weissmiller AM. Mitotic gene regulation by the N-MYC-WDR5-PDPK1 nexus. BMC Genomics 2024; 25:360. [PMID: 38605297 PMCID: PMC11007937 DOI: 10.1186/s12864-024-10282-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND During mitosis the cell depends on proper attachment and segregation of replicated chromosomes to generate two identical progeny. In cancers defined by overexpression or dysregulation of the MYC oncogene this process becomes impaired, leading to genomic instability and tumor evolution. Recently it was discovered that the chromatin regulator WDR5-a critical MYC cofactor-regulates expression of genes needed in mitosis through a direct interaction with the master kinase PDPK1. However, whether PDPK1 and WDR5 contribute to similar mitotic gene regulation in MYC-overexpressing cancers remains unclear. Therefore, to characterize the influence of WDR5 and PDPK1 on mitotic gene expression in cells with high MYC levels, we performed a comparative transcriptomic analysis in neuroblastoma cell lines defined by MYCN-amplification, which results in high cellular levels of the N-MYC protein. RESULTS Using RNA-seq analysis, we identify the genes regulated by N-MYC and PDPK1 in multiple engineered CHP-134 neuroblastoma cell lines and compare them to previously published gene expression data collected in CHP-134 cells following inhibition of WDR5. We find that as expected N-MYC regulates a multitude of genes, including those related to mitosis, but that PDPK1 regulates specific sets of genes involved in development, signaling, and mitosis. Analysis of N-MYC- and PDPK1-regulated genes reveals a small group of commonly controlled genes associated with spindle pole formation and chromosome segregation, which overlap with genes that are also regulated by WDR5. We also find that N-MYC physically interacts with PDPK1 through the WDR5-PDPK1 interaction suggesting regulation of mitotic gene expression may be achieved through a N-MYC-WDR5-PDPK1 nexus. CONCLUSIONS Overall, we identify a small group of genes highly enriched within functional gene categories related to mitotic processes that are commonly regulated by N-MYC, WDR5, and PDPK1 and suggest that a tripartite interaction between the three regulators may be responsible for setting the level of mitotic gene regulation in N-MYC amplified cell lines. This study provides a foundation for future studies to determine the exact mechanism by which N-MYC, WDR5, and PDPK1 converge on cell cycle related processes.
Collapse
Affiliation(s)
- Sarah A Streeter
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - Alexandria G Williams
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - James R Evans
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - Jing Wang
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37203, USA
| | - Alissa D Guarnaccia
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
- Department of Discovery Oncology, Genentech Inc, South San Francisco, CA, 94080, USA
| | - Andrea C Florian
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
- Department of Biology, Belmont University, Nashville, TN, 37212, USA
| | - Rafet Al-Tobasei
- Department of Computer Science, Middle Tennessee State University, Murfreesboro, TN, 32132, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37203, USA
| | - William P Tansey
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - April M Weissmiller
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN, 37132, USA.
| |
Collapse
|
3
|
Manichart N, Laosinwattana C, Somala N, Teerarak M, Chotsaeng N. Physiological mechanism of action and partial separation of herbicide-active compounds from the Diaporthe sp. extract on Amaranthus tricolor L. Sci Rep 2023; 13:18693. [PMID: 37907593 PMCID: PMC10618292 DOI: 10.1038/s41598-023-46201-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/29/2023] [Indexed: 11/02/2023] Open
Abstract
Thirteen fungi that produce compounds with herbicidal activities were isolated, identified, and extracted under the assumption that the mechanism of action occurs during seed exposure to the extract. The extracts from all the fungal strains considerably decreased the growth parameters of Amaranthus tricolor L. The EC010 strain extracts showed the greatest effect. Through ITS region gene sequencing methods, the isolated EC010 was identified as a genus of Diaporthe. The results showed a significant (p < 0.05) inhibitory effect of 91.25% on germination and a decrease in shoot and root length by 91.28% and 95.30%, respectively. The mycelium of Diaporthe sp. was extracted using sequential extraction techniques for the partial separation of the herbicidal fraction. According to the bioassay activities, the EtOAc fraction showed the highest inhibitory activity. The osmotic stress of the A. tricolor seeds was studied. Although the extract increased the accumulation of proline and soluble protein, the treated seeds showed lower imbibition. While the activity of α-amylase was dramatically decreased after treatment. A cytogenetic assay in the treated Allium cepa L. root revealed a decrease in the mitotic index, an altered mitotic phase index, and a promotion of mitotic abnormalities. Accordingly, the Diaporthe sp. may serve as a potential herbicidal compound resource.
Collapse
Affiliation(s)
- Nutcha Manichart
- Department of Plant Production Technology, School of Agricultural Technology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, 10520, Thailand
| | - Chamroon Laosinwattana
- Department of Plant Production Technology, School of Agricultural Technology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, 10520, Thailand.
| | - Naphat Somala
- Department of Plant Production Technology, School of Agricultural Technology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, 10520, Thailand
| | - Montinee Teerarak
- Department of Plant Production Technology, School of Agricultural Technology, King Mongkut's Institute of Technology Ladkrabang, Bangkok, 10520, Thailand
| | - Nawasit Chotsaeng
- Department of Chemistry, School of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok, 10520, Thailand
- Advanced Pure and Applied Chemistry Research Unit (APAC), School of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok, 10520, Thailand
| |
Collapse
|
4
|
Du X, Chen C, Xiao Y, Cui Y, Yang L, Li X, Liu X, Wang R, Tan B. Research on application of tumor treating fields in glioblastoma: A bibliometric and visual analysis. Front Oncol 2022; 12:1055366. [DOI: 10.3389/fonc.2022.1055366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
BackgroundGlioblastoma, one of the common tumors of the central nervous system (CNS), is prone to recurrence even after standard treatment protocols. As an innovative physiotherapy method emerging in recent years, the tumor treating fields (TTFields) technique has been approved for the treatment of glioblastoma due to its non-invasive and portable features. The purpose of this study is to visualize and analyze the scientific results and research trends in TTFields therapy for glioblastoma.MethodsPublications related to TTFields therapy for glioblastoma were searched in the Web of Science Core Collection (WoSCC) database in September 2022. A bibliometric and visual analysis of publications in this field was performed mainly using CiteSpace and R software for country/region, author, journal, reference and keyword.ResultsA total of 618 publications in this field were retrieved, and 248 were finally obtained according to the search criteria, including 159 articles (64.11%) and 89 reviews (37.89%). The cumulative number of publications increased year by year, with an average growth rate (AGR) of 28.50%. The test results of Pearson correlation coefficient showed a high positive correlation between publications and citations (r=0.937, p<0.001). The USA had the largest number of publications (123, 49.60%), followed by Germany (32, 12.90%) and China (30, 12.10%). As for the country/region collaborations, the USA cooperated most closely with other countries/regions, followed by Germany and China. The degree of collaboration (DC) between countries/regions was 25.81%. The institutions with the largest number of publications were Tel Aviv Univ (10), Harvard Med Sch (10) and Novocure Ltd (10). Moreover, Wong E (18) possessed the greatest number of publications, followed by Weinberg U (11) and Kirson E (10). The DC between authors was 97.58%. STUPP R (236) was the most cited author followed by KIRSON ED (164) and GILADI M (104). JOURNAL OF NEURO-ONCOLOGY (22) was the journal with the largest number of published publications (75), followed by FRONTIERS IN ONCOLOGY (15) and CANCERS (13). The top 10 keywords that occurred frequently included glioblastoma (156), tumor treating field (152), temozolomide (134), randomized phase III (48), brain (46), survivor (46), cancer (44), trial (42), alternating electric field (42) and radiotherapy (36). Furthermore, cluster analysis was performed on the basis of keyword co-occurrence, and finally 15 clusters were formed to determine the current research status and future development trend of TTFields therapy for glioblastoma.ConclusionTTFields has been increasingly known as the fourth novel physical anti-tumor therapy in addition to surgery, radiotherapy and anti-tumor drugs. Cooperation and communication between countries/regions need to be enhanced in future research. Several studies have demonstrated the therapeutic potential of TTFields in glioma, and its application alone or in combination with other treatments has become a current research hotspot.
Collapse
|
5
|
Sridhar S, Fukagawa T. Kinetochore Architecture Employs Diverse Linker Strategies Across Evolution. Front Cell Dev Biol 2022; 10:862637. [PMID: 35800888 PMCID: PMC9252888 DOI: 10.3389/fcell.2022.862637] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/23/2022] [Indexed: 01/09/2023] Open
Abstract
The assembly of a functional kinetochore on centromeric chromatin is necessary to connect chromosomes to the mitotic spindle, ensuring accurate chromosome segregation. This connecting function of the kinetochore presents multiple internal and external structural challenges. A microtubule interacting outer kinetochore and centromeric chromatin interacting inner kinetochore effectively confront forces from the external spindle and centromere, respectively. While internally, special inner kinetochore proteins, defined as "linkers," simultaneously interact with centromeric chromatin and the outer kinetochore to enable association with the mitotic spindle. With the ability to simultaneously interact with outer kinetochore components and centromeric chromatin, linker proteins such as centromere protein (CENP)-C or CENP-T in vertebrates and, additionally CENP-QOkp1-UAme1 in yeasts, also perform the function of force propagation within the kinetochore. Recent efforts have revealed an array of linker pathways strategies to effectively recruit the largely conserved outer kinetochore. In this review, we examine these linkages used to propagate force and recruit the outer kinetochore across evolution. Further, we look at their known regulatory pathways and implications on kinetochore structural diversity and plasticity.
Collapse
Affiliation(s)
- Shreyas Sridhar
- Laboratory of Chromosome Biology, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Tatsuo Fukagawa
- Laboratory of Chromosome Biology, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
6
|
Barbosa J, Sunkel CE, Conde C. The Role of Mitotic Kinases and the RZZ Complex in Kinetochore-Microtubule Attachments: Doing the Right Link. Front Cell Dev Biol 2022; 10:787294. [PMID: 35155423 PMCID: PMC8832123 DOI: 10.3389/fcell.2022.787294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/13/2022] [Indexed: 12/31/2022] Open
Abstract
During mitosis, the interaction of kinetochores (KTs) with microtubules (MTs) drives chromosome congression to the spindle equator and supports the segregation of sister chromatids. Faithful genome partition critically relies on the ability of chromosomes to establish and maintain proper amphitelic end-on attachments, a configuration in which sister KTs are connected to robust MT fibers emanating from opposite spindle poles. Because the capture of spindle MTs by KTs is error prone, cells use mechanisms that sense and correct inaccurate KT-MT interactions before committing to segregate sister chromatids in anaphase. If left unresolved, these errors can result in the unequal distribution of chromosomes and lead to aneuploidy, a hallmark of cancer. In this review, we provide an overview of the molecular strategies that monitor the formation and fine-tuning of KT-MT attachments. We describe the complex network of proteins that operates at the KT-MT interface and discuss how AURORA B and PLK1 coordinate several concurrent events so that the stability of KT-MT attachments is precisely modulated throughout mitotic progression. We also outline updated knowledge on how the RZZ complex is regulated to ensure the formation of end-on attachments and the fidelity of mitosis.
Collapse
Affiliation(s)
- João Barbosa
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Claudio E. Sunkel
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
7
|
Wheatley SP. Synchronizing Mammalian Cells for Mitotic Analysis of the Localization of Survivin. Methods Mol Biol 2022; 2415:95-103. [PMID: 34972948 DOI: 10.1007/978-1-0716-1904-9_7] [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] [Indexed: 06/14/2023]
Abstract
This paper describes an easy method to enrich the harvest of adherent mammalian cells at each stage of mitosis (from prometaphase to cytokinesis) by combining Eg5 inhibition using dimethylenastron (DMA) with mitotic shake-off, followed by timed release from the drug.
Collapse
Affiliation(s)
- Sally P Wheatley
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham, UK.
| |
Collapse
|
8
|
Arvind R, Chandana SR, Borad MJ, Pennington D, Mody K, Babiker H. Tumor-Treating Fields: A fourth modality in cancer treatment, new practice updates. Crit Rev Oncol Hematol 2021; 168:103535. [PMID: 34808377 DOI: 10.1016/j.critrevonc.2021.103535] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/12/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022] Open
Abstract
Although major innovations in treatment are advancing, cancer persists as one of the leading causes of mortality. With the rising incidence of cancer and as we treat them, patients incur short term and long-term toxicities of current traditional therapies, including chemotherapy. This imposes a significant physical, emotional, and financial burden among patients, which affects their quality of life. Tumor-Treating Fields (TTFields) is a novel innovative new treatment modality that utilizes alternating electric fields at specific intermediate frequencies to diminish tumor growth by inhibiting mitosis and thus proliferation of malignant cells. The distinguishing feature of this new treatment modality is that it is noninvasive and tolerable. In fact, TTFields is currently FDA approved for the treatment of glioblastoma multiforme (GBM) as well as malignant pleural mesothelioma (MPM). Recently, TTFields have also been found to affect immunogenic cell death resulting in stronger anti-neoplastic effects. In this review, we discuss the mechanism of action of TTFields, the plethora of clinical trials being conducted in patients with GBM, pancreatic adenocarcinoma, ovarian cancer, non-small-cell-lung-cancer (NSCLC), brain metastasis from NSCLC, and MPM and toxicity profile.
Collapse
Affiliation(s)
- Rhea Arvind
- University of Arizona, College of Science, Tucson, AZ, USA
| | - Sreenivasa R Chandana
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, Phase I Program, START Midwest, Grand Rapids, MI, USA
| | - Mitesh J Borad
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Danniel Pennington
- University of Arizona Cancer Center, Clinical Trials Office, Tucson, AZ, USA
| | - Kabir Mody
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Hani Babiker
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
9
|
Bai L, Pfeifer T, Gross W, De La Torre C, Zhao S, Liu L, Schaefer M, Herr I. Establishment of Tumor Treating Fields Combined With Mild Hyperthermia as Novel Supporting Therapy for Pancreatic Cancer. Front Oncol 2021; 11:738801. [PMID: 34804927 PMCID: PMC8597267 DOI: 10.3389/fonc.2021.738801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/18/2021] [Indexed: 12/22/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor with poor prognosis and limited therapeutic options. Alternating electrical fields with low intensity called "Tumor Treating Fields" (TTFields) are a new, non-invasive approach with almost no side effects and phase 3 trials are ongoing in advanced PDAC. We evaluated TTFields in combination with mild hyperthermia. Three established human PDAC cell lines and an immortalized pancreatic duct cell line were treated with TTFields and hyperthermia at 38.5°C, followed by microscopy, assays for MTT, migration, colony and sphere formation, RT-qPCR, FACS, Western blot, microarray and bioinformatics, and in silico analysis using the online databases GSEA, KEGG, Cytoscape-String, and Kaplan-Meier Plotter. Whereas TTFields and hyperthermia alone had weak effects, their combination strongly inhibited the viability of malignant, but not those of nonmalignant cells. Progression features and the cell cycle were impaired, and autophagy was induced. The identified target genes were key players in autophagy, the cell cycle and DNA repair. The expression profiles of part of these target genes were significantly involved in the survival of PDAC patients. In conclusion, the combination of TTFields with mild hyperthermia results in greater efficacy without increased toxicity and could be easily clinically approved as supporting therapy.
Collapse
Affiliation(s)
- Liping Bai
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Tobias Pfeifer
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Gross
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Carolina De La Torre
- Medical Research Center, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Shuyang Zhao
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital of Heidelberg, Heidelberg, Germany
| | - Li Liu
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Michael Schaefer
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Ingrid Herr
- Molecular OncoSurgery, Section Surgical Research, Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
10
|
Al-Hamashi AA, Koranne R, Dlamini S, Alqahtani A, Karaj E, Rashid MS, Knoff JR, Dunworth M, Pflum MKH, Casero RA, Perera L, Taylor WR, Tillekeratne LMV. A new class of cytotoxic agents targets tubulin and disrupts microtubule dynamics. Bioorg Chem 2021; 116:105297. [PMID: 34509798 PMCID: PMC8530978 DOI: 10.1016/j.bioorg.2021.105297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/03/2021] [Accepted: 08/17/2021] [Indexed: 01/08/2023]
Abstract
Despite the advances in treatment strategies, cancer is still the second leading cause of death in the USA. A majority of the currently used cancer drugs have limitations in their clinical use due to poor selectivity, toxic side effects and multiple drug resistance, warranting the development of new anticancer drugs of different mechanisms of action. Here we describe the design, synthesis and initial biological evaluation of a new class of antimitotic agents that modulate tubulin polymerization. Structurally, these compounds are chalcone mimics containing a 1-(1H-imidazol-2-yl)ethan-1-one moiety, which was initially introduced to act as a metal-binding group and inhibit histone deacetylase enzymes. Although several analogues selectively inhibited purified HDAC8 with IC50 values in low micromolar range, tissue culture studies suggest that HDAC inhibition is not a major mechanism responsible for cytotoxicity. The compounds demonstrated cell growth inhibition with GI50 values of upper nanomolar to low micromolar potency with significant selectively for cancer over normal cells. Interestingly, several compounds arrested HeLaM cells in mitosis and seem to target tubulin to cause mitotic arrest. For example, when combined with inhibitors of Aurora B kinase, they led to dramatic disassembly of the mitotic spindle. In-vitro tubulin polymerization studies showed that the compounds reduced the rate of polymerization of microtubules during the elongation phase and lowered the amount of polymerized tubulin during the plateau phase. Finally, in silico docking studies identified binding of IPE-7 to the colchicine site with similar affinity as the test compound D64131. These compounds represent a new antimitotic pharmacophore with limited HDAC inhibitory activity.
Collapse
Affiliation(s)
- Ayad A Al-Hamashi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA
| | - Radhika Koranne
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA
| | - Samkeliso Dlamini
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA
| | - Abdulateef Alqahtani
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA
| | - Endri Karaj
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA
| | - Maisha S Rashid
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA
| | - Joseph R Knoff
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA
| | - Matthew Dunworth
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Bunting/Blaustein Cancer Research Building 1 1650 Orleans Street - Room 551, Baltimore, MD 21231, USA
| | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA
| | - Robert A Casero
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Bunting/Blaustein Cancer Research Building 1 1650 Orleans Street - Room 551, Baltimore, MD 21231, USA
| | - Lalith Perera
- Laboratory of Genome Integrity and Structural Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - William R Taylor
- Department of Biological Sciences, College of Natural Sciences and Mathematics, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA.
| | - L M Viranga Tillekeratne
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, 2801, W. Bancroft Street, Toledo, OH-43606, USA.
| |
Collapse
|
11
|
Böhm M, Killinger K, Dudziak A, Pant P, Jänen K, Hohoff S, Mechtler K, Örd M, Loog M, Sanchez-Garcia E, Westermann S. Cdc4 phospho-degrons allow differential regulation of Ame1 CENP-U protein stability across the cell cycle. eLife 2021; 10:67390. [PMID: 34308839 PMCID: PMC8341979 DOI: 10.7554/elife.67390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/24/2021] [Indexed: 02/05/2023] Open
Abstract
Kinetochores are multi-subunit protein assemblies that link chromosomes to microtubules of the mitotic and meiotic spindle. It is still poorly understood how efficient, centromere-dependent kinetochore assembly is accomplished from hundreds of individual protein building blocks in a cell cycle-dependent manner. Here, by combining comprehensive phosphorylation analysis of native Ctf19CCAN subunits with biochemical and functional assays in the model system budding yeast, we demonstrate that Cdk1 phosphorylation activates phospho-degrons on the essential subunit Ame1CENP-U, which are recognized by the E3 ubiquitin ligase complex SCF-Cdc4. Gradual phosphorylation of degron motifs culminates in M-phase and targets the protein for degradation. Binding of the Mtw1Mis12 complex shields the proximal phospho-degron, protecting kinetochore-bound Ame1 from the degradation machinery. Artificially increasing degron strength partially suppresses the temperature sensitivity of a cdc4 mutant, while overexpression of Ame1-Okp1 is toxic in SCF mutants, demonstrating the physiological importance of this mechanism. We propose that phospho-regulated clearance of excess CCAN subunits facilitates efficient centromere-dependent kinetochore assembly. Our results suggest a novel strategy for how phospho-degrons can be used to regulate the assembly of multi-subunit complexes.
Collapse
Affiliation(s)
- Miriam Böhm
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Kerstin Killinger
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Alexander Dudziak
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Pradeep Pant
- Department of Computational Biochemistry, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Karolin Jänen
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Simone Hohoff
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Karl Mechtler
- IMP - Research Institute of Molecular Pathology, Vienna, Austria.,Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria.,Gregor Mendel Institute (GMI), Austrian Academy of Sciences, Vienna BioCenter (VBC), Vienna, Austria
| | - Mihkel Örd
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mart Loog
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Elsa Sanchez-Garcia
- Department of Computational Biochemistry, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Stefan Westermann
- Department of Molecular Genetics I, Faculty of Biology, Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
12
|
Garcia YA, Velasquez EF, Gao LW, Gholkar AA, Clutario KM, Cheung K, Williams-Hamilton T, Whitelegge JP, Torres JZ. Mapping Proximity Associations of Core Spindle Assembly Checkpoint Proteins. J Proteome Res 2021; 20:3414-3427. [PMID: 34087075 PMCID: PMC8256817 DOI: 10.1021/acs.jproteome.0c00941] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 12/25/2022]
Abstract
The spindle assembly checkpoint (SAC) is critical for sensing defective microtubule-kinetochore attachments and tension across the kinetochore and functions to arrest cells in prometaphase to allow time to repair any errors before proceeding into anaphase. Dysregulation of the SAC leads to chromosome segregation errors that have been linked to human diseases like cancer. Although much has been learned about the composition of the SAC and the factors that regulate its activity, the proximity associations of core SAC components have not been explored in a systematic manner. Here, we have taken a BioID2-proximity-labeling proteomic approach to define the proximity protein environment for each of the five core SAC proteins BUB1, BUB3, BUBR1, MAD1L1, and MAD2L1 in mitotic-enriched populations of cells where the SAC is active. These five protein association maps were integrated to generate a SAC proximity protein network that contains multiple layers of information related to core SAC protein complexes, protein-protein interactions, and proximity associations. Our analysis validated many known SAC complexes and protein-protein interactions. Additionally, it uncovered new protein associations, including the ELYS-MAD1L1 interaction that we have validated, which lend insight into the functioning of core SAC proteins and highlight future areas of investigation to better understand the SAC.
Collapse
Affiliation(s)
- Yenni A. Garcia
- Department of Chemistry and Biochemistry,
University of California, Los Angeles, California 90095,
United States
| | - Erick F. Velasquez
- Department of Chemistry and Biochemistry,
University of California, Los Angeles, California 90095,
United States
| | - Lucy W. Gao
- Pasarow Mass Spectrometry Laboratory, The Jane and
Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of
Medicine, University of California, Los Angeles, California
90095, United States
| | - Ankur A. Gholkar
- Department of Chemistry and Biochemistry,
University of California, Los Angeles, California 90095,
United States
| | - Kevin M. Clutario
- Department of Chemistry and Biochemistry,
University of California, Los Angeles, California 90095,
United States
| | - Keith Cheung
- Department of Chemistry and Biochemistry,
University of California, Los Angeles, California 90095,
United States
| | - Taylor Williams-Hamilton
- Department of Chemistry and Biochemistry,
University of California, Los Angeles, California 90095,
United States
| | - Julian P. Whitelegge
- Pasarow Mass Spectrometry Laboratory, The Jane and
Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of
Medicine, University of California, Los Angeles, California
90095, United States
- Molecular Biology Institute, University of
California, Los Angeles, California 90095, United
States
- Jonsson Comprehensive Cancer Center,
University of California, Los Angeles, California 90095,
United States
| | - Jorge Z. Torres
- Department of Chemistry and Biochemistry,
University of California, Los Angeles, California 90095,
United States
- Molecular Biology Institute, University of
California, Los Angeles, California 90095, United
States
- Jonsson Comprehensive Cancer Center,
University of California, Los Angeles, California 90095,
United States
| |
Collapse
|
13
|
Guerreiro A, De Sousa F, Liaudet N, Ivanova D, Eskat A, Meraldi P. WDR62 localizes katanin at spindle poles to ensure synchronous chromosome segregation. J Cell Biol 2021; 220:212394. [PMID: 34137788 PMCID: PMC8240857 DOI: 10.1083/jcb.202007171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 04/12/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Mutations in the WDR62 gene cause primary microcephaly, a pathological condition often associated with defective cell division that results in severe brain developmental defects. The precise function and localization of WDR62 within the mitotic spindle is, however, still under debate, as it has been proposed to act either at centrosomes or on the mitotic spindle. Here we explored the cellular functions of WDR62 in human epithelial cell lines using both short-term siRNA protein depletions and long-term CRISPR/Cas9 gene knockouts. We demonstrate that WDR62 localizes at spindle poles, promoting the recruitment of the microtubule-severing enzyme katanin. Depletion or loss of WDR62 stabilizes spindle microtubules due to insufficient microtubule minus-end depolymerization but does not affect plus-end microtubule dynamics. During chromosome segregation, WDR62 and katanin promote efficient poleward microtubule flux and favor the synchronicity of poleward movements in anaphase to prevent lagging chromosomes. We speculate that these lagging chromosomes might be linked to developmental defects in primary microcephaly.
Collapse
Affiliation(s)
- Amanda Guerreiro
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Filipe De Sousa
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Radiation Oncology Division, Geneva University Hospitals, Geneva, Switzerland
| | - Nicolas Liaudet
- Bioimaging Facility, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Daria Ivanova
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anja Eskat
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
14
|
Cell-cycle phospho-regulation of the kinetochore. Curr Genet 2021; 67:177-193. [PMID: 33221975 DOI: 10.1007/s00294-020-01127-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023]
Abstract
The kinetochore is a mega-dalton protein assembly that forms within centromeric regions of chromosomes and directs their segregation during cell division. Here we review cell cycle-mediated phosphorylation events at the kinetochore, with a focus on the budding yeast Saccharomyces cerevisiae and the insight gained from forced associations of kinases and phosphatases. The point centromeres found in the budding yeast S. cerevisiae are one of the simplest such structures found in eukaryotes. The S. cerevisiae kinetochore comprises a single nucleosome, containing a centromere-specific H3 variant Cse4CENP-A, bound to a set of kinetochore proteins that connect to a single microtubule. Despite the simplicity of the budding yeast kinetochore, the proteins are mostly homologous with their mammalian counterparts. In some cases, human proteins can complement their yeast orthologs. Like its mammalian equivalent, the regulation of the budding yeast kinetochore is complex: integrating signals from the cell cycle, checkpoints, error correction, and stress pathways. The regulatory signals from these diverse pathways are integrated at the kinetochore by post-translational modifications, notably phosphorylation and dephosphorylation, to control chromosome segregation. Here we highlight the complex interplay between the activity of the different cell-cycle kinases and phosphatases at the kinetochore, emphasizing how much more we have to understand this essential structure.
Collapse
|
15
|
Yu H, Li Y, Li L, Huang J, Wang X, Tang R, Jiang Z, Lv L, Chen F, Yu C, Yuan K. Functional reciprocity of proteins involved in mitosis and endocytosis. FEBS J 2020; 288:5850-5866. [PMID: 33300206 DOI: 10.1111/febs.15664] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/29/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
Mitosis and endocytosis are two fundamental cellular processes essential for maintaining a eukaryotic life. Mitosis partitions duplicated chromatin enveloped in the nuclear membrane into two new cells, whereas endocytosis takes in extracellular substances through membrane invagination. These two processes are spatiotemporally separated and seemingly unrelated. However, recent studies have uncovered that endocytic proteins have moonlighting functions in mitosis, and mitotic complexes manifest additional roles in endocytosis. In this review, we summarize important proteins or protein complexes that participate in both processes, compare their mechanism of action, and discuss the rationale behind this multifunctionality. We also speculate on the possible origin of the functional reciprocity from an evolutionary perspective.
Collapse
Affiliation(s)
- Haibin Yu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yinshuang Li
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Li Li
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | | | - Xujuan Wang
- The High School Attached to Hunan Normal University, Changsha, China
| | - Ruijun Tang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Zhenghui Jiang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Lu Lv
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Fang Chen
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chunhong Yu
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Oncology, Xiangya Hospital & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,The Biobank of Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Zhang Z, Bellini D, Barford D. Crystal structure of the Cenp-HIKHead-TW sub-module of the inner kinetochore CCAN complex. Nucleic Acids Res 2020; 48:11172-11184. [PMID: 32976599 PMCID: PMC7641736 DOI: 10.1093/nar/gkaa772] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/31/2020] [Accepted: 09/11/2020] [Indexed: 11/12/2022] Open
Abstract
Kinetochores are large multi-subunit complexes that attach centromeric chromatin to microtubules of the mitotic spindle, enabling sister chromatid segregation in mitosis. The inner kinetochore constitutive centromere associated network (CCAN) complex assembles onto the centromere-specific Cenp-A nucleosome (Cenp-ANuc), thereby coupling the centromere to the microtubule-binding outer kinetochore. CCAN is a conserved 14-16 subunit complex composed of discrete modules. Here, we determined the crystal structure of the Saccharomyces cerevisiae Cenp-HIKHead-TW sub-module, revealing how Cenp-HIK and Cenp-TW interact at the conserved Cenp-HIKHead-Cenp-TW interface. A major interface is formed by the C-terminal anti-parallel α-helices of the histone fold extension (HFE) of the Cenp-T histone fold domain (HFD) combining with α-helix H3 of Cenp-K to create a compact three α-helical bundle. We fitted the Cenp-HIKHead-TW sub-module to the previously determined cryo-EM map of the S. cerevisiae CCAN-Cenp-ANuc complex. This showed that the HEAT repeat domain of Cenp-IHead and C-terminal HFD of Cenp-T of the Cenp-HIKHead-TW sub-module interact with the nucleosome DNA gyre at a site close to the Cenp-ANuc dyad axis. Our structure provides a framework for understanding how Cenp-T links centromeric Cenp-ANuc to the outer kinetochore through its HFD and N-terminal Ndc80-binding motif, respectively.
Collapse
Affiliation(s)
- Ziguo Zhang
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Dom Bellini
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - David Barford
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
17
|
Cunha-Silva S, Conde C. From the Nuclear Pore to the Fibrous Corona: A MAD Journey to Preserve Genome Stability. Bioessays 2020; 42:e2000132. [PMID: 32885448 DOI: 10.1002/bies.202000132] [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: 05/31/2020] [Revised: 07/22/2020] [Indexed: 11/09/2022]
Abstract
The relationship between kinetochores and nuclear pore complexes (NPCs) is intimate but poorly understood. Several NPC components and associated proteins are relocated to mitotic kinetochores to assist in different activities that ensure faithful chromosome segregation. Such is the case of the Mad1-c-Mad2 complex, the catalytic core of the spindle assembly checkpoint (SAC), a surveillance pathway that delays anaphase until all kinetochores are attached to spindle microtubules. Mad1-c-Mad2 is recruited to discrete domains of unattached kinetochores from where it promotes the rate-limiting step in the assembly of anaphase-inhibitory complexes. SAC proficiency further requires Mad1-c-Mad2 to be anchored at NPCs during interphase. However, the mechanistic relevance of this arrangement for SAC function remains ill-defined. Recent studies uncover the molecular underpinnings that coordinate the release of Mad1-c-Mad2 from NPCs with its prompt recruitment to kinetochores. Here, current knowledge on Mad1-c-Mad2 function and spatiotemporal regulation is reviewed and the critical questions that remain unanswered are highlighted.
Collapse
Affiliation(s)
- Sofia Cunha-Silva
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal.,Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, 4050-313, Portugal
| | - Carlos Conde
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| |
Collapse
|
18
|
Ólafsson G, Thorpe PH. Polo kinase recruitment via the constitutive centromere-associated network at the kinetochore elevates centromeric RNA. PLoS Genet 2020; 16:e1008990. [PMID: 32810142 PMCID: PMC7455000 DOI: 10.1371/journal.pgen.1008990] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/28/2020] [Accepted: 07/13/2020] [Indexed: 12/23/2022] Open
Abstract
The kinetochore, a multi-protein complex assembled on centromeres, is essential to segregate chromosomes during cell division. Deficiencies in kinetochore function can lead to chromosomal instability and aneuploidy-a hallmark of cancer cells. Kinetochore function is controlled by recruitment of regulatory proteins, many of which have been documented, however their function often remains uncharacterized and many are yet to be identified. To identify candidates of kinetochore regulation we used a proteome-wide protein association strategy in budding yeast and detected many proteins that are involved in post-translational modifications such as kinases, phosphatases and histone modifiers. We focused on the Polo-like kinase, Cdc5, and interrogated which cellular components were sensitive to constitutive Cdc5 localization. The kinetochore is particularly sensitive to constitutive Cdc5 kinase activity. Targeting Cdc5 to different kinetochore subcomplexes produced diverse phenotypes, consistent with multiple distinct functions at the kinetochore. We show that targeting Cdc5 to the inner kinetochore, the constitutive centromere-associated network (CCAN), increases the levels of centromeric RNA via an SPT4 dependent mechanism.
Collapse
Affiliation(s)
- Guðjón Ólafsson
- School of Biological and Chemical Sciences, Queen Mary, University of London, London, United Kingdom
| | - Peter H. Thorpe
- School of Biological and Chemical Sciences, Queen Mary, University of London, London, United Kingdom
| |
Collapse
|
19
|
Barbosa J, Conde C, Sunkel C. RZZ-SPINDLY-DYNEIN: you got to keep 'em separated. Cell Cycle 2020; 19:1716-1726. [PMID: 32544383 PMCID: PMC7469663 DOI: 10.1080/15384101.2020.1780382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 10/24/2022] Open
Abstract
To maintain genome stability, chromosomes must be equally distributed among daughter cells at the end of mitosis. The accuracy of chromosome segregation requires sister-kinetochores to stably attach to microtubules emanating from opposite spindle poles. However, initial kinetochore-microtubule interactions are able to turnover so that defective attachment configurations that typically arise during early mitosis may be corrected. Growing evidence supports a role for the RZZ complex in preventing the stabilization of erroneous kinetochore-microtubule attachments. This inhibitory function of RZZ toward end-on attachments is relieved by DYNEIN-mediated transport of the complex as chromosomes congress and appropriate interactions with microtubules are established. However, it remains unclear how DYNEIN is antagonized to prevent premature RZZ removal. We recently described a new mechanism that sheds new light on this matter. We found that POLO kinase phosphorylates the DYNEIN adaptor SPINDLY to promote the uncoupling between RZZ and DYNEIN. Elevated POLO activity during prometaphase ensures that RZZ is retained at kinetochores to allow the dynamic turnover of kinetochore-microtubule interactions and prevent the stabilization of erroneous attachments. Here, we discuss additional interpretations to explain a model for POLO-dependent regulation of the RZZ-SPINDLY-DYNEIN module during mitosis.
Collapse
Affiliation(s)
- João Barbosa
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
| | - Carlos Conde
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
| | - Claudio Sunkel
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciência Biomédicas Abel Salazar da Universidade do Porto, Porto, Portugal
| |
Collapse
|
20
|
Rohrberg J, Van de Mark D, Amouzgar M, Lee JV, Taileb M, Corella A, Kilinc S, Williams J, Jokisch ML, Camarda R, Balakrishnan S, Shankar R, Zhou A, Chang AN, Chen B, Rugo HS, Dumont S, Goga A. MYC Dysregulates Mitosis, Revealing Cancer Vulnerabilities. Cell Rep 2020; 30:3368-3382.e7. [PMID: 32160543 PMCID: PMC7085414 DOI: 10.1016/j.celrep.2020.02.041] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/18/2019] [Accepted: 02/06/2020] [Indexed: 12/13/2022] Open
Abstract
Tumors that overexpress the MYC oncogene are frequently aneuploid, a state associated with highly aggressive cancers and tumor evolution. However, how MYC causes aneuploidy is not well understood. Here, we show that MYC overexpression induces mitotic spindle assembly defects and chromosomal instability (CIN) through effects on microtubule nucleation and organization. Attenuating MYC expression reverses mitotic defects, even in established tumor cell lines, indicating an ongoing role for MYC in CIN. MYC reprograms mitotic gene expression, and we identify TPX2 to be permissive for spindle assembly in MYC-high cells. TPX2 depletion blocks mitotic progression, induces cell death, and prevents tumor growth. Further elevating TPX2 expression reduces mitotic defects in MYC-high cells. MYC and TPX2 expression may be useful biomarkers to stratify patients for anti-mitotic therapies. Our studies implicate MYC as a regulator of mitosis and suggest that blocking MYC activity can attenuate the emergence of CIN and tumor evolution.
Collapse
Affiliation(s)
- Julia Rohrberg
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA.
| | - Daniel Van de Mark
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Meelad Amouzgar
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Joyce V Lee
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Moufida Taileb
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Alexandra Corella
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Seda Kilinc
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Jeremy Williams
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Marie-Lena Jokisch
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Roman Camarda
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Sanjeev Balakrishnan
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Rama Shankar
- Department of Pediatrics and Human Development and Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Alicia Zhou
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Bin Chen
- Department of Pediatrics and Human Development and Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Hope S Rugo
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Sophie Dumont
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Andrei Goga
- Department of Cell & Tissue Biology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Lawrimore J, Doshi A, Walker B, Bloom K. AI-Assisted Forward Modeling of Biological Structures. Front Cell Dev Biol 2019; 7:279. [PMID: 31799251 PMCID: PMC6868055 DOI: 10.3389/fcell.2019.00279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/30/2019] [Indexed: 01/01/2023] Open
Abstract
The rise of machine learning and deep learning technologies have allowed researchers to automate image classification. We describe a method that incorporates automated image classification and principal component analysis to evaluate computational models of biological structures. We use a computational model of the kinetochore to demonstrate our artificial-intelligence (AI)-assisted modeling method. The kinetochore is a large protein complex that connects chromosomes to the mitotic spindle to facilitate proper cell division. The kinetochore can be divided into two regions: the inner kinetochore, including proteins that interact with DNA; and the outer kinetochore, comprised of microtubule-binding proteins. These two kinetochore regions have been shown to have different distributions during metaphase in live budding yeast and therefore act as a test case for our forward modeling technique. We find that a simple convolutional neural net (CNN) can correctly classify fluorescent images of inner and outer kinetochore proteins and show a CNN trained on simulated, fluorescent images can detect difference in experimental images. A polymer model of the ribosomal DNA locus serves as a second test for the method. The nucleolus surrounds the ribosomal DNA locus and appears amorphous in live-cell, fluorescent microscopy experiments in budding yeast, making detection of morphological changes challenging. We show a simple CNN can detect subtle differences in simulated images of the ribosomal DNA locus, demonstrating our CNN-based classification technique can be used on a variety of biological structures.
Collapse
Affiliation(s)
- Josh Lawrimore
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ayush Doshi
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Benjamin Walker
- Department of Mathematics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kerry Bloom
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
22
|
Vukušić K, Buđa R, Tolić IM. Force-generating mechanisms of anaphase in human cells. J Cell Sci 2019; 132:132/18/jcs231985. [DOI: 10.1242/jcs.231985] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
ABSTRACT
What forces drive chromosome segregation remains one of the most challenging questions in cell division. Even though the duration of anaphase is short, it is of utmost importance for genome fidelity that no mistakes are made. Seminal studies in model organisms have revealed different mechanisms operating during chromosome segregation in anaphase, but the translation of these mechanisms to human cells is not straightforward. Recent work has shown that kinetochore fiber depolymerization during anaphase A is largely motor independent, whereas spindle elongation during anaphase B is coupled to sliding of interpolar microtubules in human cells. In this Review, we discuss the current knowledge on the mechanisms of force generation by kinetochore, interpolar and astral microtubules. By combining results from numerous studies, we propose a comprehensive picture of the role of individual force-producing and -regulating proteins. Finally, by linking key concepts of anaphase to most recent data, we summarize the contribution of all proposed mechanisms to chromosome segregation and argue that sliding of interpolar microtubules and depolymerization at the kinetochore are the main drivers of chromosome segregation during early anaphase in human cells.
Collapse
Affiliation(s)
- Kruno Vukušić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Renata Buđa
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Iva M. Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| |
Collapse
|
23
|
Akera T, Trimm E, Lampson MA. Molecular Strategies of Meiotic Cheating by Selfish Centromeres. Cell 2019; 178:1132-1144.e10. [PMID: 31402175 DOI: 10.1016/j.cell.2019.07.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/11/2019] [Accepted: 06/25/2019] [Indexed: 10/26/2022]
Abstract
Asymmetric division in female meiosis creates selective pressure favoring selfish centromeres that bias their transmission to the egg. This centromere drive can explain the paradoxical rapid evolution of both centromere DNA and centromere-binding proteins despite conserved centromere function. Here, we define a molecular pathway linking expanded centromeres to histone phosphorylation and recruitment of microtubule destabilizing factors, leading to detachment of selfish centromeres from spindle microtubules that would direct them to the polar body. Exploiting centromere divergence between species, we show that selfish centromeres in two hybrid mouse models use the same molecular pathway but modulate it differently to enrich destabilizing factors. Our results indicate that increasing microtubule destabilizing activity is a general strategy for drive in both models, but centromeres have evolved distinct mechanisms to increase that activity. Furthermore, we show that drive depends on slowing meiotic progression, suggesting that selfish centromeres can be suppressed by regulating meiotic timing.
Collapse
Affiliation(s)
- Takashi Akera
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Trimm
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael A Lampson
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
24
|
Lawrimore J, Bloom K. The regulation of chromosome segregation via centromere loops. Crit Rev Biochem Mol Biol 2019; 54:352-370. [PMID: 31573359 PMCID: PMC6856439 DOI: 10.1080/10409238.2019.1670130] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/02/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022]
Abstract
Biophysical studies of the yeast centromere have shown that the organization of the centromeric chromatin plays a crucial role in maintaining proper tension between sister kinetochores during mitosis. While centromeric chromatin has traditionally been considered a simple spring, recent work reveals the centromere as a multifaceted, tunable shock absorber. Centromeres can differ from other regions of the genome in their heterochromatin state, supercoiling state, and enrichment of structural maintenance of chromosomes (SMC) protein complexes. Each of these differences can be utilized to alter the effective stiffness of centromeric chromatin. In budding yeast, the SMC protein complexes condensin and cohesin stiffen chromatin by forming and cross-linking chromatin loops, respectively, into a fibrous structure resembling a bottlebrush. The high density of the loops compacts chromatin while spatially isolating the tension from spindle pulling forces to a subset of the chromatin. Paradoxically, the molecular crowding of chromatin via cohesin and condensin also causes an outward/poleward force. The structure allows the centromere to act as a shock absorber that buffers the variable forces generated by dynamic spindle microtubules. Based on the distribution of SMCs from bacteria to human and the conserved distance between sister kinetochores in a wide variety of organisms (0.4 to 1 micron), we propose that the bottlebrush mechanism is the foundational principle for centromere function in eukaryotes.
Collapse
Affiliation(s)
- Josh Lawrimore
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kerry Bloom
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
25
|
Vallardi G, Cordeiro MH, Saurin AT. A Kinase-Phosphatase Network that Regulates Kinetochore-Microtubule Attachments and the SAC. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:457-484. [PMID: 28840249 DOI: 10.1007/978-3-319-58592-5_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The KMN network (for KNL1, MIS12 and NDC80 complexes) is a hub for signalling at the outer kinetochore. It integrates the activities of two kinases (MPS1 and Aurora B) and two phosphatases (PP1 and PP2A-B56) to regulate kinetochore-microtubule attachments and the spindle assembly checkpoint (SAC). We will first discuss each of these enzymes separately, to describe how they are regulated at kinetochores and why this is important for their primary function in controlling either microtubule attachments or the SAC. We will then discuss why inhibiting any one of them individually produces secondary effects on all the others. This cross-talk may help to explain why all enzymes have been linked to both processes, even though the direct evidence suggests they each control only one. This chapter therefore describes how a network of kinases and phosphatases work together to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Giulia Vallardi
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Marilia Henriques Cordeiro
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian Thomas Saurin
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
26
|
|
27
|
Moura M, Conde C. Phosphatases in Mitosis: Roles and Regulation. Biomolecules 2019; 9:E55. [PMID: 30736436 PMCID: PMC6406801 DOI: 10.3390/biom9020055] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
Mitosis requires extensive rearrangement of cellular architecture and of subcellular structures so that replicated chromosomes can bind correctly to spindle microtubules and segregate towards opposite poles. This process originates two new daughter nuclei with equal genetic content and relies on highly-dynamic and tightly regulated phosphorylation of numerous cell cycle proteins. A burst in protein phosphorylation orchestrated by several conserved kinases occurs as cells go into and progress through mitosis. The opposing dephosphorylation events are catalyzed by a small set of protein phosphatases, whose importance for the accuracy of mitosis is becoming increasingly appreciated. This review will focus on the established and emerging roles of mitotic phosphatases, describe their structural and biochemical properties, and discuss recent advances in understanding the regulation of phosphatase activity and function.
Collapse
Affiliation(s)
- Margarida Moura
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal.
| | - Carlos Conde
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
28
|
Ruan W, Lim HH, Surana U. Mapping Mitotic Death: Functional Integration of Mitochondria, Spindle Assembly Checkpoint and Apoptosis. Front Cell Dev Biol 2019; 6:177. [PMID: 30687704 PMCID: PMC6335265 DOI: 10.3389/fcell.2018.00177] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/22/2018] [Indexed: 01/18/2023] Open
Abstract
Targeting the mitotic pathways of rapidly proliferating tumor cells has been an effective strategy in traditional cancer therapy. Chemotherapeutics such as taxanes and vinca alkaloids, which disrupt microtubule function, have enjoyed clinical success; however, the accompanying side effects, toxicity and multi drug resistance remain as serious concerns. The emerging classes of inhibitors targeting mitotic kinases and proteasome face their own set of challenges. It is hoped that elucidation of the regulatory interface between mitotic checkpoints, mitochondria and mitotic death will aid the development of more efficacious anti-mitotic agents and improved treatment protocols. The links between the spindle assembly checkpoint (SAC) and mitochondrial dynamics that control the progression of anti-mitotic agent-induced apoptosis have been under investigation for several years and the functional integration of these various signaling networks is now beginning to emerge. In this review, we highlight current research on the regulation of SAC, the death pathway and mitochondria with particular focus on their regulatory interconnections.
Collapse
Affiliation(s)
- Weimei Ruan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hong Hwa Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Uttam Surana
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Pharmacology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
29
|
Edwards F, Maton G, Gareil N, Canman JC, Dumont J. BUB-1 promotes amphitelic chromosome biorientation via multiple activities at the kinetochore. eLife 2018; 7:40690. [PMID: 30547880 PMCID: PMC6303103 DOI: 10.7554/elife.40690] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/13/2018] [Indexed: 12/03/2022] Open
Abstract
Accurate chromosome segregation relies on bioriented amphitelic attachments of chromosomes to microtubules of the mitotic spindle, in which sister chromatids are connected to opposite spindle poles. BUB-1 is a protein of the Spindle Assembly Checkpoint (SAC) that coordinates chromosome attachment with anaphase onset. BUB-1 is also required for accurate sister chromatid segregation independently of its SAC function, but the underlying mechanism remains unclear. Here we show that, in Caenorhabditis elegans embryos, BUB-1 accelerates the establishment of non-merotelic end-on kinetochore-microtubule attachments by recruiting the RZZ complex and its downstream partner dynein-dynactin at the kinetochore. In parallel, BUB-1 limits attachment maturation by the SKA complex. This activity opposes kinetochore-microtubule attachment stabilisation promoted by CLS-2CLASP-dependent kinetochore-microtubule assembly. BUB-1 is therefore a SAC component that coordinates the function of multiple downstream kinetochore-associated proteins to ensure accurate chromosome segregation.
Collapse
Affiliation(s)
- Frances Edwards
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Gilliane Maton
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Nelly Gareil
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University, New York, United States
| | - Julien Dumont
- Institut Jacques Monod, CNRS, UMR 7592, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
30
|
Edzuka T, Goshima G. Drosophila kinesin-8 stabilizes the kinetochore-microtubule interaction. J Cell Biol 2018; 218:474-488. [PMID: 30538142 PMCID: PMC6363442 DOI: 10.1083/jcb.201807077] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/24/2018] [Accepted: 11/28/2018] [Indexed: 02/06/2023] Open
Abstract
Kinesin-8 motor proteins control chromosome alignment in a variety of species, but the specific biochemical activity responsible is unclear. Edzuka and Goshima find that Drosophila kinesin-8 (Klp67A) exhibits both microtubule plus end–stabilizing and –destabilizing activities in vitro. In cells, Klp67A, and likely human kinesin-8 (KIF18A) as well, stabilize the kinetochore–microtubule attachment during mitosis. Kinesin-8 is required for proper chromosome alignment in a variety of animal and yeast cell types. However, it is unclear how this motor protein family controls chromosome alignment, as multiple biochemical activities, including inconsistent ones between studies, have been identified. Here, we find that Drosophila kinesin-8 (Klp67A) possesses both microtubule (MT) plus end–stabilizing and –destabilizing activity, in addition to kinesin-8's commonly observed MT plus end–directed motility and tubulin-binding activity in vitro. We further show that Klp67A is required for stable kinetochore–MT attachment during prometaphase in S2 cells. In the absence of Klp67A, abnormally long MTs interact in an “end-on” fashion with kinetochores at normal frequency. However, the interaction is unstable, and MTs frequently become detached. This phenotype is rescued by ectopic expression of the MT plus end–stabilizing factor CLASP, but not by artificial shortening of MTs. We show that human kinesin-8 (KIF18A) is also important to ensure proper MT attachment. Overall, these results suggest that the MT-stabilizing activity of kinesin-8 is critical for stable kinetochore–MT attachment.
Collapse
Affiliation(s)
- Tomoya Edzuka
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Japan.,Marine Biological Laboratory, Woods Hole, MA
| | - Gohta Goshima
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Japan .,Marine Biological Laboratory, Woods Hole, MA
| |
Collapse
|
31
|
Yan K, Zhang Z, Yang J, McLaughlin SH, Barford D. Architecture of the CBF3-centromere complex of the budding yeast kinetochore. Nat Struct Mol Biol 2018; 25:1103-1110. [PMID: 30478265 PMCID: PMC6292502 DOI: 10.1038/s41594-018-0154-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/10/2018] [Indexed: 01/01/2023]
Abstract
Kinetochores are multicomponent complexes responsible for coordinating the attachment of centromeric DNA to mitotic-spindle microtubules. The point centromeres of budding yeast are organized into three centromeric determining elements (CDEs), and are associated with the centromere-specific nucleosome Cse4. Deposition of Cse4 at CEN loci is dependent on the CBF3 complex that engages CDEIII to direct Cse4 nucleosomes to CDEII. To understand how CBF3 recognizes CDEIII and positions Cse4, we determined a cryo-EM structure of a CBF3-CEN complex. CBF3 interacts with CEN DNA as a head-to-head dimer that includes the whole of CDEIII and immediate 3' regions. Specific CEN-binding of CBF3 is mediated by a Cep3 subunit of one of the CBF3 protomers that forms major groove interactions with the conserved and essential CCG and TGT motifs of CDEIII. We propose a model for a CBF3-Cse4-CEN complex with implications for understanding CBF3-directed deposition of the Cse4 nucleosome at CEN loci.
Collapse
Affiliation(s)
- Kaige Yan
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Ziguo Zhang
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Jing Yang
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | | |
Collapse
|
32
|
Wu Y, Tan L, Chen J, Li H, Ying H, Jiang Y, Wu Q, Yu G, Tian Y, Yu J, Zeng T, Yan L, Liu C. MAD2 Combined with Mitotic Spindle Apparatus (MSA) and Anticentromere Antibody (ACA) for Diagnosis of Small Cell Lung Cancer (SCLC). Med Sci Monit 2018; 24:7541-7547. [PMID: 30346937 PMCID: PMC6354645 DOI: 10.12659/msm.909772] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND MAD2 is the gene controlling mitosis. Many studies have assessed MAD2 in various types of carcinoma. Antinuclear mitotic spindle apparatus antibody (MSA) and anticentromere antibody (ACA) are related mitotic antibodies, playing roles in autoimmune diseases and carcinomas, but the expression of MAD2, MSA, and ACA in SCLC is unclear. MATERIAL AND METHODS We enrolled 70 SCLC patients, 72 non-small cell lung cancer (NSCLC) patients, and 65 pulmonary nodule (PN) patients. MAD2 expression was measured through agarose electrophoresis and qt-PCR. Antinuclear mitotic spindle apparatus antibody (MSA) and anticentromere antibody (ACA) were detected by indirect immunofluorescence (IIF). RESULTS MAD2 was found both in SCLC and NSCLC. Interestingly, there was a significant difference found between SCLC and NSCLC using qt-PCR (P<0.05). The area under the ROC curve of MAD2 expression was 0.799, with medium diagnostic value. MAD2 expression was related to age, lymphatic metastasis, and survival time, but not with sex. The positivity for MSA and ACA by IIF assay were 37.20% and 34.00%, respectively, in the SCLC group, which were higher than in the NSCLC and pulmonary nodule groups (P<0.05). The kappa values of MSA and ACA with MAD2 expression were 0.73 and 0.65, respectively, with moderate consistency. Combining MAD2 with MSA and ACA enhanced the sensitivity and specificity for diagnosing SCLC. CONCLUSIONS MAD2 expression was found to be involved in carcinogenesis and prognosis of SCLC. The combination of MAD2 with MSA and ACA is useful for early diagnosis and shows promise in treatment of SCLC.
Collapse
Affiliation(s)
- Yang Wu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Liming Tan
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Juanjuan Chen
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Hua Li
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Houqun Ying
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Yongqing Jiang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Qiong Wu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Guofang Yu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Yongjian Tian
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Jianlin Yu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Tingting Zeng
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Laboratory Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Linxian Yan
- Department of Medical Supply, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Chuan Liu
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
33
|
Joglekar AP, Kukreja AA. How Kinetochore Architecture Shapes the Mechanisms of Its Function. Curr Biol 2018; 27:R816-R824. [PMID: 28829971 DOI: 10.1016/j.cub.2017.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The eukaryotic kinetochore is a sophisticated multi-protein machine that segregates chromosomes during cell division. To ensure accurate chromosome segregation, it performs three major functions using disparate molecular mechanisms. It operates a mechanosensitive signaling cascade known as the spindle assembly checkpoint (SAC) to detect and signal the lack of attachment to spindle microtubules, and delay anaphase onset in response. In addition, after attaching to spindle microtubules, the kinetochore generates the force necessary to move chromosomes. Finally, if the two sister kinetochores on a chromosome are both attached to microtubules emanating from the same spindle pole, they activate another mechanosensitive mechanism to correct the monopolar attachments. All three of these functions maintain genome stability during cell division. The outlines of the biochemical activities responsible for these functions are now available. How the kinetochore integrates the underlying molecular mechanisms is still being elucidated. In this Review, we discuss how the nanoscale protein organization in the kinetochore, which we refer to as kinetochore 'architecture', organizes its biochemical activities to facilitate the realization and integration of emergent mechanisms underlying its three major functions. For this discussion, we will use the relatively simple budding yeast kinetochore as a model, and extrapolate insights gained from this model to elucidate functional roles of the architecture of the much more complex human kinetochore.
Collapse
Affiliation(s)
- Ajit P Joglekar
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Biophysics, University of Michigan, Ann Arbor, MI, USA.
| | | |
Collapse
|
34
|
Kessler AF, Frömbling GE, Gross F, Hahn M, Dzokou W, Ernestus RI, Löhr M, Hagemann C. Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibition. Cell Death Discov 2018; 4:12. [PMID: 30210815 PMCID: PMC6125382 DOI: 10.1038/s41420-018-0079-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/22/2018] [Indexed: 01/22/2023] Open
Abstract
Tumor treating fields (TTFields) are approved for glioblastoma (GBM) therapy. TTFields disrupt cell division by inhibiting spindle fiber formation. Spindle assembly checkpoint (SAC) inhibition combined with antimitotic drugs synergistically decreases glioma cell growth in cell culture and mice. We hypothesized that SAC inhibition will increase TTFields efficacy. Human GBM cells (U-87 MG, GaMG) were treated with TTFields (200 kHz, 1.7 V/cm) and/or the SAC inhibitor MPS1-IN-3 (IN-3, 4 µM). Cells were counted after 24, 48, and 72 h of treatment and at 24 and 72 h after end of treatment (EOT). Flow cytometry, immunofluorescence microscopy, Annexin-V staining and TUNEL assay were used to detect alterations in cell cycle and apoptosis after 72 h of treatment. The TTFields/IN-3 combination decreased cell proliferation after 72 h compared to either treatment alone (-78.6% vs. TTFields, P = 0.0337; -52.6% vs. IN-3, P = 0.0205), and reduced the number of viable cells (62% less than seeded). There was a significant cell cycle shift from G1 to G2/M phase (P < 0.0001). The apoptotic rate increased to 44% (TTFields 14%, P = 0.0002; IN-3 4%, P < 0.0001). Cell growth recovered 24 h after EOT with TTFields and IN-3 alone, but the combination led to further decrease by 92% at 72 h EOT if IN-3 treatment was continued (P = 0.0288). The combination of TTFields and SAC inhibition led to earlier and prolonged effects that significantly augmented the efficacy of TTFields and highlights a potential new targeted multimodal treatment for GBM.
Collapse
Affiliation(s)
- Almuth F. Kessler
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Greta E. Frömbling
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Franziska Gross
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Mirja Hahn
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Wilfrid Dzokou
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Ralf-Ingo Ernestus
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| | - Carsten Hagemann
- Department of Neurosurgery, University of Würzburg, Tumorbiology Laboratory, Würzburg, Germany
| |
Collapse
|
35
|
Saurin AT. Kinase and Phosphatase Cross-Talk at the Kinetochore. Front Cell Dev Biol 2018; 6:62. [PMID: 29971233 PMCID: PMC6018199 DOI: 10.3389/fcell.2018.00062] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/31/2018] [Indexed: 01/26/2023] Open
Abstract
Multiple kinases and phosphatases act on the kinetochore to control chromosome segregation: Aurora B, Mps1, Bub1, Plk1, Cdk1, PP1, and PP2A-B56, have all been shown to regulate both kinetochore-microtubule attachments and the spindle assembly checkpoint. Given that so many kinases and phosphatases converge onto two key mitotic processes, it is perhaps not surprising to learn that they are, quite literally, entangled in cross-talk. Inhibition of any one of these enzymes produces secondary effects on all the others, which results in a complicated picture that is very difficult to interpret. This review aims to clarify this picture by first collating the direct effects of each enzyme into one overarching schematic of regulation at the Knl1/Mis12/Ndc80 (KMN) network (a major signaling hub at the outer kinetochore). This schematic will then be used to discuss the implications of the cross-talk that connects these enzymes; both in terms of why it may be needed to produce the right type of kinetochore signals and why it nevertheless complicates our interpretations about which enzymes control what processes. Finally, some general experimental approaches will be discussed that could help to characterize kinetochore signaling by dissociating the direct from indirect effect of kinase or phosphatase inhibition in vivo. Together, this review should provide a framework to help understand how a network of kinases and phosphatases cooperate to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Adrian T. Saurin
- Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
36
|
Gerhold AR, Poupart V, Labbé JC, Maddox PS. Spindle assembly checkpoint strength is linked to cell fate in the Caenorhabditis elegans embryo. Mol Biol Cell 2018; 29:1435-1448. [PMID: 29688794 PMCID: PMC6014101 DOI: 10.1091/mbc.e18-04-0215] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The spindle assembly checkpoint (SAC) is a conserved mitotic regulator that preserves genome stability by monitoring kinetochore-microtubule attachments and blocking anaphase onset until chromosome biorientation is achieved. Despite its central role in maintaining mitotic fidelity, the ability of the SAC to delay mitotic exit in the presence of kinetochore-microtubule attachment defects (SAC "strength") appears to vary widely. How different cellular aspects drive this variation remains largely unknown. Here we show that SAC strength is correlated with cell fate during development of Caenorhabditis elegans embryos, with germline-fated cells experiencing longer mitotic delays upon spindle perturbation than somatic cells. These differences are entirely dependent on an intact checkpoint and only partially attributable to differences in cell size. In two-cell embryos, cell size accounts for half of the difference in SAC strength between the larger somatic AB and the smaller germline P1 blastomeres. The remaining difference requires asymmetric cytoplasmic partitioning downstream of PAR polarity proteins, suggesting that checkpoint-regulating factors are distributed asymmetrically during early germ cell divisions. Our results indicate that SAC activity is linked to cell fate and reveal a hitherto unknown interaction between asymmetric cell division and the SAC.
Collapse
Affiliation(s)
- Abigail R Gerhold
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Vincent Poupart
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Jean-Claude Labbé
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada.,Department of Pathology and Cell Biology, Université de Montréal, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Paul S Maddox
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| |
Collapse
|
37
|
Meyer RE, Brown J, Beck L, Dawson DS. Mps1 promotes chromosome meiotic chromosome biorientation through Dam1. Mol Biol Cell 2017; 29:479-489. [PMID: 29237818 PMCID: PMC6014172 DOI: 10.1091/mbc.e17-08-0503] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 11/11/2022] Open
Abstract
During meiosis, chromosomes attach to microtubules at their kinetochores and are moved by microtubule depolymerization. The Mps1 kinase is essential for this process. Phosphorylation of Dam1 by Mps1 allows kinetochores to move processively poleward along microtubules during the biorientation process. In budding yeast meiosis, homologous chromosomes become linked by chiasmata and then move back and forth on the spindle until they are bioriented, with the kinetochores of the partners attached to microtubules from opposite spindle poles. Certain mutations in the conserved kinase, Mps1, result in catastrophic meiotic segregation errors but mild mitotic defects. We tested whether Dam1, a known substrate of Mps1, was necessary for its critical meiotic role. We found that kinetochore–microtubule attachments are established even when Dam1 is not phosphorylated by Mps1, but that Mps1 phosphorylation of Dam1 sustains those connections. But the meiotic defects when Dam1 is not phosphorylated are not nearly as catastrophic as when Mps1 is inactivated. The results demonstrate that one meiotic role of Mps1 is to stabilize connections that have been established between kinetochores and microtubles by phosphorylating Dam1.
Collapse
Affiliation(s)
- Régis E Meyer
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Jamin Brown
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Lindsay Beck
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Dean S Dawson
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 .,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
38
|
Abstract
At metaphase in mitotic cells, pulling forces at the kinetochore-microtubule interface create tension by stretching the centromeric chromatin between oppositely oriented sister kinetochores. This tension is important for stabilizing the end-on kinetochore microtubule attachment required for proper bi-orientation of sister chromosomes as well as for satisfaction of the Spindle Assembly Checkpoint and entry into anaphase. How force is coupled by proteins to kinetochore microtubules and resisted by centromere stretch is becoming better understood as many of the proteins involved have been identified. Recent application of genetically encoded fluorescent tension sensors within the mechanical linkage between the centromere and kinetochore microtubules are beginning to reveal - from live cell assays - protein specific contributions that are functionally important.
Collapse
Affiliation(s)
- Edward D Salmon
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kerry Bloom
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
39
|
Mechanisms of Chromosome Congression during Mitosis. BIOLOGY 2017; 6:biology6010013. [PMID: 28218637 PMCID: PMC5372006 DOI: 10.3390/biology6010013] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/07/2017] [Accepted: 01/28/2017] [Indexed: 12/13/2022]
Abstract
Chromosome congression during prometaphase culminates with the establishment of a metaphase plate, a hallmark of mitosis in metazoans. Classical views resulting from more than 100 years of research on this topic have attempted to explain chromosome congression based on the balance between opposing pulling and/or pushing forces that reach an equilibrium near the spindle equator. However, in mammalian cells, chromosome bi-orientation and force balance at kinetochores are not required for chromosome congression, whereas the mechanisms of chromosome congression are not necessarily involved in the maintenance of chromosome alignment after congression. Thus, chromosome congression and maintenance of alignment are determined by different principles. Moreover, it is now clear that not all chromosomes use the same mechanism for congressing to the spindle equator. Those chromosomes that are favorably positioned between both poles when the nuclear envelope breaks down use the so-called "direct congression" pathway in which chromosomes align after bi-orientation and the establishment of end-on kinetochore-microtubule attachments. This favors the balanced action of kinetochore pulling forces and polar ejection forces along chromosome arms that drive chromosome oscillatory movements during and after congression. The other pathway, which we call "peripheral congression", is independent of end-on kinetochore microtubule-attachments and relies on the dominant and coordinated action of the kinetochore motors Dynein and Centromere Protein E (CENP-E) that mediate the lateral transport of peripheral chromosomes along microtubules, first towards the poles and subsequently towards the equator. How the opposite polarities of kinetochore motors are regulated in space and time to drive congression of peripheral chromosomes only now starts to be understood. This appears to be regulated by position-dependent phosphorylation of both Dynein and CENP-E and by spindle microtubule diversity by means of tubulin post-translational modifications. This so-called "tubulin code" might work as a navigation system that selectively guides kinetochore motors with opposite polarities along specific spindle microtubule populations, ultimately leading to the congression of peripheral chromosomes. We propose an integrated model of chromosome congression in mammalian cells that depends essentially on the following parameters: (1) chromosome position relative to the spindle poles after nuclear envelope breakdown; (2) establishment of stable end-on kinetochore-microtubule attachments and bi-orientation; (3) coordination between kinetochore- and arm-associated motors; and (4) spatial signatures associated with post-translational modifications of specific spindle microtubule populations. The physiological consequences of abnormal chromosome congression, as well as the therapeutic potential of inhibiting chromosome congression are also discussed.
Collapse
|
40
|
Asbury CL. Anaphase A: Disassembling Microtubules Move Chromosomes toward Spindle Poles. BIOLOGY 2017; 6:E15. [PMID: 28218660 PMCID: PMC5372008 DOI: 10.3390/biology6010015] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/04/2017] [Accepted: 02/10/2017] [Indexed: 11/16/2022]
Abstract
The separation of sister chromatids during anaphase is the culmination of mitosis and one of the most strikingly beautiful examples of cellular movement. It consists of two distinct processes: Anaphase A, the movement of chromosomes toward spindle poles via shortening of the connecting fibers, and anaphase B, separation of the two poles from one another via spindle elongation. I focus here on anaphase A chromosome-to-pole movement. The chapter begins by summarizing classical observations of chromosome movements, which support the current understanding of anaphase mechanisms. Live cell fluorescence microscopy studies showed that poleward chromosome movement is associated with disassembly of the kinetochore-attached microtubule fibers that link chromosomes to poles. Microtubule-marking techniques established that kinetochore-fiber disassembly often occurs through loss of tubulin subunits from the kinetochore-attached plus ends. In addition, kinetochore-fiber disassembly in many cells occurs partly through 'flux', where the microtubules flow continuously toward the poles and tubulin subunits are lost from minus ends. Molecular mechanistic models for how load-bearing attachments are maintained to disassembling microtubule ends, and how the forces are generated to drive these disassembly-coupled movements, are discussed.
Collapse
Affiliation(s)
- Charles L Asbury
- Department of Physiology & Biophysics, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
41
|
Musacchio A, Desai A. A Molecular View of Kinetochore Assembly and Function. BIOLOGY 2017; 6:E5. [PMID: 28125021 PMCID: PMC5371998 DOI: 10.3390/biology6010005] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/15/2022]
Abstract
Kinetochores are large protein assemblies that connect chromosomes to microtubules of the mitotic and meiotic spindles in order to distribute the replicated genome from a mother cell to its daughters. Kinetochores also control feedback mechanisms responsible for the correction of incorrect microtubule attachments, and for the coordination of chromosome attachment with cell cycle progression. Finally, kinetochores contribute to their own preservation, across generations, at the specific chromosomal loci devoted to host them, the centromeres. They achieve this in most species by exploiting an epigenetic, DNA-sequence-independent mechanism; notable exceptions are budding yeasts where a specific sequence is associated with centromere function. In the last 15 years, extensive progress in the elucidation of the composition of the kinetochore and the identification of various physical and functional modules within its substructure has led to a much deeper molecular understanding of kinetochore organization and the origins of its functional output. Here, we provide a broad summary of this progress, focusing primarily on kinetochores of humans and budding yeast, while highlighting work from other models, and present important unresolved questions for future studies.
Collapse
Affiliation(s)
- Andrea Musacchio
- Department of Mechanistic Cell Biology, Max Planck Institute of Molecular Physiology, Otto-Hahn Straße 11, Dortmund 44227, Germany.
- Centre for Medical Biotechnology, Faculty of Biology, University Duisburg-Essen, Essen 45117, Germany.
| | - Arshad Desai
- Ludwig Institute for Cancer Research, La Jolla, CA 92093, USA.
- Department of Cellular & Molecular Medicine, 9500 Gilman Dr., La Jolla, CA 92093, USA.
| |
Collapse
|