1
|
Pluta AJ, Studniarek C, Murphy S, Norbury CJ. Cyclin-dependent kinases: Masters of the eukaryotic universe. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 15:e1816. [PMID: 37718413 PMCID: PMC10909489 DOI: 10.1002/wrna.1816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/19/2023]
Abstract
A family of structurally related cyclin-dependent protein kinases (CDKs) drives many aspects of eukaryotic cell function. Much of the literature in this area has considered individual members of this family to act primarily either as regulators of the cell cycle, the context in which CDKs were first discovered, or as regulators of transcription. Until recently, CDK7 was the only clear example of a CDK that functions in both processes. However, new data points to several "cell-cycle" CDKs having important roles in transcription and some "transcriptional" CDKs having cell cycle-related targets. For example, novel functions in transcription have been demonstrated for the archetypal cell cycle regulator CDK1. The increasing evidence of the overlap between these two CDK types suggests that they might play a critical role in coordinating the two processes. Here we review the canonical functions of cell-cycle and transcriptional CDKs, and provide an update on how these kinases collaborate to perform important cellular functions. We also provide a brief overview of how dysregulation of CDKs contributes to carcinogenesis, and possible treatment avenues. This article is categorized under: RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes RNA Processing > 3' End Processing RNA Processing > Splicing Regulation/Alternative Splicing.
Collapse
Affiliation(s)
| | | | - Shona Murphy
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Chris J. Norbury
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| |
Collapse
|
2
|
Kraus J, Travis SM, King MR, Petry S. Augmin is a Ran-regulated spindle assembly factor. J Biol Chem 2023; 299:104736. [PMID: 37086784 DOI: 10.1016/j.jbc.2023.104736] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/24/2023] Open
Abstract
Mitotic spindles are composed of microtubules (MTs) that must nucleate at the right place and time. Ran regulates this process by directly controlling the release of spindle assembly factors (SAFs) from nucleocytoplasmic shuttle proteins importin-αβ and subsequently forms a biochemical gradient of SAFs localized around chromosomes. The majority of spindle MTs are generated by branching MT nucleation, which has been shown to require an eight-subunit protein complex known as augmin. InXenopus laevis, Ran can control branching through a canonical SAF, TPX2, which is non-essential in Drosophila melanogaster embryos and HeLa cells. Thus, how Ran regulates branching MT nucleation when TPX2 is not required remains unknown. Here, we use in vitro pulldowns and TIRF microscopy to show that augmin is a Ran-regulated SAF. We demonstrate that augmin directly interacts with both importin-α and importin-β through two nuclear localization sequences on the Haus8 subunit, which overlap with the MT binding site. Moreover, we show Ran controls localization of augmin to MTs in both Xenopus egg extract and in vitro. Our results demonstrate that RanGTP directly regulates augmin, which establishes a new way by which Ran controls branching MT nucleation and spindle assembly both in the absence and presence of TPX2.
Collapse
Affiliation(s)
- Jodi Kraus
- Department of Molecular Biology; Princeton University; Princeton, NJ, 08544; USA
| | - Sophie M Travis
- Department of Molecular Biology; Princeton University; Princeton, NJ, 08544; USA
| | - Matthew R King
- Department of Molecular Biology; Princeton University; Princeton, NJ, 08544; USA
| | - Sabine Petry
- Department of Molecular Biology; Princeton University; Princeton, NJ, 08544; USA.
| |
Collapse
|
3
|
KYOGOKU H, KITAJIMA TS. The large cytoplasmic volume of oocyte. J Reprod Dev 2023; 69:1-9. [PMID: 36436912 PMCID: PMC9939283 DOI: 10.1262/jrd.2022-101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The study of the size of cells and organelles has a long history, dating back to the 1600s when cells were defined. In particular, various methods have elucidated the size of the nucleus and the mitotic spindle in several species. However, little research has been conducted on oocyte size and organelles in mammals, and many questions remain to be answered. The appropriate size is essential to cell function properly. Oocytes have a very large cytoplasm, which is more than 100 times larger than that of general somatic cells in mammals. In this review, we discuss how oocytes acquire an enormous cytoplasmic size and the adverse effects of a large cytoplasmic size on cellular functions.
Collapse
Affiliation(s)
- Hirohisa KYOGOKU
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan,Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Tomoya S KITAJIMA
- Laboratory for Chromosome Segregation, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| |
Collapse
|
4
|
Loyer P, Trembley JH. Roles of CDK/Cyclin complexes in transcription and pre-mRNA splicing: Cyclins L and CDK11 at the cross-roads of cell cycle and regulation of gene expression. Semin Cell Dev Biol 2020; 107:36-45. [PMID: 32446654 DOI: 10.1016/j.semcdb.2020.04.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/18/2022]
Abstract
Cyclin Dependent Kinases (CDKs) represent a large family of serine/threonine protein kinases that become active upon binding to a Cyclin regulatory partner. CDK/cyclin complexes recently identified, as well as "canonical" CDK/Cyclin complexes regulating cell cycle, are implicated in the regulation of gene expression via the phosphorylation of key components of the transcription and pre-mRNA processing machineries. In this review, we summarize the role of CDK/cyclin-dependent phosphorylation in the regulation of transcription and RNA splicing and highlight recent findings that indicate the involvement of CDK11/cyclin L complexes at the cross-roads of cell cycle, transcription and RNA splicing. Finally, we discuss the potential of CDK11 and Cyclins L as therapeutic targets in cancer.
Collapse
Affiliation(s)
- Pascal Loyer
- INSERM, INRAE, Univ Rennes, NuMeCan, Nutrition Metabolisms and Cancer, Rennes, France.
| | - Janeen H Trembley
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN 55417, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
5
|
Ou D, Chen L, He J, Rong Z, Gao J, Li Z, Liu L, Tang F, Li J, Deng Y, Sun L. CDK11 negatively regulates Wnt/β-catenin signaling in the endosomal compartment by affecting microtubule stability. Cancer Biol Med 2020; 17:328-342. [PMID: 32587772 PMCID: PMC7309457 DOI: 10.20892/j.issn.2095-3941.2019.0229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/18/2019] [Indexed: 12/13/2022] Open
Abstract
Objectives: Improper activation of Wnt/β-catenin signaling has been implicated in human diseases. Beyond the well-studied glycogen synthase kinase 3β (GSK3β) and casein kinase 1 (CK1), other kinases affecting Wnt/β-catenin signaling remain to be defined. Methods:To identify the kinases that modulate Wnt/β-catenin signaling, we applied a kinase small interfering RNA (siRNA) library screen approach. Luciferase assays, immunoblotting, and real-time polymerase chain reaction (PCR) were performed to confirm the regulation of the Wnt/β-catenin signaling pathway by cyclin-dependent kinase 11 (CDK11) and to investigate the underlying mechanism. Confocal immunofluorescence, coimmunoprecipitation (co-IP), and scratch wound assays were used to demonstrate colocalization, detect protein interactions, and explore the function of CDK11. Results: CDK11 was found to be a significant candidate kinase participating in the negative control of Wnt/β-catenin signaling. Down-regulation of CDK11 led to the accumulation of Wnt/β-catenin signaling receptor complexes, in a manner dependent on intact adenomatosis polyposis coli (APC) protein. Further analysis showed that CDK11 modulation of Wnt/β-catenin signaling engaged the endolysosomal machinery, and CDK11 knockdown enhanced the colocalization of Wnt/β-catenin signaling receptor complexes with early endosomes and decreased colocalization with lysosomes. Mechanistically, CDK11 was found to function in Wnt/β-catenin signaling by regulating microtubule stability. Depletion of CDK11 down-regulated acetyl-α-tubulin. Moreover, co-IP assays demonstrated that CDK11 interacts with the α-tubulin deacetylase SIRT2, whereas SIRT2 down-regulation in CDK11-depleted cells reversed the accumulation of Wnt/β-catenin signaling receptor complexes. CDK11 was found to suppress cell migration through altered Wnt/β-catenin signaling. Conclusions: CDK11 is a negative modulator of Wnt/β-catenin signaling that stabilizes microtubules, thus resulting in the dysregulation of receptor complex trafficking from early endosomes to lysosomes.
Collapse
Affiliation(s)
- Danmin Ou
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lin Chen
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiang He
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhuoxian Rong
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Gao
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhi Li
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,International Cooperation Base of Cancer Precision Therapy, Department of Science and Technology of Hunan Province, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
| | - Liyu Liu
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Feiyu Tang
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiang Li
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuezhen Deng
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,International Cooperation Base of Cancer Precision Therapy, Department of Science and Technology of Hunan Province, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China
| | - Lunquan Sun
- Department of Oncology, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.,International Cooperation Base of Cancer Precision Therapy, Department of Science and Technology of Hunan Province, Changsha 410008, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha 410008, China.,National Clinical Research Center for Geriatric Disorders, Changsha 410008, China
| |
Collapse
|
6
|
Martínez-Alonso D, Malumbres M. Mammalian cell cycle cyclins. Semin Cell Dev Biol 2020; 107:28-35. [PMID: 32334991 DOI: 10.1016/j.semcdb.2020.03.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/23/2022]
Abstract
Proper progression throughout the cell division cycle depends on the expression level of a family of proteins known as cyclins, and the subsequent activation of cyclin-dependent kinases (Cdks). Among the numerous members of the mammalian cyclin family, only a few of them, cyclins A, B, C, D and E, are known to display critical roles in the cell cycle. These functions will be reviewed here with a special focus on their relevance in different cell types in vivo and their implications in human disease.
Collapse
Affiliation(s)
- Diego Martínez-Alonso
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO) Madrid, Spain.
| | - Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO) Madrid, Spain.
| |
Collapse
|
7
|
Cyclin-dependent kinase inhibition: an opportunity to target protein-protein interactions. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 121:115-141. [PMID: 32312419 DOI: 10.1016/bs.apcsb.2019.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Cyclin-dependent kinases (CDKs) play an integral part in cellular activities. To date, most of the activities have been evaluated in the cell cycle and transcription. Several diseases are affected by abnormalities in CDKs, related-pathways, or proteins that regulate CDK activity. CDKs are primarily dependent on activation by binding other proteins, namely Cyclins. In addition, phosphorylation of key CDK residues also plays a major part in CDK activity. To date, the most successful drugs have been developed against CDK4 and CDK6 and are FDA approved for use in advanced breast cancer. However, this is likely only a small fraction of the potential for targeting CDKs as a strategy against cancer and other diseases. Based on the extensive protein-protein interactions made by CDKs with other proteins (Cyclins and others), there are numerous possibilities for targeting strategies against protein-protein interactions. Here we describe the predominant roles of CDKs in the cell, key interacting proteins, significant 3-dimensional structural characteristics, and summarize the work-to-date in inhibition of CDKs.
Collapse
|
8
|
Renshaw MJ, Panagiotou TC, Lavoie BD, Wilde A. CDK11 p58-cyclin L1β regulates abscission site assembly. J Biol Chem 2019; 294:18639-18649. [PMID: 31653703 DOI: 10.1074/jbc.ra119.009107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 10/10/2019] [Indexed: 01/22/2023] Open
Abstract
Rigorous spatiotemporal regulation of cell division is required to maintain genome stability. The final stage in cell division, when the cells physically separate (abscission), is tightly regulated to ensure that it occurs after cytokinetic events such as chromosome segregation. A key regulator of abscission timing is Aurora B kinase activity, which inhibits abscission and forms the major activity of the abscission checkpoint. This checkpoint prevents abscission until chromosomes have been cleared from the cytokinetic machinery. Here we demonstrate that the mitosis-specific CDK11p58 kinase specifically forms a complex with cyclin L1β that, in late cytokinesis, localizes to the stem body, a structure in the middle of the intercellular bridge that forms between two dividing cells. Depletion of CDK11 inhibits abscission, and rescue of this phenotype requires CDK11p58 kinase activity or inhibition of Aurora B kinase activity. Furthermore, CDK11p58 kinase activity is required for formation of endosomal sorting complex required for transport III filaments at the site of abscission. Combined, these data suggest that CDK11p58 kinase activity opposes Aurora B activity to enable abscission to proceed and result in successful completion of cytokinesis.
Collapse
Affiliation(s)
- Matthew J Renshaw
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Thomas C Panagiotou
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Brigitte D Lavoie
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Andrew Wilde
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada.
| |
Collapse
|
9
|
Abstract
A complex network precisely regulates the cell cycle through the G1, S, G2, and M phases and is the basis for cell division under physiological and pathological conditions. On the one hand, the transition from one phase to another as well as the progression within each phase is driven by the specific cyclin-dependent kinases (CDKs; e.g., CDK1, CDK2, CDK4, CDK6, and CDK7), together with their exclusive partner cyclins (e.g., cyclin A1, B1, D1–3, and E1). On the other hand, these phases are negatively regulated by endogenous CDK inhibitors such as p16ink4a, p18ink4c, p19ink4d, p21cip1, and p27kip1. In addition, several checkpoints control the commitment of cells to replicate DNA and undergo mitosis, thereby avoiding the passage of genomic errors to daughter cells. CDKs are often constitutively activated in cancer, which is characterized by the uncontrolled proliferation of transformed cells, due to genetic and epigenetic abnormalities in the genes involved in the cell cycle. Moreover, several oncogenes and defective tumor suppressors promote malignant changes by stimulating cell cycle entry and progression or disrupting DNA damage responses, including the cell cycle checkpoints, DNA repair mechanisms, and apoptosis. Thus, genes or proteins related to cell cycle regulation remain the main targets of interest in the treatment of various cancer types, including hematologic malignancies. In this context, advances in the understanding of the cell cycle regulatory machinery provide a basis for the development of novel therapeutic approaches. The present article summarizes the pathways as well as their genetic and epigenetic alterations that regulate the cell cycle; moreover, it discusses the various approved or potential therapeutic targets associated with the cell cycle, focusing on hematologic malignancies.
Collapse
|
10
|
CDK11 Loss Induces Cell Cycle Dysfunction and Death of BRAF and NRAS Melanoma Cells. Pharmaceuticals (Basel) 2019; 12:ph12020050. [PMID: 30987032 PMCID: PMC6631185 DOI: 10.3390/ph12020050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/24/2019] [Accepted: 03/24/2019] [Indexed: 12/25/2022] Open
Abstract
Cyclin dependent kinase 11 (CDK11) is a protein kinase that regulates RNA transcription, pre-mRNA splicing, mitosis, and cell death. Targeting of CDK11 expression levels is effective in the experimental treatment of breast and other cancers, but these data are lacking in melanoma. To understand CDK11 function in melanoma, we evaluated protein and RNA levels of CDK11, Cyclin L1 and Cyclin L2 in benign melanocytes and BRAF- as well as NRAS-mutant melanoma cell lines. We investigated the effectiveness of reducing expression of this survival kinase using RNA interference on viability, clonal survival, and tumorsphere formation in melanoma cell lines. We examined the impact of CDK11 loss in BRAF-mutant melanoma on more than 700 genes important in cancer signaling pathways. Follow-up analysis evaluated how CDK11 loss alters cell cycle function in BRAF- and NRAS-mutant melanoma cells. We present data on CDK11, CCNL1 and CCNL2 mRNA expression in melanoma patients, including prognosis for survival. In sum, we found that CDK11 is necessary for melanoma cell survival, and a major impact of CDK11 loss in melanoma is to cause disruption of the cell cycle distribution with accumulation of G1- and loss of G2/M-phase cancer cells.
Collapse
|
11
|
Colin A, Singaravelu P, Théry M, Blanchoin L, Gueroui Z. Actin-Network Architecture Regulates Microtubule Dynamics. Curr Biol 2018; 28:2647-2656.e4. [PMID: 30100343 DOI: 10.1016/j.cub.2018.06.028] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 03/13/2018] [Accepted: 06/14/2018] [Indexed: 11/25/2022]
Abstract
Coordination between actin filaments and microtubules is critical to complete important steps during cell division. For instance, cytoplasmic actin filament dynamics play an active role in the off-center positioning of the spindle during metaphase I in mouse oocytes [1-3] or in gathering the chromosomes to ensure proper spindle formation in starfish oocytes [4, 5], whereas cortical actin filaments control spindle rotation and positioning in adherent cells or in mouse oocytes [6-9]. Several molecular effectors have been found to facilitate anchoring between the meiotic spindle and the cortical actin [10-14]. In vitro reconstitutions have provided detailed insights in the biochemical and physical interactions between microtubules and actin filaments [15-20]. Yet how actin meshwork architecture affects microtubule dynamics is still unclear. Here, we reconstituted microtubule aster in the presence of a meshwork of actin filaments using confined actin-intact Xenopus egg extracts. We found that actin filament branching reduces the lengths and growth rates of microtubules and constrains the mobility of microtubule asters. By reconstituting the interaction between dynamic actin filaments and microtubules in a minimal system based on purified proteins, we found that the branching of actin filaments is sufficient to block microtubule growth and trigger microtubule disassembly. In a further exploration of Xenopus egg extracts, we found that dense and static branched actin meshwork perturbs monopolar spindle assembly by constraining the motion of the spindle pole. Interestingly, monopolar spindle assembly was not constrained in conditions supporting dynamic meshwork rearrangements. We propose that branched actin filament meshwork provides physical barriers that limit microtubule growth.
Collapse
Affiliation(s)
- Alexandra Colin
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Pavithra Singaravelu
- Université Grenoble-Alpes, CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, CytoMorpho Lab, 17 rue des Martyrs, 38054 Grenoble, France
| | - Manuel Théry
- Université Grenoble-Alpes, CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, CytoMorpho Lab, 17 rue des Martyrs, 38054 Grenoble, France; Université Paris Diderot, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, 1 Avenue Claude Vellefaux, 75010 Paris, France
| | - Laurent Blanchoin
- Université Grenoble-Alpes, CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire & Végétale, CytoMorpho Lab, 17 rue des Martyrs, 38054 Grenoble, France; Université Paris Diderot, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, 1 Avenue Claude Vellefaux, 75010 Paris, France.
| | - Zoher Gueroui
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France.
| |
Collapse
|
12
|
Animal Female Meiosis: The Challenges of Eliminating Centrosomes. Cells 2018; 7:cells7070073. [PMID: 29996518 PMCID: PMC6071224 DOI: 10.3390/cells7070073] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/03/2018] [Accepted: 07/03/2018] [Indexed: 01/02/2023] Open
Abstract
Sexual reproduction requires the generation of gametes, which are highly specialised for fertilisation. Female reproductive cells, oocytes, grow up to large sizes when they accumulate energy stocks and store proteins as well as mRNAs to enable rapid cell divisions after fertilisation. At the same time, metazoan oocytes eliminate their centrosomes, i.e., major microtubule-organizing centres (MTOCs), during or right after the long growth phases. Centrosome elimination poses two key questions: first, how can the centrosome be re-established after fertilisation? In general, metazoan oocytes exploit sperm components, i.e., the basal body of the sperm flagellum, as a platform to reinitiate centrosome production. Second, how do most metazoan oocytes manage to build up meiotic spindles without centrosomes? Oocytes have evolved mechanisms to assemble bipolar spindles solely around their chromosomes without the guidance of pre-formed MTOCs. Female animal meiosis involves microtubule nucleation and organisation into bipolar microtubule arrays in regulated self-assembly under the control of the Ran system and nuclear transport receptors. This review summarises our current understanding of the molecular mechanism underlying self-assembly of meiotic spindles, its spatio-temporal regulation, and the key players governing this process in animal oocytes.
Collapse
|
13
|
Williams CW, Iyer J, Liu Y, O'Connell KF. CDK-11-Cyclin L is required for gametogenesis and fertility in C. elegans. Dev Biol 2018; 441:52-66. [PMID: 29886128 DOI: 10.1016/j.ydbio.2018.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/05/2018] [Accepted: 06/05/2018] [Indexed: 11/18/2022]
Abstract
CDK11, a member of the cyclin-dependent kinase family, has been implicated in a diverse array of functions including transcription, RNA processing, sister chromatid cohesion, spindle assembly, centriole duplication and apoptosis. Despite its involvement in many essential functions, little is known about the requirements for CDK11 and its partner Cyclin L in a developing multicellular organism. Here we investigate the function of CDK11 and Cyclin L during development of the nematode Caenorhabditis elegans. Worms express two CDK11 proteins encoded by distinct loci: CDK-11.1 is essential for normal male and female fertility and is broadly expressed in the nuclei of somatic and germ line cells, while CDK-11.2 is nonessential and is enriched in hermaphrodite germ line nuclei beginning in mid pachytene. Hermaphrodites lacking CDK-11.1 develop normally but possess fewer mature sperm and oocytes and do not fully activate the RAS-ERK pathway that is required for oocyte production in response to environmental cues. Most of the sperm and eggs that are produced in cdk-11.1 null animals appear to complete development normally but fail to engage in sperm-oocyte signaling suggesting that CDK-11.1 is needed at multiple points in gametogenesis. Finally, we find that CDK-11.1 and CDK-11.2 function redundantly during embryonic and postembryonic development and likely do so in association with Cyclin L. Our results thus define multiple requirements for CDK-11-Cyclin L during animal development.
Collapse
Affiliation(s)
- Christopher W Williams
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA
| | - Jyoti Iyer
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA
| | - Yan Liu
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA
| | - Kevin F O'Connell
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA.
| |
Collapse
|
14
|
Yaguchi K, Yamamoto T, Matsui R, Tsukada Y, Shibanuma A, Kamimura K, Koda T, Uehara R. Uncoordinated centrosome cycle underlies the instability of non-diploid somatic cells in mammals. J Cell Biol 2018; 217:2463-2483. [PMID: 29712735 PMCID: PMC6028549 DOI: 10.1083/jcb.201701151] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 09/27/2017] [Accepted: 04/09/2018] [Indexed: 12/22/2022] Open
Abstract
Mammalian somatic cells are more stable as diploids, but the mechanisms underlying this stability are unclear. Yaguchi et al. show that changes in centriole licensing compromise the control of centrosome number in haploid or tetraploid human cells, suggesting that the ploidy-dependent control of the centrosome cycle explains the instability of non-diploid karyotypes. In animals, somatic cells are usually diploid and are unstable when haploid for unknown reasons. In this study, by comparing isogenic human cell lines with different ploidies, we found frequent centrosome loss specifically in the haploid state, which profoundly contributed to haploid instability through subsequent mitotic defects. We also found that the efficiency of centriole licensing and duplication changes proportionally to ploidy level, whereas that of DNA replication stays constant. This caused gradual loss or frequent overduplication of centrioles in haploid and tetraploid cells, respectively. Centriole licensing efficiency seemed to be modulated by astral microtubules, whose development scaled with ploidy level, and artificial enhancement of aster formation in haploid cells restored centriole licensing efficiency to diploid levels. The ploidy–centrosome link was observed in different mammalian cell types. We propose that incompatibility between the centrosome duplication and DNA replication cycles arising from different scaling properties of these bioprocesses upon ploidy changes underlies the instability of non-diploid somatic cells in mammals.
Collapse
Affiliation(s)
- Kan Yaguchi
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Takahiro Yamamoto
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Ryo Matsui
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Yuki Tsukada
- Division of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Atsuko Shibanuma
- Creative Research Institution, Hokkaido University, Sapporo, Japan
| | - Keiko Kamimura
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Toshiaki Koda
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan.,Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| | - Ryota Uehara
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan .,Creative Research Institution, Hokkaido University, Sapporo, Japan.,Faculty of Advanced Life Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
15
|
Abstract
Over the past two decades there has been a great deal of interest in the development of inhibitors of the cyclin-dependent kinases (CDKs). This attention initially stemmed from observations that different CDK isoforms have key roles in cancer cell proliferation through loss of regulation of the cell cycle, a hallmark feature of cancer. CDKs have now been shown to regulate other processes, particularly various aspects of transcription. The early non-selective CDK inhibitors exhibited considerable toxicity and proved to be insufficiently active in most cancers. The lack of patient selection biomarkers and an absence of understanding of the inhibitory profile required for efficacy hampered the development of these inhibitors. However, the advent of potent isoform-selective inhibitors with accompanying biomarkers has re-ignited interest. Palbociclib, a selective CDK4/6 inhibitor, is now approved for the treatment of ER+/HER2- advanced breast cancer. Current developments in the field include the identification of potent and selective inhibitors of the transcriptional CDKs; these include tool compounds that have allowed exploration of individual CDKs as cancer targets and the determination of their potential therapeutic windows. Biomarkers that allow the selection of patients likely to respond are now being discovered. Drug resistance has emerged as a major hurdle in the clinic for most protein kinase inhibitors and resistance mechanism are beginning to be identified for CDK inhibitors. This suggests that the selective inhibitors may be best used combined with standard of care or other molecularly targeted agents now in development rather than in isolation as monotherapies.
Collapse
Affiliation(s)
- Steven R Whittaker
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Aurélie Mallinger
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul Workman
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul A Clarke
- Division of Cancer Therapeutics, The Institute of Cancer Research, London SW7 3RP, United Kingdom; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SW7 3RP, United Kingdom.
| |
Collapse
|
16
|
Liu TH, Wu YF, Dong XL, Pan CX, Du GY, Yang JG, Wang W, Bao XY, Chen P, Pan MH, Lu C. Identification and characterization of the BmCyclin L1-BmCDK11A/B complex in relation to cell cycle regulation. Cell Cycle 2017; 16:861-868. [PMID: 28318374 DOI: 10.1080/15384101.2017.1304339] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cyclin proteins are the key regulatory and activity partner of cyclin-dependent kinases (CDKs), which play pivotal regulatory roles in cell cycle progression. In the present study, we identified a Cyclin L1 and 2 CDK11 2 CDK11 splice variants, CDK11A and CDK11B, from silkworm, Bombyx mori. We determined that both Cyclin L1 and CDK11A/B are nuclear proteins, and further investigations were conducted to elucidate their spatiofunctional features. Cyclin L1 forms a complex with CDK11A/B and were co-localized to the nucleus. Moreover, the dimerization of CDK11A and CDK11B and the effects of Cyclin L1 and CDK11A/B on cell cycle regulation were also investigated. Using overexpression or RNA interference experiments, we demonstrated that the abnormal expression of Cyclin L1 and CDK11A/B leads to cell cycle arrest and cell proliferation suppression. Together, these findings indicate that CDK11A/B interacts with Cyclin L1 to regulate the cell cycle.
Collapse
Affiliation(s)
- Tai-Hang Liu
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Yun-Fei Wu
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Xiao-Long Dong
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China.,b College of Animal Science and Technology , Northwest A&F University , Yangling , Shaanxi , China
| | - Cai-Xia Pan
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Guo-Yu Du
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Ji-Gui Yang
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Wei Wang
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Xi-Yan Bao
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Peng Chen
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China
| | - Min-Hui Pan
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China.,c Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry , Southwest University , Chongqing , China
| | - Cheng Lu
- a State Key Laboratory of Silkworm Genome Biology , Southwest University , Chongqing , China.,c Key Laboratory for Sericulture Functional Genomics and Biotechnology of Agricultural Ministry , Southwest University , Chongqing , China
| |
Collapse
|
17
|
Kapoor TM. Metaphase Spindle Assembly. BIOLOGY 2017; 6:biology6010008. [PMID: 28165376 PMCID: PMC5372001 DOI: 10.3390/biology6010008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 01/31/2023]
Abstract
A microtubule-based bipolar spindle is required for error-free chromosome segregation during cell division. In this review I discuss the molecular mechanisms required for the assembly of this dynamic micrometer-scale structure in animal cells.
Collapse
Affiliation(s)
- Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, the Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
18
|
Cai Z, Liu Q. Cell Cycle Regulation in Treatment of Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:251-270. [PMID: 29282688 DOI: 10.1007/978-981-10-6020-5_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cell cycle progression and cell proliferation are under precise and orchestrated control in normal cells. However, uncontrolled cell proliferation caused by aberrant cell cycle progression is a crucial characteristic of cancer. Understanding cell cycle progression and its regulation sheds light on cancer treatment. Agents targeting cell cycle regulators (such as CDKs) have been considered as promising candidates in cancer treatment. Although the first-generation pan-CDK inhibitors failed in clinical trials because of their adverse events and low efficacy, new selective CDK 4/6 inhibitors showed potent efficacy with tolerable safety in preclinical and clinical studies. Here we will review the mechanisms of cell cycle regulation and targeting key cell cycle regulators (such as CDKs) in breast cancer treatment. Particularly, we will discuss the mechanism of CDK inhibitors in disrupting cell cycle progression, the use of selective CDK4/6 inhibitors in treatment of advanced, hormone receptor (HR)-positive postmenopausal breast cancer patients, and other clinical trials that aim to extend the utilization of these agents.
Collapse
Affiliation(s)
- Zijie Cai
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China
| | - Qiang Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
19
|
Zhou Y, Shen JK, Hornicek FJ, Kan Q, Duan Z. The emerging roles and therapeutic potential of cyclin-dependent kinase 11 (CDK11) in human cancer. Oncotarget 2016; 7:40846-40859. [PMID: 27049727 PMCID: PMC5130049 DOI: 10.18632/oncotarget.8519] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/28/2016] [Indexed: 12/17/2022] Open
Abstract
Overexpression and/or hyperactivation of cyclin-dependent kinases (CDKs) are common features of most cancer types. CDKs have been shown to play important roles in tumor cell proliferation and growth by controlling cell cycle, transcription, and RNA splicing. CDK4/6 inhibitor palbociclib has been recently approved by the FDA for the treatment of breast cancer. CDK11 is a serine/threonine protein kinase in the CDK family and recent studies have shown that CDK11 also plays critical roles in cancer cell growth and proliferation. A variety of genetic and epigenetic events may cause universal overexpression of CDK11 in human cancers. Inhibition of CDK11 has been shown to lead to cancer cell death and apoptosis. Significant evidence has suggested that CDK11 may be a novel and promising therapeutic target for the treatment of cancers. This review will focus on the emerging roles of CDK11 in human cancers, and provide a proof-of-principle for continued efforts toward targeting CDK11 for effective cancer treatment.
Collapse
Affiliation(s)
- Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jacson K. Shen
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Francis J. Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhenfeng Duan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital, Boston, MA, United States of America
| |
Collapse
|
20
|
Cavazza T, Vernos I. The RanGTP Pathway: From Nucleo-Cytoplasmic Transport to Spindle Assembly and Beyond. Front Cell Dev Biol 2016; 3:82. [PMID: 26793706 PMCID: PMC4707252 DOI: 10.3389/fcell.2015.00082] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/07/2015] [Indexed: 01/03/2023] Open
Abstract
The small GTPase Ran regulates the interaction of transport receptors with a number of cellular cargo proteins. The high affinity binding of the GTP-bound form of Ran to import receptors promotes cargo release, whereas its binding to export receptors stabilizes their interaction with the cargo. This basic mechanism linked to the asymmetric distribution of the two nucleotide-bound forms of Ran between the nucleus and the cytoplasm generates a switch like mechanism controlling nucleo-cytoplasmic transport. Since 1999, we have known that after nuclear envelope breakdown (NEBD) Ran and the above transport receptors also provide a local control over the activity of factors driving spindle assembly and regulating other aspects of cell division. The identification and functional characterization of RanGTP mitotic targets is providing novel insights into mechanisms essential for cell division. Here we review our current knowledge on the RanGTP system and its regulation and we focus on the recent advances made through the characterization of its mitotic targets. We then briefly review the novel functions of the pathway that were recently described. Altogether, the RanGTP system has moonlighting functions exerting a spatial control over protein interactions that drive specific functions depending on the cellular context.
Collapse
Affiliation(s)
- Tommaso Cavazza
- Cell and Developmental Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain
| | - Isabelle Vernos
- Cell and Developmental Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain; Institució Catalana de Recerca I Estudis AvançatsBarcelona, Spain
| |
Collapse
|
21
|
Chen JWC, Barker AR, Wakefield JG. The Ran Pathway in Drosophila melanogaster Mitosis. Front Cell Dev Biol 2015; 3:74. [PMID: 26636083 PMCID: PMC4659922 DOI: 10.3389/fcell.2015.00074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/09/2015] [Indexed: 11/29/2022] Open
Abstract
Over the last two decades, the small GTPase Ran has emerged as a central regulator of both mitosis and meiosis, particularly in the generation, maintenance, and regulation of the microtubule (MT)-based bipolar spindle. Ran-regulated pathways in mitosis bear many similarities to the well-characterized functions of Ran in nuclear transport and, as with transport, the majority of these mitotic effects are mediated through affecting the physical interaction between karyopherins and Spindle Assembly Factors (SAFs)—a loose term describing proteins or protein complexes involved in spindle assembly through promoting nucleation, stabilization, and/or depolymerization of MTs, through anchoring MTs to specific structures such as centrosomes, chromatin or kinetochores, or through sliding MTs along each other to generate the force required to achieve bipolarity. As such, the Ran-mediated pathway represents a crucial functional module within the wider spindle assembly landscape. Research into mitosis using the model organism Drosophila melanogaster has contributed substantially to our understanding of centrosome and spindle function. However, in comparison to mammalian systems, very little is known about the contribution of Ran-mediated pathways in Drosophila mitosis. This article sets out to summarize our understanding of the roles of the Ran pathway components in Drosophila mitosis, focusing on the syncytial blastoderm embryo, arguing that it can provide important insights into the conserved functions on Ran during spindle formation.
Collapse
Affiliation(s)
- Jack W C Chen
- Biosciences, College of Life and Environmental Sciences, University of Exeter Exeter, UK
| | - Amy R Barker
- Biosciences, College of Life and Environmental Sciences, University of Exeter Exeter, UK ; Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London London, UK
| | - James G Wakefield
- Biosciences, College of Life and Environmental Sciences, University of Exeter Exeter, UK
| |
Collapse
|
22
|
Yokoyama H, Koch B, Walczak R, Ciray-Duygu F, González-Sánchez JC, Devos DP, Mattaj IW, Gruss OJ. The nucleoporin MEL-28 promotes RanGTP-dependent γ-tubulin recruitment and microtubule nucleation in mitotic spindle formation. Nat Commun 2015; 5:3270. [PMID: 24509916 DOI: 10.1038/ncomms4270] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 01/16/2014] [Indexed: 01/11/2023] Open
Abstract
The GTP-bound form of the Ran GTPase (RanGTP), produced around chromosomes, drives nuclear envelope and nuclear pore complex (NPC) re-assembly after mitosis. The nucleoporin MEL-28/ELYS binds chromatin in a RanGTP-regulated manner and acts to seed NPC assembly. Here we show that, upon mitotic NPC disassembly, MEL-28 dissociates from chromatin and re-localizes to spindle microtubules and kinetochores. MEL-28 directly binds microtubules in a RanGTP-regulated way via its C-terminal chromatin-binding domain. Using Xenopus egg extracts, we demonstrate that MEL-28 is essential for RanGTP-dependent microtubule nucleation and spindle assembly, independent of its function in NPC assembly. Specifically, MEL-28 interacts with the γ-tubulin ring complex and recruits it to microtubule nucleation sites. Our data identify MEL-28 as a RanGTP target that functions throughout the cell cycle. Its cell cycle-dependent binding to chromatin or microtubules discriminates MEL-28 functions in interphase and mitosis, and ensures that spindle assembly occurs only after NPC breakdown.
Collapse
Affiliation(s)
- Hideki Yokoyama
- 1] Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany [2] European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Birgit Koch
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Rudolf Walczak
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Fulya Ciray-Duygu
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | | | - Damien P Devos
- Centre for Organismal Studies (COS), Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Iain W Mattaj
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Oliver J Gruss
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| |
Collapse
|
23
|
The γ-tubulin-specific inhibitor gatastatin reveals temporal requirements of microtubule nucleation during the cell cycle. Nat Commun 2015; 6:8722. [PMID: 26503935 PMCID: PMC4640066 DOI: 10.1038/ncomms9722] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 09/24/2015] [Indexed: 11/26/2022] Open
Abstract
Inhibitors of microtubule (MT) assembly or dynamics that target α/β-tubulin are widely exploited in cancer therapy and biological research. However, specific inhibitors of the MT nucleator γ-tubulin that would allow testing temporal functions of γ-tubulin during the cell cycle are yet to be identified. By evolving β-tubulin-binding drugs we now find that the glaziovianin A derivative gatastatin is a γ-tubulin-specific inhibitor. Gatastatin decreased interphase MT dynamics of human cells without affecting MT number. Gatastatin inhibited assembly of the mitotic spindle in prometaphase. Addition of gatastatin to preformed metaphase spindles altered MT dynamics, reduced the number of growing MTs and shortened spindle length. Furthermore, gatastatin prolonged anaphase duration by affecting anaphase spindle structure, indicating the continuous requirement of MT nucleation during mitosis. Thus, gatastatin facilitates the dissection of the role of γ-tubulin during the cell cycle and reveals the sustained role of γ-tubulin. Current microtubule inhibitors target α/β-tubulin, but no specific inhibitor of γ-tubulin has been developed. Here the authors present gatastatin as a γ-tubulin inhibitor and use it to probe the role of γ-tubulin during the cell cycle.
Collapse
|
24
|
Meunier S, Vernos I. Acentrosomal Microtubule Assembly in Mitosis: The Where, When, and How. Trends Cell Biol 2015; 26:80-87. [PMID: 26475655 DOI: 10.1016/j.tcb.2015.09.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 01/08/2023]
Abstract
In mitosis the cell assembles the bipolar spindle, a microtubule (MT)-based apparatus that segregates the duplicated chromosomes into two daughter cells. Most animal cells enter mitosis with duplicated centrosomes that provide an active source of dynamic MTs. However, it is now established that spindle assembly relies on the nucleation of acentrosomal MTs occurring around the chromosomes after nuclear envelope breakdown, and on pre-existing microtubules. Where chromosome-dependent MT nucleation occurs, when MT amplification takes place and how the two pathways function are still key questions that generate some controversies. We reconcile the data and present an integrated model accounting for acentrosomal microtubule assembly in the dividing cell.
Collapse
Affiliation(s)
- Sylvain Meunier
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Doctor Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Isabelle Vernos
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Doctor Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Doctor Aiguader 88, 08003 Barcelona, Spain; Institució Catalana de Recerca I Estudis Avançats (ICREA), Passeig de Lluis Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
25
|
Chi Y, Huang S, Peng H, Liu M, Zhao J, Shao Z, Wu J. Critical role of CDK11(p58) in human breast cancer growth and angiogenesis. BMC Cancer 2015; 15:701. [PMID: 26470709 PMCID: PMC4608324 DOI: 10.1186/s12885-015-1698-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/07/2015] [Indexed: 11/24/2022] Open
Abstract
Background A capillary network is needed in cancer growth and metastasis. Induction of angiogenesis represents one of the major hallmarks of cancer. CDK11p58, a Ser/Thr kinase that belongs to the Cell Division Cycle 2-like 1 (CDC2L1) subfamily is associated with cell cycle progression, tumorigenesis, sister chromatid cohesion and apoptotic signaling. However, its role in breast cancer proliferation and angiogenesis remains unclear. Methods Tumorigenicity assays and blood vessel assessment in athymic mice were used to assess the function of CDK11p58 in tumor proliferation and angiogenesis. CCK-8 assay was used to detect breast cancer cell growth. Immunohistochemistry was used to detect the expression of vascular endothelial growth factor (VEGF), CD31 and CD34 in CDK11 positive patient breast cancer tissues. Dual-Luciferase array was used to analyze the function of CDK11p58 in the regulation of VEGF promoter activity. Western blot was used to detect related protein expression levels. Results CDK11p58 inhibited breast cancer growth and angiogenesis in breast cancer cells and in nude mice transplanted with tumors. Immunohistochemistry confirmed that CDK11p58 was negatively associated with angiogenesis-related proteins such as VEGF, CD31 and CD34 in breast cancer patients. Real-time PCR and dual-luciferase assay showed CDK11p58 inhibited the mRNA levels of VEGF and the promoter activity of VEGF. As CDK11p58 is a Ser/Thr kinase, the kinase-dead mutant failed to inhibit VEGF mRNA and promoter activity. Western blot analysis showed the same pattern of related protein expression. The data suggested angiogenesis inhibition was dependent on CDK11p58 kinase activity. Conclusion This study indicates that CDK11p58 inhibits the growth and angiogenesis of breast cancer dependent on its kinase activity. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1698-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yayun Chi
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Sheng Huang
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Haojie Peng
- School of Biomedical Engineering, hanghai Jiao Tong University, Shanghai, 200240, China.
| | - Mengying Liu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Jun Zhao
- School of Biomedical Engineering, hanghai Jiao Tong University, Shanghai, 200240, China.
| | - Zhiming Shao
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Jiong Wu
- Department of Breast Surgery, Breast Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| |
Collapse
|
26
|
Forbes DJ, Travesa A, Nord MS, Bernis C. Reprint of "Nuclear transport factors: global regulation of mitosis". Curr Opin Cell Biol 2015. [PMID: 26196321 DOI: 10.1016/j.ceb.2015.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The unexpected repurposing of nuclear transport proteins from their function in interphase to an equally vital and very different set of functions in mitosis was very surprising. The multi-talented cast when first revealed included the import receptors, importin alpha and beta, the small regulatory GTPase RanGTP, and a subset of nuclear pore proteins. In this review, we report that recent years have revealed new discoveries in each area of this expanding story in vertebrates: (a) The cast of nuclear import receptors playing a role in mitotic spindle regulation has expanded: both transportin, a nuclear import receptor, and Crm1/Xpo1, an export receptor, are involved in different aspects of spindle assembly. Importin beta and transportin also regulate nuclear envelope and pore assembly. (b) The role of nucleoporins has grown to include recruiting the key microtubule nucleator – the γ-TuRC complex – and the exportin Crm1 to the mitotic kinetochores of humans. Together they nucleate microtubule formation from the kinetochores toward the centrosomes. (c) New research finds that the original importin beta/RanGTP team have been further co-opted by evolution to help regulate other cellular and organismal activities, ranging from the actual positioning of the spindle within the cell perimeter, to regulation of a newly discovered spindle microtubule branching activity, to regulation of the interaction of microtubule structures with specific actin structures. (d) Lastly, because of the multitudinous roles of karyopherins throughout the cell cycle, a recent large push toward testing their potential as chemotherapeutic targets has begun to yield burgeoning progress in the clinic.
Collapse
Affiliation(s)
- Douglass J Forbes
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States.
| | - Anna Travesa
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Matthew S Nord
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Cyril Bernis
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| |
Collapse
|
27
|
Forbes DJ, Travesa A, Nord MS, Bernis C. Nuclear transport factors: global regulation of mitosis. Curr Opin Cell Biol 2015; 35:78-90. [PMID: 25982429 DOI: 10.1016/j.ceb.2015.04.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/01/2015] [Accepted: 04/17/2015] [Indexed: 12/22/2022]
Abstract
The unexpected repurposing of nuclear transport proteins from their function in interphase to an equally vital and very different set of functions in mitosis was very surprising. The multi-talented cast when first revealed included the import receptors, importin alpha and beta, the small regulatory GTPase RanGTP, and a subset of nuclear pore proteins. In this review, we report that recent years have revealed new discoveries in each area of this expanding story in vertebrates: (a) The cast of nuclear import receptors playing a role in mitotic spindle regulation has expanded: both transportin, a nuclear import receptor, and Crm1/Xpo1, an export receptor, are involved in different aspects of spindle assembly. Importin beta and transportin also regulate nuclear envelope and pore assembly. (b) The role of nucleoporins has grown to include recruiting the key microtubule nucleator - the γ-TuRC complex - and the exportin Crm1 to the mitotic kinetochores of humans. Together they nucleate microtubule formation from the kinetochores toward the centrosomes. (c) New research finds that the original importin beta/RanGTP team have been further co-opted by evolution to help regulate other cellular and organismal activities, ranging from the actual positioning of the spindle within the cell perimeter, to regulation of a newly discovered spindle microtubule branching activity, to regulation of the interaction of microtubule structures with specific actin structures. (d) Lastly, because of the multitudinous roles of karyopherins throughout the cell cycle, a recent large push toward testing their potential as chemotherapeutic targets has begun to yield burgeoning progress in the clinic.
Collapse
Affiliation(s)
- Douglass J Forbes
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States.
| | - Anna Travesa
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Matthew S Nord
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| | - Cyril Bernis
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, Room 2124A Pacific Hall, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0347, United States
| |
Collapse
|
28
|
Christodoulou A, Yokoyama H. Purification of nuclear localization signal-containing proteins and its application to investigation of the mechanisms of the cell division cycle. Small GTPases 2015; 6:20-7. [PMID: 25862163 PMCID: PMC4601338 DOI: 10.4161/21541248.2014.978162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The GTP bound form of the Ran GTPase (RanGTP) in the nucleus promotes nuclear import of the proteins bearing nuclear localization signals (NLS). When nuclear envelopes break down during mitosis, RanGTP is locally produced around chromosomes and drives the assembly of the spindle early in mitosis and the nuclear envelope (NE) later. RanGTP binds to the heterodimeric nuclear transport receptor importin α/β and releases NLS proteins from the receptor. Liberated NLS proteins around chromosomes have been shown to play distinct, essential roles in spindle and NE assembly. Here we provide a highly specific protocol to purify NLS proteins from crude cell lysates. The pure NLS fraction is an excellent resource to investigate the NLS protein function and identify new mitotic regulators, uncovering fundamental mechanisms of the cell division cycle. It takes 2–3 days to obtain the NLS fraction.
Collapse
|
29
|
Abstract
Summary Cyclin-dependent kinases (CDKs) are protein kinases characterized by needing a separate subunit - a cyclin - that provides domains essential for enzymatic activity. CDKs play important roles in the control of cell division and modulate transcription in response to several extra- and intracellular cues. The evolutionary expansion of the CDK family in mammals led to the division of CDKs into three cell-cycle-related subfamilies (Cdk1, Cdk4 and Cdk5) and five transcriptional subfamilies (Cdk7, Cdk8, Cdk9, Cdk11 and Cdk20). Unlike the prototypical Cdc28 kinase of budding yeast, most of these CDKs bind one or a few cyclins, consistent with functional specialization during evolution. This review summarizes how, although CDKs are traditionally separated into cell-cycle or transcriptional CDKs, these activities are frequently combined in many family members. Not surprisingly, deregulation of this family of proteins is a hallmark of several diseases, including cancer, and drug-targeted inhibition of specific members has generated very encouraging results in clinical trials.
Collapse
|
30
|
Kren BT, Unger GM, Abedin MJ, Vogel RI, Henzler CM, Ahmed K, Trembley JH. Preclinical evaluation of cyclin dependent kinase 11 and casein kinase 2 survival kinases as RNA interference targets for triple negative breast cancer therapy. Breast Cancer Res 2015; 17:19. [PMID: 25837326 PMCID: PMC4344788 DOI: 10.1186/s13058-015-0524-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 01/27/2015] [Indexed: 12/27/2022] Open
Abstract
Introduction Targeted therapies for aggressive breast cancers like triple negative breast cancer (TNBC) are needed. The use of small interfering RNAs (siRNAs) to disable expression of survival genes provides a tool for killing these cancer cells. Cyclin dependent kinase 11 (CDK11) is a survival protein kinase that regulates RNA transcription, splicing and mitosis. Casein kinase 2 (CK2) is a survival protein kinase that suppresses cancer cell death. Eliminating the expression of these genes has potential therapeutic utility for breast cancer. Methods Expression levels of CDK11 and CK2 mRNAs and associated proteins were examined in breast cancer cell lines and tissue arrays. RNA expression levels of CDC2L1, CDC2L2, CCNL1, CCNL2, CSNK2A1, CSNK2A2, and CSNK2B genes in breast cancer subtypes were analyzed. Effects following transfection of siRNAs against CDK11 and CK2 in cultured cells were examined by viability and clonal survival assays and by RNA and protein measures. Uptake of tenfibgen (TBG) nanocapsules by TNBC cells was analyzed by fluorescence-activated cell sorting. TBG nanocapsules delivered siRNAs targeting CDK11 or CK2 in mice carrying TNBC xenograft tumors. Transcript cleavage and response parameters were evaluated. Results We found strong CDK11 and CK2 mRNA and protein expression in most human breast cancer cells. Immunohistochemical analysis of TNBC patient tissues showed 100% of tumors stained positive for CDK11 with high nuclear intensity compared to normal tissue. The Cancer Genome Atlas analysis comparing basal to other breast cancer subtypes and to normal breast revealed statistically significant differences. Down-regulation of CDK11 and/or CK2 in breast cancer cells caused significant loss of cell viability and clonal survival, reduced relevant mRNA and protein expression, and induced cell death changes. TBG nanocapsules were taken up by TNBC cells both in culture and in xenograft tumors. Treatment with TBG- siRNA to CDK11 or TBG- siRNA to CK2αα’ nanocapsules induced appropriate cleavage of CDK11 and CK2α transcripts in TNBC tumors, and caused MDA-MB-231 tumor reduction, loss of proliferation, and decreased expression of targeted genes. Conclusions CDK11 and CK2 expression are individually essential for breast cancer cell survival, including TNBC. These genes serve as promising new targets for therapeutic development in breast cancer.
Collapse
Affiliation(s)
- Betsy T Kren
- Research Service (151), Minneapolis VA Health Care System, One Veterans Drive, Minneapolis, MN 55417 USA ; Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street, SE, Minneapolis, MN USA ; Masonic Cancer Center, University of Minnesota, 717 Delaware Street SE Room 130, Minneapolis, MN 55414 USA
| | | | - Md J Abedin
- Research Service (151), Minneapolis VA Health Care System, One Veterans Drive, Minneapolis, MN 55417 USA ; Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street, SE, Minneapolis, MN USA
| | - Rachel I Vogel
- Masonic Cancer Center, University of Minnesota, 717 Delaware Street SE Room 130, Minneapolis, MN 55414 USA
| | - Christine M Henzler
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street, SE, Minneapolis, MN USA ; Minnesota Supercomputing Institute, University of Minnesota, 117 Pleasant Street SE, Minneapolis, MN 55455 USA
| | - Khalil Ahmed
- Research Service (151), Minneapolis VA Health Care System, One Veterans Drive, Minneapolis, MN 55417 USA ; Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street, SE, Minneapolis, MN USA ; Masonic Cancer Center, University of Minnesota, 717 Delaware Street SE Room 130, Minneapolis, MN 55414 USA ; Department of Urology, University of Minnesota, 420 Delaware St. SE, Minneapolis, MN 55455 USA
| | - Janeen H Trembley
- Research Service (151), Minneapolis VA Health Care System, One Veterans Drive, Minneapolis, MN 55417 USA ; Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street, SE, Minneapolis, MN USA ; Masonic Cancer Center, University of Minnesota, 717 Delaware Street SE Room 130, Minneapolis, MN 55414 USA
| |
Collapse
|
31
|
Abstract
In this article, we will discuss the biochemistry of mitosis in eukaryotic cells. We will focus on conserved principles that, importantly, are adapted to the biology of the organism. It is vital to bear in mind that the structural requirements for division in a rapidly dividing syncytial Drosophila embryo, for example, are markedly different from those in a unicellular yeast cell. Nevertheless, division in both systems is driven by conserved modules of antagonistic protein kinases and phosphatases, underpinned by ubiquitin-mediated proteolysis, which create molecular switches to drive each stage of division forward. These conserved control modules combine with the self-organizing properties of the subcellular architecture to meet the specific needs of the cell. Our discussion will draw on discoveries in several model systems that have been important in the long history of research on mitosis, and we will try to point out those principles that appear to apply to all cells, compared with those in which the biochemistry has been specifically adapted in a particular organism.
Collapse
Affiliation(s)
- Samuel Wieser
- The Gurdon Institute, Cambridge CB2 1QN, United Kingdom
| | | |
Collapse
|
32
|
Scrofani J, Sardon T, Meunier S, Vernos I. Microtubule nucleation in mitosis by a RanGTP-dependent protein complex. Curr Biol 2014; 25:131-140. [PMID: 25532896 DOI: 10.1016/j.cub.2014.11.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/23/2014] [Accepted: 11/07/2014] [Indexed: 01/04/2023]
Abstract
BACKGROUND The γ-tubulin ring complex (γTuRC) is a multisubunit complex responsible for microtubule (MT) nucleation in eukaryotic cells. During mitosis, its spatial and temporal regulation promotes MT nucleation through different pathways. One of them is triggered around the chromosomes by RanGTP. Chromosomal MTs are essential for functional spindle assembly, but the mechanism by which RanGTP activates MT nucleation has not yet been resolved. RESULTS We used a combination of Xenopus egg extracts and in vitro experiments to dissect the mechanism by which RanGTP triggers MT nucleation. In egg extracts, NEDD1-coated beads promote MT nucleation only in the presence of RanGTP. We show that RanGTP promotes a direct interaction between one of its targets, TPX2, and XRHAMM that defines a specific γTuRC subcomplex. Through depletion/add-back experiments using mutant forms of TPX2 and NEDD1, we show that the activation of MT nucleation by RanGTP requires both NEDD1 phosphorylation on S405 by the TPX2-activated Aurora A and the recruitment of the complex through a TPX2-dependent mechanism. CONCLUSIONS The XRHAMM-γTuRC complex is the target for activation by RanGTP that promotes an interaction between TPX2 and XRHAMM. The resulting TPX2-RHAMM-γTuRC supracomplex fulfills the two essential requirements for the activation of MT nucleation by RanGTP: NEDD1 phosphorylation on S405 by the TPX2-activated Aurora A and the recruitment of the complex onto a TPX2-dependent scaffold. Our data identify TPX2 as the only direct RanGTP target and NEDD1 as the only Aurora A substrate essential for the activation of the RanGTP-dependent MT nucleation pathway.
Collapse
Affiliation(s)
- Jacopo Scrofani
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Doctor Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Teresa Sardon
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Doctor Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Sylvain Meunier
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Doctor Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Doctor Aiguader 88, 08003 Barcelona, Spain.
| | - Isabelle Vernos
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Doctor Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Doctor Aiguader 88, 08003 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluis Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
33
|
Regulators of carcinogenesis: emerging roles beyond their primary functions. Cancer Lett 2014; 357:75-82. [PMID: 25448403 DOI: 10.1016/j.canlet.2014.11.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/23/2014] [Accepted: 11/24/2014] [Indexed: 12/20/2022]
Abstract
Cancers are characterized by aberrant cell signaling that results in accelerated proliferation, suppressed cell death, and reprogrammed metabolism to provide sufficient energy and intermediate metabolites for macromolecular biosynthesis. Here, we summarize the emerging "unconventional" roles of these regulators based on their newly identified interaction partners, different subcellular localizations, and/or structural variants. For example, the epidermal growth factor receptor (EGFR) regulates DNA synthesis, microRNA maturation and drug resistance by interacting with previously undescribed partners; cyclins and cyclin-dependent kinases (CDKs) crosstalk with multiple canonical pathways by phosphorylating novel substrates or by functioning as transcriptional factors; apoptosis executioners play extensive roles in necroptosis, autophagy, and in the self-renewal of stem cells; and various metabolic enzymes and their mutants control carcinogenesis independently of their enzymatic activity. These recent findings will supplement the systemic functional annotation of cancer regulators and provide new rationales for potential molecular targeted cancer treatments.
Collapse
|
34
|
Tegha-Dunghu J, Bausch E, Neumann B, Wuensche A, Walter T, Ellenberg J, Gruss OJ. MAP1S controls microtubule stability throughout the cell cycle in human cells. J Cell Sci 2014; 127:5007-13. [PMID: 25300793 DOI: 10.1242/jcs.136457] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Summary Understanding the molecular basis for proper cell division requires a detailed functional analysis of microtubule (MT)-associated proteins. MT-associated protein 1S (MAP1S), the most ubiquitously expressed MAP1 family member, is required for accurate cell division. Here, using quantitative analysis of MT plus-end tracking, we show that MAP1S knockdown alters MT dynamics throughout the cell cycle. Surprisingly, MAP1S downregulation results in faster growing, yet short-lived, MTs in all cell cycle stages and in a global loss of MT acetylation. These aberrations correlate with severe defects in the final stages of cell division. In monopolar cytokinesis assays, we demonstrate that MAP1S guides MT-dependent initiation of cytokinesis. Our data underline the key role of MAP1S as a global regulator of MT stability and demonstrate a new primary function of MAP1S to regulate MT dynamics at the onset of cytokinesis.
Collapse
Affiliation(s)
- Justus Tegha-Dunghu
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Elena Bausch
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Beate Neumann
- European Molecular Biology Laboratory (EMBL), Advanced Light Microscopy Facility Programme, Meyerhostr.1, 69117 Heidelberg, Germany
| | - Annelie Wuensche
- European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Programme, Meyerhostr.1, 69117 Heidelberg, Germany
| | - Thomas Walter
- European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Programme, Meyerhostr.1, 69117 Heidelberg, Germany
| | - Jan Ellenberg
- European Molecular Biology Laboratory (EMBL), Cell Biology and Biophysics Programme, Meyerhostr.1, 69117 Heidelberg, Germany
| | - Oliver J Gruss
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| |
Collapse
|
35
|
Bernis C, Swift-Taylor B, Nord M, Carmona S, Chook YM, Forbes DJ. Transportin acts to regulate mitotic assembly events by target binding rather than Ran sequestration. Mol Biol Cell 2014; 25:992-1009. [PMID: 24478460 PMCID: PMC3967982 DOI: 10.1091/mbc.e13-08-0506] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Transportin-specific molecular tools are used to show that the mitotic cell contains importin β and transportin “global positioning system” pathways that are mechanistically parallel. Transportin works to control where the spindle, nuclear membrane, and nuclear pores are formed by directly affecting assembly factor function. The nuclear import receptors importin β and transportin play a different role in mitosis: both act phenotypically as spatial regulators to ensure that mitotic spindle, nuclear membrane, and nuclear pore assembly occur exclusively around chromatin. Importin β is known to act by repressing assembly factors in regions distant from chromatin, whereas RanGTP produced on chromatin frees factors from importin β for localized assembly. The mechanism of transportin regulation was unknown. Diametrically opposed models for transportin action are as follows: 1) indirect action by RanGTP sequestration, thus down-regulating release of assembly factors from importin β, and 2) direct action by transportin binding and inhibiting assembly factors. Experiments in Xenopus assembly extracts with M9M, a superaffinity nuclear localization sequence that displaces cargoes bound by transportin, or TLB, a mutant transportin that can bind cargo and RanGTP simultaneously, support direct inhibition. Consistently, simple addition of M9M to mitotic cytosol induces microtubule aster assembly. ELYS and the nucleoporin 107–160 complex, components of mitotic kinetochores and nuclear pores, are blocked from binding to kinetochores in vitro by transportin, a block reversible by M9M. In vivo, 30% of M9M-transfected cells have spindle/cytokinesis defects. We conclude that the cell contains importin β and transportin “global positioning system”or “GPS” pathways that are mechanistically parallel.
Collapse
Affiliation(s)
- Cyril Bernis
- Section of Cell and Developmental Biology, Division of Biological Sciences 0347, University of California-San Diego, La Jolla, CA 92093-0347 Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041
| | | | | | | | | | | |
Collapse
|
36
|
Ran GTPase in nuclear envelope formation and cancer metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 773:323-51. [PMID: 24563355 DOI: 10.1007/978-1-4899-8032-8_15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ran is a small ras-related GTPase that controls the nucleocytoplasmic exchange of macromolecules across the nuclear envelope. It binds to chromatin early during nuclear formation and has important roles during the eukaryotic cell cycle, where it regulates mitotic spindle assembly, nuclear envelope formation and cell cycle checkpoint control. Like other GTPases, Ran relies on the cycling between GTP-bound and GDP-bound conformations to interact with effector proteins and regulate these processes. In nucleocytoplasmic transport, Ran shuttles across the nuclear envelope through nuclear pores. It is concentrated in the nucleus by an active import mechanism where it generates a high concentration of RanGTP by nucleotide exchange. It controls the assembly and disassembly of a range of complexes that are formed between Ran-binding proteins and cellular cargo to maintain rapid nuclear transport. Ran also has been identified as an essential protein in nuclear envelope formation in eukaryotes. This mechanism is dependent on importin-β, which regulates the assembly of further complexes important in this process, such as Nup107-Nup160. A strong body of evidence is emerging implicating Ran as a key protein in the metastatic progression of cancer. Ran is overexpressed in a range of tumors, such as breast and renal, and these perturbed levels are associated with local invasion, metastasis and reduced patient survival. Furthermore, tumors with oncogenic KRAS or PIK3CA mutations are addicted to Ran expression, which yields exciting future therapeutic opportunities.
Collapse
|
37
|
Alawi F, Lin P. Dyskerin localizes to the mitotic apparatus and is required for orderly mitosis in human cells. PLoS One 2013; 8:e80805. [PMID: 24303026 PMCID: PMC3841160 DOI: 10.1371/journal.pone.0080805] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 10/15/2013] [Indexed: 11/18/2022] Open
Abstract
Dyskerin is a highly conserved, nucleolar RNA-binding protein with established roles in small nuclear ribonucleoprotein biogenesis, telomerase and telomere maintenance and precursor rRNA processing. Telomerase is functional during S phase and the bulk of rRNA maturation occurs during G1 and S phases; both processes are inactivated during mitosis. Yet, we show that during the course of cell cycle progression, human dyskerin expression peaks during G2/M in parallel with the upregulation of pro-mitotic factors. Dyskerin redistributed from the nucleolus in interphase cells to the perichromosomal region during prometaphase, metaphase and anaphase. With continued anaphase progression, dyskerin also localized to the cytoplasm within the mid-pole region. Loss of dyskerin function via siRNA-mediated depletion promoted G2/M accumulation and this was accompanied by an increased mitotic index and activation of the spindle assembly checkpoint. Live cell imaging further revealed an array of mitotic defects including delayed prometaphase progression, a significantly increased incidence of multi-polar spindles, and anaphase bridges culminating in micronucleus formation. Together, these findings suggest that dyskerin is a highly dynamic protein throughout the cell cycle and increases the repertoire of fundamental cellular processes that are disrupted by absence of its normal function.
Collapse
Affiliation(s)
- Faizan Alawi
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Ping Lin
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
38
|
Yokoyama H, Nakos K, Santarella-Mellwig R, Rybina S, Krijgsveld J, Koffa MD, Mattaj IW. CHD4 is a RanGTP-dependent MAP that stabilizes microtubules and regulates bipolar spindle formation. Curr Biol 2013; 23:2443-51. [PMID: 24268414 DOI: 10.1016/j.cub.2013.09.062] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 08/19/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Production of the GTP-bound form of the Ran GTPase (RanGTP) around chromosomes induces spindle assembly by activating nuclear localization signal (NLS)-containing proteins. Several NLS proteins have been identified as spindle assembly factors, but the complexity of the process led us to search for additional proteins with distinct roles in spindle assembly. RESULTS We identify a chromatin-remodeling ATPase, CHD4, as a RanGTP-dependent microtubule (MT)-associated protein (MAP). MT binding occurs via the region containing an NLS and chromatin-binding domains. In Xenopus egg extracts and cultured cells, CHD4 largely dissociates from mitotic chromosomes and partially localizes to the spindle. Immunodepletion of CHD4 from egg extracts significantly reduces the quantity of MTs produced around chromatin and prevents spindle assembly. CHD4 RNAi in both HeLa and Drosophila S2 cells induces defects in spindle assembly and chromosome alignment in early mitosis, leading to chromosome missegregation. Further analysis in egg extracts and in HeLa cells reveals that CHD4 is a RanGTP-dependent MT stabilizer. Moreover, the CHD4-containing NuRD complex promotes organization of MTs into bipolar spindles in egg extracts. Importantly, this function of CHD4 is independent of chromatin remodeling. CONCLUSIONS Our results uncover a new role for CHD4 as a MAP required for MT stabilization and involved in generating spindle bipolarity.
Collapse
Affiliation(s)
- Hideki Yokoyama
- European Molecular Biology Laboratory, Meyerhofstrasse 1, Heidelberg 69117, Germany.
| | - Konstantinos Nakos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis 68100, Greece
| | | | - Sofia Rybina
- European Molecular Biology Laboratory, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Jeroen Krijgsveld
- European Molecular Biology Laboratory, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Maria D Koffa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus Dragana, Alexandroupolis 68100, Greece.
| | - Iain W Mattaj
- European Molecular Biology Laboratory, Meyerhofstrasse 1, Heidelberg 69117, Germany
| |
Collapse
|
39
|
Bonnemay L, Hostachy S, Hoffmann C, Gautier J, Gueroui Z. Engineering spatial gradients of signaling proteins using magnetic nanoparticles. NANO LETTERS 2013; 13:5147-52. [PMID: 24111679 DOI: 10.1021/nl402356b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Intracellular biochemical reactions are often localized in space and time, inducing gradients of enzymatic activity that may play decisive roles in determining cell's fate and functions. However, the techniques available to examine such enzymatic gradients of activity remain limited. Here, we propose a new method to engineer a spatial gradient of signaling protein concentration within Xenopus egg extracts using superparamagnetic nanoparticles. We show that, upon the application of a magnetic field, a concentration gradient of nanoparticles with a tunable length extension is established within confined egg extracts. We then conjugate the nanoparticles to RanGTP, a small G-protein controlling microtubule assembly. We found that the generation of an artificial gradient of Ran-nanoparticles modifies the spatial positioning of microtubule assemblies. Furthermore, the spatial control of the level of Ran concentration allows us to correlate the local fold increase in Ran-nanoparticle concentration with the spatial positioning of the microtubule-asters. Our assay provides a bottom-up approach to examine the minimum ingredients generating polarization and symmetry breaking within cells. More generally, these results show how magnetic nanoparticles and magnetogenetic tools can be used to control the spatiotemporal dynamics of signaling pathways.
Collapse
Affiliation(s)
- L Bonnemay
- Département de Chimie, Ecole Normale Supérieure, UMR 8640 CNRS-ENS-UPMC , 24, rue Lhomond, 75005 Paris, France
| | | | | | | | | |
Collapse
|
40
|
Bärenz F, Inoue D, Yokoyama H, Tegha-Dunghu J, Freiss S, Draeger S, Mayilo D, Cado I, Merker S, Klinger M, Hoeckendorf B, Pilz S, Hupfeld K, Steinbeisser H, Lorenz H, Ruppert T, Wittbrodt J, Gruss OJ. The centriolar satellite protein SSX2IP promotes centrosome maturation. ACTA ACUST UNITED AC 2013; 202:81-95. [PMID: 23816619 PMCID: PMC3704989 DOI: 10.1083/jcb.201302122] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SSX2IP promotes centrosome maturation and maintenance at the onset of vertebrate development, preserving centrosome integrity and mitosis during rapid cleavage divisions and in somatic cells. Meiotic maturation in vertebrate oocytes is an excellent model system for microtubule reorganization during M-phase spindle assembly. Here, we surveyed changes in the pattern of microtubule-interacting proteins upon Xenopus laevis oocyte maturation by quantitative proteomics. We identified the synovial sarcoma X breakpoint protein (SSX2IP) as a novel spindle protein. Using X. laevis egg extracts, we show that SSX2IP accumulated at spindle poles in a Dynein-dependent manner and interacted with the γ-tubulin ring complex (γ-TuRC) and the centriolar satellite protein PCM-1. Immunodepletion of SSX2IP impeded γ-TuRC loading onto centrosomes. This led to reduced microtubule nucleation and spindle assembly failure. In rapidly dividing blastomeres of medaka (Oryzias latipes) and in somatic cells, SSX2IP knockdown caused fragmentation of pericentriolar material and chromosome segregation errors. We characterize SSX2IP as a novel centrosome maturation and maintenance factor that is expressed at the onset of vertebrate development. It preserves centrosome integrity and faithful mitosis during the rapid cleavage division of blastomeres and in somatic cells.
Collapse
Affiliation(s)
- Felix Bärenz
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Drogat J, Migeot V, Mommaerts E, Mullier C, Dieu M, van Bakel H, Hermand D. Cdk11-cyclinL controls the assembly of the RNA polymerase II mediator complex. Cell Rep 2012; 2:1068-76. [PMID: 23122962 DOI: 10.1016/j.celrep.2012.09.027] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 08/30/2012] [Accepted: 09/24/2012] [Indexed: 10/27/2022] Open
Abstract
The large Mediator (L-Mediator) is a general coactivator of RNA polymerase II transcription and is formed by the reversible association of the small Mediator (S-Mediator) and the kinase-module-harboring Cdk8. It is not known how the kinase module association/dissociation is regulated. We describe the fission yeast Cdk11-L-type cyclin pombe (Lcp1) complex and show that its inactivation alters the global expression profile in a manner very similar to that of mutations of the kinase module. Cdk11 is broadly distributed onto chromatin and phosphorylates the Med27 and Med4 Mediator subunits on conserved residues. The association of the kinase module and the S-Mediator is strongly decreased by the inactivation of either Cdk11 or the mutation of its target residues on the Mediator. These results show that Cdk11-Lcp1 regulates the association of the kinase module and the S-Mediator to form the L-Mediator complex.
Collapse
Affiliation(s)
- Julie Drogat
- Namur Research College (NARC), The University of Namur, Namur 5000 Belgium
| | | | | | | | | | | | | |
Collapse
|
42
|
Meunier S, Vernos I. Microtubule assembly during mitosis - from distinct origins to distinct functions? J Cell Sci 2012; 125:2805-14. [PMID: 22736044 DOI: 10.1242/jcs.092429] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The mitotic spindle is structurally and functionally defined by its main component, the microtubules (MTs). The MTs making up the spindle have various functions, organization and dynamics: astral MTs emanate from the centrosome and reach the cell cortex, and thus have a major role in spindle positioning; interpolar MTs are the main constituent of the spindle and are key for the establishment of spindle bipolarity, chromosome congression and central spindle assembly; and kinetochore-fibers are MT bundles that connect the kinetochores with the spindle poles and segregate the sister chromatids during anaphase. The duplicated centrosomes were long thought to be the origin of all of these MTs. However, in the last decade, a number of studies have contributed to the identification of non-centrosomal pathways that drive MT assembly in dividing cells. These pathways are now known to be essential for successful spindle assembly and to participate in various processes such as K-fiber formation and central spindle assembly. In this Commentary, we review the recent advances in the field and discuss how different MT assembly pathways might cooperate to successfully form the mitotic spindle.
Collapse
Affiliation(s)
- Sylvain Meunier
- Microtubule Function and Cell Division group, Cell and Developmental Biology Program, Centre for Genomic Regulation (CRG) and UPF, Dr. Aiguader 88, 08003 Barcelona, Spain
| | | |
Collapse
|
43
|
Abstract
Mitosis is the process by which eukaryotic cells organize and segregate their chromosomes in preparation for cell division. It is accomplished by a cellular machine composed largely of microtubules (MTs) and their associated proteins. This article reviews literature on mitosis from a biophysical point of view, drawing attention to the assembly and motility processes required to do this complex job with precision. Work from both the recent and the older literature is integrated into a description of relevant biological events and the experiments that probe their mechanisms. Theoretical work on specific subprocesses is also reviewed. Our goal is to provide a document that will expose biophysicists to the fascination of this quite amazing process and provide them with a good background from which they can pursue their own research interests in the subject.
Collapse
|
44
|
Abstract
'…in Italy, for thirty years under the Borgias, they had warfare, terror, murder and bloodshed, but they produced Michelangelo, Leonardo da Vinci and the Renaissance. In Switzerland, they had brotherly love, they had five hundred years of democracy and peace-and what did that produce? The cuckoo clock'. Orson Welles as Harry Lime: The Third Man. Orson Welles might have been a little unfair on the Swiss, after all cuckoo clocks were developed in the Schwartzwald, but, more importantly, Swiss democracy gives remarkably stable government with considerable decision-making at the local level. The alternative is the battling city-states of Renaissance Italy: culturally rich but chaotic at a higher level of organization. As our understanding of the cell cycle improves, it appears that the cell is organized more along the lines of Switzerland than Renaissance Italy, and one major challenge is to determine how local decisions are made and coordinated to produce the robust cell cycle mechanisms that we observe in the cell as a whole.
Collapse
Affiliation(s)
- Jonathon Pines
- Gurdon Institute, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Iain Hagan
- Paterson Institute for Cancer Research, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
45
|
Wilkinson S, Croft DR, O'Prey J, Meedendorp A, O'Prey M, Dufès C, Ryan KM. The cyclin-dependent kinase PITSLRE/CDK11 is required for successful autophagy. Autophagy 2011; 7:1295-301. [PMID: 21808150 PMCID: PMC3242795 DOI: 10.4161/auto.7.11.16646] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 07/13/2011] [Accepted: 07/18/2011] [Indexed: 12/12/2022] Open
Abstract
(Macro)autophagy is a membrane-trafficking process that serves to sequester cellular constituents in organelles termed autophagosomes, which target their degradation in the lysosome. Autophagy operates at basal levels in all cells where it serves as a homeostatic mechanism to maintain cellular integrity. The levels and cargoes of autophagy can, however, change in response to a variety of stimuli, and perturbations in autophagy are known to be involved in the aetiology of various human diseases. Autophagy must therefore be tightly controlled. We report here that the Drosophila cyclin-dependent kinase PITSLRE is a modulator of autophagy. Loss of the human PITSLRE orthologue, CDK11, initially appears to induce autophagy, but at later time points CDK11 is critically required for autophagic flux and cargo digestion. Since PITSLRE/CDK11 regulates autophagy in both Drosophila and human cells, this kinase represents a novel phylogenetically conserved component of the autophagy machinery.
Collapse
Affiliation(s)
- Simon Wilkinson
- Tumour Cell Death Laboratory, Beatson Institute for Cancer Research, Glasgow, UK.
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The basic biology of the cell division cycle and its control by protein kinases was originally studied through genetic and biochemical studies in yeast and other model organisms. The major regulatory mechanisms identified in this pioneer work are conserved in mammals. However, recent studies in different cell types or genetic models are now providing a new perspective on the function of these major cell cycle regulators in different tissues. Here, we review the physiological relevance of mammalian cell cycle kinases such as cyclin-dependent kinases (Cdks), Aurora and Polo-like kinases, and mitotic checkpoint regulators (Bub1, BubR1, and Mps1) as well as other less-studied enzymes such as Cdc7, Nek proteins, or Mastl and their implications in development, tissue homeostasis, and human disease. Among these functions, the control of self-renewal or asymmetric cell division in stem/progenitor cells and the ability to regenerate injured tissues is a central issue in current research. In addition, many of these proteins play previously unexpected roles in metabolism, cardiovascular function, or neuron biology. The modulation of their enzymatic activity may therefore have multiple therapeutic benefits in human disease.
Collapse
Affiliation(s)
- Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre, Madrid, Spain.
| |
Collapse
|
47
|
Kang J, Goodman B, Zheng Y, Tantin D. Dynamic regulation of Oct1 during mitosis by phosphorylation and ubiquitination. PLoS One 2011; 6:e23872. [PMID: 21897860 PMCID: PMC3163677 DOI: 10.1371/journal.pone.0023872] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 07/27/2011] [Indexed: 01/03/2023] Open
Abstract
Background Transcription factor Oct1 regulates multiple cellular processes. It is known to be phosphorylated during the cell cycle and by stress, however the upstream kinases and downstream consequences are not well understood. One of these modified forms, phosphorylated at S335, lacks the ability to bind DNA. Other modification states besides phosphorylation have not been described. Methodology/Principal Findings We show that Oct1 is phosphorylated at S335 in the Oct1 DNA binding domain during M-phase by the NIMA-related kinase Nek6. Phospho-Oct1 is also ubiquitinated. Phosphorylation excludes Oct1 from mitotic chromatin. Instead, Oct1pS335 concentrates at centrosomes, mitotic spindle poles, kinetochores and the midbody. Oct1 siRNA knockdown diminishes the signal at these locations. Both Oct1 ablation and overexpression result in abnormal mitoses. S335 is important for the overexpression phenotype, implicating this residue in mitotic regulation. Oct1 depletion causes defects in spindle morphogenesis in Xenopus egg extracts, establishing a mitosis-specific function of Oct1. Oct1 colocalizes with lamin B1 at the spindle poles and midbody. At the midbody, both proteins are mutually required to correctly localize the other. We show that phospho-Oct1 is modified late in mitosis by non-canonical K11-linked polyubiquitin chains. Ubiquitination requires the anaphase-promoting complex, and we further show that the anaphase-promoting complex large subunit APC1 and Oct1pS335 interact. Conclusions/Significance These findings reveal mechanistic coupling between Oct1 phosphorylation and ubquitination during mitotic progression, and a role for Oct1 in mitosis.
Collapse
Affiliation(s)
- Jinsuk Kang
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Ben Goodman
- Department of Embryology, Carnegie Institution of Washington/HHMI, Baltimore, Maryland, United States of America
| | - Yixian Zheng
- Department of Embryology, Carnegie Institution of Washington/HHMI, Baltimore, Maryland, United States of America
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
48
|
Gopinathan L, Ratnacaram CK, Kaldis P. Established and novel Cdk/cyclin complexes regulating the cell cycle and development. Results Probl Cell Differ 2011; 53:365-89. [PMID: 21630153 DOI: 10.1007/978-3-642-19065-0_16] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The identification of new members in the Cdk and cyclin families, functions for many of which are still emerging, has added new facets to the cell cycle regulatory network. With roles extending beyond the classical regulation of cell cycle progression, these new players are involved in diverse processes such as transcription, neuronal function, and ion transport. Members closely related to Cdks and cyclins such as the Speedy/RINGO proteins offer fresh insights and hope for filling in the missing gaps in our understanding of cell division. This chapter will present a broad outlook on the cell cycle and its key regulators with special emphasis on the less-studied members and their emerging roles.
Collapse
Affiliation(s)
- Lakshmi Gopinathan
- Cell Division and Cancer Laboratory (PRK), Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos #03-09, Singapore
| | | | | |
Collapse
|
49
|
Mislocalization of CDK11/PITSLRE, a regulator of the G2/M phase of the cell cycle, in Alzheimer disease. Cell Mol Biol Lett 2011; 16:359-72. [PMID: 21461981 PMCID: PMC3153952 DOI: 10.2478/s11658-011-0011-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 03/28/2011] [Indexed: 11/20/2022] Open
Abstract
Post-mitotic neurons are typically terminally differentiated and in a quiescent status. However, in Alzheimer disease (AD), many neurons display ectopic re-expression of cell cycle-related proteins. Cyclin-dependent kinase 11 (CDK11) mRNA produces a 110-kDa protein (CDK11p110) throughout the cell cycle, a 58-kDa protein (CDK11p58) that is specifically translated from an internal ribosome entry site and expressed only in the G2/M phase of the cell cycle, and a 46-kDa protein (CDK11p46) that is considered to be apoptosis specific. CDK11 is required for sister chromatid cohesion and the completion of mitosis. In this study, we found that the expression patterns of CDK11 vary such that cytoplasmic CDK11 is increased in AD cellular processes, compared to a pronounced nuclear expression pattern in most controls. We also investigated the effect of amyloid precursor protein (APP) on CDK11 expression in vitro by using M17 cells overexpressing wild-type APP and APP Swedish mutant phenotype and found increased CDK11 expression compared to empty vector. In addition, amyloid-β25–35 resulted in increased CDK11 in M17 cells. These data suggest that CDK11 may play a vital role in cell cycle re-entry in AD neurons in an APP-dependent manner, thus presenting an intriguing novel function of the APP signaling pathway in AD.
Collapse
|
50
|
Kaláb P, Solc P, Motlík J. The role of RanGTP gradient in vertebrate oocyte maturation. Results Probl Cell Differ 2011; 53:235-67. [PMID: 21630149 DOI: 10.1007/978-3-642-19065-0_12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The maturation of vertebrate oocyte into haploid gamete, the egg, consists of two specialized asymmetric cell divisions with no intervening S-phase. Ran GTPase has an essential role in relaying the active role of chromosomes in their own segregation by the meiotic process. In addition to its conserved role as a key regulator of macromolecular transport between nucleus and cytoplasm, Ran has important functions during cell division, including in mitotic spindle assembly and in the assembly of nuclear envelope at the exit from mitosis. The cellular functions of Ran are mediated by RanGTP interactions with nuclear transport receptors (NTRs) related to importin β and depend on the existence of chromosome-centered RanGTP gradient. Live imaging with FRET biosensors indeed revealed the existence of RanGTP gradient throughout mouse oocyte maturation. NTR-dependent transport of cell cycle regulators including cyclin B1, Wee2, and Cdc25B between the oocyte cytoplasm and germinal vesicle (GV) is required for normal resumption of meiosis. After GVBD in mouse oocytes, RanGTP gradient is required for timely meiosis I (MI) spindle assembly and provides long-range signal directing egg cortex differentiation. However, RanGTP gradient is not required for MI spindle migration and may be dispensable for MI spindle function in chromosome segregation. In contrast, MII spindle assembly and function in maturing mouse and Xenopus laevis eggs depend on RanGTP gradient, similar to X. laevis MII-derived egg extracts.
Collapse
Affiliation(s)
- Petr Kaláb
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, NIH, Bethesda, MD 20892-4256, USA.
| | | | | |
Collapse
|