1
|
Kasera H, Sanghi S, Singh P. PLK4 Homodimerization is Required for CEP152 Centrosome Localization and Spindle Organization. J Mol Biol 2025; 437:169152. [PMID: 40222413 DOI: 10.1016/j.jmb.2025.169152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/18/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
The centrosome-specific Polo-Like Kinase 4 (PLK4) is a unique serine/threonine kinase family member that homodimerizes using its cryptic polo-box (CPB) region. PLK4 homodimerization causes transphosphorylation, which activates its ubiquitin-mediated degradation. The same CPB interacts with upstream centrosome recruiters, CEP152 and CEP192 in human cells. However, the involvement of PLK4 homodimerization with the CEP192-CEP152 network remains unexplored. This work identified a cancerous PLK4 variant, which truncated the protein to disrupt the CPB at 774 residue. The truncated PLK4 is unable to homodimerize or interact with CEP152 or CEP192. During the S-phase, CEP152 recruits PLK4 to centrosomes, and the homodimerization of PLK4 is needed to maintain CEP152 at centrosomes. The reduction in levels of CEP152 on PLK4 homodimerization mutant expression correlates to pericentrin at S-phase centrosomes, which causes unfocussed spindles at the M-phase and reduces cell viability. The work shows a cross-dependency between CEP152 and PLK4 homodimerization for centrosome functioning, which is disrupted in cancer.
Collapse
Affiliation(s)
- Harshita Kasera
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342037, Jodhpur, Rajasthan, India
| | - Srishti Sanghi
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342037, Jodhpur, Rajasthan, India
| | - Priyanka Singh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 62, Nagaur Road, Karwar 342037, Jodhpur, Rajasthan, India.
| |
Collapse
|
2
|
Hamzah M, Meitinger F, Ohta M. PLK4: Master Regulator of Centriole Duplication and Its Therapeutic Potential. Cytoskeleton (Hoboken) 2025. [PMID: 40257113 DOI: 10.1002/cm.22031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/10/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
Centrosomes catalyze the assembly of a microtubule-based bipolar spindle, essential for the precise chromosome segregation during cell division. At the center of this process lies Polo-Like Kinase 4 (PLK4), the master regulator that controls the duplication of the centriolar core to ensure the correct balance of two centrosomes per dividing cell. Disruptions in centrosome number or function can lead to genetic disorders such as primary microcephaly or drive tumorigenesis via centrosome amplification. In this context, several chemical inhibitors of PLK4 have emerged as promising therapeutic candidates. The inhibition of PLK4 results in the emergence of acentrosomal cells, which undergo prolonged and error-prone mitosis. This aberrant mitotic duration triggers a "mitotic stopwatch" mechanism that activates the tumor suppressor p53, halting cellular proliferation. However, in a multitude of cancers, the efficacy of this mitotic surveillance mechanism is compromised by mutations that incapacitate p53. Recent investigations have unveiled p53-independent vulnerabilities in cancers characterized by chromosomal gain or amplification of 17q23, which encodes for the ubiquitin ligase TRIM37, in response to PLK4 inhibition, particularly in neuroblastoma and breast cancer. This review encapsulates the latest advancements in our understanding of centriole duplication and acentrosomal cell division in the context of TRIM37 amplification, positioning PLK4 as a compelling target for innovative cancer therapeutics.
Collapse
Affiliation(s)
- Muhammad Hamzah
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Franz Meitinger
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Midori Ohta
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
3
|
Curinha A, Huang Z, Anglen T, Strong MA, Gliech CR, Jewett CE, Friskes A, Phan TP, Nicholas Z, Holland AJ. Centriole structural integrity defects are a crucial feature of hydrolethalus syndrome. J Cell Biol 2025; 224:e202403022. [PMID: 40009365 PMCID: PMC11864076 DOI: 10.1083/jcb.202403022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 11/16/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Hydrolethalus syndrome (HLS) is a lethal, autosomal recessive ciliopathy caused by the mutation of the conserved centriole protein HYLS1. How HYLS1 controls centriole function is poorly understood. Here, we show that mice harboring the HYLS1 disease mutation die shortly after birth and exhibit developmental defects that recapitulate several manifestations of HLS. These phenotypes arise from a loss of centriole integrity that causes tissue-specific defects in cilia assembly and function. We show that HYLS1 is recruited to the centriole by CEP120 and stabilizes the localization of centriole inner scaffold proteins that ensure the integrity of the centriolar microtubule wall. The HLS disease mutation reduced the centriole localization of HYLS1 and caused degeneration of the centriole distal end. We propose that tissue-specific defects in centriole integrity caused by the HYLS1 mutation prevent ciliogenesis and contribute to HLS phenotypes.
Collapse
Affiliation(s)
- Ana Curinha
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhaoyu Huang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Taylor Anglen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Margaret A. Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Colin R. Gliech
- Department of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cayla E. Jewett
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anoek Friskes
- Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Thao P. Phan
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Zachary Nicholas
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew J. Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Brunet M, Thomas J, Lapart JA, Krüttli L, Laporte MH, Riparbelli MG, Callaini G, Durand B, Morel V. Drosophila Alms1 proteins regulate centriolar cartwheel assembly by enabling Plk4-Ana2 amplification loop. EMBO J 2025; 44:2366-2395. [PMID: 40021845 PMCID: PMC12000580 DOI: 10.1038/s44318-025-00382-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 01/06/2025] [Accepted: 01/24/2025] [Indexed: 03/03/2025] Open
Abstract
Centrioles play a central role in cell division by recruiting pericentriolar material (PCM) to form the centrosome. Alterations in centriole number or function lead to various diseases including cancer or microcephaly. Centriole duplication is a highly conserved mechanism in eukaryotes. Here, we show that the two Drosophila orthologs of the Alström syndrome protein 1 (Alms1a and Alms1b) are unexpected novel players of centriole duplication in fly. Using Ultrastructure Expansion Microscopy, we reveal that Alms1a is a PCM protein that is loaded proximally on centrioles at the onset of procentriole formation, whereas Alms1b caps the base of mature centrioles. We demonstrate that chronic loss of Alms1 proteins (with RNA null alleles) affects PCM maturation, whereas their acute loss (in RNAi KD) completely disrupts procentriole formation before Sas-6 cartwheel assembly. We establish that Alms1 proteins are required for the amplification of the Plk4-Ana2 pool at the duplication site and the subsequent Sas-6 recruitment. Thus, Alms1 proteins are novel critical but highly buffered regulators of PCM and cartwheel assembly in flies.
Collapse
Affiliation(s)
- Marine Brunet
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Joëlle Thomas
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Jean-André Lapart
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Léo Krüttli
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Marine H Laporte
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | | | | | - Bénédicte Durand
- Universite Claude BERNARD Lyon 1, Lyon, France.
- MeLiS-CNRS-UMR5284, Lyon, France.
- INSERM-U1314, Lyon, France.
| | - Véronique Morel
- Universite Claude BERNARD Lyon 1, Lyon, France.
- MeLiS-CNRS-UMR5284, Lyon, France.
- INSERM-U1314, Lyon, France.
| |
Collapse
|
5
|
Kasera H, Singh P. Harnessing Structure Prediction of Polo-Like Kinase 4 for Drug Repurposing. Cytoskeleton (Hoboken) 2025. [PMID: 40110897 DOI: 10.1002/cm.22020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/13/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
Polo-like kinase 4 (PLK4) is a centrosome-specific kinase aberrantly expressed in cancers. Drugs inhibiting its catalytic kinase domain are under clinical phase-1/2 trials in patients with different leukemia types. However, the kinase domain of PLK4 shows structural similarity with other kinases. Therefore, drugs targeting the unique C-terminal polo-box domain (PBD) of PLK4 could provide better specificity. The knowledge of domain orientation in a full-length PLK4 structure is imperative for drug discovery. In this work, we utilized ab initio and threading approaches to predict the full-length structure of human PLK4, which was employed for virtually screening the ChEMBL library. Among the hit compounds targeting the unique regions in PLK4, we identified Alectinib, which affects centrosome numbers corresponding to PLK4 levels at centrosomes. The FT-IR analysis also confirmed Alectinib interaction with the PBD. Therefore, this work identifies a chemical scaffold that could be repurposed to target the unique regions of PLK4.
Collapse
Affiliation(s)
- Harshita Kasera
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, India
| | - Priyanka Singh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, India
| |
Collapse
|
6
|
Dwivedi D, Meraldi P. Balancing Plk1 activity levels: The secret of synchrony between the cell and the centrosome cycle. Bioessays 2024; 46:e2400048. [PMID: 39128131 DOI: 10.1002/bies.202400048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
The accuracy of cell division requires precise regulation of the cellular machinery governing DNA/genome duplication, ensuring its equal distribution among the daughter cells. The control of the centrosome cycle is crucial for the formation of a bipolar spindle, ensuring error-free segregation of the genome. The cell and centrosome cycles operate in close synchrony along similar principles. Both require a single duplication round in every cell cycle, and both are controlled by the activity of key protein kinases. Nevertheless, our comprehension of the precise cellular mechanisms and critical regulators synchronizing these two cycles remains poorly defined. Here, we present our hypothesis that the spatiotemporal regulation of a dynamic equilibrium of mitotic kinases activities forms a molecular clock that governs the synchronous progression of both the cell and the centrosome cycles.
Collapse
Affiliation(s)
- Devashish Dwivedi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Translational Research Centre in Onco-haematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
7
|
Kiermaier E, Stötzel I, Schapfl MA, Villunger A. Amplified centrosomes-more than just a threat. EMBO Rep 2024; 25:4153-4167. [PMID: 39285247 PMCID: PMC11467336 DOI: 10.1038/s44319-024-00260-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/05/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Centrosomes are major organizing components of the tubulin-based cytoskeleton. In recent years, we have gained extensive knowledge about their structure, biogenesis, and function from single cells, cell-cell interactions to tissue homeostasis, including their role in human diseases. Centrosome abnormalities are linked to, among others primary microcephaly, birth defects, ciliopathies, and tumorigenesis. Centrosome amplification, a state where two or more centrosomes are present in the G1 phase of the cell cycle, correlates in cancer with karyotype alterations, clinical aggressiveness, and lymph node metastasis. However, amplified centrosomes also appear in healthy tissues and, independent of their established role, in multi-ciliation. One example is the liver where hepatocytes carry amplified centrosomes owing to whole-genome duplication events during organogenesis. More recently, amplified centrosomes have been found in neuronal progenitors and several cell types of hematopoietic origin in which they enhance cellular effector functions. These findings suggest that extra centrosomes do not necessarily pose a risk for genome integrity and are harnessed for physiological processes. Here, we compare established and emerging 'non-canonical functions' of amplified centrosomes in cancerous and somatic cells and discuss their role in cellular physiology.
Collapse
Affiliation(s)
- Eva Kiermaier
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany.
| | - Isabel Stötzel
- Life and Medical Sciences Institute, Immune and Tumor Biology, University of Bonn, Bonn, Germany
| | - Marina A Schapfl
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Lazarettgasse 14, 1090, Vienna, Austria.
| |
Collapse
|
8
|
Rajam SM, Varghese PC, Shirude MB, Syed KM, Devarajan A, Natarajan K, Dutta D. Kinase activity of histone chaperone APLF maintains steady state of centrosomes in mouse embryonic stem cells. Eur J Cell Biol 2024; 103:151439. [PMID: 38968704 DOI: 10.1016/j.ejcb.2024.151439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/07/2024] Open
Abstract
Our recent studies revealed the role of mouse Aprataxin PNK-like Factor (APLF) in development. Nevertheless, the comprehensive characterization of mouse APLF remains entirely unexplored. Based on domain deletion studies, here we report that mouse APLF's Acidic Domain and Fork Head Associated (FHA) domain can chaperone histones and repair DNA like the respective human orthologs. Immunofluorescence studies in mouse embryonic stem cells showed APLF co-localized with γ-tubulin within and around the centrosomes and govern the number and integrity of centrosomes via PLK4 phosphorylation. Enzymatic analysis established mouse APLF as a kinase. Docking studies identified three putative ATP binding sites within the FHA domain. Site-directed mutagenesis showed that R37 residue within the FHA domain is indispensable for the kinase activity of APLF thereby regulating the centrosome number. These findings might assist us comprehend APLF in different pathological and developmental conditions and reveal non-canonical kinase activity of proteins harbouring FHA domains that might impact multiple cellular processes.
Collapse
Affiliation(s)
- Sruthy Manuraj Rajam
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695014, India; Manipal Academy of Higher Education, Manipal, Karnataka State 576104, India
| | - Pallavi Chinnu Varghese
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695014, India
| | - Mayur Balkrishna Shirude
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695014, India; Manipal Academy of Higher Education, Manipal, Karnataka State 576104, India
| | - Khaja Mohieddin Syed
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695014, India
| | - Anjali Devarajan
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695014, India
| | - Kathiresan Natarajan
- Rajiv Gandhi Centre for Biotechnology (RGCB), Transdisciplinary Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695014, India
| | - Debasree Dutta
- Rajiv Gandhi Centre for Biotechnology (RGCB), Regenerative Biology Program, Thycaud PO, Poojappura, Thiruvananthapuram, Kerala 695014, India.
| |
Collapse
|
9
|
Ryniawec JM, Amoiroglou A, Rogers GC. Generating CRISPR-edited clonal lines of cultured Drosophila S2 cells. Biol Methods Protoc 2024; 9:bpae059. [PMID: 39206452 PMCID: PMC11357795 DOI: 10.1093/biomethods/bpae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/26/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
CRISPR/Cas9 genome editing is a pervasive research tool due to its relative ease of use. However, some systems are not amenable to generating edited clones due to genomic complexity and/or difficulty in establishing clonal lines. For example, Drosophila Schneider 2 (S2) cells possess a segmental aneuploid genome and are challenging to single-cell select. Here, we describe a streamlined CRISPR/Cas9 methodology for knock-in and knock-out experiments in S2 cells, whereby an antibiotic resistance gene is inserted in-frame with the coding region of a gene-of-interest. By using selectable markers, we have improved the ease and efficiency for the positive selection of null cells using antibiotic selection in feeder layers followed by cell expansion to generate clonal lines. Using this method, we generated the first acentrosomal S2 cell lines by knocking-out centriole genes Polo-like Kinase 4/Plk4 or Ana2 as proof of concept. These strategies for generating gene-edited clonal lines will add to the collection of CRISPR tools available for cultured Drosophila cells by making CRISPR more practical and therefore improving gene function studies.
Collapse
Affiliation(s)
- John M Ryniawec
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, United States
| | - Anastasia Amoiroglou
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, United States
| | - Gregory C Rogers
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, United States
| |
Collapse
|
10
|
Skinner MW, Simington CJ, López-Jiménez P, Baran KA, Xu J, Dayani Y, Pryzhkova MV, Page J, Gómez R, Holland AJ, Jordan PW. Spermatocytes have the capacity to segregate chromosomes despite centriole duplication failure. EMBO Rep 2024; 25:3373-3405. [PMID: 38943004 PMCID: PMC11316026 DOI: 10.1038/s44319-024-00187-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/30/2024] Open
Abstract
Centrosomes are the canonical microtubule organizing centers (MTOCs) of most mammalian cells, including spermatocytes. Centrosomes comprise a centriole pair within a structurally ordered and dynamic pericentriolar matrix (PCM). Unlike in mitosis, where centrioles duplicate once per cycle, centrioles undergo two rounds of duplication during spermatogenesis. The first duplication is during early meiotic prophase I, and the second is during interkinesis. Using mouse mutants and chemical inhibition, we have blocked centriole duplication during spermatogenesis and determined that non-centrosomal MTOCs (ncMTOCs) can mediate chromosome segregation. This mechanism is different from the acentriolar MTOCs that form bipolar spindles in oocytes, which require PCM components, including gamma-tubulin and CEP192. From an in-depth analysis, we identified six microtubule-associated proteins, TPX2, KIF11, NuMA, and CAMSAP1-3, that localized to the non-centrosomal MTOC. These factors contribute to a mechanism that ensures bipolar MTOC formation and chromosome segregation during spermatogenesis when centriole duplication fails. However, despite the successful completion of meiosis and round spermatid formation, centriole inheritance and PLK4 function are required for normal spermiogenesis and flagella assembly, which are critical to ensure fertility.
Collapse
Affiliation(s)
- Marnie W Skinner
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Carter J Simington
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Pablo López-Jiménez
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
- MRC Laboratory of Medical Sciences, London, W12 0NN, UK
| | - Kerstin A Baran
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jingwen Xu
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yaron Dayani
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Marina V Pryzhkova
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jesús Page
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Rocío Gómez
- Department of Biology, Autonomous University of Madrid, Madrid, Spain
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip W Jordan
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
11
|
Martinez A, Stemm-Wolf AJ, Sheridan RM, Taliaferro MJ, Pearson CG. The Unkempt RNA binding protein reveals a local translation program in centriole overduplication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605660. [PMID: 39131325 PMCID: PMC11312568 DOI: 10.1101/2024.07.29.605660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Excess centrosomes cause defects in mitosis, cell-signaling, and cell migration, and therefore their assembly is tightly regulated. Plk4 controls centriole duplication at the heart of centrosome assembly, and elevation of Plk4 promotes centrosome amplification (CA), a founding event of tumorigenesis. Here, we investigate the transcriptional consequences of elevated Plk4 and find Unkempt, a gene encoding an RNA binding protein with roles in translational regulation, to be one of only two upregulated mRNAs. Unk protein localizes to centrosomes and Cep131-positive centriolar satellites and is required for Plk4-induced centriole overduplication in an RNA-binding dependent manner. Translation is enriched at centrosomes and centriolar satellites with Unk and Cep131 promoting this localized translation. A transient centrosomal downregulation of translation occurs early in Plk4-induced CA. CNOT9, an Unk interactor and component of the translational inhibitory CCR4-NOT complex, localizes to centrosomes at this time. In summary, centriolar satellites and Unk promote local translation as part of a translational program that ensures centriole duplication.
Collapse
Affiliation(s)
- Abraham Martinez
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Alexander J. Stemm-Wolf
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Ryan M. Sheridan
- RNA Bioscience Initiative (RBI), University of Colorado, Anschutz Medical Campus, Aurora CO 80045
| | - Matthew J. Taliaferro
- RNA Bioscience Initiative (RBI), University of Colorado, Anschutz Medical Campus, Aurora CO 80045
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Chad G. Pearson
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
12
|
Grossmann J, Kratz AS, Kordonsky A, Prag G, Hoffmann I. CRL4 DCAF1 ubiquitin ligase regulates PLK4 protein levels to prevent premature centriole duplication. Life Sci Alliance 2024; 7:e202402668. [PMID: 38490717 PMCID: PMC10942865 DOI: 10.26508/lsa.202402668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024] Open
Abstract
Centrioles play important roles in the assembly of centrosomes and cilia. Centriole duplication occurs once per cell cycle and is dependent on polo-like kinase 4 (PLK4). To prevent centriole amplification, which is a hallmark of cancer, PLK4 protein levels need to be tightly regulated. Here, we show that the Cullin4A/B-DDB1-DCAF1, CRL4DCAF1, E3 ligase targets PLK4 for degradation in human cells. DCAF1 binds and ubiquitylates PLK4 in the G2 phase to prevent premature centriole duplication in mitosis. In contrast to the regulation of PLK4 by SCFβ-TrCP, the interaction between PLK4 and DCAF1 is independent of PLK4 kinase activity and mediated by polo-boxes 1 and 2 of PLK4, suggesting that DCAF1 promotes PLK4 ubiquitylation independently of β-TrCP. Thus, the SCFSlimb/β-TrCP pathway, targeting PLK4 for ubiquitylation based on its phosphorylation state and CRL4DCAF1, which ubiquitylates PLK4 by binding to the conserved PB1-PB2 domain, appear to be complementary ways to control PLK4 abundance to prevent centriole overduplication.
Collapse
Affiliation(s)
- Josina Grossmann
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anne-Sophie Kratz
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alina Kordonsky
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gali Prag
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ingrid Hoffmann
- Cell Cycle Control and Carcinogenesis, German Cancer Research Center, DKFZ, Heidelberg, Germany
| |
Collapse
|
13
|
Curinha A, Huang Z, Anglen T, Strong MA, Gliech CR, Jewett CE, Friskes A, Holland AJ. Centriole structural integrity defects are a crucial feature of Hydrolethalus Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583733. [PMID: 38496445 PMCID: PMC10942441 DOI: 10.1101/2024.03.06.583733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Hydrolethalus Syndrome (HLS) is a lethal, autosomal recessive ciliopathy caused by the mutation of the conserved centriole protein HYLS1. However, how HYLS1 facilitates the centriole-based templating of cilia is poorly understood. Here, we show that mice harboring the HYLS1 disease mutation die shortly after birth and exhibit developmental defects that recapitulate several manifestations of the human disease. These phenotypes arise from tissue-specific defects in cilia assembly and function caused by a loss of centriole integrity. We show that HYLS1 is recruited to the centriole by CEP120 and functions to recruit centriole inner scaffold proteins that stabilize the centriolar microtubule wall. The HLS mutation disrupts the interaction of HYLS1 with CEP120 leading to HYLS1 displacement and degeneration of the centriole distal end. We propose that tissue-specific defects in centriole integrity caused by the HYLS1 mutation prevent ciliogenesis and drive HLS phenotypes.
Collapse
Affiliation(s)
- Ana Curinha
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhaoyu Huang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Taylor Anglen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Margaret A Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Colin R Gliech
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cayla E Jewett
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anoek Friskes
- Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Lei Q, Yu Q, Yang N, Xiao Z, Song C, Zhang R, Yang S, Liu Z, Deng H. Therapeutic potential of targeting polo-like kinase 4. Eur J Med Chem 2024; 265:116115. [PMID: 38199166 DOI: 10.1016/j.ejmech.2023.116115] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
Polo-like kinase 4 (PLK4), a highly conserved serine/threonine kinase, masterfully regulates centriole duplication in a spatiotemporal manner to ensure the fidelity of centrosome duplication and proper mitosis. Abnormal expression of PLK4 contributes to genomic instability and associates with a poor prognosis in cancer. Inhibition of PLK4 is demonstrated to exhibit significant efficacy against various types of human cancers, further highlighting its potential as a promising therapeutic target for cancer treatment. As such, numerous small-molecule inhibitors with distinct chemical scaffolds targeting PLK4 have been extensively investigated for the treatment of different human cancers, with several undergoing clinical evaluation (e.g., CFI-400945). Here, we review the structure, distribution, and biological functions of PLK4, encapsulate its intricate regulatory mechanisms of expression, and highlighting its multifaceted roles in cancer development and metastasis. Moreover, the recent advancements of PLK4 inhibitors in patent or literature are summarized, and their therapeutic potential as monotherapies or combination therapies with other anticancer agents are also discussed.
Collapse
Affiliation(s)
- Qian Lei
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Quanwei Yu
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Na Yang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhaolin Xiao
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chao Song
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rui Zhang
- Department of Pharmacy, Guizhou Provincial People's Hospital, Guizhou, Guiyang, 550002, China
| | - Shuxin Yang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Zhihao Liu
- Department of Emergency Medicine and Laboratory of Emergency Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Hui Deng
- Department of Respiratory and Critical Care Medicine, West China Hospital and Targeted Tracer Research and Development Laboratory, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
15
|
Medley JC, Yim RN, DiPanni J, Sebou B, Shaffou B, Cramer E, Wu C, Kabara M, Song MH. Site-specific phosphorylation of ZYG-1 regulates ZYG-1 stability and centrosome number. iScience 2023; 26:108410. [PMID: 38034351 PMCID: PMC10687292 DOI: 10.1016/j.isci.2023.108410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/21/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023] Open
Abstract
Spindle bipolarity is critical for genomic integrity. As centrosome number often dictates bipolarity, tight control of centrosome assembly is vital for faithful cell division. The master centrosome regulator ZYG-1/Plk4 plays a pivotal role in this process. In C. elegans, casein kinase II (CK2) negatively regulates centrosome duplication by controlling centrosome-associated ZYG-1 levels. Here, we investigated CK2 as a regulator of ZYG-1 and its impact on centrosome assembly. We show that CK2 phosphorylates ZYG-1 in vitro and physically interacts with ZYG-1 in vivo. Depleting CK2 or blocking ZYG-1 phosphorylation at CK2 target sites leads to centrosome amplification. Non-phosphorylatable ZYG-1 mutants exhibit elevated ZYG-1 levels, leading to increased ZYG-1 and downstream factors at centrosomes, thus driving centrosome amplification. Moreover, inhibiting the 26S proteasome prevents degradation of the phospho-mimetic ZYG-1. Our findings suggest that CK2-dependent phosphorylation of ZYG-1 controls ZYG-1 levels via proteasomal degradation to limit centrosome number.
Collapse
Affiliation(s)
- Jeffrey C. Medley
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Rachel N. Yim
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Joseph DiPanni
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Brandon Sebou
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Blake Shaffou
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Evan Cramer
- Department of Chemistry, Oakland University, Rochester, MI, USA
| | - Colin Wu
- Department of Chemistry, Oakland University, Rochester, MI, USA
| | - Megan Kabara
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
- University of Connecticut School of Medicine, Office of Graduate Medical Education, Farmington, CT, USA
| | - Mi Hye Song
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| |
Collapse
|
16
|
Scott P, Curinha A, Gliech C, Holland AJ. PLK4 self-phosphorylation drives the selection of a single site for procentriole assembly. J Cell Biol 2023; 222:e202301069. [PMID: 37773039 PMCID: PMC10541313 DOI: 10.1083/jcb.202301069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/02/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Polo-like kinase 4 (PLK4) is a key regulator of centriole biogenesis, but how PLK4 selects a single site for procentriole assembly remains unclear. Using ultrastructure expansion microscopy, we show that PLK4 localizes to discrete sites along the wall of parent centrioles. While there is variation in the number of sites PLK4 occupies on the parent centriole, most PLK4 localize at a dominant site that directs procentriole assembly. Inhibition of PLK4 activity leads to stable binding of PLK4 to the centriole and increases occupancy to a maximum of nine sites. We show that self-phosphorylation of an unstructured linker promotes the release of active PLK4 from the centriole to drive the selection of a single site for procentriole assembly. Preventing linker phosphorylation blocks PLK4 turnover, leading to supernumerary sites of PLK4 localization and centriole amplification. Therefore, self-phosphorylation is a major driver of the spatial patterning of PLK4 at the centriole and plays a critical role in selecting a single centriole duplication site.
Collapse
Affiliation(s)
- Phillip Scott
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ana Curinha
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Colin Gliech
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew J. Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Gregorczyk M, Parkes EE. Targeting mitotic regulators in cancer as a strategy to enhance immune recognition. DNA Repair (Amst) 2023; 132:103583. [PMID: 37871511 DOI: 10.1016/j.dnarep.2023.103583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
Eukaryotic DNA has evolved to be enclosed within the nucleus to protect the cellular genome from autoinflammatory responses driven by the immunogenic nature of cytoplasmic DNA. Cyclic GMP-AMP Synthase (cGAS) is the cytoplasmic dsDNA sensor, which upon activation of Stimulator of Interferon Genes (STING), mediates production of pro-inflammatory interferons (IFNs) and interferon stimulated genes (ISGs). However, although this pathway is crucial in detection of viral and microbial genetic material, cytoplasmic DNA is not always of foreign origin. It is now recognised that specifically in genomic instability, a hallmark of cancer, extranuclear material in the form of micronuclei (MN) can be generated as a result of unresolved DNA lesions during mitosis. Activation of cGAS-STING in cancer has been shown to regulate numerous tumour-immune interactions such as acquisition of 'immunologically hot' phenotype which stimulates immune-mediated elimination of transformed cells. Nonetheless, a significant percentage of poorly prognostic cancers is 'immunologically cold'. As this state has been linked with low proportion of tumour-infiltrating lymphocytes (TILs), improving immunogenicity of cold tumours could be clinically relevant by exhibiting synergy with immunotherapy. This review aims to present how inhibition of vital mitotic regulators could provoke cGAS-STING response in cancer and improve the efficacy of current immunotherapy regimens.
Collapse
Affiliation(s)
- Mateusz Gregorczyk
- Oxford Centre for Immuno-Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Eileen E Parkes
- Oxford Centre for Immuno-Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom.
| |
Collapse
|
18
|
Carden S, Vitiello E, Rosa E Silva I, Holder J, Quarantotti V, Kishore K, Roamio Franklin VN, D'Santos C, Ochi T, van Breugel M, Gergely F. Proteomic profiling of centrosomes across multiple mammalian cell and tissue types by an affinity capture method. Dev Cell 2023; 58:2393-2410.e9. [PMID: 37852252 DOI: 10.1016/j.devcel.2023.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
Centrosomes are the major microtubule-organizing centers in animals and play fundamental roles in many cellular processes. Understanding how their composition varies across diverse cell types and how it is altered in disease are major unresolved questions, yet currently available centrosome isolation protocols are cumbersome and time-consuming, and they lack scalability. Here, we report the development of centrosome affinity capture (CAPture)-mass spectrometry (MS), a powerful one-step purification method to obtain high-resolution centrosome proteomes from mammalian cells. Utilizing a synthetic peptide derived from CCDC61 protein, CAPture specifically isolates intact centrosomes. Importantly, as a bead-based affinity method, it enables rapid sample processing and multiplexing unlike conventional approaches. Our study demonstrates the power of CAPture-MS to elucidate cell-type-dependent heterogeneity in centrosome composition, dissect hierarchical interactions, and identify previously unknown centrosome components. Overall, CAPture-MS represents a transformative tool to unveil temporal, regulatory, cell-type- and tissue-specific changes in centrosome proteomes in health and disease.
Collapse
Affiliation(s)
- Sarah Carden
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Elisa Vitiello
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - James Holder
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Valentina Quarantotti
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Kamal Kishore
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | | | - Clive D'Santos
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Takashi Ochi
- MRC Laboratory of Molecular Biology, Cambridge, UK; The Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK.
| | - Mark van Breugel
- MRC Laboratory of Molecular Biology, Cambridge, UK; School of Biological & Behavioural Sciences, Queen Mary University of London, London, UK.
| | - Fanni Gergely
- CRUK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK; Department of Biochemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Wilmott ZM, Goriely A, Raff JW. A simple Turing reaction-diffusion model explains how PLK4 breaks symmetry during centriole duplication and assembly. PLoS Biol 2023; 21:e3002391. [PMID: 37983248 PMCID: PMC10659181 DOI: 10.1371/journal.pbio.3002391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/18/2023] [Indexed: 11/22/2023] Open
Abstract
Centrioles duplicate when a mother centriole gives birth to a daughter that grows from its side. Polo-like-kinase 4 (PLK4), the master regulator of centriole duplication, is recruited symmetrically around the mother centriole, but it then concentrates at a single focus that defines the daughter centriole assembly site. How PLK4 breaks symmetry is unclear. Here, we propose that phosphorylated and unphosphorylated species of PLK4 form the 2 components of a classical Turing reaction-diffusion system. These 2 components bind to/unbind from the surface of the mother centriole at different rates, allowing a slow-diffusing activator species of PLK4 to accumulate at a single site on the mother, while a fast-diffusing inhibitor species of PLK4 suppresses activator accumulation around the rest of the centriole. This "short-range activation/long-range inhibition," inherent to Turing systems, can drive PLK4 symmetry breaking on a either a continuous or compartmentalised Plk4-binding surface, with PLK4 overexpression producing multiple PLK4 foci and PLK4 kinase inhibition leading to a lack of symmetry-breaking and PLK4 accumulation-as observed experimentally.
Collapse
Affiliation(s)
- Zachary M. Wilmott
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Jordan W. Raff
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Galletta BJ, Varadarajan R, Fagerstrom CJ, Yang B, Haase KP, McJunkin K, Rusan NM. The E3 ligase Poe promotes Pericentrin degradation. Mol Biol Cell 2023; 34:br15. [PMID: 37342879 PMCID: PMC10398894 DOI: 10.1091/mbc.e22-11-0534] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023] Open
Abstract
Centrosomes are essential parts of diverse cellular processes, and precise regulation of the levels of their constituent proteins is critical for their function. One such protein is Pericentrin (PCNT) in humans and Pericentrin-like protein (PLP) in Drosophila. Increased PCNT expression and its protein accumulation are linked to clinical conditions including cancer, mental disorders, and ciliopathies. However, the mechanisms by which PCNT levels are regulated remain underexplored. Our previous study demonstrated that PLP levels are sharply down-regulated during early spermatogenesis and this regulation is essential to spatially position PLP on the proximal end of centrioles. We hypothesized that the sharp drop in PLP protein was a result of rapid protein degradation during the male germ line premeiotic G2 phase. Here, we show that PLP is subject to ubiquitin-mediated degradation and identify multiple proteins that promote the reduction of PLP levels in spermatocytes, including the UBR box containing E3 ligase Poe (UBR4), which we show binds to PLP. Although protein sequences governing posttranslational regulation of PLP are not restricted to a single region of the protein, we identify a region that is required for Poe-mediated degradation. Experimentally stabilizing PLP, via internal PLP deletions or loss of Poe, leads to PLP accumulation in spermatocytes, its mispositioning along centrioles, and defects in centriole docking in spermatids.
Collapse
Affiliation(s)
- Brian J. Galletta
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, and
| | - Ramya Varadarajan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, and
| | - Carey J. Fagerstrom
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, and
| | - Bing Yang
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Karen Plevock Haase
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, and
| | - Katherine McJunkin
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Nasser M. Rusan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, and
| |
Collapse
|
21
|
Ryniawec JM, Buster DW, Slevin LK, Boese CJ, Amoiroglou A, Dean SM, Slep KC, Rogers GC. Polo-like kinase 4 homodimerization and condensate formation regulate its own protein levels but are not required for centriole assembly. Mol Biol Cell 2023; 34:ar80. [PMID: 37163316 PMCID: PMC10398880 DOI: 10.1091/mbc.e22-12-0572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/11/2023] Open
Abstract
Polo-like kinase 4 (Plk4) is the master-regulator of centriole assembly, and cell cycle-dependent regulation of its activity maintains proper centrosome number. During most of the cell cycle, Plk4 levels are nearly undetectable due to its ability to autophosphorylate and trigger its own ubiquitin-mediated degradation. However, during mitotic exit, Plk4 forms a single aggregate on the centriole surface to stimulate centriole duplication. Whereas most Polo-like kinase family members are monomeric, Plk4 is unique because it forms homodimers. Notably, Plk4 trans-autophosphorylates a degron near its kinase domain, a critical step in autodestruction. While it is thought that the purpose of homodimerization is to promote trans-autophosphorylation, this has not been tested. Here, we generated separation-of-function Plk4 mutants that fail to dimerize and show that homodimerization creates a binding site for the Plk4 activator, Asterless. Surprisingly, however, Plk4 dimer mutants are catalytically active in cells, promote centriole assembly, and can trans-autophosphorylate through concentration-dependent condensate formation. Moreover, we mapped and then deleted the weak-interacting regions within Plk4 that mediate condensation and conclude that dimerization and condensation are not required for centriole assembly. Our findings suggest that Plk4 dimerization and condensation function simply to down-regulate Plk4 and suppress centriole overduplication.
Collapse
Affiliation(s)
- John M. Ryniawec
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Daniel W. Buster
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Lauren K. Slevin
- Department of Biology, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599
| | - Cody J. Boese
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Anastasia Amoiroglou
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Spencer M. Dean
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| | - Kevin C. Slep
- Department of Biology, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599
| | - Gregory C. Rogers
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
22
|
Medley JC, Yim N, DiPanni J, Sebou B, Shaffou B, Cramer E, Wu C, Kabara M, Song MH. Site-Specific Phosphorylation of ZYG-1 Regulates ZYG-1 Stability and Centrosome Number. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.07.539463. [PMID: 37333374 PMCID: PMC10274923 DOI: 10.1101/2023.05.07.539463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Spindle bipolarity is critical for genomic integrity. Given that centrosome number often dictates mitotic bipolarity, tight control of centrosome assembly is vital for the fidelity of cell division. The kinase ZYG-1/Plk4 is a master centrosome factor that is integral for controlling centrosome number and is modulated by protein phosphorylation. While autophosphorylation of Plk4 has been extensively studied in other systems, the mechanism of ZYG-1 phosphorylation in C. elegans remains largely unexplored. In C. elegans, Casein Kinase II (CK2) negatively regulates centrosome duplication by controlling centrosome-associated ZYG-1 levels. In this study, we investigated ZYG-1 as a potential substrate of CK2 and the functional impact of ZYG-1 phosphorylation on centrosome assembly. First, we show that CK2 directly phosphorylates ZYG-1 in vitro and physically interacts with ZYG-1 in vivo. Intriguingly, depleting CK2 or blocking ZYG-1 phosphorylation at putative CK2 target sites leads to centrosome amplification. In the non-phosphorylatable (NP)-ZYG-1 mutant embryo, the overall levels of ZYG-1 are elevated, leading to an increase in centrosomal ZYG-1 and downstream factors, providing a possible mechanism of the NP-ZYG-1 mutation to drive centrosome amplification. Moreover, inhibiting the 26S proteasome blocks degradation of the phospho-mimetic (PM)-ZYG-1, while the NP-ZYG-1 mutant shows partial resistance to proteasomal degradation. Our findings suggest that site-specific phosphorylation of ZYG-1, partly mediated by CK2, controls ZYG-1 levels via proteasomal degradation, limiting centrosome number. We provide a mechanism linking CK2 kinase activity to centrosome duplication through direct phosphorylation of ZYG-1, which is critical for the integrity of centrosome number.
Collapse
Affiliation(s)
| | - Nahyun Yim
- Department of Biological Sciences, Oakland University, MI, USA
| | - Joseph DiPanni
- Department of Biological Sciences, Oakland University, MI, USA
| | - Brandon Sebou
- Department of Biological Sciences, Oakland University, MI, USA
| | - Blake Shaffou
- Department of Biological Sciences, Oakland University, MI, USA
| | - Evan Cramer
- Department of Chemistry, Oakland University, MI, USA
| | - Colin Wu
- Department of Chemistry, Oakland University, MI, USA
| | - Megan Kabara
- Department of Biological Sciences, Oakland University, MI, USA
- University of Connecticut School of Medicine, Office of Graduate Medical Education, Farmington, CT, USA
| | - Mi Hye Song
- Department of Biological Sciences, Oakland University, MI, USA
| |
Collapse
|
23
|
Zhang C, Ma X, Wei G, Zhu X, Hu P, Chen X, Wang D, Li Y, Ruan T, Zhang W, Tao K, Wu C. Centrosomal protein 120 promotes centrosome amplification and gastric cancer progression via USP54-mediated deubiquitination of PLK4. iScience 2022; 26:105745. [PMID: 36590171 PMCID: PMC9800543 DOI: 10.1016/j.isci.2022.105745] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/09/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Centrosomal protein 120 (CEP120) is a 120 kDa centrosome protein that plays an important role in centrosome replication. Overexpression of CEP120 can lead to centrosome duplicate abnormality, which is closely associated with tumorigenesis and development. However, there are no reports on the relationship between CEP120 and tumors. In our study, overexpression of CEP120 promoted centrosome amplification in gastric cancer (GC), and the role of CEP120 in promoting GC progression was demonstrated in vitro and in vivo. We demonstrated that CEP120 promotes centrosome amplification and GC progression by promoting the expression and centrosome aggregation of the deubiquitinating enzyme USP54, maintaining the stability of PLK4 and reducing its ubiquitination degradation. In conclusion, the CEP120-USP54-PLK4 axis may play an important role in promoting centrosome amplification and GC progression, thus providing a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Chenggang Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Xianxiong Ma
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Guanxin Wei
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Xiuxian Zhu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Peng Hu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Xiang Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Dianshi Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Yuan Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Tuo Ruan
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Weikang Zhang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Chuanqing Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
- Corresponding author
| |
Collapse
|
24
|
Phan TP, Boatwright CA, Drown CG, Skinner MW, Strong MA, Jordan PW, Holland AJ. Upstream open reading frames control PLK4 translation and centriole duplication in primordial germ cells. Genes Dev 2022; 36:718-736. [PMID: 35772791 PMCID: PMC9296005 DOI: 10.1101/gad.349604.122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/09/2022] [Indexed: 11/24/2022]
Abstract
Centrosomes are microtubule-organizing centers comprised of a pair of centrioles and the surrounding pericentriolar material. Abnormalities in centriole number are associated with cell division errors and can contribute to diseases such as cancer. Centriole duplication is limited to once per cell cycle and is controlled by the dosage-sensitive Polo-like kinase 4 (PLK4). Here, we show that PLK4 abundance is translationally controlled through conserved upstream open reading frames (uORFs) in the 5' UTR of the mRNA. Plk4 uORFs suppress Plk4 translation and prevent excess protein synthesis. Mice with homozygous knockout of Plk4 uORFs (Plk4 Δu/Δu ) are viable but display dramatically reduced fertility because of a significant depletion of primordial germ cells (PGCs). The remaining PGCs in Plk4 Δu/Δu mice contain extra centrioles and display evidence of increased mitotic errors. PGCs undergo hypertranscription and have substantially more Plk4 mRNA than somatic cells. Reducing Plk4 mRNA levels in mice lacking Plk4 uORFs restored PGC numbers and fully rescued fertility. Together, our data uncover a specific requirement for uORF-dependent control of PLK4 translation in counterbalancing the increased Plk4 transcription in PGCs. Thus, uORF-mediated translational suppression of PLK4 has a critical role in preventing centriole amplification and preserving the genomic integrity of future gametes.
Collapse
Affiliation(s)
- Thao P Phan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Christina A Boatwright
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Chelsea G Drown
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Marnie W Skinner
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Margaret A Strong
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Philip W Jordan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, USA
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
25
|
Iyer J, Gentry LK, Bergwell M, Smith A, Guagliardo S, Kropp PA, Sankaralingam P, Liu Y, Spooner E, Bowerman B, O’Connell KF. The chromatin remodeling protein CHD-1 and the EFL-1/DPL-1 transcription factor cooperatively down regulate CDK-2 to control SAS-6 levels and centriole number. PLoS Genet 2022; 18:e1009799. [PMID: 35377871 PMCID: PMC9009770 DOI: 10.1371/journal.pgen.1009799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 04/14/2022] [Accepted: 03/17/2022] [Indexed: 11/24/2022] Open
Abstract
Centrioles are submicron-scale, barrel-shaped organelles typically found in pairs, and play important roles in ciliogenesis and bipolar spindle assembly. In general, successful execution of centriole-dependent processes is highly reliant on the ability of the cell to stringently control centriole number. This in turn is mainly achieved through the precise duplication of centrioles during each S phase. Aberrations in centriole duplication disrupt spindle assembly and cilia-based signaling and have been linked to cancer, primary microcephaly and a variety of growth disorders. Studies aimed at understanding how centriole duplication is controlled have mainly focused on the post-translational regulation of two key components of this pathway: the master regulatory kinase ZYG-1/Plk4 and the scaffold component SAS-6. In contrast, how transcriptional control mechanisms might contribute to this process have not been well explored. Here we show that the chromatin remodeling protein CHD-1 contributes to the regulation of centriole duplication in the C. elegans embryo. Specifically, we find that loss of CHD-1 or inactivation of its ATPase activity can restore embryonic viability and centriole duplication to a strain expressing insufficient ZYG-1 activity. Interestingly, loss of CHD-1 is associated with increases in the levels of two ZYG-1-binding partners: SPD-2, the centriole receptor for ZYG-1 and SAS-6. Finally, we explore transcriptional regulatory networks governing centriole duplication and find that CHD-1 and a second transcription factor, EFL-1/DPL-1 cooperate to down regulate expression of CDK-2, which in turn promotes SAS-6 protein levels. Disruption of this regulatory network results in the overexpression of SAS-6 and the production of extra centrioles. Centrioles are cellular constituents that play an important role in cell reproduction, signaling and movement. To properly function, centrioles must be present in the cell at precise numbers. Errors in maintaining centriole number result in cell division defects and diseases such as cancer and microcephaly. How the cell maintains proper centriole copy number is not entirely understood. Here we show that two transcription factors, EFL-1/DPL-1 and CHD-1 cooperate to reduce expression of CDK-2, a master regulator of the cell cycle. We find that CDK-2 in turn promotes expression of SAS-6, a major building block of centrioles. When EFL-1/DPL-1 and CHD-1 are inhibited, CDK-2 is overexpressed. This leads to increased levels of SAS-6 and excess centrioles. Our work thus demonstrates a novel mechanism for controlling centriole number and is thus relevant to those human diseases caused by defects in centriole copy number control.
Collapse
Affiliation(s)
- Jyoti Iyer
- Department of Chemistry and Biochemistry, University of Tulsa, Tulsa, Oklahoma, United States of America
- * E-mail: (JI); (KFO)
| | - Lindsey K. Gentry
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Mary Bergwell
- Department of Chemistry and Biochemistry, University of Tulsa, Tulsa, Oklahoma, United States of America
| | - Amy Smith
- Department of Chemistry and Biochemistry, University of Tulsa, Tulsa, Oklahoma, United States of America
| | - Sarah Guagliardo
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Peter A. Kropp
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Prabhu Sankaralingam
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Yan Liu
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
| | - Eric Spooner
- Proteomics Core Facility, Whitehead Institute for Biomedical Research, Cambridge Massachusetts, United States of America
| | - Bruce Bowerman
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Kevin F. O’Connell
- Laboratory of Biochemistry and Genetics, National Institutes of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, United States of America
- * E-mail: (JI); (KFO)
| |
Collapse
|
26
|
Hoffmann I. Role of Polo-like Kinases Plk1 and Plk4 in the Initiation of Centriole Duplication-Impact on Cancer. Cells 2022; 11:786. [PMID: 35269408 PMCID: PMC8908989 DOI: 10.3390/cells11050786] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Centrosomes nucleate and anchor microtubules and therefore play major roles in spindle formation and chromosome segregation during mitosis. Duplication of the centrosome occurs, similar to DNA, only once during the cell cycle. Aberration of the centrosome number is common in human tumors. At the core of centriole duplication is the conserved polo-like kinase 4, Plk4, and two structural proteins, STIL and Sas-6. In this review, I summarize and discuss developments in our understanding of the first steps of centriole duplication and their regulation.
Collapse
Affiliation(s)
- Ingrid Hoffmann
- F045, Cell Cycle Control and Carcinogenesis, Im Neuenheimer Feld 242, 69115 Heidelberg, Germany
| |
Collapse
|
27
|
Cunningham NHJ, Bouhlel IB, Conduit PT. Daughter centrioles assemble preferentially towards the nuclear envelope in Drosophila syncytial embryos. Open Biol 2022; 12:210343. [PMID: 35042404 PMCID: PMC8767211 DOI: 10.1098/rsob.210343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Centrosomes are important organizers of microtubules within animal cells. They comprise a pair of centrioles surrounded by the pericentriolar material, which nucleates and organizes the microtubules. To maintain centrosome numbers, centrioles must duplicate once and only once per cell cycle. During S-phase, a single new ‘daughter’ centriole is built orthogonally on one side of each radially symmetric ‘mother’ centriole. Mis-regulation of duplication can result in the simultaneous formation of multiple daughter centrioles around a single mother centriole, leading to centrosome amplification, a hallmark of cancer. It remains unclear how a single duplication site is established. It also remains unknown whether this site is pre-defined or randomly positioned around the mother centriole. Here, we show that within Drosophila syncytial embryos daughter centrioles preferentially assemble on the side of the mother facing the nuclear envelope, to which the centrosomes are closely attached. This positional preference is established early during duplication and remains stable throughout daughter centriole assembly, but is lost in centrosomes forced to lose their connection to the nuclear envelope. This shows that non-centrosomal cues influence centriole duplication and raises the possibility that these external cues could help establish a single duplication site.
Collapse
Affiliation(s)
- Neil H J Cunningham
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Imène B Bouhlel
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Paul T Conduit
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK.,Université de Paris, CNRS, Institut Jacques Monod, 75006 Paris, France
| |
Collapse
|
28
|
Keep Calm and Carry on with Extra Centrosomes. Cancers (Basel) 2022; 14:cancers14020442. [PMID: 35053604 PMCID: PMC8774008 DOI: 10.3390/cancers14020442] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Precise chromosome segregation during mitosis is a vital event orchestrated by formation of bipolar spindle poles. Supernumerary centrosomes, caused by centrosome amplification, deteriorates mitotic processes, resulting in segregation defects leading to chromosomal instability (CIN). Centrosome amplification is frequently observed in various types of cancer and considered as a significant contributor to destabilization of chromosomes. This review provides a comprehensive overview of causes and consequences of centrosome amplification thoroughly describing molecular mechanisms. Abstract Aberrations in the centrosome number and structure can readily be detected at all stages of tumor progression and are considered hallmarks of cancer. Centrosome anomalies are closely linked to chromosome instability and, therefore, are proposed to be one of the driving events of tumor formation and progression. This concept, first posited by Boveri over 100 years ago, has been an area of interest to cancer researchers. We have now begun to understand the processes by which these numerical and structural anomalies may lead to cancer, and vice-versa: how key events that occur during carcinogenesis could lead to amplification of centrosomes. Despite the proliferative advantages that having extra centrosomes may confer, their presence can also lead to loss of essential genetic material as a result of segregational errors and cancer cells must deal with these deadly consequences. Here, we review recent advances in the current literature describing the mechanisms by which cancer cells amplify their centrosomes and the methods they employ to tolerate the presence of these anomalies, focusing particularly on centrosomal clustering.
Collapse
|
29
|
Blanco-Ameijeiras J, Lozano-Fernández P, Martí E. Centrosome maturation - in tune with the cell cycle. J Cell Sci 2022; 135:274149. [PMID: 35088834 DOI: 10.1242/jcs.259395] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Centrosomes are the main microtubule-organizing centres, playing essential roles in the organization of the cytoskeleton during interphase, and in the mitotic spindle, which controls chromosome segregation, during cell division. Centrosomes also act as the basal body of cilia, regulating cilium length and affecting extracellular signal reception as well as the integration of intracellular signalling pathways. Centrosomes are self-replicative and duplicate once every cell cycle to generate two centrosomes. The core support structure of the centrosome consists of two molecularly distinct centrioles. The mother (mature) centriole exhibits accessory appendages and is surrounded by both pericentriolar material and centriolar satellites, structures that the daughter (immature) centriole lacks. In this Review, we discuss what is currently known about centrosome duplication, its dialogue with the cell cycle and the sequential acquisition of specific components during centriole maturation. We also describe our current understanding of the mature centriolar structures that are required to build a cilium. Altogether, the built-in centrosome asymmetries that stem from the two centrosomes inheriting molecularly different centrioles sets the foundation for cell division being an intrinsically asymmetric process.
Collapse
Affiliation(s)
- Jose Blanco-Ameijeiras
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Pilar Lozano-Fernández
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Baldiri i Reixac 20, Barcelona 08028, Spain
| | - Elisa Martí
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Baldiri i Reixac 20, Barcelona 08028, Spain
| |
Collapse
|
30
|
Kinterová V, Kaňka J, Bartková A, Toralová T. SCF Ligases and Their Functions in Oogenesis and Embryogenesis-Summary of the Most Important Findings throughout the Animal Kingdom. Cells 2022; 11:234. [PMID: 35053348 PMCID: PMC8774150 DOI: 10.3390/cells11020234] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 12/10/2022] Open
Abstract
SCF-dependent proteolysis was first discovered via genetic screening of budding yeast almost 25 years ago. In recent years, more and more functions of SCF (Skp1-Cullin 1-F-box) ligases have been described, and we can expect the number of studies on this topic to increase. SCF ligases, which are E3 ubiquitin multi-protein enzymes, catalyse protein ubiquitination and thus allow protein degradation mediated by the 26S proteasome. They play a crucial role in the degradation of cell cycle regulators, regulation of the DNA repair and centrosome cycle and play an important role in several diseases. SCF ligases seem to be needed during all phases of development, from oocyte formation through fertilization, activation of the embryonic genome to embryo implantation. In this review, we summarize known data on SCF ligase-mediated degradation during oogenesis and embryogenesis. In particular, SCFβTrCP and SCFSEL-10/FBXW7 are among the most important and best researched ligases during early development. SCFβTrCP is crucial for the oogenesis of Xenopus and mouse and also in Xenopus and Drosophila embryogenesis. SCFSEL-10/FBXW7 participates in the degradation of several RNA-binding proteins and thereby affects the regulation of gene expression during the meiosis of C. elegans. Nevertheless, a large number of SCF ligases that are primarily involved in embryogenesis remain to be elucidated.
Collapse
Affiliation(s)
- Veronika Kinterová
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
| | - Jiří Kaňka
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
| | - Alexandra Bartková
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
- Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
| | - Tereza Toralová
- Laboratory of Developmental Biology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 27721 Libechov, Czech Republic; (J.K.); (A.B.); (T.T.)
| |
Collapse
|
31
|
Mofatteh M, Echegaray-Iturra F, Alamban A, Dalla Ricca F, Bakshi A, Aydogan MG. Autonomous clocks that regulate organelle biogenesis, cytoskeletal organization, and intracellular dynamics. eLife 2021; 10:e72104. [PMID: 34586070 PMCID: PMC8480978 DOI: 10.7554/elife.72104] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/14/2021] [Indexed: 12/27/2022] Open
Abstract
How do cells perceive time? Do cells use temporal information to regulate the production/degradation of their enzymes, membranes, and organelles? Does controlling biological time influence cytoskeletal organization and cellular architecture in ways that confer evolutionary and physiological advantages? Potential answers to these fundamental questions of cell biology have historically revolved around the discussion of 'master' temporal programs, such as the principal cyclin-dependent kinase/cyclin cell division oscillator and the circadian clock. In this review, we provide an overview of the recent evidence supporting an emerging concept of 'autonomous clocks,' which under normal conditions can be entrained by the cell cycle and/or the circadian clock to run at their pace, but can also run independently to serve their functions if/when these major temporal programs are halted/abrupted. We begin the discussion by introducing recent developments in the study of such clocks and their roles at different scales and complexities. We then use current advances to elucidate the logic and molecular architecture of temporal networks that comprise autonomous clocks, providing important clues as to how these clocks may have evolved to run independently and, sometimes at the cost of redundancy, have strongly coupled to run under the full command of the cell cycle and/or the circadian clock. Next, we review a list of important recent findings that have shed new light onto potential hallmarks of autonomous clocks, suggestive of prospective theoretical and experimental approaches to further accelerate their discovery. Finally, we discuss their roles in health and disease, as well as possible therapeutic opportunities that targeting the autonomous clocks may offer.
Collapse
Affiliation(s)
- Mohammad Mofatteh
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Fabio Echegaray-Iturra
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Andrew Alamban
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Francesco Dalla Ricca
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Anand Bakshi
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Mustafa G Aydogan
- Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
32
|
Nabais C, Pessoa D, de-Carvalho J, van Zanten T, Duarte P, Mayor S, Carneiro J, Telley IA, Bettencourt-Dias M. Plk4 triggers autonomous de novo centriole biogenesis and maturation. J Cell Biol 2021; 220:211915. [PMID: 33760919 PMCID: PMC7995200 DOI: 10.1083/jcb.202008090] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/14/2020] [Accepted: 02/18/2021] [Indexed: 12/23/2022] Open
Abstract
Centrioles form centrosomes and cilia. In most proliferating cells, centrioles assemble through canonical duplication, which is spatially, temporally, and numerically regulated by the cell cycle and the presence of mature centrioles. However, in certain cell types, centrioles assemble de novo, yet by poorly understood mechanisms. Herein, we established a controlled system to investigate de novo centriole biogenesis, using Drosophila melanogaster egg explants overexpressing Polo-like kinase 4 (Plk4), a trigger for centriole biogenesis. We show that at a high Plk4 concentration, centrioles form de novo, mature, and duplicate, independently of cell cycle progression and of the presence of other centrioles. Plk4 concentration determines the temporal onset of centriole assembly. Moreover, our results suggest that distinct biochemical kinetics regulate de novo and canonical biogenesis. Finally, we investigated which other factors modulate de novo centriole assembly and found that proteins of the pericentriolar material (PCM), and in particular γ-tubulin, promote biogenesis, likely by locally concentrating critical components.
Collapse
Affiliation(s)
| | | | | | | | - Paulo Duarte
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Satyajit Mayor
- National Centre for Biological Sciences, Bangalore, India
| | | | - Ivo A Telley
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | |
Collapse
|
33
|
Thompson LL, Rutherford KA, Lepage CC, McManus KJ. The SCF Complex Is Essential to Maintain Genome and Chromosome Stability. Int J Mol Sci 2021; 22:8544. [PMID: 34445249 PMCID: PMC8395177 DOI: 10.3390/ijms22168544] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 12/20/2022] Open
Abstract
The SKP1, CUL1, F-box protein (SCF) complex encompasses a group of 69 SCF E3 ubiquitin ligase complexes that primarily modify protein substrates with poly-ubiquitin chains to target them for proteasomal degradation. These SCF complexes are distinguishable by variable F-box proteins, which determine substrate specificity. Although the function(s) of each individual SCF complex remain largely unknown, those that have been characterized regulate a wide array of cellular processes, including gene transcription and the cell cycle. In this regard, the SCF complex regulates transcription factors that modulate cell signaling and ensures timely degradation of primary cell cycle regulators for accurate replication and segregation of genetic material. SCF complex members are aberrantly expressed in a myriad of cancer types, with altered expression or function of the invariable core SCF components expected to have a greater impact on cancer pathogenesis than that of the F-box proteins. Accordingly, this review describes the normal roles that various SCF complexes have in maintaining genome stability before discussing the impact that aberrant SCF complex expression and/or function have on cancer pathogenesis. Further characterization of the SCF complex functions is essential to identify and develop therapeutic approaches to exploit aberrant SCF complex expression and function.
Collapse
Affiliation(s)
- Laura L. Thompson
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Kailee A. Rutherford
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Chloe C. Lepage
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Kirk J. McManus
- CancerCare Manitoba Research Institute, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; (L.L.T.); (K.A.R.); (C.C.L.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
34
|
Pereira SG, Dias Louro MA, Bettencourt-Dias M. Biophysical and Quantitative Principles of Centrosome Biogenesis and Structure. Annu Rev Cell Dev Biol 2021; 37:43-63. [PMID: 34314592 DOI: 10.1146/annurev-cellbio-120219-051400] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The centrosome is a main orchestrator of the animal cellular microtubule cytoskeleton. Dissecting its structure and assembly mechanisms has been a goal of cell biologists for over a century. In the last two decades, a good understanding of the molecular constituents of centrosomes has been achieved. Moreover, recent breakthroughs in electron and light microscopy techniques have enabled the inspection of the centrosome and the mapping of its components with unprecedented detail. However, we now need a profound and dynamic understanding of how these constituents interact in space and time. Here, we review the latest findings on the structural and molecular architecture of the centrosome and how its biogenesis is regulated, highlighting how biophysical techniques and principles as well as quantitative modeling are changing our understanding of this enigmatic cellular organelle. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
|
35
|
Medley JC, DiPanni JR, Schira L, Shaffou BM, Sebou BM, Song MH. APC/CFZR-1 regulates centrosomal ZYG-1 to limit centrosome number. J Cell Sci 2021; 134:jcs253088. [PMID: 34308970 PMCID: PMC8349554 DOI: 10.1242/jcs.253088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 06/23/2021] [Indexed: 12/28/2022] Open
Abstract
Aberrant centrosome numbers are associated with human cancers. The levels of centrosome regulators positively correlate with centrosome number. Thus, tight control of centrosome protein levels is critical. In Caenorhabditis elegans, the anaphase-promoting complex/cyclosome and its co-activator FZR-1 (APC/CFZR-1), a ubiquitin ligase, negatively regulates centrosome assembly through SAS-5 degradation. In this study, we report the C. elegans ZYG-1 (Plk4 in humans) as a potential substrate of APC/CFZR-1. Inhibiting APC/CFZR-1 or mutating a ZYG-1 destruction (D)-box leads to elevated ZYG-1 levels at centrosomes, restoring bipolar spindles and embryonic viability to zyg-1 mutants, suggesting that APC/CFZR-1 influences centrosomal ZYG-1 via the D-box motif. We also show the Slimb/βTrCP-binding (SB) motif is critical for ZYG-1 degradation, substantiating a conserved mechanism by which ZYG-1/Plk4 stability is regulated by the SKP1-CUL1-F-box (Slimb/βTrCP)-protein complex (SCFSlimb/βTrCP)-dependent proteolysis via the conserved SB motif in C. elegans. Furthermore, we show that co-mutating ZYG-1 SB and D-box motifs stabilizes ZYG-1 in an additive manner, suggesting that the APC/CFZR-1 and SCFSlimb/βTrCP ubiquitin ligases function cooperatively for timely ZYG-1 destruction in C. elegans embryos where ZYG-1 activity remains at threshold level to ensure normal centrosome number.
Collapse
Affiliation(s)
| | | | | | | | | | - Mi Hye Song
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
| |
Collapse
|
36
|
Badarudeen B, Anand U, Mukhopadhyay S, Manna TK. Ubiquitin signaling in the control of centriole duplication. FEBS J 2021; 289:4830-4849. [PMID: 34115927 DOI: 10.1111/febs.16069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/22/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022]
Abstract
The centrosome plays an essential role in maintaining genetic stability, ciliogenesis and cell polarisation. The core of the centrosome is made up of two centrioles that duplicate precisely once during every cell cycle to generate two centrosomes that are required for bipolar spindle assembly and chromosome segregation. Abundance of centriole proteins at optimal levels and their recruitment to the centrosome are tightly regulated in time and space in order to restrict aberrant duplication of centrioles, a phenomenon that is observed in many cancers. Recent advances have conclusively shown that dedicated ubiquitin ligase-dependent protein degradation machineries are involved in governing centriole duplication. These studies revealed intricate mechanistic insights into how the ubiquitin ligases target different centriole proteins. In certain cases, a specific ubiquitin ligase targets a number of substrate proteins that co-regulate centriole assembly, prompting the possibility that substrate-targeting occurs during formation of the sub-centriolar structures. There are also instances where a specific centriole duplication protein is targeted by several ubiquitin ligases at different stages of the cell cycle, suggesting synchronised actions. Recent evidence also indicated a direct association of E3 ubiquitin ligase with the centrioles, supporting the notion that substrate-targeting occurs in the organelle itself. In this review, we highlight these advances by underlining the mechanisms of how different ubiquitin ligase machineries control centriole duplication and discuss our views on their coordination.
Collapse
Affiliation(s)
- Binshad Badarudeen
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| | - Ushma Anand
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| | - Swarnendu Mukhopadhyay
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Vithura, India
| |
Collapse
|
37
|
Gurkaslar HK, Culfa E, Arslanhan MD, Lince-Faria M, Firat-Karalar EN. CCDC57 Cooperates with Microtubules and Microcephaly Protein CEP63 and Regulates Centriole Duplication and Mitotic Progression. Cell Rep 2021; 31:107630. [PMID: 32402286 DOI: 10.1016/j.celrep.2020.107630] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/08/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
Centrosomes function in key cellular processes ranging from cell division to cellular signaling. Their dysfunction is linked to cancer and developmental disorders. Here, we identify CCDC57 as a pleiotropic regulator of centriole duplication, mitosis, and ciliogenesis. Combining proximity mapping with superresolution imaging, we show that CCDC57 localizes to the proximal end of centrioles and interacts with the microcephaly protein CEP63, centriolar satellite proteins, and microtubules. Loss of CCDC57 causes defects in centriole duplication and results in a failure to localize CEP63 and CEP152 to the centrosome. Additionally, CCDC57 depletion perturbs mitotic progression both in wild-type and centriole-less cells. Importantly, its centrosome-targeting region is required for its interaction with CEP63 and functions during centriole duplication and cilium assembly, whereas the microtubule-targeting region is required for its mitotic functions. Together, our results identify CCDC57 as a critical interface between centrosome and microtubule-mediated cellular processes that are deregulated in microcephaly.
Collapse
Affiliation(s)
- H Kubra Gurkaslar
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey
| | - Efraim Culfa
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey
| | - Melis D Arslanhan
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey
| | - Mariana Lince-Faria
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras 2780-156, Portugal
| | - Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Koç University, Sarıyer, İstanbul 34450, Turkey.
| |
Collapse
|
38
|
Zhang Z, Xing X, Guan P, Song S, You G, Xia C, Liu T. Recent progress in agents targeting polo-like kinases: Promising therapeutic strategies. Eur J Med Chem 2021; 217:113314. [PMID: 33765606 DOI: 10.1016/j.ejmech.2021.113314] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022]
Abstract
Polo-like kinases (PLKs) play important roles in regulating multiple aspects of cell cycle and cell proliferation. In many cancer types, PLK family members are often dysregulated, which can lead to uncontrolled cell proliferation and aberrant cell division and has been shown to associate with poor prognosis of cancers. The key roles of PLK kinases in cancers lead to an enhanced interest in them as promising targets for anticancer drug development. In consideration of PLK inhibitors and some other anticancer agents, such as BRD4, EEF2K and Aurora inhibitors, exert synergy effects in cancer cells, dual-targeting of PLK and other cancer-related targets is regarded as an rational and potent strategy to enhance the effectiveness of single-targeting therapy for cancer treatment. This review introduces the PLK family members at first and then focuses on the recent advances of single-target PLK inhibitors and summarizes the corresponding SARs of them. Moreover, we discuss the synergisms between PLK and other anti-tumor targets, and sum up the current dual-target agents based on them.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| | - Xiaolan Xing
- Yangtze River Pharmaceutical Group Shanghai Haini Pharmaceutical Co., Ltd. Pudong, Shanghai, 201100, PR China
| | - Peng Guan
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, PR China
| | - Shubin Song
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Guirong You
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| | - Chengcai Xia
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| | - Tingting Liu
- Department of Medicinal Chemistry, School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China.
| |
Collapse
|
39
|
Dias Louro MA, Bettencourt-Dias M, Carneiro J. A first-takes-all model of centriole copy number control based on cartwheel elongation. PLoS Comput Biol 2021; 17:e1008359. [PMID: 33970906 PMCID: PMC8136855 DOI: 10.1371/journal.pcbi.1008359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 05/20/2021] [Accepted: 04/06/2021] [Indexed: 11/18/2022] Open
Abstract
How cells control the numbers of subcellular components is a fundamental question in biology. Given that biosynthetic processes are fundamentally stochastic it is utterly puzzling that some structures display no copy number variation within a cell population. Centriole biogenesis, with each centriole being duplicated once and only once per cell cycle, stands out due to its remarkable fidelity. This is a highly controlled process, which depends on low-abundance rate-limiting factors. How can exactly one centriole copy be produced given the variation in the concentration of these key factors? Hitherto, tentative explanations of this control evoked lateral inhibition- or phase separation-like mechanisms emerging from the dynamics of these rate-limiting factors but how strict centriole number is regulated remains unsolved. Here, a novel solution to centriole copy number control is proposed based on the assembly of a centriolar scaffold, the cartwheel. We assume that cartwheel building blocks accumulate around the mother centriole at supercritical concentrations, sufficient to assemble one or more cartwheels. Our key postulate is that once the first cartwheel is formed it continues to elongate by stacking the intermediate building blocks that would otherwise form supernumerary cartwheels. Using stochastic models and simulations, we show that this mechanism may ensure formation of one and only one cartwheel robustly over a wide range of parameter values. By comparison to alternative models, we conclude that the distinctive signatures of this novel mechanism are an increasing assembly time with cartwheel numbers and the translation of stochasticity in building block concentrations into variation in cartwheel numbers or length.
Collapse
Affiliation(s)
| | | | - Jorge Carneiro
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova, Oeiras, Portugal
| |
Collapse
|
40
|
Marcinkowski M, Pilžys T, Garbicz D, Piwowarski J, Mielecki D, Nowaczyk G, Taube M, Gielnik M, Kozak M, Winiewska-Szajewska M, Szołajska E, Dębski J, Maciejewska AM, Przygońska K, Ferenc K, Grzesiuk E, Poznański J. Effect of Posttranslational Modifications on the Structure and Activity of FTO Demethylase. Int J Mol Sci 2021; 22:ijms22094512. [PMID: 33925955 PMCID: PMC8123419 DOI: 10.3390/ijms22094512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
The FTO protein is involved in a wide range of physiological processes, including adipogenesis and osteogenesis. This two-domain protein belongs to the AlkB family of 2-oxoglutarate (2-OG)- and Fe(II)-dependent dioxygenases, displaying N6-methyladenosine (N6-meA) demethylase activity. The aim of the study was to characterize the relationships between the structure and activity of FTO. The effect of cofactors (Fe2+/Mn2+ and 2-OG), Ca2+ that do not bind at the catalytic site, and protein concentration on FTO properties expressed in either E. coli (ECFTO) or baculovirus (BESFTO) system were determined using biophysical methods (DSF, MST, SAXS) and biochemical techniques (size-exclusion chromatography, enzymatic assay). We found that BESFTO carries three phosphoserines (S184, S256, S260), while there were no such modifications in ECFTO. The S256D mutation mimicking the S256 phosphorylation moderately decreased FTO catalytic activity. In the presence of Ca2+, a slight stabilization of the FTO structure was observed, accompanied by a decrease in catalytic activity. Size exclusion chromatography and MST data confirmed the ability of FTO from both expression systems to form homodimers. The MST-determined dissociation constant of the FTO homodimer was consistent with their in vivo formation in human cells. Finally, a low-resolution structure of the FTO homodimer was built based on SAXS data.
Collapse
Affiliation(s)
- Michał Marcinkowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Tomaš Pilžys
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Damian Garbicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Jan Piwowarski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Damian Mielecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Grzegorz Nowaczyk
- NanoBioMedical Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznan, Poland;
| | - Michał Taube
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznanskiego 2, 61-614 Poznan, Poland; (M.T.); (M.G.); (M.K.)
| | - Maciej Gielnik
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznanskiego 2, 61-614 Poznan, Poland; (M.T.); (M.G.); (M.K.)
| | - Maciej Kozak
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, Uniwersytetu Poznanskiego 2, 61-614 Poznan, Poland; (M.T.); (M.G.); (M.K.)
- National Synchrotron Radiation Centre SOLARIS, Jagiellonian University, Czerwone Maki 98, 30-392 Kraków, Poland
| | - Maria Winiewska-Szajewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Ewa Szołajska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Janusz Dębski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Agnieszka M. Maciejewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Kaja Przygońska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
| | - Karolina Ferenc
- Veterinary Research Centre, Department of Large Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797 Warsaw, Poland;
| | - Elżbieta Grzesiuk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
- Correspondence: (E.G.); (J.P.)
| | - Jarosław Poznański
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland; (M.M.); (T.P.); (D.G.); (J.P.); (D.M.); (M.W.-S.); (E.S.); (J.D.); (A.M.M.); (K.P.)
- Correspondence: (E.G.); (J.P.)
| |
Collapse
|
41
|
Garvey DR, Chhabra G, Ndiaye MA, Ahmad N. Role of Polo-Like Kinase 4 (PLK4) in Epithelial Cancers and Recent Progress in its Small Molecule Targeting for Cancer Management. Mol Cancer Ther 2021; 20:632-640. [PMID: 33402398 PMCID: PMC8026525 DOI: 10.1158/1535-7163.mct-20-0741] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/02/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022]
Abstract
The polo-like kinases (PLKs) are a family of serine/threonine kinases traditionally linked to cell-cycle regulation. A structurally unique member of this family, PLK4, has been shown to regulate centriole duplication during the cell cycle via interactions with a variety of centrosomal proteins. Recent findings suggest that PLK4 is overexpressed in various human cancers and associated with poor cancer prognosis. Although several studies have shown that PLK4 inhibition may lead to cancer cell death, the underlying mechanisms are largely unknown. In this review, we discuss the structure, localization, and function of PLK4, along with the functional significance of PLK4 in epithelial cancers and some preliminary work suggesting a role for PLK4 in the key cancer progression process epithelial-mesenchymal transition. We also discuss the potential of PLK4 as a druggable target for anticancer drug development based on critical analysis of the available data of PLK4 inhibitors in preclinical development and clinical trials. Overall, the emerging data suggest that PLK4 plays an essential role in epithelial cancers and should be further explored as a potential biomarker and/or therapeutic target. Continued detailed exploration of available and next-generation PLK4 inhibitors may provide a new dimension for novel cancer therapeutics following successful clinical trials.
Collapse
Affiliation(s)
- Debra R Garvey
- Department of Dermatology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mary A Ndiaye
- Department of Dermatology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin-Madison, Madison, Wisconsin.
- William S. Middleton VA Medical Center, Madison, Wisconsin
| |
Collapse
|
42
|
Li J, Urabe G, Huang Y, Zhang M, Wang B, Marcho L, Shen H, Kent KC, Guo LW. A Role for Polo-Like Kinase 4 in Vascular Fibroblast Cell-Type Transition. JACC Basic Transl Sci 2021; 6:257-283. [PMID: 33778212 PMCID: PMC7987547 DOI: 10.1016/j.jacbts.2020.12.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 11/08/2022]
Abstract
PLK4, previously known as a centriole-associated factor, regulates the transcription factor activity of serum response factor. PLK4 inhibition blocks the profibrogenic cell state transition of vascular fibroblasts. PLK4’s activation and gene expression are regulated by PDGF receptor and epigenetic reader BRD4, respectively. Periadventitial administration of a PLK4 inhibitor mitigates vascular fibrosis.
Polo-like kinase 4 (PLK4) is canonically known for its cytoplasmic function in centriole duplication. Here we show a noncanonical PLK4 function of regulating the transcription factor SRF’s nuclear activity and associated myofibroblast-like cell-type transition. In this context, we have further found that PLK4’s phosphorylation and transcription are respectively regulated by PDGF receptor and epigenetic factor BRD4. Furthermore, in vivo experiments suggest PLK4 inhibition as a potential approach to mitigating vascular fibrosis.
Collapse
Key Words
- AA, PDGF-AA
- BET, bromo/extraterminal domain–containing protein
- BRD4
- BRD4, bromodomain protein 4
- CenB, centrinone-B
- EEL, external elastic lamina
- JQ1, a BET family–selective epigenetic modulator drug
- MRTF-A, myocardin-related transcription factor A
- PDGF receptor
- PDGF, platelet-derived growth factor
- PDGFR, PDGF receptor
- PLK, polo-like kinase
- PLK4
- SRF
- SRF, serum response factor
- fibroblast cell-type transition
- αSMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Go Urabe
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Yitao Huang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Mengxue Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Cellular and Molecular Pathology Graduate Program, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Bowen Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Lynn Marcho
- Davis Heart and Lung Research Institute, Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
| | - Hongtao Shen
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - K Craig Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
43
|
Zhang X, Wei C, Liang H, Han L. Polo-Like Kinase 4's Critical Role in Cancer Development and Strategies for Plk4-Targeted Therapy. Front Oncol 2021; 11:587554. [PMID: 33777739 PMCID: PMC7994899 DOI: 10.3389/fonc.2021.587554] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Polo-like kinases (Plks) are critical regulatory molecules during the cell cycle process. This family has five members: Plk1, 2, 3, 4, and 5. Plk4 has been identified as a master regulator of centriole replication, and its aberrant expression is closely associated with cancer development. In this review, we depict the DNA, mRNA, and protein structure of Plk4, and the regulation of Plk4 at a molecular level. Then we list the downstream targets of Plk4 and the hallmarks of cancer associated with these targets. The role of Plk4 in different cancers is also summarized. Finally, we review the inhibitors that target Plk4 in the hope of discovering effective anticancer drugs. From authors' perspective, Plk4 might represent a valuable tumor biomarker and critical target for cancer diagnosis and therapy.
Collapse
Affiliation(s)
| | | | | | - Lei Han
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
44
|
Niwa T, Akaike Y, Watanabe K, Chibazakura T. Hyperactivation of cyclin A-CDK induces centrosome overduplication and chromosome tetraploidization in mouse cells. Biochem Biophys Res Commun 2021; 549:91-97. [PMID: 33667714 DOI: 10.1016/j.bbrc.2021.02.079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 02/18/2021] [Indexed: 11/17/2022]
Abstract
Mammalian cyclin A-CDK (cyclin-dependent kinase) activity during mitotic exit is regulated by two redundant pathways, cyclin degradation and CDK inhibitors (CKIs). Ectopic expression of a destruction box-truncated (thereby stabilized) mutant of cyclin A in the mouse embryonic fibroblasts nullizygous for three CKIs (p21, p27, and p107) results in constitutive activation ("hyperactivation") of cyclin A-CDK and induces rapid tetraploidization, suggesting loss of the two redundant pathways causes genomic instability. To elucidate the mechanism underlying teraploidization by hyperactive cyclin A-CDK, we first examined if the induction of tetraploidization depends on specific cell cycle stage(s). Arresting the cell cycle at either S phase or M phase blocked the induction of tetraploidization, which was restored by subsequent release from the arrest. These results suggest that both S- and M-phase progressions are necessary for the tetraploidization by hyperactive cyclin A-CDK and that the tetraploidization is not caused by chromosome endoreduplication but by mitotic failure. We also observed that the induction of tetraploidization is associated with excessive duplication of centrosomes, which was suppressed by S-phase but not M-phase block, suggesting that hyperactive cyclin A-CDK promotes centrosome overduplication during S phase. Time-lapse microscopy revealed that hyperactive cyclin A-CDK can lead cells to bypass cell division and enter pseudo-G1 state. These observations implicate that hyperactive cyclin A-CDK causes centrosome overduplication, which leads to mitotic slippage and subsequent tetraploidization.
Collapse
Affiliation(s)
- Tetsuo Niwa
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Yasunori Akaike
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Kaichi Watanabe
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Taku Chibazakura
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan.
| |
Collapse
|
45
|
Raab CA, Raab M, Becker S, Strebhardt K. Non-mitotic functions of polo-like kinases in cancer cells. Biochim Biophys Acta Rev Cancer 2021; 1875:188467. [PMID: 33171265 DOI: 10.1016/j.bbcan.2020.188467] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Inhibitors of mitotic protein kinases are currently being developed as non-neurotoxic alternatives of microtubule-targeting agents (taxanes, vinca alkaloids) which provide a substantial survival benefit for patients afflicted with different types of solid tumors. Among the mitotic kinases, the cyclin-dependent kinases, the Aurora kinases, the kinesin spindle protein and Polo-like kinases (PLKs) have emerged as attractive targets of cancer therapeutics. The functions of mammalian PLK1-5 are traditionally linked to the regulation of the cell cycle and to the stress response. Especially the key role of PLK1 and PLK4 in cellular growth and proliferation, their overexpression in multiple types of human cancer and their druggability, make them appealing targets for cancer therapy. Inhibitors for PLK1 and PLK4 are currently being tested in multiple cancer trials. The clinical success of microtubule-targeting agents is attributed not solely to the induction of a mitotic arrest in cancer cells, but also to non-mitotic effects like targeting intracellular trafficking on microtubules. This raises the question whether new cancer targets like PLK1 and PLK4 regulate critical non-mitotic functions in tumor cells. In this article we summarize the important roles of PLK1-5 for the regulation of non-mitotic signaling. Due to these functions it is conceivable that inhibitors for PLK1 or PLK4 can target interphase cells, which underscores their attractive potential as cancer drug targets. Moreover, we also describe the contribution of the tumor-suppressors PLK2, PLK3 and PLK5 to cancer cell signaling outside of mitosis. These observations highlight the urgent need to develop highly specific ATP-competitive inhibitors for PLK4 and for PLK1 like the 3rd generation PLK-inhibitor Onvansertib to prevent the inhibition of tumor-suppressor PLKs in- and outside of mitosis. The remarkable feature of PLKs to encompass a unique druggable domain, the polo-box-domain (PBD) that can be found only in PLKs offers the opportunity for the development of inhibitors that target PLKs exclusively. Beyond the development of mono-specific ATP-competitive PLK inhibitors, the PBD as drug target will support the design of new drugs that eradicate cancer cells based on the mitotic and non-mitotic function of PLK1 and PLK4.
Collapse
Affiliation(s)
| | - Monika Raab
- Department of Gynecology, Goethe-University, Frankfurt, Germany
| | - Sven Becker
- Department of Gynecology, Goethe-University, Frankfurt, Germany
| | - Klaus Strebhardt
- Department of Gynecology, Goethe-University, Frankfurt, Germany; German Cancer Consortium (DKTK), German Cancer Research Center, Partner Site Frankfurt am Main, Frankfurt, Germany.
| |
Collapse
|
46
|
Ong JY, Bradley MC, Torres JZ. Phospho-regulation of mitotic spindle assembly. Cytoskeleton (Hoboken) 2020; 77:558-578. [PMID: 33280275 PMCID: PMC7898546 DOI: 10.1002/cm.21649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/08/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022]
Abstract
The assembly of the bipolar mitotic spindle requires the careful orchestration of a myriad of enzyme activities like protein posttranslational modifications. Among these, phosphorylation has arisen as the principle mode for spatially and temporally activating the proteins involved in early mitotic spindle assembly processes. Here, we review key kinases, phosphatases, and phosphorylation events that regulate critical aspects of these processes. We highlight key phosphorylation substrates that are important for ensuring the fidelity of centriole duplication, centrosome maturation, and the establishment of the bipolar spindle. We also highlight techniques used to understand kinase-substrate relationships and to study phosphorylation events. We conclude with perspectives on the field of posttranslational modifications in early mitotic spindle assembly.
Collapse
Affiliation(s)
- Joseph Y Ong
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Michelle C Bradley
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA
| | - Jorge Z Torres
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| |
Collapse
|
47
|
Use of the Polo-like kinase 4 (PLK4) inhibitor centrinone to investigate intracellular signalling networks using SILAC-based phosphoproteomics. Biochem J 2020; 477:2451-2475. [PMID: 32501498 PMCID: PMC7338032 DOI: 10.1042/bcj20200309] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/27/2020] [Accepted: 06/05/2020] [Indexed: 12/22/2022]
Abstract
Polo-like kinase 4 (PLK4) is the master regulator of centriole duplication in metazoan organisms. Catalytic activity and protein turnover of PLK4 are tightly coupled in human cells, since changes in PLK4 concentration and catalysis have profound effects on centriole duplication and supernumerary centrosomes, which are associated with aneuploidy and cancer. Recently, PLK4 has been targeted with a variety of small molecule kinase inhibitors exemplified by centrinone, which rapidly induces inhibitory effects on PLK4 and leads to on-target centrosome depletion. Despite this, relatively few PLK4 substrates have been identified unequivocally in human cells, and PLK4 signalling outside centriolar networks remains poorly characterised. We report an unbiased mass spectrometry (MS)-based quantitative analysis of cellular protein phosphorylation in stable PLK4-expressing U2OS human cells exposed to centrinone. PLK4 phosphorylation was itself sensitive to brief exposure to the compound, resulting in PLK4 stabilisation. Analysing asynchronous cell populations, we report hundreds of centrinone-regulated cellular phosphoproteins, including centrosomal and cell cycle proteins and a variety of likely 'non-canonical' substrates. Surprisingly, sequence interrogation of ∼300 significantly down-regulated phosphoproteins reveals an extensive network of centrinone-sensitive [Ser/Thr]Pro phosphorylation sequence motifs, which based on our analysis might be either direct or indirect targets of PLK4. In addition, we confirm that NMYC and PTPN12 are PLK4 substrates, both in vitro and in human cells. Our findings suggest that PLK4 catalytic output directly controls the phosphorylation of a diverse set of cellular proteins, including Pro-directed targets that are likely to be important in PLK4-mediated cell signalling.
Collapse
|
48
|
Yamamoto S, Kitagawa D. Emerging insights into symmetry breaking in centriole duplication: updated view on centriole duplication theory. Curr Opin Struct Biol 2020; 66:8-14. [PMID: 32956908 DOI: 10.1016/j.sbi.2020.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 11/15/2022]
Abstract
Centriole duplication occurs once per cell cycle. Since only a single daughter centriole is assembled adjacent to each mother centriole, symmetry around the mother centriole must be broken in the process of centriole duplication. Recent studies have established that Plk4, a master kinase for centriole duplication, can self-assemble into condensates, and have suggested that this Plk4 self-assembly is the key to symmetry breaking. Here, we present the current hypotheses for how Plk4 could break symmetry around the mother centriole via autonomous regulation. After this initial symmetry-breaking process, the ring-to-dot conversion of Plk4 around the mother centriole completes the selection of the site for procentriole formation. We also discuss how this dynamic transition contributes to the strict regulation of centriole duplication.
Collapse
Affiliation(s)
- Shohei Yamamoto
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Tokyo 113-0033, Japan
| | - Daiju Kitagawa
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Tokyo 113-0033, Japan.
| |
Collapse
|
49
|
Panda P, Kovacs L, Dzhindzhev N, Fatalska A, Persico V, Geymonat M, Riparbelli MG, Callaini G, Glover DM. Tissue specific requirement of Drosophila Rcd4 for centriole duplication and ciliogenesis. J Cell Biol 2020; 219:151861. [PMID: 32543652 PMCID: PMC7401805 DOI: 10.1083/jcb.201912154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/01/2020] [Accepted: 05/13/2020] [Indexed: 12/22/2022] Open
Abstract
Rcd4 is a poorly characterized Drosophila centriole component whose mammalian counterpart, PPP1R35, is suggested to function in centriole elongation and conversion to centrosomes. Here, we show that rcd4 mutants exhibit fewer centrioles, aberrant mitoses, and reduced basal bodies in sensory organs. Rcd4 interacts with the C-terminal part of Ana3, which loads onto the procentriole during interphase, ahead of Rcd4 and before mitosis. Accordingly, depletion of Ana3 prevents Rcd4 recruitment but not vice versa. We find that neither Ana3 nor Rcd4 participates directly in the mitotic conversion of centrioles to centrosomes, but both are required to load Ana1, which is essential for such conversion. Whereas ana3 mutants are male sterile, reflecting a requirement for Ana3 for centriole development in the male germ line, rcd4 mutants are fertile and have male germ line centrioles of normal length. Thus, Rcd4 is essential in somatic cells but is not absolutely required in spermatogenesis, indicating tissue-specific roles in centriole and basal body formation.
Collapse
Affiliation(s)
- Pallavi Panda
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Levente Kovacs
- Department of Genetics, University of Cambridge, Cambridge, UK
| | | | - Agnieszka Fatalska
- Department of Genetics, University of Cambridge, Cambridge, UK.,Institute of Biochemistry and Biophysics, Polish Academy of Science, Warsaw, Poland.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Veronica Persico
- Department of Genetics, University of Cambridge, Cambridge, UK.,Department of Life Sciences, University of Siena, Siena, Italy
| | - Marco Geymonat
- Department of Genetics, University of Cambridge, Cambridge, UK
| | | | | | - David M Glover
- Department of Genetics, University of Cambridge, Cambridge, UK.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| |
Collapse
|
50
|
Kazazian K, Haffani Y, Ng D, Lee CMM, Johnston W, Kim M, Xu R, Pacholzyk K, Zih FSW, Tan J, Smrke A, Pollett A, Wu HST, Swallow CJ. FAM46C/TENT5C functions as a tumor suppressor through inhibition of Plk4 activity. Commun Biol 2020; 3:448. [PMID: 32807875 PMCID: PMC7431843 DOI: 10.1038/s42003-020-01161-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
Polo like kinase 4 (Plk4) is a tightly regulated serine threonine kinase that governs centriole duplication. Increased Plk4 expression, which is a feature of many common human cancers, causes centriole overduplication, mitotic irregularities, and chromosomal instability. Plk4 can also promote cancer invasion and metastasis through regulation of the actin cytoskeleton. Herein we demonstrate physical interaction of Plk4 with FAM46C/TENT5C, a conserved protein of unknown function until recently. FAM46C localizes to centrioles, inhibits Plk4 kinase activity, and suppresses Plk4-induced centriole duplication. Interference with Plk4 function by FAM46C was independent of the latter's nucleotidyl transferase activity. In addition, FAM46C restrained cancer cell invasion and suppressed MDA MB-435 cancer growth in a xenograft model, opposing the effect of Plk4. We demonstrate loss of FAM46C in patient-derived colorectal cancer tumor tissue that becomes more profound with advanced clinical stage. These results implicate FAM46C as a tumor suppressor that acts by inhibiting Plk4 activity.
Collapse
Affiliation(s)
- Karineh Kazazian
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada.,Department of Surgical Oncology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Yosr Haffani
- Laboratory of Physiopathology, Alimentation and Biomolecules LR17ES03, Higher Institute of Biotechnology, Sidi Thabet, University of Manouba, Ariana, 2020, Tunisia
| | - Deanna Ng
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Chae Min Michelle Lee
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Wendy Johnston
- Department of Radiation Oncology, University of Toronto, Toronto, ON, M5T 1P5, Canada
| | - Minji Kim
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Roland Xu
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Karina Pacholzyk
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Francis Si-Wah Zih
- Department of Surgical Oncology, University of Toronto, Toronto, ON, M5G 2M9, Canada
| | - Julie Tan
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Alannah Smrke
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Aaron Pollett
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Hannah Sun-Tsi Wu
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Carol Jane Swallow
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada. .,Department of Surgical Oncology, University of Toronto, Toronto, ON, M5G 2M9, Canada.
| |
Collapse
|