1
|
Kono Y, Shimi T. Crosstalk between mitotic reassembly and repair of the nuclear envelope. Nucleus 2024; 15:2352203. [PMID: 38780365 PMCID: PMC11123513 DOI: 10.1080/19491034.2024.2352203] [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: 09/01/2023] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
In eukaryotic cells, the nuclear envelope (NE) is a membrane partition between the nucleus and the cytoplasm to compartmentalize nuclear contents. It plays an important role in facilitating nuclear functions including transcription, DNA replication and repair. In mammalian cells, the NE breaks down and then reforms during cell division, and in interphase it is restored shortly after the NE rupture induced by mechanical force. In this way, the partitioning effect is regulated through dynamic processes throughout the cell cycle. A failure in rebuilding the NE structure triggers the mixing of nuclear and cytoplasmic contents, leading to catastrophic consequences for the nuclear functions. Whereas the precise details of molecular mechanisms for NE reformation during cell division and NE restoration in interphase are still being investigated, here, we mostly focus on mammalian cells to describe key aspects that have been identified and to discuss the crosstalk between them.
Collapse
Affiliation(s)
- Yohei Kono
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Takeshi Shimi
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| |
Collapse
|
2
|
Jarusintanakorn S, Mastrobattista E, Yamabhai M. Ectoine enhances recombinant antibody production in Chinese hamster ovary cells by promoting cell cycle arrest. N Biotechnol 2024; 83:56-65. [PMID: 38945523 DOI: 10.1016/j.nbt.2024.06.006] [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: 02/07/2024] [Revised: 06/08/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Chinese hamster ovary (CHO) cells represent the most preferential host cell system for therapeutic monoclonal antibody (mAb) production. Enhancing mAb production in CHO cells can be achieved by adding chemical compounds that regulate the cell cycle and cell survival pathways. This study investigated the impact of ectoine supplementation on mAb production in CHO cells. The results showed that adding ectoine at a concentration of 100 mM on the 3rd day of cultivation improved mAb production by improving cell viability and extending the culture duration. RNA sequencing analysis revealed differentially expressed genes associated with cell cycle regulation, cell proliferation, and cellular homeostasis, in particular promotion of cell cycle arrest, which was then confirmed by flow cytometry analysis. Ectoine-treated CHO cells exhibited an increase in the number of cells in the G0/G1 phase. In addition, the cell diameter was also increased. These findings support the hypothesis that ectoine enhances mAb production in CHO cells through mechanisms involving cell cycle arrest and cellular homeostasis. Overall, this study highlights the potential of ectoine as a promising supplementation strategy to enhance mAb production not only in CHO cells but also in other cell lines.
Collapse
Affiliation(s)
- Salinthip Jarusintanakorn
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, 447, Sri-Ayuthaya Road, Rachathevi, Bangkok 10400, Thailand
| | - Enrico Mastrobattista
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands.
| | - Montarop Yamabhai
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
3
|
Lemaire S, Ferreira M, Claes Z, Derua R, Lake M, Van der Hoeven G, Withof F, Cao X, Greiner EC, Kettenbach AN, Van Eynde A, Bollen M. PPP1R2 stimulates protein phosphatase-1 through stabilisation of dynamic subunit interactions. Nat Commun 2024; 15:9822. [PMID: 39537675 PMCID: PMC11561318 DOI: 10.1038/s41467-024-54256-4] [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: 05/17/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Protein Ser/Thr phosphatase PP1 is always associated with one or two regulatory subunits or RIPPOs. One of the earliest evolved RIPPOs is PPP1R2, also known as Inhibitor-2. Since its discovery nearly 5 decades ago, PPP1R2 has been variously described as an inhibitor, activator or (metal) chaperone of PP1, but it is still unknown how PPP1R2 affects the function of PP1 in intact cells. Here, using specific research tools, we demonstrate that PPP1R2 stabilises a subgroup of PP1 holoenzymes, exemplified by PP1:RepoMan, thereby promoting the dephosphorylation of their substrates. Mechanistically, the recruitment of PPP1R2 disrupts an inhibitory, fuzzy interaction between the C-terminal tail and catalytic domain of PP1, and generates an additional C-terminal RepoMan-interaction site. The resulting holoenzyme is further stabilized by a direct PPP1R2:RepoMan interaction, which renders it refractory to competitive disruption by RIPPOs that do not interact with PPP1R2. Our data demonstrate that PPP1R2 modulates the function of PP1 by altering the balance between holoenzymes through stabilisation of specific subunit interactions.
Collapse
Affiliation(s)
- Sarah Lemaire
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Mónica Ferreira
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Zander Claes
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation & Proteomics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Madryn Lake
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Gerd Van der Hoeven
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Fabienne Withof
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Xinyu Cao
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Elora C Greiner
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | - Aleyde Van Eynde
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Mathieu Bollen
- Laboratory of Biosignaling & Therapeutics, KU Leuven Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Xiang S, Craig KC, Luo X, Welch DL, Ferreira RB, Lawrence HR, Lawrence NJ, Reed DR, Alexandrow MG. Identification of ATP-Competitive Human CMG Helicase Inhibitors for Cancer Intervention that Disrupt CMG-Replisome Function. Mol Cancer Ther 2024; 23:1568-1585. [PMID: 38982858 PMCID: PMC11532780 DOI: 10.1158/1535-7163.mct-23-0904] [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: 12/19/2023] [Revised: 03/25/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
The human CMG helicase (Cdc45-MCM-GINS) is a novel target for anticancer therapy. Tumor-specific weaknesses in the CMG are caused by oncogene-driven changes that adversely affect CMG function, and CMG activity is required for recovery from replicative stresses such as chemotherapy. Herein, we developed an orthogonal biochemical screening approach and identified CMG inhibitors (CMGi) that inhibit ATPase and helicase activities in an ATP-competitive manner at low micromolar concentrations. Structure-activity information, in silico docking, and testing with synthetic chemical compounds indicate that CMGi require specific chemical elements and occupy ATP-binding sites and channels within minichromosome maintenance (MCM) subunits leading to the ATP clefts, which are likely used for ATP/ADP ingress or egress. CMGi are therefore MCM complex inhibitors (MCMi). Biologic testing shows that CMGi/MCMi inhibit cell growth and DNA replication using multiple molecular mechanisms distinct from other chemotherapy agents. CMGi/MCMi block helicase assembly steps that require ATP binding/hydrolysis by the MCM complex, specifically MCM ring assembly on DNA and GINS recruitment to DNA-loaded MCM hexamers. During the S-phase, inhibition of MCM ATP binding/hydrolysis by CMGi/MCMi causes a "reverse allosteric" dissociation of Cdc45/GINS from the CMG that destabilizes replisome components Ctf4, Mcm10, and DNA polymerase-α, -δ, and -ε, resulting in DNA damage. CMGi/MCMi display selective toxicity toward multiple solid tumor cell types with K-Ras mutations, targeting the CMG and inducing DNA damage, Parp cleavage, and loss of viability. This new class of CMGi/MCMi provides a basis for small chemical development of CMG helicase-targeted anticancer compounds with distinct mechanisms of action.
Collapse
Affiliation(s)
- Shengyan Xiang
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Molecular Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kendall C. Craig
- Molecular Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Drug Discovery Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xingju Luo
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Molecular Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Darcy L. Welch
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Renan B. Ferreira
- Drug Discovery Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Chemical Biology Core Facility, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Harshani R. Lawrence
- Drug Discovery Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Chemical Biology Core Facility, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nicholas J. Lawrence
- Drug Discovery Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Damon R. Reed
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mark G. Alexandrow
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Molecular Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
5
|
Xiang S, Luo X, Welch D, Reed DR, Alexandrow MG. Identification of Selective ATP-Competitive CMG Helicase Inhibitors for Cancer Intervention that Disrupt CMG-Replisome Function. RESEARCH SQUARE 2023:rs.3.rs-3182731. [PMID: 37609279 PMCID: PMC10441460 DOI: 10.21203/rs.3.rs-3182731/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The human CMG helicase (Cdc45-MCM-GINS) is a novel target for anti-cancer therapy due to tumor-specific weaknesses in CMG function induced by oncogenic changes and the need for CMG function during recovery from replicative stresses such as chemotherapy. Here, we developed an orthogonal biochemical screening approach and identified selective CMG inhibitors (CMGi) that inhibit ATPase and helicase activities in an ATP-competitive manner at low micromolar concentrations. Structure-activity information and in silico docking indicate that CMGi occupy ATP binding sites and channels within MCM subunits leading to the ATP clefts, which are likely used for ATP/ADP ingress or egress. CMGi inhibit cell growth and DNA replication using multiple molecular mechanisms. CMGi block helicase assembly steps that require ATP binding/hydrolysis by the MCM complex, specifically MCM ring assembly on DNA and GINS recruitment to DNA-loaded MCM hexamers. During S-phase, inhibition of MCM ATP binding/hydrolysis by CMGi causes a 'reverse allosteric' dissociation of Cdc45/GINS from the CMG that destabilizes the replisome and disrupts interactions with Ctf4, Mcm10, and DNA polymerase-α, -δ, -ε, resulting in DNA damage. These novel CMGi are selectively toxic toward tumor cells and define a new class of CMG helicase-targeted anti-cancer compounds with distinct mechanisms of action.
Collapse
Affiliation(s)
- Shengyan Xiang
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
- Molecular Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Xingju Luo
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
- Molecular Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Darcy Welch
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Damon R. Reed
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
- Department of Individualized Cancer Management, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Mark G. Alexandrow
- Cancer Biology and Evolution Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
- Molecular Oncology Department, Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| |
Collapse
|
6
|
Camlin NJ, Venkatachalam I, Evans JP. Oscillations in PP1 activity are essential for accurate progression through mammalian oocyte meiosis. Cell Cycle 2023; 22:1614-1636. [PMID: 37340734 PMCID: PMC10361142 DOI: 10.1080/15384101.2023.2225924] [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: 08/25/2022] [Revised: 04/17/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
Tightly controlled fluctuations in kinase and phosphatase activity play important roles in regulating M-phase transitions. Protein Phosphatase 1 (PP1) is one of these phosphatases, with oscillations in PP1 activity driving mitotic M-phase. Evidence from a variety of experimental systems also points to roles in meiosis. Here, we report that PP1 is important for M-phase transitions through mouse oocyte meiosis. We employed a unique small-molecule approach to inhibit or activate PP1 at distinct phases of mouse oocyte meiosis. These studies show that temporal control of PP1 activity is essential for the G2/M transition, metaphase I/anaphase I transition, and the formation of a normal metaphase II oocyte. Our data also reveal that inappropriate activation of PP1 is more deleterious at the G2/M transition than at prometaphase I-to-metaphase I, and that an active pool of PP1 during prometaphase is vital for metaphase I/anaphase I transition and metaphase II chromosome alignment. Taken together, these results establish that loss of oscillations in PP1 activity causes a range of severe meiotic defects, pointing to essential roles for PP1 in female fertility, and more broadly, M-phase regulation.
Collapse
Affiliation(s)
- Nicole J. Camlin
- Department of Biological Sciences, Purdue University, West Lafayette, INUnited States
| | - Ilakkiya Venkatachalam
- Department of Biological Sciences, Purdue University, West Lafayette, INUnited States
- Department of Human Genetics, University of Michigan, Ann Arbor, MIUnited States
| | - Janice P. Evans
- Department of Biological Sciences, Purdue University, West Lafayette, INUnited States
| |
Collapse
|
7
|
Alam SK, Wang L, Zhu Z, Hoeppner LH. IKKα promotes lung adenocarcinoma growth through ERK signaling activation via DARPP-32-mediated inhibition of PP1 activity. NPJ Precis Oncol 2023; 7:33. [PMID: 36966223 PMCID: PMC10039943 DOI: 10.1038/s41698-023-00370-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/08/2023] [Indexed: 03/27/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 80-85% cases of lung cancer cases. Diagnosis at advanced stages is common, after which therapy-refractory disease progression frequently occurs. Therefore, a better understanding of the molecular mechanisms that control NSCLC progression is necessary to develop new therapies. Overexpression of IκB kinase α (IKKα) in NSCLC correlates with poor patient survival. IKKα is an NF-κB-activating kinase that is important in cell survival and differentiation, but its regulation of oncogenic signaling is not well understood. We recently demonstrated that IKKα promotes NSCLC cell migration by physically interacting with dopamine- and cyclic AMP-regulated phosphoprotein, Mr 32000 (DARPP-32), and its truncated splice variant, t-DARPP. Here, we show that IKKα phosphorylates DARPP-32 at threonine 34, resulting in DARPP-32-mediated inhibition of protein phosphatase 1 (PP1), subsequent inhibition of PP1-mediated dephosphorylation of ERK, and activation of ERK signaling to promote lung oncogenesis. Correspondingly, IKKα ablation in human lung adenocarcinoma cells reduced their anchorage-independent growth in soft agar. Mice challenged with IKKα-ablated HCC827 cells exhibited less lung tumor growth than mice orthotopically administered control HCC827 cells. Our findings suggest that IKKα drives NSCLC growth through the activation of ERK signaling via DARPP-32-mediated inhibition of PP1 activity.
Collapse
Affiliation(s)
- Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
| | - Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Zhu Zhu
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
8
|
St. Louis BM, Quagliato SM, Lee PC. Bacterial effector kinases and strategies to identify their target host substrates. Front Microbiol 2023; 14:1113021. [PMID: 36846793 PMCID: PMC9950578 DOI: 10.3389/fmicb.2023.1113021] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/25/2023] [Indexed: 02/12/2023] Open
Abstract
Post-translational modifications (PTMs) are critical in regulating protein function by altering chemical characteristics of proteins. Phosphorylation is an integral PTM, catalyzed by kinases and reversibly removed by phosphatases, that modulates many cellular processes in response to stimuli in all living organisms. Consequently, bacterial pathogens have evolved to secrete effectors capable of manipulating host phosphorylation pathways as a common infection strategy. Given the importance of protein phosphorylation in infection, recent advances in sequence and structural homology search have significantly expanded the discovery of a multitude of bacterial effectors with kinase activity in pathogenic bacteria. Although challenges exist due to complexity of phosphorylation networks in host cells and transient interactions between kinases and substrates, approaches are continuously being developed and applied to identify bacterial effector kinases and their host substrates. In this review, we illustrate the importance of exploiting phosphorylation in host cells by bacterial pathogens via the action of effector kinases and how these effector kinases contribute to virulence through the manipulation of diverse host signaling pathways. We also highlight recent developments in the identification of bacterial effector kinases and a variety of techniques to characterize kinase-substrate interactions in host cells. Identification of host substrates provides new insights for regulation of host signaling during microbial infection and may serve as foundation for developing interventions to treat infection by blocking the activity of secreted effector kinases.
Collapse
Affiliation(s)
- Brendyn M. St. Louis
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI, United States
| | - Sydney M. Quagliato
- Department of Biological Sciences, College of Liberal Arts and Sciences, Wayne State University, Detroit, MI, United States
| | | |
Collapse
|
9
|
Panda M, Kalita E, Rao A, Prajapati VK. Mechanism of cell cycle regulation and cell proliferation during human viral infection. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:497-525. [PMID: 37061340 DOI: 10.1016/bs.apcsb.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Over the history of the coevolution of Host viral interaction, viruses have customized the host cellular machinery into their use for viral genome replication, causing effective infection and ultimately aiming for survival. They do so by inducing subversions to the host cellular pathways like cell cycle via dysregulation of important cell cycle checkpoints by viral encoded proteins, arresting the cell cycle machinery, blocking cytokinesis as well as targeting subnuclear bodies, thus ultimately disorienting the cell proliferation. Both DNA and RNA viruses have been active participants in such manipulation resulting in serious outcomes of cancer. They achieve this by employing different mechanisms-Protein-protein interaction, protein-phosphorylation, degradation, redistribution, viral homolog, and viral regulation of APC at different stages of cell cycle events. Several DNA viruses cause the quiescent staged cells to undergo cell cycle which increases nucleotide pools logistically significantly persuading viral replication whereas few other viruses arrest a particular stage of cell cycle. This allows the latter group to sustain the infection which allows them to escape host immune response and support viral multiplication. Mechanical study of signaling such viral mediated pathways could give insight into understanding the etiology of tumorigenesis and progression. Overall this chapter highlights the possible strategies employed by DNA/RNA viral families which impact the normal cell cycle but facilitate viral infected cell replication. Such information could contribute to comprehending viral infection-associated disorders to further depth.
Collapse
Affiliation(s)
- Mamta Panda
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan, India
| | - Elora Kalita
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan, India
| | - Abhishek Rao
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandarsindri, Kishangarh, Ajmer, Rajasthan, India; Department of Biochemistry, School of Biological Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
10
|
Compartmentalized control of Cdk1 drives mitotic spindle assembly. Cell Rep 2022; 38:110305. [DOI: 10.1016/j.celrep.2022.110305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/14/2021] [Accepted: 01/06/2022] [Indexed: 12/28/2022] Open
|
11
|
Swartz SZ, Nguyen HT, McEwan BC, Adamo ME, Cheeseman IM, Kettenbach AN. Selective dephosphorylation by PP2A-B55 directs the meiosis I-meiosis II transition in oocytes. eLife 2021; 10:70588. [PMID: 34342579 PMCID: PMC8370769 DOI: 10.7554/elife.70588] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Meiosis is a specialized cell cycle that requires sequential changes to the cell division machinery to facilitate changing functions. To define the mechanisms that enable the oocyte-to-embryo transition, we performed time-course proteomics in synchronized sea star oocytes from prophase I through the first embryonic cleavage. Although we found that protein levels were broadly stable, our analysis reveals that dynamic waves of phosphorylation underlie each meiotic stage. We found that the phosphatase PP2A-B55 is reactivated at the meiosis I/meiosis II (MI/MII) transition, resulting in the preferential dephosphorylation of threonine residues. Selective dephosphorylation is critical for directing the MI/MII transition as altering PP2A-B55 substrate preferences disrupts key cell cycle events after MI. In addition, threonine to serine substitution of a conserved phosphorylation site in the substrate INCENP prevents its relocalization at anaphase I. Thus, through its inherent phospho-threonine preference, PP2A-B55 imposes specific phosphoregulated behaviors that distinguish the two meiotic divisions.
Collapse
Affiliation(s)
- S Zachary Swartz
- Whitehead Institute for Biomedical Research, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Hieu T Nguyen
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, United States
| | - Brennan C McEwan
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, United States
| | - Mark E Adamo
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, United States
| | - Iain M Cheeseman
- Whitehead Institute for Biomedical Research, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, United States.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, United States
| |
Collapse
|
12
|
Protein phosphatase 1 regulates atypical mitotic and meiotic division in Plasmodium sexual stages. Commun Biol 2021; 4:760. [PMID: 34145386 PMCID: PMC8213788 DOI: 10.1038/s42003-021-02273-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
PP1 is a conserved eukaryotic serine/threonine phosphatase that regulates many aspects of mitosis and meiosis, often working in concert with other phosphatases, such as CDC14 and CDC25. The proliferative stages of the malaria parasite life cycle include sexual development within the mosquito vector, with male gamete formation characterized by an atypical rapid mitosis, consisting of three rounds of DNA synthesis, successive spindle formation with clustered kinetochores, and a meiotic stage during zygote to ookinete development following fertilization. It is unclear how PP1 is involved in these unusual processes. Using real-time live-cell and ultrastructural imaging, conditional gene knockdown, RNA-seq and proteomic approaches, we show that Plasmodium PP1 is implicated in both mitotic exit and, potentially, establishing cell polarity during zygote development in the mosquito midgut, suggesting that small molecule inhibitors of PP1 should be explored for blocking parasite transmission.
Collapse
|
13
|
Huang H, Lee MH, Liu K, Dong Z, Ryoo Z, Kim MO. PBK/TOPK: An Effective Drug Target with Diverse Therapeutic Potential. Cancers (Basel) 2021; 13:cancers13092232. [PMID: 34066486 PMCID: PMC8124186 DOI: 10.3390/cancers13092232] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Cancer is a major public health problem worldwide, and addressing its morbidity, mortality, and prevalence is the first step towards appropriate control measures. Over the past several decades, many pharmacologists have worked to identify anti-cancer targets and drug development strategies. Within this timeframe, many natural compounds have been developed to inhibit cancer growth by targeting kinases, such as AKT, AURKA, and TOPK. Kinase assays and computer modeling are considered to be effective and powerful tools for target screening, as they can predict physical interactions between small molecules and their bio-molecular targets. In the present review, we summarize the inhibitors and compounds that target TOPK and describe its role in cancer progression. The extensive body of research that has investigated the contribution of TOPK to cancer suggests that it may be a promising target for cancer therapy. Abstract T-lymphokine-activated killer cell-originated protein kinase (TOPK, also known as PDZ-binding kinase or PBK) plays a crucial role in cell cycle regulation and mitotic progression. Abnormal overexpression or activation of TOPK has been observed in many cancers, including colorectal cancer, triple-negative breast cancer, and melanoma, and it is associated with increased development, dissemination, and poor clinical outcomes and prognosis in cancer. Moreover, TOPK phosphorylates p38, JNK, ERK, and AKT, which are involved in many cellular functions, and participates in the activation of multiple signaling pathways related to MAPK, PI3K/PTEN/AKT, and NOTCH1; thus, the direct or indirect interactions of TOPK make it a highly attractive yet elusive target for cancer therapy. Small molecule inhibitors targeting TOPK have shown great therapeutic potential in the treatment of cancer both in vitro and in vivo, even in combination with chemotherapy or radiotherapy. Therefore, targeting TOPK could be an important approach for cancer prevention and therapy. Thus, the purpose of the present review was to consider and analyze the role of TOPK as a drug target in cancer therapy and describe the recent findings related to its role in tumor development. Moreover, this review provides an overview of the current progress in the discovery and development of TOPK inhibitors, considering future clinical applications.
Collapse
Affiliation(s)
- Hai Huang
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea;
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju, Jeollanamdo 58245, Korea;
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
- Department of Pathophysiology, School of Basic Medical Sciences, The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou 450001, China
| | - Zigang Dong
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
- Department of Pathophysiology, School of Basic Medical Sciences, The Academy of Medical Science, College of Medical, Zhengzhou University, Zhengzhou 450001, China
| | - Zeayoung Ryoo
- School of Life Science, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (Z.R.); (M.O.K.); Tel.: +82-54-530-1234 (M.O.K.)
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, ITRD, Kyungpook National University, Sangju 37224, Korea;
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450008, China; (K.L.); (Z.D.)
- Correspondence: (Z.R.); (M.O.K.); Tel.: +82-54-530-1234 (M.O.K.)
| |
Collapse
|
14
|
Vagnarelli P. Back to the new beginning: Mitotic exit in space and time. Semin Cell Dev Biol 2021; 117:140-148. [PMID: 33810980 DOI: 10.1016/j.semcdb.2021.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 11/16/2022]
Abstract
The ultimate goal of cell division is to generate two identical daughter cells that resemble the mother cell from which they derived. Once all the proper attachments to the spindle have occurred, the chromosomes have aligned at the metaphase plate and the spindle assembly checkpoint (a surveillance mechanism that halts cells form progressing in the cell cycle in case of spindle - microtubule attachment errors) has been satisfied, mitotic exit will occur. Mitotic exit has the purpose of completing the separation of the genomic material but also to rebuild the cellular structures necessary for the new cell cycle. This stage of mitosis received little attention until a decade ago, therefore our knowledge is much patchier than the molecular details we now have for the early stages of mitosis. However, it is emerging that mitotic exit is not just the simple reverse of mitotic entry and it is highly regulated in space and time. In this review I will discuss the main advances in the field that provided us with a better understanding on the key role of protein phosphorylation/de-phosphorylation in this transition together with the concept of their spatial regulation. As this field is much younger, I will highlight general consensus, contrasting views together with the outstanding questions awaiting for answers.
Collapse
Affiliation(s)
- Paola Vagnarelli
- College of Medicine, Health and Life Science, Centre for Genomic Engineering and Maintenance (CenGEM), Brunel University London, Uxbridge UB8 3PH, UK.
| |
Collapse
|
15
|
MARK2 phosphorylates eIF2α in response to proteotoxic stress. PLoS Biol 2021; 19:e3001096. [PMID: 33705388 PMCID: PMC7951919 DOI: 10.1371/journal.pbio.3001096] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
The regulation of protein synthesis is essential for maintaining cellular homeostasis, especially during stress responses, and its dysregulation could underlie the development of human diseases. The critical step during translation regulation is the phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α). Here we report the identification of a direct kinase of eIF2α, microtubule affinity-regulating kinase 2 (MARK2), which phosphorylates eIF2α in response to proteotoxic stress. The activity of MARK2 was confirmed in the cells lacking the 4 previously known eIF2α kinases. MARK2 itself was found to be a substrate of protein kinase C delta (PKCδ), which serves as a sensor for protein misfolding stress through a dynamic interaction with heat shock protein 90 (HSP90). Both MARK2 and PKCδ are activated via phosphorylation in proteotoxicity-associated neurodegenerative mouse models and in human patients with amyotrophic lateral sclerosis (ALS). These results reveal a PKCδ-MARK2-eIF2α cascade that may play a critical role in cellular proteotoxic stress responses and human diseases. The regulation of protein translation is vital for cellular stress responses and human diseases. This study identifies a new pathway that regulates the key step of translation initiation, with MARK2 directly phosphorylating eIF2α and acting downstream of PKCδ. This pathway is activated in conditions of cellular stress and in proteotoxicity-associated neurodegeneration.
Collapse
|
16
|
Kamenz J, Gelens L, Ferrell JE. Bistable, Biphasic Regulation of PP2A-B55 Accounts for the Dynamics of Mitotic Substrate Phosphorylation. Curr Biol 2020; 31:794-808.e6. [PMID: 33357450 PMCID: PMC7904671 DOI: 10.1016/j.cub.2020.11.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/20/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
Abstract
The phosphorylation of mitotic proteins is bistable, which contributes to the decisiveness of the transitions into and out of M phase. The bistability in substrate phosphorylation has been attributed to bistability in the activation of the cyclin-dependent kinase Cdk1. However, more recently it has been suggested that bistability also arises from positive feedback in the regulation of the Cdk1-counteracting phosphatase PP2A-B55. Here, we demonstrate biochemically using Xenopus laevis egg extracts that the Cdk1-counter-acting phosphatase PP2A-B55 functions as a bistable switch, even when the bistability of Cdk1 activation is suppressed. In addition, Cdk1 regulates PP2A-B55 in a biphasic manner; low concentrations of Cdk1 activate PP2A-B55 and high concentrations inactivate it. As a consequence of this incoherent feedforward regulation, PP2A-B55 activity rises concurrently with Cdk1 activity during interphase and suppresses substrate phosphorylation. PP2A-B55 activity is then sharply downregulated at the onset of mitosis. During mitotic exit, Cdk1 activity initially falls with no obvious change in substrate phosphorylation; dephosphorylation then commences once PP2A-B55 spikes in activity. These findings suggest that changes in Cdk1 activity are permissive for mitotic entry and exit but that the changes in PP2A-B55 activity are the ultimate trigger. Mitotic transitions are accompanied by drastic changes in the phosphorylation state of proteins. Kamenz et al. demonstrate biochemically that the major mitotic phosphatase PP2A-B55 is regulated by incoherent feedforward and double-negative feedback loops to promote rapid and switch-like mitotic entry and exit.
Collapse
Affiliation(s)
- Julia Kamenz
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA.
| | - Lendert Gelens
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA; Laboratory of Dynamics in Biological Systems, KU Leuven, Department of Cellular and Molecular Medicine, University of Leuven, B-3000 Leuven, Belgium
| | - James E Ferrell
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305-5174, USA; Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305-5307, USA.
| |
Collapse
|
17
|
Hume S, Dianov GL, Ramadan K. A unified model for the G1/S cell cycle transition. Nucleic Acids Res 2020; 48:12483-12501. [PMID: 33166394 PMCID: PMC7736809 DOI: 10.1093/nar/gkaa1002] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 01/01/2023] Open
Abstract
Efficient S phase entry is essential for development, tissue repair, and immune defences. However, hyperactive or expedited S phase entry causes replication stress, DNA damage and oncogenesis, highlighting the need for strict regulation. Recent paradigm shifts and conflicting reports demonstrate the requirement for a discussion of the G1/S transition literature. Here, we review the recent studies, and propose a unified model for the S phase entry decision. In this model, competition between mitogen and DNA damage signalling over the course of the mother cell cycle constitutes the predominant control mechanism for S phase entry of daughter cells. Mitogens and DNA damage have distinct sensing periods, giving rise to three Commitment Points for S phase entry (CP1-3). S phase entry is mitogen-independent in the daughter G1 phase, but remains sensitive to DNA damage, such as single strand breaks, the most frequently-occurring lesions that uniquely threaten DNA replication. To control CP1-3, dedicated hubs integrate the antagonistic mitogenic and DNA damage signals, regulating the stoichiometric cyclin: CDK inhibitor ratio for ultrasensitive control of CDK4/6 and CDK2. This unified model for the G1/S cell cycle transition combines the findings of decades of study, and provides an updated foundation for cell cycle research.
Collapse
Affiliation(s)
- Samuel Hume
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Grigory L Dianov
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Lavrentieva 10, 630090 Novosibirsk, Russian Federation
- Novosibirsk State University, 630090 Novosibirsk, Russian Federation
| | - Kristijan Ramadan
- Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
18
|
Yin Z, Guo B, Ma S, Sun Y, Mi Z, Zheng Z. DReSS: a method to quantitatively describe the influence of structural perturbations on state spaces of genetic regulatory networks. Brief Bioinform 2020; 22:6032613. [PMID: 33313791 DOI: 10.1093/bib/bbaa315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/23/2020] [Accepted: 10/16/2020] [Indexed: 11/14/2022] Open
Abstract
Structures of genetic regulatory networks are not fixed. These structural perturbations can cause changes to the reachability of systems' state spaces. As system structures are related to genotypes and state spaces are related to phenotypes, it is important to study the relationship between structures and state spaces. However, there is still no method can quantitively describe the reachability differences of two state spaces caused by structural perturbations. Therefore, Difference in Reachability between State Spaces (DReSS) is proposed. DReSS index family can quantitively describe differences of reachability, attractor sets between two state spaces and can help find the key structure in a system, which may influence system's state space significantly. First, basic properties of DReSS including non-negativity, symmetry and subadditivity are proved. Then, typical examples are shown to explain the meaning of DReSS and the differences between DReSS and traditional graph distance. Finally, differences of DReSS distribution between real biological regulatory networks and random networks are compared. Results show most structural perturbations in biological networks tend to affect reachability inside and between attractor basins rather than to affect attractor set itself when compared with random networks, which illustrates that most genotype differences tend to influence the proportion of different phenotypes and only a few ones can create new phenotypes. DReSS can provide researchers with a new insight to study the relation between genotypes and phenotypes.
Collapse
Affiliation(s)
- Ziqiao Yin
- Shenyuan Honors College and School of Mathematical Sciences, Beihang University, and Key Laboratory of Mathematics, Informatics and Behavioral Semantics, Ministry of Education. He currently works as a visiting scholar at Yale University
| | - Binghui Guo
- Artificial Intelligence Institute, Beijing Advanced Innovation Center for Big Data and Brain Computing, LMIB, NLSDE, School of Mathematical Sciences, Beihang University, and Peng Cheng Laboratory
| | - Shuangge Ma
- Department of Biostatistics, Yale University
| | - Yifan Sun
- School of Statistics, Renmin University of China
| | - Zhilong Mi
- Key Laboratory of Mathematics, Informatics and Behavioral Semantics, Ministry of Education, and School of Mathematical Sciences from Beihang University
| | - Zhiming Zheng
- Artificial Intelligence Institute, Beijing Advanced Innovation Center for Big Data and Brain Computing, LMIB, NLSDE, School of Mathematical Sciences, Beihang University, and Peng Cheng Laboratory
| |
Collapse
|
19
|
Targeting Bfl-1 via acute CDK9 inhibition overcomes intrinsic BH3-mimetic resistance in lymphomas. Blood 2020; 137:2947-2957. [PMID: 33259592 DOI: 10.1182/blood.2020008528] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/21/2020] [Indexed: 12/30/2022] Open
Abstract
BH3 mimetics like venetoclax target prosurvival Bcl-2 family proteins and are important therapeutics in the treatment of hematological malignancies. We demonstrate that endogenous Bfl-1 expression can render preclinical lymphoma tumor models insensitive to Mcl-1 and Bcl-2 inhibitors. However, suppression of Bfl-1 alone was insufficient to fully induce apoptosis in Bfl-1-expressing lymphomas, highlighting the need for targeting additional prosurvival proteins in this context. Importantly, we demonstrated that cyclin-dependent kinase 9 (CDK9) inhibitors rapidly downregulate both Bfl-1 and Mcl-1, inducing apoptosis in BH3-mimetic-resistant lymphoma cell lines in vitro and driving in vivo tumor regressions in diffuse large B-cell lymphoma patient-derived xenograft models expressing Bfl-1. These data underscore the need to clinically develop CDK9 inhibitors, like AZD4573, for the treatment of lymphomas using Bfl-1 as a selection biomarker.
Collapse
|
20
|
Salvi F, Hoermann B, Del Pino García J, Fontanillo M, Derua R, Beullens M, Bollen M, Barabas O, Köhn M. Towards Dissecting the Mechanism of Protein Phosphatase-1 Inhibition by Its C-Terminal Phosphorylation. Chembiochem 2020; 22:834-838. [PMID: 33085143 PMCID: PMC7984433 DOI: 10.1002/cbic.202000669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/21/2020] [Indexed: 01/23/2023]
Abstract
Phosphoprotein phosphatase‐1 (PP1) is a key player in the regulation of phospho‐serine (pSer) and phospho‐threonine (pThr) dephosphorylation and is involved in a large fraction of cellular signaling pathways. Aberrant activity of PP1 has been linked to many diseases, including cancer and heart failure. Besides a well‐established activity control by regulatory proteins, an inhibitory function for phosphorylation (p) of a Thr residue in the C‐terminal intrinsically disordered tail of PP1 has been demonstrated. The associated phenotype of cell‐cycle arrest was repeatedly proposed to be due to autoinhibition of PP1 through either conformational changes or substrate competition. Here, we use PP1 variants created by mutations and protein semisynthesis to differentiate between these hypotheses. Our data support the hypothesis that pThr exerts its inhibitory function by mediating protein complex formation rather than by a direct mechanism of structural changes or substrate competition.
Collapse
Affiliation(s)
- Francesca Salvi
- Genome Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Bernhard Hoermann
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| | - Javier Del Pino García
- Laboratory of Biosignaling and Therapeutics Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Miriam Fontanillo
- Genome Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.,SyBioMa, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Monique Beullens
- Laboratory of Biosignaling and Therapeutics Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Mathieu Bollen
- Laboratory of Biosignaling and Therapeutics Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Orsolya Barabas
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Maja Köhn
- Genome Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104, Freiburg, Germany
| |
Collapse
|
21
|
Xin G, Fu J, Luo J, Deng Z, Jiang Q, Zhang C. Aurora B regulates PP1γ-Repo-Man interactions to maintain the chromosome condensation state. J Biol Chem 2020; 295:14780-14788. [PMID: 32938714 DOI: 10.1074/jbc.ac120.012772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 09/12/2020] [Indexed: 11/06/2022] Open
Abstract
The mitotic kinase Aurora B regulates the condensation of chromatin into chromosomes by phosphorylating chromatin proteins during early mitosis, whereas the phosphatase PP1γ performs the opposite function. The roles of Aurora B and PP1γ must be tightly coordinated to maintain chromosomes at a high phosphorylation state, but the precise mechanisms regulating their function remain largely unclear. Here, mainly through immunofluorescence microscopy and co-immunoprecipitation assays, we find that dissociation of PP1γ from chromosomes is essential for maintaining chromosome phosphorylation. We uncover that PP1γ is recruited to mitotic chromosomes by its regulatory subunit Repo-Man in the absence of Aurora B activity and that Aurora B regulates dissociation of PP1γ by phosphorylating and disrupting PP1γ-Repo-Man interactions on chromatin. Overexpression of Repo-Man mutants that cannot be phosphorylated or inhibition of Aurora B kinase activity resulted in the retention of PP1γ on chromatin and prolonged the chromatin condensation process; a similar outcome was caused by the ectopic targeting of PP1γ to chromatin. Together, our findings reveal a novel regulation mechanism of chromatin condensation in which Aurora B counteracts PP1γ activity by releasing PP1γ from Repo-Man and may have important implications for understanding the regulations of dynamic structural changes of the chromosomes in mitosis.
Collapse
Affiliation(s)
- Guangwei Xin
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Jingyan Fu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Jia Luo
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Zhaoxuan Deng
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Qing Jiang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China
| | - Chuanmao Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
22
|
Khalife J, Fréville A, Gnangnon B, Pierrot C. The Multifaceted Role of Protein Phosphatase 1 in Plasmodium. Trends Parasitol 2020; 37:154-164. [PMID: 33036936 DOI: 10.1016/j.pt.2020.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 11/29/2022]
Abstract
Protein phosphatase type 1 (PP1) forms a wide range of Ser/Thr-specific phosphatase holoenzymes which contain one catalytic subunit (PP1c), present in all eukaryotic cells, associated with variable subunits known as regulatory proteins. It has recently been shown that regulators take a leading role in the organization and the control of PP1 functions. Many studies have addressed the role of these regulators in diverse organisms, including humans, and investigated their link to diseases. In this review we summarize recent advances on the role of PP1c in Plasmodium, its interactome and regulators. As a proof of concept, peptides interfering with the regulator binding capacity of PP1c were shown to inhibit the growth of P. falciparum, suggesting their potential as drug precursors.
Collapse
Affiliation(s)
- Jamal Khalife
- Center for Infection and Immunity of Lille, Biology of Apicomplexan Parasites, UMR 9017 CNRS, U1019 INSERM, Université de Lille, Institut Pasteur de Lille, Lille, France.
| | - Aline Fréville
- Center for Infection and Immunity of Lille, Biology of Apicomplexan Parasites, UMR 9017 CNRS, U1019 INSERM, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Bénédicte Gnangnon
- Center for Infection and Immunity of Lille, Biology of Apicomplexan Parasites, UMR 9017 CNRS, U1019 INSERM, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Christine Pierrot
- Center for Infection and Immunity of Lille, Biology of Apicomplexan Parasites, UMR 9017 CNRS, U1019 INSERM, Université de Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
23
|
Bancroft J, Holder J, Geraghty Z, Alfonso-Pérez T, Murphy D, Barr FA, Gruneberg U. PP1 promotes cyclin B destruction and the metaphase-anaphase transition by dephosphorylating CDC20. Mol Biol Cell 2020; 31:2315-2330. [PMID: 32755477 PMCID: PMC7851957 DOI: 10.1091/mbc.e20-04-0252] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/30/2022] Open
Abstract
Ubiquitin-dependent proteolysis of cyclin B and securin initiates sister chromatid segregation and anaphase. The anaphase-promoting complex/cyclosome and its coactivator CDC20 (APC/CCDC20) form the main ubiquitin E3 ligase for these two proteins. APC/CCDC20 is regulated by CDK1-cyclin B and counteracting PP1 and PP2A family phosphatases through modulation of both activating and inhibitory phosphorylation. Here, we report that PP1 promotes cyclin B destruction at the onset of anaphase by removing specific inhibitory phosphorylation in the N-terminus of CDC20. Depletion or chemical inhibition of PP1 stabilizes cyclin B and results in a pronounced delay at the metaphase-to-anaphase transition after chromosome alignment. This requirement for PP1 is lost in cells expressing CDK1 phosphorylation-defective CDC206A mutants. These CDC206A cells show a normal spindle checkpoint response and rapidly destroy cyclin B once all chromosomes have aligned and enter into anaphase in the absence of PP1 activity. PP1 therefore facilitates the metaphase-to-anaphase transition by promoting APC/CCDC20-dependent destruction of cyclin B in human cells.
Collapse
Affiliation(s)
- James Bancroft
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - James Holder
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Zoë Geraghty
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | | | - Daniel Murphy
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Francis A. Barr
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| |
Collapse
|
24
|
Lemonnier T, Dupré A, Jessus C. The G2-to-M transition from a phosphatase perspective: a new vision of the meiotic division. Cell Div 2020; 15:9. [PMID: 32508972 PMCID: PMC7249327 DOI: 10.1186/s13008-020-00065-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022] Open
Abstract
Cell division is orchestrated by the phosphorylation and dephosphorylation of thousands of proteins. These post-translational modifications underlie the molecular cascades converging to the activation of the universal mitotic kinase, Cdk1, and entry into cell division. They also govern the structural events that sustain the mechanics of cell division. While the role of protein kinases in mitosis has been well documented by decades of investigations, little was known regarding the control of protein phosphatases until the recent years. However, the regulation of phosphatase activities is as essential as kinases in controlling the activation of Cdk1 to enter M-phase. The regulation and the function of phosphatases result from post-translational modifications but also from the combinatorial association between conserved catalytic subunits and regulatory subunits that drive their substrate specificity, their cellular localization and their activity. It now appears that sequential dephosphorylations orchestrated by a network of phosphatase activities trigger Cdk1 activation and then order the structural events necessary for the timely execution of cell division. This review discusses a series of recent works describing the important roles played by protein phosphatases for the proper regulation of meiotic division. Many breakthroughs in the field of cell cycle research came from studies on oocyte meiotic divisions. Indeed, the meiotic division shares most of the molecular regulators with mitosis. The natural arrests of oocytes in G2 and in M-phase, the giant size of these cells, the variety of model species allowing either biochemical or imaging as well as genetics approaches explain why the process of meiosis has served as an historical model to decipher signalling pathways involved in the G2-to-M transition. The review especially highlights how the phosphatase PP2A-B55δ critically orchestrates the timing of meiosis resumption in amphibian oocytes. By opposing the kinase PKA, PP2A-B55δ controls the release of the G2 arrest through the dephosphorylation of their substrate, Arpp19. Few hours later, the inhibition of PP2A-B55δ by Arpp19 releases its opposing kinase, Cdk1, and triggers M-phase. In coordination with a variety of phosphatases and kinases, the PP2A-B55δ/Arpp19 duo therefore emerges as the key effector of the G2-to-M transition.
Collapse
Affiliation(s)
- Tom Lemonnier
- Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Sorbonne Université, CNRS, 75005 Paris, France
| | - Aude Dupré
- Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Sorbonne Université, CNRS, 75005 Paris, France
| | - Catherine Jessus
- Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Sorbonne Université, CNRS, 75005 Paris, France
| |
Collapse
|
25
|
Bi A, An W, Wang C, Hua Y, Fang F, Dong X, Chen R, Zhang Z, Luo L. SCR-1693 inhibits tau phosphorylation and improves insulin resistance associated cognitive deficits. Neuropharmacology 2020; 168:108027. [DOI: 10.1016/j.neuropharm.2020.108027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 12/29/2022]
|
26
|
Feizbakhsh O, Pontheaux F, Glippa V, Morales J, Ruchaud S, Cormier P, Roch F. A Peak of H3T3 Phosphorylation Occurs in Synchrony with Mitosis in Sea Urchin Early Embryos. Cells 2020; 9:cells9040898. [PMID: 32272587 PMCID: PMC7226724 DOI: 10.3390/cells9040898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/31/2022] Open
Abstract
The sea urchin embryo provides a valuable system to analyse the molecular mechanisms orchestrating cell cycle progression and mitosis in a developmental context. However, although it is known that the regulation of histone activity by post-translational modification plays an important role during cell division, the dynamics and the impact of these modifications have not been characterised in detail in a developing embryo. Using different immuno-detection techniques, we show that the levels of Histone 3 phosphorylation at Threonine 3 oscillate in synchrony with mitosis in Sphaerechinus granularis early embryos. We present, in addition, the results of a pharmacological study aimed at analysing the role of this key histone post-translational modification during sea urchin early development.
Collapse
|
27
|
Lemmens B, Lindqvist A. DNA replication and mitotic entry: A brake model for cell cycle progression. J Cell Biol 2019; 218:3892-3902. [PMID: 31712253 PMCID: PMC6891093 DOI: 10.1083/jcb.201909032] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/31/2019] [Accepted: 10/31/2019] [Indexed: 12/22/2022] Open
Abstract
Lemmens and Lindqvist discuss how DNA replication and mitosis are coordinated and propose a cell cycle model controlled by brakes. The core function of the cell cycle is to duplicate the genome and divide the duplicated DNA into two daughter cells. These processes need to be carefully coordinated, as cell division before DNA replication is complete leads to genome instability and cell death. Recent observations show that DNA replication, far from being only a consequence of cell cycle progression, plays a key role in coordinating cell cycle activities. DNA replication, through checkpoint kinase signaling, restricts the activity of cyclin-dependent kinases (CDKs) that promote cell division. The S/G2 transition is therefore emerging as a crucial regulatory step to determine the timing of mitosis. Here we discuss recent observations that redefine the coupling between DNA replication and cell division and incorporate these insights into an updated cell cycle model for human cells. We propose a cell cycle model based on a single trigger and sequential releases of three molecular brakes that determine the kinetics of CDK activation.
Collapse
Affiliation(s)
- Bennie Lemmens
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet and Science for Life Laboratory, Stockholm, Sweden
| | - Arne Lindqvist
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Ding J, Fang Z, Liu X, Zhu Z, Wen C, Wang H, Gu J, Li QR, Zeng R, Li H, Jin Y. CDK11 safeguards the identity of human embryonic stem cells via fine-tuning signaling pathways. J Cell Physiol 2019; 235:4279-4290. [PMID: 31612516 DOI: 10.1002/jcp.29305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/27/2019] [Indexed: 11/07/2022]
Abstract
Signaling pathways transmit extracellular cues into cells and regulate transcriptome and epigenome to maintain or change the cell identity. Protein kinases and phosphatases are critical for signaling transduction and regulation. Here, we report that CDK11, a member of the CDK family, is required for the maintenance of human embryonic stem cell (hESC) self-renewal. Our results show that, among the three main isoforms of CDK11, CDK11p46 is the main isoform safeguarding the hESC identity. Mechanistically, CDK11 constrains two important mitogen-activated protein kinase (MAPK) signaling pathways (JNK and p38 signaling) through modulating the activity of protein phosphatase 1. Furthermore, CDK11 knockdown activates transforming growth factor β (TGF-β)/SMAD2/3 signaling and upregulates certain nonneural differentiation-associated genes. Taken together, this study uncovers a kinase required for hESC self-renewal through fine-tuning MAPK and TGF-β signaling at appropriate levels. The kinase-phosphatase axis reported here may shed new light on the molecular mechanism sustaining the identity of hESCs.
Collapse
Affiliation(s)
- Jianyi Ding
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Chinese Academy of Sciences, Shanghai, China
| | - Zhuoqing Fang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Chinese Academy of Sciences, Shanghai, China
| | - Xinyuan Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Chinese Academy of Sciences, Shanghai, China
| | - Zhexin Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Chinese Academy of Sciences, Shanghai, China
| | - Chunsheng Wen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Chinese Academy of Sciences, Shanghai, China
| | - Han Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Chinese Academy of Sciences, Shanghai, China
| | - Junjie Gu
- Basic Clinical Research Center, Renji Hospital, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Run Li
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Rong Zeng
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Hui Li
- Basic Clinical Research Center, Renji Hospital, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Chinese Academy of Sciences, Shanghai, China.,Basic Clinical Research Center, Renji Hospital, Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
29
|
Crncec A, Hochegger H. Triggering mitosis. FEBS Lett 2019; 593:2868-2888. [PMID: 31602636 DOI: 10.1002/1873-3468.13635] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/07/2019] [Accepted: 10/07/2019] [Indexed: 12/28/2022]
Abstract
Entry into mitosis is triggered by the activation of cyclin-dependent kinase 1 (Cdk1). This simple reaction rapidly and irreversibly sets the cell up for division. Even though the core step in triggering mitosis is so simple, the regulation of this cellular switch is highly complex, involving a large number of interconnected signalling cascades. We do have a detailed knowledge of most of the components of this network, but only a poor understanding of how they work together to create a precise and robust system that ensures that mitosis is triggered at the right time and in an orderly fashion. In this review, we will give an overview of the literature that describes the Cdk1 activation network and then address questions relating to the systems biology of this switch. How is the timing of the trigger controlled? How is mitosis insulated from interphase? What determines the sequence of events, following the initial trigger of Cdk1 activation? Which elements ensure robustness in the timing and execution of the switch? How has this system been adapted to the high levels of replication stress in cancer cells?
Collapse
Affiliation(s)
- Adrijana Crncec
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| | - Helfrid Hochegger
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| |
Collapse
|
30
|
Hayward D, Alfonso-Pérez T, Gruneberg U. Orchestration of the spindle assembly checkpoint by CDK1-cyclin B1. FEBS Lett 2019; 593:2889-2907. [PMID: 31469407 DOI: 10.1002/1873-3468.13591] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/01/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022]
Abstract
In mitosis, the spindle assembly checkpoint (SAC) monitors the formation of microtubule-kinetochore attachments during capture of chromosomes by the mitotic spindle. Spindle assembly is complete once there are no longer any unattached kinetochores. Here, we will discuss the mechanism and key components of spindle checkpoint signalling. Unattached kinetochores bind the principal spindle checkpoint kinase monopolar spindle 1 (MPS1). MPS1 triggers the recruitment of other spindle checkpoint proteins and the formation of a soluble inhibitor of anaphase, thus preventing exit from mitosis. On microtubule attachment, kinetochores become checkpoint silent due to the actions of PP2A-B56 and PP1. This SAC responsive period has to be coordinated with mitotic spindle formation to ensure timely mitotic exit and accurate chromosome segregation. We focus on the molecular mechanisms by which the SAC permissive state is created, describing a central role for CDK1-cyclin B1 and its counteracting phosphatase PP2A-B55. Furthermore, we discuss how CDK1-cyclin B1, through its interaction with MAD1, acts as an integral component of the SAC, and actively orchestrates checkpoint signalling and thus contributes to the faithful execution of mitosis.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, UK
| | | | | |
Collapse
|
31
|
Kim HJ, Shin J, Lee S, Kim TW, Jang H, Suh MY, Kim JH, Hwang IY, Hwang DS, Cho EJ, Youn HD. Cyclin-dependent kinase 1 activity coordinates the chromatin associated state of Oct4 during cell cycle in embryonic stem cells. Nucleic Acids Res 2019; 46:6544-6560. [PMID: 29901724 PMCID: PMC6061841 DOI: 10.1093/nar/gky371] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 04/30/2018] [Indexed: 11/23/2022] Open
Abstract
Cyclin-dependent kinase 1 (Cdk1) is indispensable for embryonic stem cell (ESC) maintenance and embryo development. Even though some reports have described a connection between Cdk1 and Oct4, there is no evidence that Cdk1 activity is directly linked to the ESC pluripotency transcription program. We recently reported that Aurkb/PP1-mediated Oct4 resetting is important to cell cycle maintenance and pluripotency in mouse ESCs (mESCs). In this study, we show that Cdk1 is an upstream regulator of the Oct4 phosphorylation state during cell cycle progression, and it coordinates the chromatin associated state of Oct4 for pluripotency-related gene expression within the cell cycle. Upon entry into mitosis, Aurkb in the chromosome passenger complex becomes fully activated and PP1 activity is inhibited downstream of Cdk1 activation, leading to sustaining Oct4(S229) phosphorylation and dissociation of Oct4 from chromatin during the mitotic phase. Cdk1 inhibition at the mitotic phase abnormally results in Oct4 dephosphorylation, chromosome decondensation and chromatin association of Oct4, even in replicated chromosome. Our study results suggest a molecular mechanism by which Cdk1 directly links the cell cycle to the pluripotency transcription program in mESCs.
Collapse
Affiliation(s)
- Hye Ji Kim
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jihoon Shin
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sangho Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Republic of Korea
| | - Tae Wan Kim
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyonchol Jang
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Min Young Suh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Republic of Korea
| | - Jae-Hwan Kim
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - In-Young Hwang
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Deog Su Hwang
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Jung Cho
- College of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hong-Duk Youn
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Republic of Korea.,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
32
|
Interplay between Phosphatases and the Anaphase-Promoting Complex/Cyclosome in Mitosis. Cells 2019; 8:cells8080814. [PMID: 31382469 PMCID: PMC6721574 DOI: 10.3390/cells8080814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/25/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Accurate division of cells into two daughters is a process that is vital to propagation of life. Protein phosphorylation and selective degradation have emerged as two important mechanisms safeguarding the delicate choreography of mitosis. Protein phosphatases catalyze dephosphorylation of thousands of sites on proteins, steering the cells through establishment of the mitotic phase and exit from it. A large E3 ubiquitin ligase, the anaphase-promoting complex/cyclosome (APC/C) becomes active during latter stages of mitosis through G1 and marks hundreds of proteins for destruction. Recent studies have revealed the complex interregulation between these two classes of enzymes. In this review, we highlight the direct and indirect mechanisms by which phosphatases and the APC/C mutually influence each other to ensure accurate spatiotemporal and orderly progression through mitosis, with a particular focus on recent insights and conceptual advances.
Collapse
|
33
|
Huguet F, Flynn S, Vagnarelli P. The Role of Phosphatases in Nuclear Envelope Disassembly and Reassembly and Their Relevance to Pathologies. Cells 2019; 8:cells8070687. [PMID: 31284660 PMCID: PMC6678589 DOI: 10.3390/cells8070687] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 12/20/2022] Open
Abstract
The role of kinases in the regulation of cell cycle transitions is very well established, however, over the past decade, studies have identified the ever-growing importance of phosphatases in these processes. It is well-known that an intact or otherwise non-deformed nuclear envelope (NE) is essential for maintaining healthy cells and any deviation from this can result in pathological conditions. This review aims at assessing the current understanding of how phosphatases contribute to the remodelling of the nuclear envelope during its disassembling and reformation after cell division and how errors in this process may lead to the development of diseases.
Collapse
Affiliation(s)
- Florentin Huguet
- College of Health and Life Science, Research Institute for Environment Health and Society, Brunel University London, London UB8 3PH, UK
| | - Shane Flynn
- College of Health and Life Science, Research Institute for Environment Health and Society, Brunel University London, London UB8 3PH, UK
| | - Paola Vagnarelli
- College of Health and Life Science, Research Institute for Environment Health and Society, Brunel University London, London UB8 3PH, UK.
| |
Collapse
|
34
|
Brautigan DL, Shenolikar S. Protein Serine/Threonine Phosphatases: Keys to Unlocking Regulators and Substrates. Annu Rev Biochem 2019; 87:921-964. [PMID: 29925267 DOI: 10.1146/annurev-biochem-062917-012332] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein serine/threonine phosphatases (PPPs) are ancient enzymes, with distinct types conserved across eukaryotic evolution. PPPs are segregated into types primarily on the basis of the unique interactions of PPP catalytic subunits with regulatory proteins. The resulting holoenzymes dock substrates distal to the active site to enhance specificity. This review focuses on the subunit and substrate interactions for PPP that depend on short linear motifs. Insights about these motifs from structures of holoenzymes open new opportunities for computational biology approaches to elucidate PPP networks. There is an expanding knowledge base of posttranslational modifications of PPP catalytic and regulatory subunits, as well as of their substrates, including phosphorylation, acetylation, and ubiquitination. Cross talk between these posttranslational modifications creates PPP-based signaling. Knowledge of PPP complexes, signaling clusters, as well as how PPPs communicate with each other in response to cellular signals should unlock the doors to PPP networks and signaling "clouds" that orchestrate and coordinate different aspects of cell physiology.
Collapse
Affiliation(s)
- David L Brautigan
- Center for Cell Signaling and Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA;
| | - Shirish Shenolikar
- Signature Research Programs in Cardiovascular and Metabolic Disorders and Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore 169857
| |
Collapse
|
35
|
Vallardi G, Cordeiro MH, Saurin AT. A Kinase-Phosphatase Network that Regulates Kinetochore-Microtubule Attachments and the SAC. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:457-484. [PMID: 28840249 DOI: 10.1007/978-3-319-58592-5_19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The KMN network (for KNL1, MIS12 and NDC80 complexes) is a hub for signalling at the outer kinetochore. It integrates the activities of two kinases (MPS1 and Aurora B) and two phosphatases (PP1 and PP2A-B56) to regulate kinetochore-microtubule attachments and the spindle assembly checkpoint (SAC). We will first discuss each of these enzymes separately, to describe how they are regulated at kinetochores and why this is important for their primary function in controlling either microtubule attachments or the SAC. We will then discuss why inhibiting any one of them individually produces secondary effects on all the others. This cross-talk may help to explain why all enzymes have been linked to both processes, even though the direct evidence suggests they each control only one. This chapter therefore describes how a network of kinases and phosphatases work together to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Giulia Vallardi
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Marilia Henriques Cordeiro
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Adrian Thomas Saurin
- Division of Cancer Research, School of Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
36
|
Hayward D, Alfonso-Pérez T, Cundell MJ, Hopkins M, Holder J, Bancroft J, Hutter LH, Novak B, Barr FA, Gruneberg U. CDK1-CCNB1 creates a spindle checkpoint-permissive state by enabling MPS1 kinetochore localization. J Cell Biol 2019; 218:1182-1199. [PMID: 30674582 PMCID: PMC6446832 DOI: 10.1083/jcb.201808014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/19/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022] Open
Abstract
Spindle checkpoint signaling is initiated by recruitment of the kinase MPS1 to unattached kinetochores during mitosis. We show that CDK1-CCNB1 and a counteracting phosphatase PP2A-B55 regulate the engagement of human MPS1 with unattached kinetochores by controlling the phosphorylation status of S281 in the kinetochore-binding domain. This regulation is essential for checkpoint signaling, since MPS1S281A is not recruited to unattached kinetochores and fails to support the recruitment of other checkpoint proteins. Directly tethering MPS1S281A to the kinetochore protein Mis12 bypasses this regulation and hence the requirement for S281 phosphorylation in checkpoint signaling. At the metaphase-anaphase transition, MPS1 S281 dephosphorylation is delayed because PP2A-B55 is negatively regulated by CDK1-CCNB1 and only becomes fully active once CCNB1 concentration falls below a characteristic threshold. This mechanism prolongs the checkpoint-responsive period when MPS1 can localize to kinetochores and enables a response to late-stage spindle defects. By acting together, CDK1-CCNB1 and PP2A-B55 thus create a spindle checkpoint-permissive state and ensure the fidelity of mitosis.
Collapse
Affiliation(s)
- Daniel Hayward
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, England, UK
| | - Tatiana Alfonso-Pérez
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Michael J Cundell
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Michael Hopkins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - James Holder
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - James Bancroft
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, England, UK
| | - Lukas H Hutter
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Bela Novak
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Francis A Barr
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, England, UK
| | - Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, England, UK
| |
Collapse
|
37
|
Moura M, Conde C. Phosphatases in Mitosis: Roles and Regulation. Biomolecules 2019; 9:E55. [PMID: 30736436 PMCID: PMC6406801 DOI: 10.3390/biom9020055] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
Mitosis requires extensive rearrangement of cellular architecture and of subcellular structures so that replicated chromosomes can bind correctly to spindle microtubules and segregate towards opposite poles. This process originates two new daughter nuclei with equal genetic content and relies on highly-dynamic and tightly regulated phosphorylation of numerous cell cycle proteins. A burst in protein phosphorylation orchestrated by several conserved kinases occurs as cells go into and progress through mitosis. The opposing dephosphorylation events are catalyzed by a small set of protein phosphatases, whose importance for the accuracy of mitosis is becoming increasingly appreciated. This review will focus on the established and emerging roles of mitotic phosphatases, describe their structural and biochemical properties, and discuss recent advances in understanding the regulation of phosphatase activity and function.
Collapse
Affiliation(s)
- Margarida Moura
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal.
| | - Carlos Conde
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
- i3S-Instituto de Investigação e Inovação em Saúde da Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
38
|
Structure-Guided Exploration of SDS22 Interactions with Protein Phosphatase PP1 and the Splicing Factor BCLAF1. Structure 2019; 27:507-518.e5. [PMID: 30661852 DOI: 10.1016/j.str.2018.12.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 10/09/2018] [Accepted: 12/03/2018] [Indexed: 01/29/2023]
Abstract
SDS22 is an ancient regulator of protein phosphatase-1 (PP1). Our crystal structure of SDS22 shows that its twelve leucine-rich repeats adopt a banana-shaped fold that is shielded from solvent by capping domains at its extremities. Subsequent modeling and biochemical studies revealed that the concave side of SDS22 likely interacts with PP1 helices α5 and α6, which are distal from the binding sites of many previously described PP1 interactors. Accordingly, we found that SDS22 acts as a "third" subunit of multiple PP1 holoenzymes. The crystal structure of SDS22 also revealed a large basic surface patch that enables binding of a phosphorylated form of splicing factor BCLAF1. Taken together, our data provide insights into the formation of PP1:SDS22 and the recruitment of additional interaction proteins, such as BCLAF1.
Collapse
|
39
|
Vavrdová T, ˇSamaj J, Komis G. Phosphorylation of Plant Microtubule-Associated Proteins During Cell Division. FRONTIERS IN PLANT SCIENCE 2019; 10:238. [PMID: 30915087 PMCID: PMC6421500 DOI: 10.3389/fpls.2019.00238] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/12/2019] [Indexed: 05/20/2023]
Abstract
Progression of mitosis and cytokinesis depends on the reorganization of cytoskeleton, with microtubules driving the segregation of chromosomes and their partitioning to two daughter cells. In dividing plant cells, microtubules undergo global reorganization throughout mitosis and cytokinesis, and with the aid of various microtubule-associated proteins (MAPs), they form unique systems such as the preprophase band (PPB), the acentrosomal mitotic spindle, and the phragmoplast. Such proteins include nucleators of de novo microtubule formation, plus end binding proteins involved in the regulation of microtubule dynamics, crosslinking proteins underlying microtubule bundle formation and members of the kinesin superfamily with microtubule-dependent motor activities. The coordinated function of such proteins not only drives the continuous remodeling of microtubules during mitosis and cytokinesis but also assists the positioning of the PPB, the mitotic spindle, and the phragmoplast, affecting tissue patterning by controlling cell division plane (CDP) orientation. The affinity and the function of such proteins is variably regulated by reversible phosphorylation of serine and threonine residues within the microtubule binding domain through a number of protein kinases and phosphatases which are differentially involved throughout cell division. The purpose of the present review is to provide an overview of the function of protein kinases and protein phosphatases involved in cell division regulation and to identify cytoskeletal substrates relevant to the progression of mitosis and cytokinesis and the regulation of CDP orientation.
Collapse
|
40
|
Nilsson J. Protein phosphatases in the regulation of mitosis. J Cell Biol 2018; 218:395-409. [PMID: 30446607 PMCID: PMC6363451 DOI: 10.1083/jcb.201809138] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/29/2018] [Accepted: 10/29/2018] [Indexed: 12/15/2022] Open
Abstract
The accurate segregation of genetic material to daughter cells during mitosis depends on the precise coordination and regulation of hundreds of proteins by dynamic phosphorylation. Mitotic kinases are major regulators of protein function, but equally important are protein phosphatases that balance their actions, their coordinated activity being essential for accurate chromosome segregation. Phosphoprotein phosphatases (PPPs) that dephosphorylate phosphoserine and phosphothreonine residues are increasingly understood as essential regulators of mitosis. In contrast to kinases, the lack of a pronounced peptide-binding cleft on the catalytic subunit of PPPs suggests that these enzymes are unlikely to be specific. However, recent exciting insights into how mitotic PPPs recognize specific substrates have revealed that they are as specific as kinases. Furthermore, the activities of PPPs are tightly controlled at many levels to ensure that they are active only at the proper time and place. Here, I will discuss substrate selection and regulation of mitotic PPPs focusing mainly on animal cells and explore how these actions control mitosis, as well as important unanswered questions.
Collapse
Affiliation(s)
- Jakob Nilsson
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Du S, Sarver JG, Trabbic CJ, Erhardt PW, Schroering A, Maltese WA. 6-MOMIPP, a novel brain-penetrant anti-mitotic indolyl-chalcone, inhibits glioblastoma growth and viability. Cancer Chemother Pharmacol 2018; 83:237-254. [PMID: 30426158 DOI: 10.1007/s00280-018-3726-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 11/03/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE 3-(6-Methoxy-2-methyl-1H-indol-3-yl)-1-(4-pyridinyl)-2-propene-1-one (6-MOMIPP) is a novel indole-based chalcone that disrupts microtubules. The present study aims to define the mechanism through which 6-MOMIPP induces cell death and to evaluate the efficacy of the compound in penetrating the blood-brain barrier and inhibiting growth of glioblastoma xenografts. METHODS The effects of 6-MOMIPP were evaluated in cultured U251 glioblastoma cells, using viability, flow cytometry, and tubulin polymerization assays. Scintillation proximity and tubulin crosslinking methods were used to identify the binding site of 6-MOMIPP on tubulin, and western blots were performed to define the signaling pathways that contribute to cell death. LC/MS assays were used to study the pharmacokinetic behavior of 6-MOMIPP in mice. Subcutaneous and intracerebral xenograft models were utilized to assess the effects of 6-MOMIPP on growth of U251 glioblastoma in vivo. RESULTS The findings indicate that 6-MOMIPP targets the colchicine site on β-tubulin. At concentrations ≥ 250 nm, 6-MOMIPP induces mitotic arrest, caspase activation and loss of cell viability. Cells are protected by caspase inhibitors, pointing to an apoptotic mechanism of cell death. Loss of cell viability is preceded by activation of Cdk1(Cdc2) and phosphorylation of Bcl-2 and Bcl-xL. Inhibition of both events with a Cdk1 inhibitor prevents cell death. 6-MOMIPP has broad activity against the viability of multiple glioblastoma, melanoma and lung carcinoma cell lines. Viability of normal cells, including differentiated neurons, is not significantly affected at a drug concentration (1 µM) that reduces viability in most cancer lines. Pharmacokinetic studies in mice show that concentrations of 6-MOMIPP in the brain mirror those in the plasma, indicating that 6-MOMIPP readily penetrates the blood-brain barrier. Studies with mice bearing human U251 glioblastoma xenografts demonstrate that 6-MOMIPP is effective in suppressing growth of subcutaneous and intracerebral tumors without causing general toxicity. CONCLUSIONS The results indicate that 6-MOMIPP is a novel microtubule disruptor that targets the colchicine binding site on β-tubulin to induce mitotic arrest and cell death. The ability of 6-MOMIPP to penetrate the blood-brain barrier and inhibit growth of glioblastoma xenografts suggests that it warrants further preclinical evaluation as potential small-molecule therapeutic that may have advantages in treating primary and metastatic brain tumors.
Collapse
Affiliation(s)
- Shengnan Du
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - Jeffrey G Sarver
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2810 W. Bancroft Street, Toledo, OH, 43606, USA
| | - Christopher J Trabbic
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2810 W. Bancroft Street, Toledo, OH, 43606, USA
| | - Paul W Erhardt
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2810 W. Bancroft Street, Toledo, OH, 43606, USA
| | - Allen Schroering
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA
| | - William A Maltese
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Transverse Drive, Toledo, OH, 43614, USA.
| |
Collapse
|
42
|
Wu D, De Wever V, Derua R, Winkler C, Beullens M, Van Eynde A, Bollen M. A substrate-trapping strategy for protein phosphatase PP1 holoenzymes using hypoactive subunit fusions. J Biol Chem 2018; 293:15152-15162. [PMID: 30115685 DOI: 10.1074/jbc.ra118.004132] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/13/2018] [Indexed: 01/24/2023] Open
Abstract
The protein Ser/Thr phosphatase PP1 catalyzes an important fraction of protein dephosphorylation events and forms highly specific holoenzymes through an association with regulatory interactors of protein phosphatase one (RIPPOs). The functional characterization of individual PP1 holoenzymes is hampered by the lack of straightforward strategies for substrate mapping. Because efficient substrate recruitment often involves binding to both PP1 and its associated RIPPO, here we examined whether PP1-RIPPO fusions can be used to trap substrates for further analysis. Fusions of an hypoactive point mutant of PP1 and either of four tested RIPPOs accumulated in HEK293T cells with their associated substrates and were co-immunoprecipitated for subsequent identification of the substrates by immunoblotting or MS analysis. Hypoactive fusions were also used to study RIPPOs themselves as substrates for associated PP1. In contrast, substrate trapping was barely detected with active PP1-RIPPO fusions or with nonfused PP1 or RIPPO subunits. Our results suggest that hypoactive fusions of PP1 subunits represent an easy-to-use tool for substrate identification of individual holoenzymes.
Collapse
Affiliation(s)
- Dan Wu
- From the Laboratory of Biosignaling and Therapeutics
| | | | - Rita Derua
- the Protein Phosphorylation and Proteomics Lab, KU Leuven Department of Cellular and Molecular Medicine, and.,SyBioMa, University of Leuven, 3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
43
|
Saurin AT. Kinase and Phosphatase Cross-Talk at the Kinetochore. Front Cell Dev Biol 2018; 6:62. [PMID: 29971233 PMCID: PMC6018199 DOI: 10.3389/fcell.2018.00062] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 05/31/2018] [Indexed: 01/26/2023] Open
Abstract
Multiple kinases and phosphatases act on the kinetochore to control chromosome segregation: Aurora B, Mps1, Bub1, Plk1, Cdk1, PP1, and PP2A-B56, have all been shown to regulate both kinetochore-microtubule attachments and the spindle assembly checkpoint. Given that so many kinases and phosphatases converge onto two key mitotic processes, it is perhaps not surprising to learn that they are, quite literally, entangled in cross-talk. Inhibition of any one of these enzymes produces secondary effects on all the others, which results in a complicated picture that is very difficult to interpret. This review aims to clarify this picture by first collating the direct effects of each enzyme into one overarching schematic of regulation at the Knl1/Mis12/Ndc80 (KMN) network (a major signaling hub at the outer kinetochore). This schematic will then be used to discuss the implications of the cross-talk that connects these enzymes; both in terms of why it may be needed to produce the right type of kinetochore signals and why it nevertheless complicates our interpretations about which enzymes control what processes. Finally, some general experimental approaches will be discussed that could help to characterize kinetochore signaling by dissociating the direct from indirect effect of kinase or phosphatase inhibition in vivo. Together, this review should provide a framework to help understand how a network of kinases and phosphatases cooperate to regulate two key mitotic processes.
Collapse
Affiliation(s)
- Adrian T. Saurin
- Jacqui Wood Cancer Centre, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
44
|
HIV-1 Protein Tat 1-72 Impairs Neuronal Dendrites via Activation of PP1 and Regulation of the CREB/BDNF Pathway. Virol Sin 2018; 33:261-269. [PMID: 29737506 DOI: 10.1007/s12250-018-0031-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/02/2018] [Indexed: 12/18/2022] Open
Abstract
Despite the success of combined antiretroviral therapy in recent years, the prevalence of human immunodeficiency virus (HIV)-associated neurocognitive disorders in people living with HIV-1 is increasing, significantly reducing the health-related quality of their lives. Although neurons cannot be infected by HIV-1, shed viral proteins such as transactivator of transcription (Tat) can cause dendritic damage. However, the detailed molecular mechanism of Tat-induced neuronal impairment remains unknown. In this study, we first showed that recombinant Tat (1-72 aa) induced neurotoxicity in primary cultured mouse neurons. Second, exposure to Tat1-72 was shown to reduce the length and number of dendrites in cultured neurons. Third, Tat1-72 (0-6 h) modulates protein phosphatase 1 (PP1) expression and enhances its activity by decreasing the phosphorylation level of PP1 at Thr320. Finally, Tat1-72 (24 h) downregulates CREB activity and CREB-mediated gene (BDNF, c-fos, Egr-1) expression. Together, these findings suggest that Tat1-72 might impair cognitive function by regulating the activity of PP1 and the CREB/BDNF pathway.
Collapse
|
45
|
Nasa I, Kettenbach AN. Coordination of Protein Kinase and Phosphoprotein Phosphatase Activities in Mitosis. Front Cell Dev Biol 2018; 6:30. [PMID: 29623276 PMCID: PMC5874294 DOI: 10.3389/fcell.2018.00030] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/08/2018] [Indexed: 01/09/2023] Open
Abstract
Dynamic changes in protein phosphorylation govern the transitions between different phases of the cell division cycle. A "tug of war" between highly conserved protein kinases and the family of phosphoprotein phosphatases (PPP) establishes the phosphorylation state of proteins, which controls their function. More than three-quarters of all proteins are phosphorylated at one or more sites in human cells, with the highest occupancy of phosphorylation sites seen in mitosis. Spatial and temporal regulation of opposing kinase and PPP activities is crucial for accurate execution of the mitotic program. The role of mitotic kinases has been the focus of many studies, while the contribution of PPPs was for a long time underappreciated and is just emerging. Misconceptions regarding the specificity and activity of protein phosphatases led to the belief that protein kinases are the primary determinants of mitotic regulation, leaving PPPs out of the limelight. Recent studies have shown that protein phosphatases are specific and selective enzymes, and that their activity is tightly regulated. In this review, we discuss the emerging roles of PPPs in mitosis and their regulation of and by mitotic kinases, as well as mechanisms that determine PPP substrate recognition and specificity.
Collapse
Affiliation(s)
- Isha Nasa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
46
|
Whittaker SR, Barlow C, Martin MP, Mancusi C, Wagner S, Self A, Barrie E, Te Poele R, Sharp S, Brown N, Wilson S, Jackson W, Fischer PM, Clarke PA, Walton MI, McDonald E, Blagg J, Noble M, Garrett MD, Workman P. Molecular profiling and combinatorial activity of CCT068127: a potent CDK2 and CDK9 inhibitor. Mol Oncol 2018; 12:287-304. [PMID: 29063678 PMCID: PMC5830651 DOI: 10.1002/1878-0261.12148] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 10/05/2017] [Accepted: 10/07/2017] [Indexed: 01/18/2023] Open
Abstract
Deregulation of the cyclin-dependent kinases (CDKs) has been implicated in the pathogenesis of multiple cancer types. Consequently, CDKs have garnered intense interest as therapeutic targets for the treatment of cancer. We describe herein the molecular and cellular effects of CCT068127, a novel inhibitor of CDK2 and CDK9. Optimized from the purine template of seliciclib, CCT068127 exhibits greater potency and selectivity against purified CDK2 and CDK9 and superior antiproliferative activity against human colon cancer and melanoma cell lines. X-ray crystallography studies reveal that hydrogen bonding with the DFG motif of CDK2 is the likely mechanism of greater enzymatic potency. Commensurate with inhibition of CDK activity, CCT068127 treatment results in decreased retinoblastoma protein (RB) phosphorylation, reduced phosphorylation of RNA polymerase II, and induction of cell cycle arrest and apoptosis. The transcriptional signature of CCT068127 shows greatest similarity to other small-molecule CDK and also HDAC inhibitors. CCT068127 caused a dramatic loss in expression of DUSP6 phosphatase, alongside elevated ERK phosphorylation and activation of MAPK pathway target genes. MCL1 protein levels are rapidly decreased by CCT068127 treatment and this associates with synergistic antiproliferative activity after combined treatment with CCT068127 and ABT263, a BCL2 family inhibitor. These findings support the rational combination of this series of CDK2/9 inhibitors and BCL2 family inhibitors for the treatment of human cancer.
Collapse
Affiliation(s)
- Steven R. Whittaker
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Clare Barlow
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Mathew P. Martin
- Northern Institute for Cancer ResearchUniversity of Newcastle upon TyneMedical SchoolNewcastle upon TyneUK
| | - Caterina Mancusi
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Steve Wagner
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Annette Self
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Elaine Barrie
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Robert Te Poele
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Swee Sharp
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Nathan Brown
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Stuart Wilson
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Wayne Jackson
- Cyclacel Ltd.DundeeUK
- Present address:
Samuel Lister AcademyBingleyWest YorkshireBD16 1TZUK
| | - Peter M. Fischer
- Cyclacel Ltd.DundeeUK
- Present address:
School of Pharmacy and Centre for Biomolecular SciencesUniversity of Nottingham, University ParkNottinghamNG7 2RDUK
| | - Paul A. Clarke
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Michael I. Walton
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Edward McDonald
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Martin Noble
- Northern Institute for Cancer ResearchUniversity of Newcastle upon TyneMedical SchoolNewcastle upon TyneUK
| | - Michelle D. Garrett
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
- Present address:
School of BiosciencesUniversity of KentCanterburyKentCT2 7NJUK
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics UnitDivision of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| |
Collapse
|
47
|
Liu X, Huang S, Liu C, Liu X, Shen Y, Cui Z. PPP1CC is associated with astrocyte and microglia proliferation after traumatic spinal cord injury in rats. Pathol Res Pract 2017; 213:1355-1364. [DOI: 10.1016/j.prp.2017.09.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 10/18/2022]
|
48
|
Trotta AP, Gelles JD, Serasinghe MN, Loi P, Arbiser JL, Chipuk JE. Disruption of mitochondrial electron transport chain function potentiates the pro-apoptotic effects of MAPK inhibition. J Biol Chem 2017; 292:11727-11739. [PMID: 28546431 DOI: 10.1074/jbc.m117.786442] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/22/2017] [Indexed: 12/30/2022] Open
Abstract
The mitochondrial network is a major site of ATP production through the coupled integration of the electron transport chain (ETC) with oxidative phosphorylation. In melanoma arising from the V600E mutation in the kinase v-RAF murine sarcoma viral oncogene homolog B (BRAFV600E), oncogenic signaling enhances glucose-dependent metabolism while reducing mitochondrial ATP production. Likewise, when BRAFV600E is pharmacologically inhibited by targeted therapies (e.g. PLX-4032/vemurafenib), glucose metabolism is reduced, and cells increase mitochondrial ATP production to sustain survival. Therefore, collateral inhibition of oncogenic signaling and mitochondrial respiration may help enhance the therapeutic benefit of targeted therapies. Honokiol (HKL) is a well tolerated small molecule that disrupts mitochondrial function; however, its underlying mechanisms and potential utility with targeted anticancer therapies remain unknown. Using wild-type BRAF and BRAFV600E melanoma model systems, we demonstrate here that HKL administration rapidly reduces mitochondrial respiration by broadly inhibiting ETC complexes I, II, and V, resulting in decreased ATP levels. The subsequent energetic crisis induced two cellular responses involving cyclin-dependent kinases (CDKs). First, loss of CDK1-mediated phosphorylation of the mitochondrial division GTPase dynamin-related protein 1 promoted mitochondrial fusion, thus coupling mitochondrial energetic status and morphology. Second, HKL decreased CDK2 activity, leading to G1 cell cycle arrest. Importantly, although pharmacological inhibition of oncogenic MAPK signaling increased ETC activity, co-treatment with HKL ablated this response and vastly enhanced the rate of apoptosis. Collectively, these findings integrate HKL action with mitochondrial respiration and shape and substantiate a pro-survival role of mitochondrial function in melanoma cells after oncogenic MAPK inhibition.
Collapse
Affiliation(s)
- Andrew P Trotta
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jesse D Gelles
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Madhavika N Serasinghe
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Patrick Loi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jack L Arbiser
- Atlanta Veterans Administration Medical Center, Winship Cancer Institute, Atlanta, Georgia 30322; Emory University, School of Medicine, Department of Dermatology, Atlanta, Georgia 30322
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York 10029; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029.
| |
Collapse
|
49
|
Rai R, Hu C, Broton C, Chen Y, Lei M, Chang S. NBS1 Phosphorylation Status Dictates Repair Choice of Dysfunctional Telomeres. Mol Cell 2017; 65:801-817.e4. [PMID: 28216226 PMCID: PMC5639704 DOI: 10.1016/j.molcel.2017.01.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/22/2016] [Accepted: 01/05/2017] [Indexed: 01/07/2023]
Abstract
Telomeres employ TRF2 to protect chromosome ends from activating the DNA damage sensor MRE11-RAD50-NBS1 (MRN), thereby repressing ATM-dependent DNA damage checkpoint responses. How TRF2 prevents MRN activation at dysfunctional telomeres is unclear. Here, we show that the phosphorylation status of NBS1 determines the repair pathway choice of dysfunctional telomeres. The crystal structure of the TRF2-NBS1 complex at 3.0 Å resolution shows that the NBS1 429YQLSP433 motif interacts specifically with the TRF2TRFH domain. Phosphorylation of NBS1 serine 432 by CDK2 in S/G2 dissociates NBS1 from TRF2, promoting TRF2-Apollo/SNM1B complex formation and the protection of leading-strand telomeres. Classical-NHEJ-mediated repair of telomeres lacking TRF2 requires phosphorylated NBS1S432 to activate ATM, while interaction of de-phosphorylated NBS1S432 with TRF2 promotes alternative-NHEJ repair of telomeres lacking POT1-TPP1. Our work advances understanding of how the TRF2TRFH domain orchestrates telomere end protection and reveals how the phosphorylation status of the NBS1S432 dictates repair pathway choice of dysfunctional telomeres.
Collapse
Affiliation(s)
- Rekha Rai
- Department of Laboratory Medicine, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Chunyi Hu
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Cayla Broton
- Department of Laboratory Medicine, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Yong Chen
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Shanghai Research Center, Chinese Academy of Sciences, Shanghai 200031, China, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA
| | - Ming Lei
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 201210, China, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Shanghai Research Center, Chinese Academy of Sciences, Shanghai 200031, China, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA.
| | - Sandy Chang
- Department of Laboratory Medicine, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Department of Pathology, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA; Molecular Biophysics and Biochemistry, Yale University School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
50
|
Heim A, Rymarczyk B, Mayer TU. Regulation of Cell Division. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 953:83-116. [PMID: 27975271 DOI: 10.1007/978-3-319-46095-6_3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The challenging task of mitotic cell divisions is to generate two genetically identical daughter cells from a single precursor cell. To accomplish this task, a complex regulatory network evolved, which ensures that all events critical for the duplication of cellular contents and their subsequent segregation occur in the correct order, at specific intervals and with the highest possible fidelity. Transitions between cell cycle stages are triggered by changes in the phosphorylation state and levels of components of the cell cycle machinery. Entry into S-phase and M-phase are mediated by cyclin-dependent kinases (Cdks), serine-threonine kinases that require a regulatory cyclin subunit for their activity. Resetting the system to the interphase state is mediated by protein phosphatases (PPs) that counteract Cdks by dephosphorylating their substrates. To avoid futile cycles of phosphorylation and dephosphorylation, Cdks and PPs must be regulated in a manner such that their activities are mutually exclusive.
Collapse
Affiliation(s)
- Andreas Heim
- Department of Biology and Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Beata Rymarczyk
- Department of Biology and Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany
| | - Thomas U Mayer
- Department of Biology and Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Universitätsstr. 10, 78457, Konstanz, Germany.
| |
Collapse
|