1
|
Cao R, Jones DTD, Pan L, Yang A, Wang S, Padi SKR, Rawson S, Aster JC, Blacklow SC. Molecular mechanism of PP2A/B55α phosphatase inhibition by IER5. Cell Chem Biol 2025; 32:631-642.e7. [PMID: 40209703 PMCID: PMC12067903 DOI: 10.1016/j.chembiol.2025.03.004] [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: 09/10/2024] [Revised: 02/06/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025]
Abstract
PP2A serine/threonine phosphatases are heterotrimeric complexes that execute many essential physiologic functions. These activities are modulated by additional regulatory proteins, such as ARPP19, FAM122A, and IER5. Here, we report the cryoelectron microscopy (cryo-EM) structure of a complex of PP2A/B55α with the N-terminal structured region of IER5 (IER5-N50), which occludes a surface on B55α used for substrate recruitment, and show that IER5-N50 inhibits PP2A/B55α catalyzed dephosphorylation of pTau in biochemical assays. Mutations of full-length IER5 that disrupt its PP2A/B55α interface interfere with co-immunoprecipitation of PP2A/B55α. IER5 antagonism of B55α in keratinocytes is required for expression of KRT1, a differentiation marker. Mini-IER5 composed of IER5-N50 and a nuclear localization sequence restores this activity in IER5 knockout cells. Using structural bioinformatics, we identify homology of IER5-N50 with SERTA (SEI-1, RBT-1, and TARA) domain containing proteins. These studies define the molecular basis of PP2A/B55α nuclear inhibition by IER5 and suggest a roadmap for selective pharmacologic modulation of PP2A/B55α complexes.
Collapse
Affiliation(s)
- Ruili Cao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Daniel T D Jones
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Li Pan
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Annie Yang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shumei Wang
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sathish K R Padi
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
2
|
Di Talia S. Developmental Control of Cell Cycle and Signaling. Cold Spring Harb Perspect Biol 2025; 17:a041499. [PMID: 38858070 PMCID: PMC11864111 DOI: 10.1101/cshperspect.a041499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
In most species, the earliest stages of embryogenesis are characterized by rapid proliferation, which must be tightly controlled with other cellular processes across the large scale of the embryo. The study of this coordination has recently revealed new mechanisms of regulation of morphogenesis. Here, I discuss progress on how the integration of biochemical and mechanical signals leads to the proper positioning of cellular components, how signaling waves ensure the synchronization of the cell cycle, and how cell cycle transitions are properly timed. Similar concepts are emerging in the control of morphogenesis of other tissues, highlighting both common and unique features of early embryogenesis.
Collapse
Affiliation(s)
- Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
3
|
Lin L, Ding J, Liu S, Liu C, Li Q, Gao X, Niu Y, Tong WM. Protein Phosphatase 2ACα Regulates ATR-Mediated Endogenous DNA Damage Response Against Microcephaly. Mol Neurobiol 2025; 62:1266-1281. [PMID: 38976130 DOI: 10.1007/s12035-024-04301-6] [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: 11/29/2022] [Accepted: 06/11/2024] [Indexed: 07/09/2024]
Abstract
Protein phosphatase 2A (PP2A) is an abundant heterotrimeric holoenzyme in eukaryotic cells coordinating with specific kinases to regulate spatial-temporal protein dephosphorylation in various biological processes. However, the function of PP2A in cortical neurogenesis remains largely unknown. Here, we report that neuronal-specific deletion of Pp2acα in mice displayed microcephaly, with significantly smaller brains and defective learning and memory ability. Mechanistically, neuronal Pp2acα deficiency resulted in elevated endogenous DNA damage and activation of ATR/CHK1 signaling. It was further induced by the loss of direct interaction between PP2AC and ATR as well as the function of PP2AC to dephosphorylate ATR. Importantly, ATR/CHK1 signaling dysregulation altered both the expression and activity of several critical downstream factors including P53, P21, Bcl2, and Bax, which led to decreased proliferation of cortical progenitor cells and increased apoptosis in developing cortical neurons. Taken together, our results indicate an essential function of PP2ACα in endogenous DNA damage response-mediated ATR signaling during neurogenesis, and defective PP2ACα in neurons contributes to microcephaly.
Collapse
Affiliation(s)
- Lin Lin
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jing Ding
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Simeng Liu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Department of Pathology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chunying Liu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Qing Li
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Xiang Gao
- Model Animal Research Center and MOE Key Laboratory of Model Animal for Disease Study, Nanjing University, Nanjing, China
| | - Yamei Niu
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wei-Min Tong
- Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
- Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
4
|
Klemm C, Ólafsson G, Wood HR, Mellor C, Zabet NR, Thorpe PH. Proteome-wide forced interactions reveal a functional map of cell-cycle phospho-regulation in S. cerevisiae. Nucleus 2024; 15:2420129. [PMID: 39618027 PMCID: PMC11622623 DOI: 10.1080/19491034.2024.2420129] [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: 07/25/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 12/08/2024] Open
Abstract
Dynamic protein phosphorylation and dephosphorylation play an essential role in cell cycle progression. Kinases and phosphatases are generally highly conserved across eukaryotes, underlining their importance for post-translational regulation of substrate proteins. In recent years, advances in phospho-proteomics have shed light on protein phosphorylation dynamics throughout the cell cycle, and ongoing progress in bioinformatics has significantly improved annotation of specific phosphorylation events to a given kinase. However, the functional impact of individual phosphorylation events on cell cycle progression is often unclear. To address this question, we used the Synthetic Physical Interactions (SPI) method, which enables the systematic recruitment of phospho-regulators to most yeast proteins. Using this method, we identified several putative novel targets involved in chromosome segregation and cytokinesis. The SPI method monitors cell growth and, therefore, serves as a tool to determine the impact of protein phosphorylation on cell cycle progression.
Collapse
Affiliation(s)
- Cinzia Klemm
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Bioengineering, Imperial College London, London, UK
| | - Guðjón Ólafsson
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Henry Richard Wood
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Caitlin Mellor
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Nicolae Radu Zabet
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Peter Harold Thorpe
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| |
Collapse
|
5
|
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
|
6
|
Xue W, Chu H, Wang J, Sun Y, Qiu X, Song C, Tan L, Ding C, Liao Y. Coronavirus nucleocapsid protein enhances the binding of p-PKCα to RACK1: Implications for inhibition of nucleocytoplasmic trafficking and suppression of the innate immune response. PLoS Pathog 2024; 20:e1012097. [PMID: 39602452 PMCID: PMC11633972 DOI: 10.1371/journal.ppat.1012097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 12/11/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
The hallmark of coronavirus infection lies in its ability to evade host immune defenses, a process intricately linked to the nuclear entry of transcription factors crucial for initiating the expression of antiviral genes. Central to this evasion strategy is the manipulation of the nucleocytoplasmic trafficking system, which serves as an effective target for the virus to modulate the expression of immune response-related genes. In this investigation, we discovered that infection with the infectious bronchitis virus (IBV) dynamically impedes the nuclear translocation of several transcription factors such as IRF3, STAT1, STAT2, NF-κB p65, and the p38 MAPK, leading to compromised transcriptional induction of key antiviral genes such as IFNβ, IFITM3, and IL-8. Further examination revealed that during the infection process, components of the nuclear pore complex (NPC), particularly FG-Nups (such as NUP62, NUP153, NUP42, and TPR), undergo cytosolic dispersion from the nuclear envelope; NUP62 undergoes phosphorylation, and NUP42 exhibits a mobility shift in size. These observations suggest a disruption in nucleocytoplasmic trafficking. Screening efforts identified the IBV nucleocapsid (N) protein as the agent responsible for the cytoplasmic distribution of FG-Nups, subsequently hindering the nuclear entry of transcription factors and suppressing the expression of antiviral genes. Interactome analysis further revealed that the IBV N protein interacts with the scaffold protein RACK1, facilitating the recruitment of activated protein kinase C alpha (p-PKCα) to RACK1 and relocating the p-PKCα-RACK1 complex to the cytoplasm. These observations are conserved across diverse coronaviruses N proteins. Concurrently, the presence of both RACK1 and PKCα/β proved essential for the phosphorylation and cytoplasmic dispersion of NUP62, the suppression of antiviral cytokine expression, and efficient virus replication. These findings unveil a novel, highly effective, and evolutionarily conserved mechanism.
Collapse
Affiliation(s)
- Wenxiang Xue
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Hongyan Chu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Jiehuang Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Xusheng Qiu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Lei Tan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, P. R. China
| | - Ying Liao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, P. R. China
| |
Collapse
|
7
|
Mu B, Zeng Y, Luo L, Wang K. Oxidative stress-mediated protein sulfenylation in human diseases: Past, present, and future. Redox Biol 2024; 76:103332. [PMID: 39217848 PMCID: PMC11402764 DOI: 10.1016/j.redox.2024.103332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Reactive Oxygen Species (ROS) refer to a variety of derivatives of molecular oxygen that play crucial roles in regulating a wide range of physiological and pathological processes. Excessive ROS levels can cause oxidative stress, leading to cellular damage and even cell demise. However, moderately elevated levels of ROS can mediate the oxidative post-translational modifications (oxPTMs) of redox-sensitive proteins, thereby affecting protein functions and regulating various cellular signaling pathways. Among the oxPTMs, ROS-induced reversible protein sulfenylation represents the initial form of cysteine oxidation for sensing redox signaling. In this review, we will summarize the discovery, chemical formation, and detection approaches of protein sulfenylation. In addition, we will highlight recent findings for the roles of protein sulfenylation in various diseases, including thrombotic disorders, diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer.
Collapse
Affiliation(s)
- Baoquan Mu
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Zeng
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Luo
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, 610041, China.
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
9
|
Cao R, Jones DTD, Pan L, Yang A, Wang S, Padi SKR, Rawson S, Aster JC, Blacklow SC. Molecular Mechanism of PP2A/B55α Phosphatase Inhibition by IER5. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.29.555174. [PMID: 37693604 PMCID: PMC10491241 DOI: 10.1101/2023.08.29.555174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
PP2A serine/threonine phosphatases are heterotrimeric complexes that execute many essential physiologic functions. These activities are modulated by additional regulatory proteins, such as ARPP19, FAM122A, and IER5. Here, we report the cryoelectron microscopy structure of a complex of PP2A/B55α with the N-terminal structured region of IER5 (IER5-N50), which occludes a surface on B55α used for substrate recruitment, and show that IER5-N50 inhibits PP2A/B55α catalyzed dephosphorylation of pTau in biochemical assays. Mutations of full-length IER5 that disrupt its PP2A/B55α interface interfere with co-immunoprecipitation of PP2A/B55α. These mutations and deletions that remove the nuclear localization sequence of IER5 suppress cellular events such as KRT1 expression that depend on association of IER5 with PP2A/B55α. Querying the Alphafold2 predicted structure database identified SERTA domain proteins as high-confidence PP2A/B55α-binding structural homologs of IER5-N50. These studies define the molecular basis of PP2A/B55α inhibition by IER5-family proteins and suggest a roadmap for selective pharmacologic modulation of PP2A/B55α complexes.
Collapse
Affiliation(s)
- Ruili Cao
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Daniel TD Jones
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Li Pan
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Annie Yang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shumei Wang
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sathish K. R. Padi
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Shaun Rawson
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jon C Aster
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Stephen C Blacklow
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
10
|
Walker RL, Hornicek FJ, Duan Z. Transcriptional regulation and therapeutic potential of cyclin-dependent kinase 9 (CDK9) in sarcoma. Biochem Pharmacol 2024; 226:116342. [PMID: 38848777 DOI: 10.1016/j.bcp.2024.116342] [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: 04/01/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Sarcomas include various subtypes comprising two significant groups - soft tissue and bone sarcomas. Although the survival rate for some sarcoma subtypes has improved over time, the current methods of treatment remain efficaciously limited, as recurrent, and metastatic diseases remain a major obstacle. There is a need for better options and therapeutic strategies in treating sarcoma. Cyclin dependent kinase 9 (CDK9) is a transcriptional kinase and has emerged as a promising target for treating various cancers. The aberrant expression and activation of CDK9 have been observed in several sarcoma subtypes, including rhabdomyosarcoma, synovial sarcoma, osteosarcoma, Ewing sarcoma, and chordoma. Enhanced CDK9 expression has also been correlated with poorer prognosis in sarcoma patients. As a master regulator of transcription, CDK9 promotes transcription elongation by phosphorylation and releasing RNA polymerase II (RNAPII) from its promoter proximal pause. Release of RNAPII from this pause induces transcription of critical genes in the tumor cell. Overexpression and activation of CDK9 have been observed to lead to the expression of oncogenes, including MYC and MCL-1, that aid sarcoma development and progression. Inhibition of CDK9 in sarcoma has been proven to reduce these oncogenes' expression and decrease proliferation and growth in different sarcoma cells. Currently, there are several CDK9 inhibitors in preclinical and clinical investigations. This review aims to highlight the recent discovery and results on the transcriptional role and therapeutic potential of CDK9 in sarcoma.
Collapse
Affiliation(s)
- Robert L Walker
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA.
| |
Collapse
|
11
|
Wu F, Akbar H, Wang C, Yuan X, Dou Z, Mullen M, Niu L, Zhang L, Zang J, Wang Z, Yao X, Song X, Liu X. Sgo1 interacts with CENP-A to guide accurate chromosome segregation in mitosis. J Mol Cell Biol 2024; 15:mjad061. [PMID: 37777834 PMCID: PMC11181942 DOI: 10.1093/jmcb/mjad061] [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: 09/05/2022] [Revised: 02/21/2023] [Accepted: 09/29/2023] [Indexed: 10/02/2023] Open
Abstract
Shugoshin-1 (Sgo1) is necessary for maintaining sister centromere cohesion and ensuring accurate chromosome segregation during mitosis. It has been reported that the localization of Sgo1 at the centromere is dependent on Bub1-mediated phosphorylation of histone H2A at T120. However, it remains uncertain whether other centromeric proteins play a role in regulating the localization and function of Sgo1 during mitosis. Here, we show that CENP-A interacts with Sgo1 and determines the localization of Sgo1 to the centromere during mitosis. Further biochemical characterization revealed that lysine and arginine residues in the C-terminal domain of Sgo1 are critical for binding CENP-A. Interestingly, the replacement of these basic amino acids with acidic amino acids perturbed the localization of Sgo1 and Aurora B to the centromere, resulting in aberrant chromosome segregation and premature chromatid separation. Taken together, these findings reveal a previously unrecognized but direct link between Sgo1 and CENP-A in centromere plasticity control and illustrate how the Sgo1-CENP-A interaction guides accurate cell division.
Collapse
Affiliation(s)
- Fengge Wu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Hameed Akbar
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Chunyue Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Xiao Yuan
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Zhen Dou
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
- Keck Center for Cellular Dynamics, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - McKay Mullen
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Keck Center for Cellular Dynamics, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Liwen Niu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Liang Zhang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Jianye Zang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
| | - Zhikai Wang
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Xiaoyu Song
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics, Center for Advanced Interdisciplinary Science and Biomedicine of Institute of Health and Medicine (IHM), University of Science and Technology of China School of Life Sciences, Hefei 230026, China
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, Hefei National Center for Cross-disciplinary Sciences, Hefei 230027, China
| |
Collapse
|
12
|
Zhang H, Xie P. The mechanisms of microcystin-LR-induced genotoxicity and neurotoxicity in fish and mammals: Bibliometric analysis and meta-analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167018. [PMID: 37709090 DOI: 10.1016/j.scitotenv.2023.167018] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/03/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Microcystin-leucine arginine (MC-LR) is a typical cyanobacterial toxin, and the threat of this toxin is increasing among organisms. Despite extensive toxicological studies on MC-LR, there is no comprehensive analysis based on previously published data. Therefore, we conducted bibliometric analysis and meta-analysis to identify research hotspots and to elucidate the key mechanism of the relationship between MC-LR and genotoxicity and neurotoxicity among fish and mammals. One of the hotspots is toxic mechanisms (indicated by the frequent appearance of oxidative stress, DNA damage, apoptosis, neurotoxicity, genotoxicity, ROS, comet assay, signalling pathway, and gene expression indicate as keywords). The density visualization shows a high frequency of "microcystin-LR" and "toxicology," and the overlay visualization emphasizes the prominence of "neurotoxicity" in recent years. These findings confirm the importance of studying MC-LR toxicity. Meta-analysis indicated that in both fish and mammals, MC-LR exposure increased ROS levels by 294 % and increased DNA damage biomarkers by 174 % but decreased neurotoxicity biomarkers by 9 %. Intergroup comparisons revealed that the exposure concentration of MC-LR was significantly correlated with genotoxicity and neurotoxicity levels in both fish and mammals (p < 0.05). Furthermore, the random forest (RF) model revealed that exposure concentration was the primary determinant associated with the induction of ROS, genotoxicity, and neurotoxicity induced by MC-LR. This is likely the dominant mechanism by which excessive ROS production induced by MC-LR causes oxidative stress, ultimately leading to genotoxicity and neurotoxicity in both fish and mammals.
Collapse
Affiliation(s)
- Huixia Zhang
- Institute for Ecological Research and Pollution Control of Plateau Lakes; School of Ecology and Environmental Sciences, Yunnan University, Kunming 650500, PR China
| | - Ping Xie
- Institute for Ecological Research and Pollution Control of Plateau Lakes; School of Ecology and Environmental Sciences, Yunnan University, Kunming 650500, PR China; Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, PR China.
| |
Collapse
|
13
|
Keaton JM, Workman BG, Xie L, Paulson JR. Analog-sensitive Cdk1 as a tool to study mitotic exit: protein phosphatase 1 is required downstream from Cdk1 inactivation in budding yeast. Chromosome Res 2023; 31:27. [PMID: 37690059 DOI: 10.1007/s10577-023-09736-6] [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: 04/06/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023]
Abstract
We show that specific inactivation of the protein kinase Cdk1/cyclin B (Cdc28/Clb2) triggers exit from mitosis in the budding yeast Saccharomyces cerevisiae. Cells carrying the allele cdc28-as1, which makes Cdk1 (Cdc28) uniquely sensitive to the ATP analog 1NM-PP1, were arrested with spindle poisons and then treated with 1NM-PP1 to inhibit Cdk1. This caused the cells to leave mitosis and enter G1-phase as shown by initiation of rebudding (without cytokinesis), induction of mating projections ("shmoos") by α-factor, stabilization of Sic1, and degradation of Clb2. It is known that Cdk1 must be inactivated for cells to exit mitosis, but our results show that inactivation of Cdk1 is not only necessary but also sufficient to initiate the transition from mitosis to G1-phase. This result suggests a system in which to test requirements for particular gene products downstream from Cdk1 inactivation, for example, by combining cdc28-as1 with conditional mutations in the genes of interest. Using this approach, we demonstrate that protein phosphatase 1 (PPase1; Glc7 in S. cerevisiae) is required for mitotic exit and reestablishment of interphase following Cdk1 inactivation. This system could be used to test the need for other protein phosphatases downstream from Cdk1 inactivation, such as PPase 2A and Cdc14, and it could be combined with phosphoproteomics to gain information about the substrates that the various phosphatases act upon during mitotic exit.
Collapse
Affiliation(s)
- Jason M Keaton
- Acacia Safety Consulting, Inc, P.O. Box 342603, Milwaukee, WI, 53234, USA
- Department of Chemistry, University of Wisconsin-Oshkosh, Oshkosh, WI, 54901, USA
| | - Benjamin G Workman
- Department of Chemistry, University of Wisconsin-Oshkosh, Oshkosh, WI, 54901, USA
| | - Linfeng Xie
- Department of Chemistry, University of Wisconsin-Oshkosh, Oshkosh, WI, 54901, USA
| | - James R Paulson
- Department of Chemistry, University of Wisconsin-Oshkosh, Oshkosh, WI, 54901, USA.
| |
Collapse
|
14
|
Partscht P, Schiebel E. The diverging role of CDC14B: from mitotic exit in yeast to cell fate control in humans. EMBO J 2023; 42:e114364. [PMID: 37493185 PMCID: PMC10425841 DOI: 10.15252/embj.2023114364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/22/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023] Open
Abstract
CDC14, originally identified as crucial mediator of mitotic exit in budding yeast, belongs to the family of dual-specificity phosphatases (DUSPs) that are present in most eukaryotes. Contradicting data have sparked a contentious discussion whether a cell cycle role is conserved in the human paralogs CDC14A and CDC14B but possibly masked due to redundancy. Subsequent studies on CDC14A and CDC14B double knockouts in human and mouse demonstrated that CDC14 activity is dispensable for mitotic progression in higher eukaryotes and instead suggested functional specialization. In this review, we provide a comprehensive overview of our current understanding of how faithful cell division is linked to phosphorylation and dephosphorylation and compare functional similarities and divergences between the mitotic phosphatases CDC14, PP2A, and PP1 from yeast and higher eukaryotes. Furthermore, we review the latest discoveries on CDC14B, which identify this nuclear phosphatase as a key regulator of gene expression and reveal its role in neuronal development. Finally, we discuss CDC14B functions in meiosis and possible implications in other developmental processes.
Collapse
Affiliation(s)
- Patrick Partscht
- Zentrum für Molekulare BiologieUniversität Heidelberg, DKFZ‐ZMBH AllianzHeidelbergGermany
| | - Elmar Schiebel
- Zentrum für Molekulare BiologieUniversität Heidelberg, DKFZ‐ZMBH AllianzHeidelbergGermany
| |
Collapse
|
15
|
Heidari B, Nemie-Feyissa D, Lillo C. Distinct Clades of Protein Phosphatase 2A Regulatory B'/B56 Subunits Engage in Different Physiological Processes. Int J Mol Sci 2023; 24:12255. [PMID: 37569631 PMCID: PMC10418862 DOI: 10.3390/ijms241512255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Protein phosphatase 2A (PP2A) is a strongly conserved and major protein phosphatase in all eukaryotes. The canonical PP2A complex consists of a catalytic (C), scaffolding (A), and regulatory (B) subunit. Plants have three groups of evolutionary distinct B subunits: B55, B' (B56), and B''. Here, the Arabidopsis B' group is reviewed and compared with other eukaryotes. Members of the B'α/B'β clade are especially important for chromatid cohesion, and dephosphorylation of transcription factors that mediate brassinosteroid (BR) signaling in the nucleus. Other B' subunits interact with proteins at the cell membrane to dampen BR signaling or harness immune responses. The transition from vegetative to reproductive phase is influenced differentially by distinct B' subunits; B'α and B'β being of little importance, whereas others (B'γ, B'ζ, B'η, B'θ, B'κ) promote transition to flowering. Interestingly, the latter B' subunits have three motifs in a conserved manner, i.e., two docking sites for protein phosphatase 1 (PP1), and a POLO consensus phosphorylation site between these motifs. This supports the view that a conserved PP1-PP2A dephosphorelay is important in a variety of signaling contexts throughout eukaryotes. A profound understanding of these regulators may help in designing future crops and understand environmental issues.
Collapse
Affiliation(s)
| | | | - Cathrine Lillo
- IKBM, Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036 Stavanger, Norway; (B.H.); (D.N.-F.)
| |
Collapse
|
16
|
Vertyshev AY, Akberdin IR, Kolpakov FA. Numerous Trigger-like Interactions of Kinases/Protein Phosphatases in Human Skeletal Muscles Can Underlie Transient Processes in Activation of Signaling Pathways during Exercise. Int J Mol Sci 2023; 24:11223. [PMID: 37446402 DOI: 10.3390/ijms241311223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Optimizing physical training regimens to increase muscle aerobic capacity requires an understanding of the internal processes that occur during exercise that initiate subsequent adaptation. During exercise, muscle cells undergo a series of metabolic events that trigger downstream signaling pathways and induce the expression of many genes in working muscle fibers. There are a number of studies that show the dependence of changes in the activity of AMP-activated protein kinase (AMPK), one of the mediators of cellular signaling pathways, on the duration and intensity of single exercises. The activity of various AMPK isoforms can change in different directions, increasing for some isoforms and decreasing for others, depending on the intensity and duration of the load. This review summarizes research data on changes in the activity of AMPK, Ca2+/calmodulin-dependent protein kinase II (CaMKII), and other components of the signaling pathways in skeletal muscles during exercise. Based on these data, we hypothesize that the observed changes in AMPK activity may be largely related to metabolic and signaling transients rather than exercise intensity per se. Probably, the main events associated with these transients occur at the beginning of the exercise in a time window of about 1-10 min. We hypothesize that these transients may be partly due to putative trigger-like kinase/protein phosphatase interactions regulated by feedback loops. In addition, numerous dynamically changing factors, such as [Ca2+], metabolite concentration, and reactive oxygen and nitrogen species (RONS), can shift the switching thresholds and change the states of these triggers, thereby affecting the activity of kinases (in particular, AMPK and CaMKII) and phosphatases. The review considers the putative molecular mechanisms underlying trigger-like interactions. The proposed hypothesis allows for a reinterpretation of the experimental data available in the literature as well as the generation of ideas to optimize future training regimens.
Collapse
Affiliation(s)
| | - Ilya R Akberdin
- Department of Computational Biology, Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Biosoft.Ru, Ltd., 630058 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Fedor A Kolpakov
- Department of Computational Biology, Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Biosoft.Ru, Ltd., 630058 Novosibirsk, Russia
- Federal Research Center for Information and Computational Technologies, 630090 Novosibirsk, Russia
| |
Collapse
|
17
|
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
|
18
|
Keaton JM, Workman BG, Xie L, Paulson JR. Exit from Mitosis in Budding Yeast: Protein Phosphatase 1 is Required Downstream from Cdk1 Inactivation. RESEARCH SQUARE 2023:rs.3.rs-2787001. [PMID: 37090579 PMCID: PMC10120774 DOI: 10.21203/rs.3.rs-2787001/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
We show that inactivation of the protein kinase Cdk1/Cyclin B (Cdc28/Clb 2 in the budding yeast Saccharomyces cerevisiae ) is not only necessary for cells to leave mitosis, as is well known, but also sufficient to trigger mitotic exit. Cells carrying the mutation cdc28-as1 , which makes Cdc28 (Cdk1) uniquely sensitive to the ATP analog 1NM-PP1, were arrested with spindle poisons and then treated with 1NM-PP1 to inhibit Cdk1. This treatment caused the cells to exit mitosis and enter G1-phase as shown by initiation of rebudding (without cytokinesis), production of "shmoos" (when α-factor was present), stabilization of Sic1, and degradation of Clb2. This result provides a system in which to test whether particular gene products are required downstream from Cdk1 inactivation in exit from mitosis. In this system, the mutation cdc28-as1 is combined with a conditional mutation in the gene of interest. Using this approach, we demonstrate that Protein Phosphatase 1 (PPase1; Glc7 in S. cerevisiae ) is required for reestablishment of G1-phase following Cdk1 inactivation. This system could be used to test whether other protein phosphatases are also needed downstream from Cdk1 inactivation, and it could be combined with phosphoproteomics to gain information about the substrates those phosphatases act on during mitotic exit.
Collapse
|
19
|
The TFIIS N-terminal domain (TND): a transcription assembly module at the interface of order and disorder. Biochem Soc Trans 2023; 51:125-135. [PMID: 36651856 PMCID: PMC9987994 DOI: 10.1042/bst20220342] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023]
Abstract
Interaction scaffolds that selectively recognize disordered protein strongly shape protein interactomes. An important scaffold of this type that contributes to transcription is the TFIIS N-terminal domain (TND). The TND is a five-helical bundle that has no known enzymatic activity, but instead selectively reads intrinsically disordered sequences of other proteins. Here, we review the structural and functional properties of TNDs and their cognate disordered ligands known as TND-interacting motifs (TIMs). TNDs or TIMs are found in prominent members of the transcription machinery, including TFIIS, super elongation complex, SWI/SNF, Mediator, IWS1, SPT6, PP1-PNUTS phosphatase, elongin, H3K36me3 readers, the transcription factor MYC, and others. We also review how the TND interactome contributes to the regulation of transcription. Because the TND is the most significantly enriched fold among transcription elongation regulators, TND- and TIM-driven interactions have widespread roles in the regulation of many transcriptional processes.
Collapse
|
20
|
Sun X, Wang D, Li W, Gao Q, Tao J, Liu H. Comprehensive analysis of nonsurrounded nucleolus and surrounded nucleolus oocytes on chromatin accessibility using ATAC-seq. Mol Reprod Dev 2023; 90:87-97. [PMID: 36598871 DOI: 10.1002/mrd.23668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/13/2022] [Accepted: 12/25/2022] [Indexed: 01/05/2023]
Abstract
Mouse germinal vesicle (GV) oocytes are divided into surrounded nucleolus (SN) and nonsurrounded nucleolus (NSN) oocytes based on chromatin morphology. NSN oocytes spontaneously transform into SN oocytes after accumulating enough maternal transcripts. SN oocytes show transcriptional silencing. When oocyte maturation is abnormal or takes place in vitro, NSN oocytes do not go through SN stage before proceeding to MII. Nontransitive oocytes show developmental retardation, a low fertilization rate, and arrest at the two-cell embryo stage in mice. Here, chromatin-binding ribonucleic acid polymerase II (RNAP II) activity, newly synthesized RNA, and chromatin accessibility in GV oocytes were examined. In SN oocytes, RNAP II did not bind to DNA, neo-RNA was not generated in nuclei, and the phosphorylation state of RNAP II did not affect the chromatin-binding activity. The number of accessible genes in SN oocytes was remarkably lower than that in NSN oocytes. The accessibility of different functional genes was also different between the two types of oocytes. Thus, low chromatin accessibility leads to transcriptional silencing in SN oocytes.
Collapse
Affiliation(s)
- Xiaofan Sun
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Dayu Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Weijian Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qian Gao
- Laboratory Animal Center, College of Veterinary Medicine, Nanjing Agriculture University, Nanjing, China
| | - Jingli Tao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Honglin Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
21
|
Villarroya‐Beltri C, Martins AFB, García A, Giménez D, Zarzuela E, Novo M, del Álamo C, González‐Martínez J, Bonel‐Pérez GC, Díaz I, Guillamot M, Chiesa M, Losada A, Graña‐Castro O, Rovira M, Muñoz J, Salazar‐Roa M, Malumbres M. Mammalian CDC14 phosphatases control exit from stemness in pluripotent cells. EMBO J 2023; 42:e111251. [PMID: 36326833 PMCID: PMC9811616 DOI: 10.15252/embj.2022111251] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 10/17/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
Maintenance of stemness is tightly linked to cell cycle regulation through protein phosphorylation by cyclin-dependent kinases (CDKs). However, how this process is reversed during differentiation is unknown. We report here that exit from stemness and differentiation of pluripotent cells along the neural lineage are controlled by CDC14, a CDK-counteracting phosphatase whose function in mammals remains obscure. Lack of the two CDC14 family members, CDC14A and CDC14B, results in deficient development of the neural system in the mouse and impairs neural differentiation from embryonic stem cells (ESCs). Mechanistically, CDC14 directly dephosphorylates specific proline-directed Ser/Thr residues of undifferentiated embryonic transcription Factor 1 (UTF1) during the exit from stemness, triggering its proteasome-dependent degradation. Multiomic single-cell analysis of transcription and chromatin accessibility in differentiating ESCs suggests that increased UTF1 levels in the absence of CDC14 prevent the proper firing of bivalent promoters required for differentiation. CDC14 phosphatases are dispensable for mitotic exit, suggesting that CDC14 phosphatases have evolved to control stemness rather than cell cycle exit and establish the CDK-CDC14 axis as a critical molecular switch for linking cell cycle regulation and self-renewal.
Collapse
Affiliation(s)
| | - Ana Filipa B Martins
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Alejandro García
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | | | | | - Mónica Novo
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Cristina del Álamo
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | | | - Gloria C Bonel‐Pérez
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Irene Díaz
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - María Guillamot
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Massimo Chiesa
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Ana Losada
- Chromosome Dynamics groupCNIOMadridSpain
| | - Osvaldo Graña‐Castro
- Bioinformatics UnitCNIOMadridSpain
- Present address:
Department of Basic Medical Sciences, Institute of Applied Molecular Medicine (IMMA‐Nemesio Díez), School of MedicineSan Pablo‐CEU University, CEU UniversitiesBoadilla del MonteSpain
| | - Meritxell Rovira
- Department of Physiological Science, School of Medicine, L'Hospitalet de LlobregatUniversity of Barcelona (UB)BarcelonaSpain
- Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, P‐CMR[C]Institut d'Investigació Biomèdica de Bellvitge—IDIBELL, L'Hospitalet de LlobregatBarcelonaSpain
| | | | - María Salazar‐Roa
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
- Present address:
Advanced Therapies and Cancer Group, Faculty of BiologyComplutense UniversityMadridSpain
| | - Marcos Malumbres
- Cell Division and Cancer groupSpanish National Cancer Research Centre (CNIO)MadridSpain
| |
Collapse
|
22
|
Morgado-Palacin L. Elvan Böke: Long live the oocyte. J Cell Biol 2022; 221:213560. [PMID: 36264230 PMCID: PMC9587376 DOI: 10.1083/jcb.202210049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Elvan Böke investigates the mechanisms that preserve the viability of dormant oocytes.
Collapse
|
23
|
Francois L, Boskovic P, Knerr J, He W, Sigismondo G, Schwan C, More TH, Schlotter M, Conway ME, Krijgsveld J, Hiller K, Grosse R, Lichter P, Radlwimmer B. BCAT1 redox function maintains mitotic fidelity. Cell Rep 2022; 41:111524. [PMID: 36260995 DOI: 10.1016/j.celrep.2022.111524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
The metabolic enzyme branched-chain amino acid transaminase 1 (BCAT1) drives cell proliferation in aggressive cancers such as glioblastoma. Here, we show that BCAT1 localizes to mitotic structures and has a non-metabolic function as a mitotic regulator. Furthermore, BCAT1 is required for chromosome segregation in cancer and induced pluripotent stem cells and tumor growth in human cerebral organoid and mouse syngraft models. Applying gene knockout and rescue strategies, we show that the BCAT1 CXXC redox motif is crucial for controlling cysteine sulfenylation specifically in mitotic cells, promoting Aurora kinase B localization to centromeres, and securing accurate chromosome segregation. These findings offer an explanation for the well-established role of BCAT1 in promoting cancer cell proliferation. In summary, our data establish BCAT1 as a component of the mitotic apparatus that safeguards mitotic fidelity through a moonlighting redox functionality.
Collapse
Affiliation(s)
- Liliana Francois
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Pavle Boskovic
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Julian Knerr
- Institute of Pharmacology, University of Freiburg, 79102 Freiburg, Germany
| | - Wei He
- Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, and Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, 38092 Braunschweig, Germany
| | - Gianluca Sigismondo
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Carsten Schwan
- Institute of Pharmacology, University of Freiburg, 79102 Freiburg, Germany
| | - Tushar H More
- Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, and Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, 38092 Braunschweig, Germany
| | - Magdalena Schlotter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Myra E Conway
- College of Science and Engineering, University of Derby, Derby DE22 1GB, UK
| | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Karsten Hiller
- Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, and Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, 38092 Braunschweig, Germany
| | - Robert Grosse
- Institute of Pharmacology, University of Freiburg, 79102 Freiburg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Bernhard Radlwimmer
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
24
|
Archambault V, Li J, Emond-Fraser V, Larouche M. Dephosphorylation in nuclear reassembly after mitosis. Front Cell Dev Biol 2022; 10:1012768. [PMID: 36268509 PMCID: PMC9576876 DOI: 10.3389/fcell.2022.1012768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
In most animal cell types, the interphase nucleus is largely disassembled during mitotic entry. The nuclear envelope breaks down and chromosomes are compacted into separated masses. Chromatin organization is also mostly lost and kinetochores assemble on centromeres. Mitotic protein kinases play several roles in inducing these transformations by phosphorylating multiple effector proteins. In many of these events, the mechanistic consequences of phosphorylation have been characterized. In comparison, how the nucleus reassembles at the end of mitosis is less well understood in mechanistic terms. In recent years, much progress has been made in deciphering how dephosphorylation of several effector proteins promotes nuclear envelope reassembly, chromosome decondensation, kinetochore disassembly and interphase chromatin organization. The precise roles of protein phosphatases in this process, in particular of the PP1 and PP2A groups, are emerging. Moreover, how these enzymes are temporally and spatially regulated to ensure that nuclear reassembly progresses in a coordinated manner has been partly uncovered. This review provides a global view of nuclear reassembly with a focus on the roles of dephosphorylation events. It also identifies important open questions and proposes hypotheses.
Collapse
Affiliation(s)
- Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
- *Correspondence: Vincent Archambault,
| | - Jingjing Li
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Virginie Emond-Fraser
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Myreille Larouche
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
25
|
Deng DJ, Wang X, Yue KY, Wang Y, Jin QW. Analysis of the potential role of fission yeast PP2A in spindle assembly checkpoint inactivation. FASEB J 2022; 36:e22524. [PMID: 36006032 DOI: 10.1096/fj.202101884r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/11/2022]
Abstract
As a surveillance mechanism, the activated spindle assembly checkpoint (SAC) potently inhibits the E3 ubiquitin ligase APC/C (anaphase-promoting complex/cyclosome) to ensure accurate chromosome segregation. Although the protein phosphatase 2A (PP2A) has been proposed to be both, directly and indirectly, involved in spindle assembly checkpoint inactivation in mammalian cells, whether it is similarly operating in the fission yeast Schizosaccharomycer pombe has never been demonstrated. Here, we investigated whether fission yeast PP2A is involved in SAC silencing by following the rate of cyclin B (Cdc13) destruction at SPBs during the recovery phase in nda3-KM311 cells released from the inhibition of APC/C by the activated spindle checkpoint. The timing of the SAC inactivation is only slightly delayed when two B56 regulatory subunits (Par1 and Par2) of fission yeast PP2A are absent. Overproduction of individual PP2A subunits either globally in the nda3-KM311 arrest-and-release system or locally in the synthetic spindle checkpoint activation system only slightly suppresses the SAC silencing defects in PP1 deletion (dis2Δ) cells. Our study thus demonstrates that the fission yeast PP2A is not a key regulator actively involved in SAC inactivation.
Collapse
Affiliation(s)
- Da-Jie Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Xi Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Kai-Ye Yue
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yamei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Quan-Wen Jin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
26
|
Chica N, Portantier M, Nyquist-Andersen M, Espada-Burriel S, Lopez-Aviles S. Uncoupling of Mitosis and Cytokinesis Upon a Prolonged Arrest in Metaphase Is Influenced by Protein Phosphatases and Mitotic Transcription in Fission Yeast. Front Cell Dev Biol 2022; 10:876810. [PMID: 35923846 PMCID: PMC9340479 DOI: 10.3389/fcell.2022.876810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/11/2022] [Indexed: 11/22/2022] Open
Abstract
Depletion of the Anaphase-Promoting Complex/Cyclosome (APC/C) activator Cdc20 arrests cells in metaphase with high levels of the mitotic cyclin (Cyclin B) and the Separase inhibitor Securin. In mammalian cells this arrest has been exploited for the treatment of cancer with drugs that engage the spindle assembly checkpoint and, recently, with chemical inhibitors of the APC/C. While most cells arrested in mitosis for prolonged periods undergo apoptosis, others skip cytokinesis and enter G1 with unsegregated chromosomes. This process, known as mitotic slippage, generates aneuploidy and increases genomic instability in the cancer cell. Here, we analyze the behavior of fission yeast cells arrested in mitosis through the transcriptional silencing of the Cdc20 homolog slp1. While depletion of slp1 readily halts cells in metaphase, this arrest is only transient and a majority of cells eventually undergo cytokinesis and show steady mitotic dephosphorylation. Notably, this occurs in the absence of Cyclin B (Cdc13) degradation. We investigate the involvement of phosphatase activity in these events and demonstrate that PP2A-B55Pab1 is required to prevent septation and, during the arrest, its CDK-mediated inhibition facilitates the induction of cytokinesis. In contrast, deletion of PP2A-B56Par1 completely abrogates septation. We show that this effect is partly due to this mutant entering mitosis with reduced CDK activity. Interestingly, both PP2A-B55Pab1 and PP2A-B56Par1, as well as Clp1 (the homolog of the budding yeast mitotic phosphatase Cdc14) are required for the dephosphorylation of mitotic substrates during the escape. Finally, we show that the mitotic transcriptional wave controlled by the RFX transcription factor Sak1 facilitates the induction of cytokinesis and also requires the activity of PP2A-B56Par1 in a mechanism independent of CDK.
Collapse
Affiliation(s)
- Nathalia Chica
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL partnership, Faculty of Medicine, University of Oslo, Oslo, Norway
- *Correspondence: Sandra Lopez-Aviles, ; Nathalia Chica,
| | - Marina Portantier
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL partnership, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Mari Nyquist-Andersen
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL partnership, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Silvia Espada-Burriel
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL partnership, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sandra Lopez-Aviles
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL partnership, Faculty of Medicine, University of Oslo, Oslo, Norway
- Institute of Biosciences (IBV), Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- *Correspondence: Sandra Lopez-Aviles, ; Nathalia Chica,
| |
Collapse
|
27
|
Basu S, Greenwood J, Jones AW, Nurse P. Core control principles of the eukaryotic cell cycle. Nature 2022; 607:381-386. [PMID: 35676478 PMCID: PMC9279155 DOI: 10.1038/s41586-022-04798-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/25/2022] [Indexed: 12/23/2022]
Abstract
Cyclin-dependent kinases (CDKs) lie at the heart of eukaryotic cell cycle control, with different cyclin-CDK complexes initiating DNA replication (S-CDKs) and mitosis (M-CDKs)1,2. However, the principles on which cyclin-CDK complexes organize the temporal order of cell cycle events are contentious3. One model proposes that S-CDKs and M-CDKs are functionally specialized, with substantially different substrate specificities to execute different cell cycle events4-6. A second model proposes that S-CDKs and M-CDKs are redundant with each other, with both acting as sources of overall CDK activity7,8. In this model, increasing CDK activity, rather than CDK substrate specificity, orders cell cycle events9,10. Here we reconcile these two views of core cell cycle control. Using phosphoproteomic assays of in vivo CDK activity in fission yeast, we find that S-CDK and M-CDK substrate specificities are remarkably similar, showing that S-CDKs and M-CDKs are not completely specialized for S phase and mitosis alone. Normally, S-CDK cannot drive mitosis but can do so when protein phosphatase 1 is removed from the centrosome. Thus, increasing S-CDK activity in vivo is sufficient to overcome substrate specificity differences between S-CDK and M-CDK, and allows S-CDK to carry out M-CDK function. Therefore, we unite the two opposing views of cell cycle control, showing that the core cell cycle engine is largely based on a quantitative increase in CDK activity through the cell cycle, combined with minor and surmountable qualitative differences in catalytic specialization of S-CDKs and M-CDKs.
Collapse
Affiliation(s)
- Souradeep Basu
- Cell Cycle Laboratory, The Francis Crick Institute, London, UK. .,DeepMind, London, UK.
| | | | - Andrew W Jones
- Cell Cycle Laboratory, The Francis Crick Institute, London, UK
| | - Paul Nurse
- Cell Cycle Laboratory, The Francis Crick Institute, London, UK.,Laboratory of Yeast Genetics and Cell Biology, Rockefeller University, New York, NY, USA
| |
Collapse
|
28
|
Cell cycle involvement in cancer therapy; WEE1 kinase, a potential target as therapeutic strategy. Mutat Res 2022; 824:111776. [PMID: 35247630 DOI: 10.1016/j.mrfmmm.2022.111776] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/22/2022]
Abstract
Mitosis is the process of cell division and is regulated by checkpoints in the cell cycle. G1-S, S, and G2-M are the three main checkpoints that prevent initiation of the next phase of the cell cycle phase until previous phase has completed. DNA damage leads to activation of the G2-M checkpoint, which can trigger a downstream DNA damage response (DDR) pathway to induce cell cycle arrest while the damage is repaired. If the DNA damage cannot be repaired, the replication stress response (RSR) pathway finally leads to cell death by apoptosis, in this case called mitotic catastrophe. Many cancer treatments (chemotherapy and radiotherapy) cause DNA damages based on SSBs (single strand breaks) or DSBs (double strand breaks), which cause cell death through mitotic catastrophe. However, damaged cells can activate WEE1 kinase (as a part of the DDR and RSR pathways), which prevents apoptosis and cell death by inducing cell cycle arrest at G2 phase. Therefore, inhibition of WEE1 kinase could sensitize cancer cells to chemotherapeutic drugs. This review focuses on the role of WEE1 kinase (as a biological macromolecule which has a molecular mass of 96 kDa) in the cell cycle, and its interactions with other regulatory pathways. In addition, we discuss the potential of WEE1 inhibition as a new therapeutic approach in the treatment of various cancers, such as melanoma, breast cancer, pancreatic cancer, cervical cancer, etc.
Collapse
|
29
|
Yamada C, Morooka A, Miyazaki S, Nagai M, Mase S, Iemura K, Tasnin MN, Takuma T, Nakamura S, Morshed S, Koike N, Mostofa MG, Rahman MA, Sharmin T, Katsuta H, Ohara K, Tanaka K, Ushimaru T. TORC1 inactivation promotes APC/C-dependent mitotic slippage in yeast and human cells. iScience 2022; 25:103675. [PMID: 35141499 PMCID: PMC8814761 DOI: 10.1016/j.isci.2021.103675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 10/20/2021] [Accepted: 12/20/2021] [Indexed: 12/31/2022] Open
Abstract
Unsatisfied kinetochore-microtubule attachment activates the spindle assembly checkpoint to inhibit the metaphase-anaphase transition. However, some cells eventually override mitotic arrest by mitotic slippage. Here, we show that inactivation of TORC1 kinase elicits mitotic slippage in budding yeast and human cells. Yeast mitotic slippage was accompanied with aberrant aspects, such as degradation of the nucleolar protein Net1, release of phosphatase Cdc14, and anaphase-promoting complex/cyclosome (APC/C)-Cdh1-dependent degradation of securin and cyclin B in metaphase. This mitotic slippage caused chromosome instability. In human cells, mammalian TORC1 (mTORC1) inactivation also invoked mitotic slippage, indicating that TORC1 inactivation-induced mitotic slippage is conserved from yeast to mammalian cells. However, the invoked mitotic slippage in human cells was not dependent on APC/C-Cdh1. This study revealed an unexpected involvement of TORC1 in mitosis and provides information on undesirable side effects of the use of TORC1 inhibitors as immunosuppressants and anti-tumor drugs. Yeast TORC1 inhibition promotes Net1 degradation and Cdc14 release Yeast TORC1 inhibition invokes mitotic slippage in an APC/C-Cdh1-dependent manner Human mTORC1 inhibition also elicits mitotic slippage
Collapse
Affiliation(s)
- Chihiro Yamada
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8021, Japan
| | - Aya Morooka
- Department of Biological Science, Faculty of Science, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan
| | - Seira Miyazaki
- Department of Biological Science, Faculty of Science, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan
| | - Masayoshi Nagai
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8021, Japan.,Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Satoru Mase
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8021, Japan
| | - Kenji Iemura
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Most Naoshia Tasnin
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8021, Japan
| | - Tsuneyuki Takuma
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8021, Japan
| | - Shotaro Nakamura
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8021, Japan
| | - Shamsul Morshed
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8021, Japan
| | - Naoki Koike
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8021, Japan
| | - Md Golam Mostofa
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8021, Japan
| | - Muhammad Arifur Rahman
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8021, Japan
| | - Tasnuva Sharmin
- Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8021, Japan
| | - Haruko Katsuta
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8021, Japan
| | - Kotaro Ohara
- Department of Biological Science, Faculty of Science, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Takashi Ushimaru
- Department of Science, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka 422-8021, Japan.,Department of Biological Science, Faculty of Science, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka 422-8529, Japan.,Graduate School of Science and Technology, Shizuoka University, Ohya 836, Suruga-ku, Shizuoka 422-8021, Japan
| |
Collapse
|
30
|
Gillani SQ, Reshi I, Nabi N, Un Nisa M, Sarwar Z, Bhat S, Roberts TM, Higgins JMG, Andrabi S. PCTAIRE1 promotes mitotic progression and resistance against antimitotic and apoptotic signals. J Cell Sci 2022; 135:jcs258831. [PMID: 35044463 PMCID: PMC8918779 DOI: 10.1242/jcs.258831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 12/29/2021] [Indexed: 10/24/2022] Open
Abstract
PCTAIRE1 (also known as CDK16) is a serine-threonine kinase implicated in physiological processes like neuronal development, vesicle trafficking, spermatogenesis and cell proliferation. However, its exact role in cell division remains unclear. In this study, using a library screening approach, we identified PCTAIRE1 among several candidates that resisted mitotic arrest and mitotic cell death induced by polyomavirus small T (PolST) expression in mammalian cells. Our study showed that PCTAIRE1 is a mitotic kinase that localizes at centrosomes during G2 and at spindle poles as the cells enter mitosis, and then at the midbody during cytokinesis. We also report that PCTAIRE1 protein levels fluctuate through the cell cycle and reach their peak at mitosis, during which there is an increase in PCTAIRE1 phosphorylation as well. Interestingly, knockdown of PCTAIRE1 resulted in aberrant mitosis by interfering with spindle assembly and chromosome segregation. Further, we found that PCTAIRE1 promotes resistance of cancer cells to antimitotic drugs, and this underscores the significance of PCTAIRE1 as a potential drug target for overcoming chemotherapeutic resistance. Taken together, these studies establish PCTAIRE1 as a critical mediator of mitotic progression and highlight its role in chemotherapeutic resistance. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Irfana Reshi
- Department of Biotechnology, University of Kashmir, Srinagar 190006, India
| | - Nusrat Nabi
- Department of Biochemistry, University of Kashmir, Srinagar 190006, India
| | - Misbah Un Nisa
- Department of Biochemistry, University of Kashmir, Srinagar 190006, India
| | - Zarka Sarwar
- Department of Biochemistry, University of Kashmir, Srinagar 190006, India
| | - Sameer Bhat
- Department of Biotechnology, University of Kashmir, Srinagar 190006, India
| | - Thomas M. Roberts
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan M. G. Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University,Newcastle upon Tyne NE2 4HH, UK
| | - Shaida Andrabi
- Department of Biochemistry, University of Kashmir, Srinagar 190006, India
| |
Collapse
|
31
|
Sergienko NM, Donner DG, Delbridge LMD, McMullen JR, Weeks KL. Protein phosphatase 2A in the healthy and failing heart: New insights and therapeutic opportunities. Cell Signal 2021; 91:110213. [PMID: 34902541 DOI: 10.1016/j.cellsig.2021.110213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/02/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
Protein phosphatases have emerged as critical regulators of phosphoprotein homeostasis in settings of health and disease. Protein phosphatase 2A (PP2A) encompasses a large subfamily of enzymes that remove phosphate groups from serine/threonine residues within phosphoproteins. The heterogeneity in PP2A structure, which arises from the grouping of different catalytic, scaffolding and regulatory subunit isoforms, creates distinct populations of catalytically active enzymes (i.e. holoenzymes) that localise to different parts of the cell. This structural complexity, combined with other regulatory mechanisms, such as interaction of PP2A heterotrimers with accessory proteins and post-translational modification of the catalytic and/or regulatory subunits, enables PP2A holoenzymes to target phosphoprotein substrates in a highly specific manner. In this review, we summarise the roles of PP2A in cardiac physiology and disease. PP2A modulates numerous processes that are vital for heart function including calcium handling, contractility, β-adrenergic signalling, metabolism and transcription. Dysregulation of PP2A has been observed in human cardiac disease settings, including heart failure and atrial fibrillation. Efforts are underway, particularly in the cancer field, to develop therapeutics targeting PP2A activity. The development of small molecule activators of PP2A (SMAPs) and other compounds that selectively target specific PP2A holoenzymes (e.g. PP2A/B56α and PP2A/B56ε) will improve understanding of the function of different PP2A species in the heart, and may lead to the development of therapeutics for normalising aberrant protein phosphorylation in settings of cardiac remodelling and dysfunction.
Collapse
Affiliation(s)
- Nicola M Sergienko
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Central Clinical School, Monash University, Clayton VIC 3800, Australia
| | - Daniel G Donner
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia
| | - Lea M D Delbridge
- Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Physiology and Department of Medicine Alfred Hospital, Monash University, Clayton VIC 3800, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora VIC 3086, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| | - Kate L Weeks
- Baker Heart and Diabetes Institute, Melbourne VIC 3004, Australia; Department of Anatomy and Physiology, The University of Melbourne, Parkville VIC 3010, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville VIC 3010, Australia; Department of Diabetes, Central Clinical School, Monash University, Clayton VIC 3800, Australia.
| |
Collapse
|
32
|
Gräf R, Grafe M, Meyer I, Mitic K, Pitzen V. The Dictyostelium Centrosome. Cells 2021; 10:cells10102657. [PMID: 34685637 PMCID: PMC8534566 DOI: 10.3390/cells10102657] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022] Open
Abstract
The centrosome of Dictyostelium amoebae contains no centrioles and consists of a cylindrical layered core structure surrounded by a corona harboring microtubule-nucleating γ-tubulin complexes. It is the major centrosomal model beyond animals and yeasts. Proteomics, protein interaction studies by BioID and superresolution microscopy methods led to considerable progress in our understanding of the composition, structure and function of this centrosome type. We discuss all currently known components of the Dictyostelium centrosome in comparison to other centrosomes of animals and yeasts.
Collapse
|
33
|
Lara-Gonzalez P, Pines J, Desai A. Spindle assembly checkpoint activation and silencing at kinetochores. Semin Cell Dev Biol 2021; 117:86-98. [PMID: 34210579 PMCID: PMC8406419 DOI: 10.1016/j.semcdb.2021.06.009] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 01/01/2023]
Abstract
The spindle assembly checkpoint (SAC) is a surveillance mechanism that promotes accurate chromosome segregation in mitosis. The checkpoint senses the attachment state of kinetochores, the proteinaceous structures that assemble onto chromosomes in mitosis in order to mediate their interaction with spindle microtubules. When unattached, kinetochores generate a diffusible inhibitor that blocks the activity of the anaphase-promoting complex/cyclosome (APC/C), an E3 ubiquitin ligase required for sister chromatid separation and exit from mitosis. Work from the past decade has greatly illuminated our understanding of the mechanisms by which the diffusible inhibitor is assembled and how it inhibits the APC/C. However, less is understood about how SAC proteins are recruited to kinetochores in the absence of microtubule attachment, how the kinetochore catalyzes formation of the diffusible inhibitor, and how attachments silence the SAC at the kinetochore. Here, we summarize current understanding of the mechanisms that activate and silence the SAC at kinetochores and highlight open questions for future investigation.
Collapse
Affiliation(s)
- Pablo Lara-Gonzalez
- Ludwig Institute for Cancer Research, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| | | | - Arshad Desai
- Ludwig Institute for Cancer Research, USA; Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
34
|
Chen C, Huang FW, Huang SS, Huang JS. IGFBP-3 and TGF-β inhibit growth in epithelial cells by stimulating type V TGF-β receptor (TβR-V)-mediated tumor suppressor signaling. FASEB Bioadv 2021; 3:709-729. [PMID: 34485840 PMCID: PMC8409558 DOI: 10.1096/fba.2021-00016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/06/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
The TGF-β type V receptor (TβR-V) mediates growth inhibition by IGFBP-3 and TGF-β in epithelial cells and loss of TβR-V expression in these cells leads to development of carcinoma. The mechanisms by which TβR-V mediates growth inhibition (tumor suppressor) signaling remain elusive. Previous studies revealed that IGFBP-3 and TGF-β inhibit growth in epithelial cells by stimulating TβR-V-mediated IRS-1/2-dependent activation and cytoplasm-to-nucleus translocation of IGFBP-3- or TGF-β-stimulated protein phosphatase (PPase), resulting in dephosphorylation of pRb-related proteins (p107, p130) or pRb, and growth arrest. To define the signaling, we characterized/identified the IGFBP-3- and TGF-β-stimulated PPases in cell lysates and nucleus fractions in Mv1Lu cells treated with IGFBP-3 and TGF-β, using a cell-free assay with 32P-labeled casein as a substrate. Both IGFBP-3- and TGF-β-stimulated PPase activities in cell lysates are abolished when cells are co-treated with TGF-β/IGFBP-3 antagonist or RAP (LRP-1/TβR-V antagonist). However, the IGFBP-3-stimulated PPase activity, but not TGF-β-stimulated PPase activity, is sensitive to inhibition by okadaic acid (OA). In addition, OA or PP2Ac siRNA reverses IGFBP-3 growth inhibition, but not TGF-β growth inhibition, in Mv1Lu and 32D cells. These suggest that IGFBP-3- and TGF-β-stimulated PPases are identical to PP2A and PP1, respectively. By Western blot/phosphorimager/immunofluorescence-microscopy analyses, IGFBP-3 and TGF-β stimulate TβR-V-mediated IRS-2-dependent activation and cytoplasm-to-nucleus translocation of PP2Ac and PP1c, resulting in dephosphorylation of p130/p107 and pRb, respectively, and growth arrest. Small molecule TGF-β enhancers, which potentiate TGF-β growth inhibition by enhancing TβR-I-TβR-II-mediated canonical signaling and thus activating TβR-V-mediated tumor suppressor signaling cascade (TβR-V/IRS-2/PP1/pRb), could be used to prevent and treat carcinoma.
Collapse
Affiliation(s)
- Chun‐Lin Chen
- Department of Biological ScienceNational Sun Yat‐sen UniversityKaohsiungTaiwan
- Departments of Biochemistry and Molecular BiologySaint Louis University School of MedicineSt. LouisMOUSA
| | - Franklin W. Huang
- Division of Hematology and OncologyDepartment of MedicineUniversity of CaliforniaSan FranciscoCAUSA
| | | | - Jung San Huang
- Departments of Biochemistry and Molecular BiologySaint Louis University School of MedicineSt. LouisMOUSA
| |
Collapse
|
35
|
Aurora B Tension Sensing Mechanisms in the Kinetochore Ensure Accurate Chromosome Segregation. Int J Mol Sci 2021; 22:ijms22168818. [PMID: 34445523 PMCID: PMC8396173 DOI: 10.3390/ijms22168818] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 11/29/2022] Open
Abstract
The accurate segregation of chromosomes is essential for the survival of organisms and cells. Mistakes can lead to aneuploidy, tumorigenesis and congenital birth defects. The spindle assembly checkpoint ensures that chromosomes properly align on the spindle, with sister chromatids attached to microtubules from opposite poles. Here, we review how tension is used to identify and selectively destabilize incorrect attachments, and thus serves as a trigger of the spindle assembly checkpoint to ensure fidelity in chromosome segregation. Tension is generated on properly attached chromosomes as sister chromatids are pulled in opposing directions but resisted by centromeric cohesin. We discuss the role of the Aurora B kinase in tension-sensing and explore the current models for translating mechanical force into Aurora B-mediated biochemical signals that regulate correction of chromosome attachments to the spindle.
Collapse
|
36
|
Mangione MC, Chen JS, Gould KL. Cdk1 phosphorylation of fission yeast paxillin inhibits its cytokinetic ring localization. Mol Biol Cell 2021; 32:1534-1544. [PMID: 34133210 PMCID: PMC8351747 DOI: 10.1091/mbc.e20-12-0807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 11/11/2022] Open
Abstract
Divisions of the genetic material and cytoplasm are coordinated spatially and temporally to ensure genome integrity. This coordination is mediated in part by the major cell cycle regulator cyclin-dependent kinase (Cdk1). Cdk1 activity peaks during mitosis, but during mitotic exit/cytokinesis Cdk1 activity is reduced, and phosphorylation of its substrates is reversed by various phosphatases including Cdc14, PP1, PP2A, and PP2B. Cdk1 is known to phosphorylate several components of the actin- and myosin-based cytokinetic ring (CR) that mediates division of yeast and animal cells. Here we show that Cdk1 also phosphorylates the Schizosaccharomyces pombe CR component paxillin Pxl1. We determined that both the Cdc14 phosphatase Clp1 and the PP1 phosphatase Dis2 contribute to Pxl1 dephosphorylation at mitotic exit, but PP2B/calcineurin does not. Preventing Pxl1 phosphorylation by Cdk1 results in increased Pxl1 levels, precocious Pxl1 recruitment to the division site, and increased duration of CR constriction. In vitro Cdk1-mediated phosphorylation of Pxl1 inhibits its interaction with the F-BAR domain of the cytokinetic scaffold Cdc15, thereby disrupting a major mechanism of Pxl1 recruitment. Thus, Pxl1 is a novel substrate through which S. pombe Cdk1 and opposing phosphatases coordinate mitosis and cytokinesis.
Collapse
Affiliation(s)
- MariaSanta C. Mangione
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| | - Jun-Song Chen
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| | - Kathleen L. Gould
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240
| |
Collapse
|
37
|
Remnant L, Kochanova NY, Reid C, Cisneros-Soberanis F, Earnshaw WC. The intrinsically disorderly story of Ki-67. Open Biol 2021; 11:210120. [PMID: 34375547 PMCID: PMC8354752 DOI: 10.1098/rsob.210120] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/13/2021] [Indexed: 01/14/2023] Open
Abstract
Ki-67 is one of the most famous marker proteins used by histologists to identify proliferating cells. Indeed, over 30 000 articles referring to Ki-67 are listed on PubMed. Here, we review some of the current literature regarding the protein. Despite its clinical importance, our knowledge of the molecular biology and biochemistry of Ki-67 is far from complete, and its exact molecular function(s) remain enigmatic. Furthermore, reports describing Ki-67 function are often contradictory, and it has only recently become clear that this proliferation marker is itself dispensable for cell proliferation. We discuss the unusual organization of the protein and its mRNA and how they relate to various models for its function. In particular, we focus on ways in which the intrinsically disordered structure of Ki-67 might aid in the assembly of the still-mysterious mitotic chromosome periphery compartment by controlling liquid-liquid phase separation of nucleolar proteins and RNAs.
Collapse
Affiliation(s)
- Lucy Remnant
- Wellcome Centre for Cell Biology, University of Edinburgh, ICB, Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Natalia Y. Kochanova
- Wellcome Centre for Cell Biology, University of Edinburgh, ICB, Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Caitlin Reid
- Wellcome Centre for Cell Biology, University of Edinburgh, ICB, Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - Fernanda Cisneros-Soberanis
- Wellcome Centre for Cell Biology, University of Edinburgh, ICB, Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| | - William C. Earnshaw
- Wellcome Centre for Cell Biology, University of Edinburgh, ICB, Michael Swann Building, King's Buildings, Max Born Crescent, Edinburgh EH9 3BF, UK
| |
Collapse
|
38
|
Abstract
Understanding the mechanisms of embryonic cell cycles is a central goal of developmental biology, as the regulation of the cell cycle must be closely coordinated with other events during early embryogenesis. Quantitative imaging approaches have recently begun to reveal how the cell cycle oscillator is controlled in space and time, and how it is integrated with mechanical signals to drive morphogenesis. Here, we discuss how the Drosophila embryo has served as an excellent model for addressing the molecular and physical mechanisms of embryonic cell cycles, with comparisons to other model systems to highlight conserved and species-specific mechanisms. We describe how the rapid cleavage divisions characteristic of most metazoan embryos require chemical waves and cytoplasmic flows to coordinate morphogenesis across the large expanse of the embryo. We also outline how, in the late cleavage divisions, the cell cycle is inter-regulated with the activation of gene expression to ensure a reliable maternal-to-zygotic transition. Finally, we discuss how precise transcriptional regulation of the timing of mitosis ensures that tissue morphogenesis and cell proliferation are tightly controlled during gastrulation.
Collapse
Affiliation(s)
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27705, USA
| |
Collapse
|
39
|
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: 16] [Impact Index Per Article: 4.0] [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
|
40
|
Vervoort SJ, Welsh SA, Devlin JR, Barbieri E, Knight DA, Offley S, Bjelosevic S, Costacurta M, Todorovski I, Kearney CJ, Sandow JJ, Fan Z, Blyth B, McLeod V, Vissers JHA, Pavic K, Martin BP, Gregory G, Demosthenous E, Zethoven M, Kong IY, Hawkins ED, Hogg SJ, Kelly MJ, Newbold A, Simpson KJ, Kauko O, Harvey KF, Ohlmeyer M, Westermarck J, Gray N, Gardini A, Johnstone RW. The PP2A-Integrator-CDK9 axis fine-tunes transcription and can be targeted therapeutically in cancer. Cell 2021; 184:3143-3162.e32. [PMID: 34004147 PMCID: PMC8567840 DOI: 10.1016/j.cell.2021.04.022] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/27/2020] [Accepted: 04/14/2021] [Indexed: 12/18/2022]
Abstract
Gene expression by RNA polymerase II (RNAPII) is tightly controlled by cyclin-dependent kinases (CDKs) at discrete checkpoints during the transcription cycle. The pausing checkpoint following transcription initiation is primarily controlled by CDK9. We discovered that CDK9-mediated, RNAPII-driven transcription is functionally opposed by a protein phosphatase 2A (PP2A) complex that is recruited to transcription sites by the Integrator complex subunit INTS6. PP2A dynamically antagonizes phosphorylation of key CDK9 substrates including DSIF and RNAPII-CTD. Loss of INTS6 results in resistance to tumor cell death mediated by CDK9 inhibition, decreased turnover of CDK9 phospho-substrates, and amplification of acute oncogenic transcriptional responses. Pharmacological PP2A activation synergizes with CDK9 inhibition to kill both leukemic and solid tumor cells, providing therapeutic benefit in vivo. These data demonstrate that fine control of gene expression relies on the balance between kinase and phosphatase activity throughout the transcription cycle, a process dysregulated in cancer that can be exploited therapeutically.
Collapse
Affiliation(s)
- Stephin J Vervoort
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia.
| | - Sarah A Welsh
- The Wistar Institute, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jennifer R Devlin
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | | | - Deborah A Knight
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Sarah Offley
- The Wistar Institute, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stefan Bjelosevic
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Matteo Costacurta
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Izabela Todorovski
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Conor J Kearney
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Jarrod J Sandow
- The Walter and Eliza Hall Institute, Parkville 3010, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Zheng Fan
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Benjamin Blyth
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia
| | - Victoria McLeod
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia
| | - Joseph H A Vissers
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia; Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Karolina Pavic
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku FI-20014, Finland; Institute of Biomedicine, University of Turku, Turku FI-20014, Finland
| | - Ben P Martin
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Gareth Gregory
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia; School of Clinical Sciences at Monash Health, Monash University, Clayton 3168, VIC, Australia
| | | | - Magnus Zethoven
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia
| | - Isabella Y Kong
- The Walter and Eliza Hall Institute, Parkville 3010, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Edwin D Hawkins
- The Walter and Eliza Hall Institute, Parkville 3010, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Simon J Hogg
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Madison J Kelly
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | - Andrea Newbold
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia
| | | | - Otto Kauko
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku FI-20014, Finland; Institute of Biomedicine, University of Turku, Turku FI-20014, Finland
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia; Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton 3168, VIC, Australia
| | - Michael Ohlmeyer
- Mount Sinai School of Medicine, New York, NY 10029, USA; Atux Iskay LLC, Plainsboro, NJ 08536, USA
| | - Jukka Westermarck
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku FI-20014, Finland; Institute of Biomedicine, University of Turku, Turku FI-20014, Finland
| | | | | | - Ricky W Johnstone
- Peter MacCallum Cancer Centre, Melbourne 3000, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, VIC, Australia.
| |
Collapse
|
41
|
Hernansaiz-Ballesteros RD, Földi C, Cardelli L, Nagy LG, Csikász-Nagy A. Evolution of opposing regulatory interactions underlies the emergence of eukaryotic cell cycle checkpoints. Sci Rep 2021; 11:11122. [PMID: 34045495 PMCID: PMC8159995 DOI: 10.1038/s41598-021-90384-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 05/11/2021] [Indexed: 02/04/2023] Open
Abstract
In eukaryotes the entry into mitosis is initiated by activation of cyclin-dependent kinases (CDKs), which in turn activate a large number of protein kinases to induce all mitotic processes. The general view is that kinases are active in mitosis and phosphatases turn them off in interphase. Kinases activate each other by cross- and self-phosphorylation, while phosphatases remove these phosphate groups to inactivate kinases. Crucial exceptions to this general rule are the interphase kinase Wee1 and the mitotic phosphatase Cdc25. Together they directly control CDK in an opposite way of the general rule of mitotic phosphorylation and interphase dephosphorylation. Here we investigate why this opposite system emerged and got fixed in almost all eukaryotes. Our results show that this reversed action of a kinase-phosphatase pair, Wee1 and Cdc25, on CDK is particularly suited to establish a stable G2 phase and to add checkpoints to the cell cycle. We show that all these regulators appeared together in LECA (Last Eukaryote Common Ancestor) and co-evolved in eukaryotes, suggesting that this twist in kinase-phosphatase regulation was a crucial step happening at the emergence of eukaryotes.
Collapse
Affiliation(s)
- Rosa D Hernansaiz-Ballesteros
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK
- Faculty of Medicine, Institute for Computational Biomedicine, Bioquant, Heidelberg University, 69120, Heidelberg, Germany
| | - Csenge Földi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, 6726, Hungary
| | - Luca Cardelli
- Department of Computer Science, University of Oxford, Wolfson Building, Parks Road, Oxford, OX1 3QD, UK
| | - László G Nagy
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, 6726, Hungary
| | - Attila Csikász-Nagy
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, SE1 1UL, UK.
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50/A, Budapest, 1083, Hungary.
| |
Collapse
|
42
|
Cossa G, Parua PK, Eilers M, Fisher RP. Protein phosphatases in the RNAPII transcription cycle: erasers, sculptors, gatekeepers, and potential drug targets. Genes Dev 2021; 35:658-676. [PMID: 33888562 PMCID: PMC8091971 DOI: 10.1101/gad.348315.121] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In this review, Cossa et al. discuss the current knowledge and outstanding questions about phosphatases in the context of the RNAPII transcription cycle. The transcription cycle of RNA polymerase II (RNAPII) is governed at multiple points by opposing actions of cyclin-dependent kinases (CDKs) and protein phosphatases, in a process with similarities to the cell division cycle. While important roles of the kinases have been established, phosphatases have emerged more slowly as key players in transcription, and large gaps remain in understanding of their precise functions and targets. Much of the earlier work focused on the roles and regulation of sui generis and often atypical phosphatases—FCP1, Rtr1/RPAP2, and SSU72—with seemingly dedicated functions in RNAPII transcription. Decisive roles in the transcription cycle have now been uncovered for members of the major phosphoprotein phosphatase (PPP) family, including PP1, PP2A, and PP4—abundant enzymes with pleiotropic roles in cellular signaling pathways. These phosphatases appear to act principally at the transitions between transcription cycle phases, ensuring fine control of elongation and termination. Much is still unknown, however, about the division of labor among the PPP family members, and their possible regulation by or of the transcriptional kinases. CDKs active in transcription have recently drawn attention as potential therapeutic targets in cancer and other diseases, raising the prospect that the phosphatases might also present opportunities for new drug development. Here we review the current knowledge and outstanding questions about phosphatases in the context of the RNAPII transcription cycle.
Collapse
Affiliation(s)
- Giacomo Cossa
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Pabitra K Parua
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Martin Eilers
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | - Robert P Fisher
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
43
|
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: 11] [Impact Index Per Article: 2.8] [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
|
44
|
Zhang X, Park JE, Kim EH, Hong J, Hwang KT, Kim YA, Jang CY. Wip1 controls the translocation of the chromosomal passenger complex to the central spindle for faithful mitotic exit. Cell Mol Life Sci 2021; 78:2821-2838. [PMID: 33067654 PMCID: PMC11072438 DOI: 10.1007/s00018-020-03665-x] [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: 03/10/2020] [Revised: 08/12/2020] [Accepted: 10/05/2020] [Indexed: 10/23/2022]
Abstract
Dramatic cellular reorganization in mitosis critically depends on the timely and temporal phosphorylation of a broad range of proteins, which is mediated by the activation of the mitotic kinases and repression of counteracting phosphatases. The mitosis-to-interphase transition, which is termed mitotic exit, involves the removal of mitotic phosphorylation by protein phosphatases. Although protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) drive this reversal in animal cells, the phosphatase network associated with ordered bulk dephosphorylation in mitotic exit is not fully understood. Here, we describe a new mitotic phosphatase relay in which Wip1/PPM1D phosphatase activity is essential for chromosomal passenger complex (CPC) translocation to the anaphase central spindle after release from the chromosome via PP1-mediated dephosphorylation of histone H3T3. Depletion of endogenous Wip1 and overexpression of the phosphatase-dead mutant disturbed CPC translocation to the central spindle, leading to failure of cytokinesis. While Wip1 was degraded in early mitosis, its levels recovered in anaphase and the protein functioned as a Cdk1-counteracting phosphatase at the anaphase central spindle and midbody. Mechanistically, Wip1 dephosphorylated Thr-59 in inner centromere protein (INCENP), which, subsequently bound to MKLP2 and recruited other components to the central spindle. Furthermore, Wip1 overexpression is associated with the overall survival rate of patients with breast cancer, suggesting that Wip1 not only functions as a weak oncogene in the DNA damage network but also as a tumor suppressor in mitotic exit. Altogether, our findings reveal that sequential dephosphorylation of mitotic phosphatases provides spatiotemporal regulation of mitotic exit to prevent tumor initiation and progression.
Collapse
Affiliation(s)
- Xianghua Zhang
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ji Eun Park
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Catholic University of Daegu, Daegu, 42472, Republic of Korea
| | - Jihee Hong
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ki-Tae Hwang
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Young A Kim
- Department of Pathology, Seoul National University Boramae Medical Center, Seoul, 07061, Republic of Korea.
| | - Chang-Young Jang
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
45
|
Tao SC, Huang JY, Li ZX, Zhan S, Guo SC. Small extracellular vesicles with LncRNA H19 "overload": YAP Regulation as a Tendon Repair Therapeutic Tactic. iScience 2021; 24:102200. [PMID: 33733065 PMCID: PMC7937563 DOI: 10.1016/j.isci.2021.102200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/10/2021] [Accepted: 02/12/2021] [Indexed: 01/20/2023] Open
Abstract
Functional healing of tendon injuries remains a great challenge. Small extracellular vesicles (sEVs) have received attention as pro-regenerative agents. H19 overexpression could bring tendon regenerative ability, but the mechanism is still not fully elucidated, and reliable method for delivery of long non-coding RNAs (LncRNAs) was demanded. We identified the downstream mechanism of H19, the activation of yes-associated protein (YAP) via the H19-PP1-YAP axis. We established tendon stem/progenitor cells (TSPCs) stably overexpressing H19 with CRISPR-dCas9-based hnRNP A2/B1 activation (H19-CP-TSPCs). H19-OL-sEVs (H19 “overloading” sEVs) could be produced effectively from H19-CP-TSPCs. Only H19-OL-sEVs were able to significantly load large amounts of H19 rather than other competitors, and the potential of H19-OL-sEVs to promote tendon healing was far better than that of other competitors. Our study established a relatively reliable method for enrichment of LncRNAs into sEVs, providing new hints for modularized sEV-based therapies, and modularized sEVs represented a potential strategy for tendon regeneration. H19 overexpression enhances tendon regeneration H19 dephosphorylates and activates YAP hnRNP A2/B1 assists the enrichment of H19 into sEVs H19-OL-sEVs promote tendon regeneration
Collapse
Affiliation(s)
- Shi-Cong Tao
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Ji-Yan Huang
- Department of Stomatology, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 185 Pu'an Road, Shanghai 200021, China
| | - Zi-Xiang Li
- Department of Medicine, Soochou University, Suzhou, Jiangsu 215123, China
| | - Shi Zhan
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Shang-Chun Guo
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China.,Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
46
|
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
|
47
|
Schutt KL, Moseley JB. The phosphatase inhibitor Sds23 promotes symmetric spindle positioning in fission yeast. Cytoskeleton (Hoboken) 2020; 77:544-557. [PMID: 33280247 PMCID: PMC8195570 DOI: 10.1002/cm.21648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/05/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022]
Abstract
A hallmark of cell division in eukaryotic cells is the formation and elongation of a microtubule (MT)-based mitotic spindle. Proper positioning of the spindle is critical to ensure equal segregation of the genetic material to the resulting daughter cells. Both the timing of spindle elongation and constriction of the actomyosin contractile ring must be precisely coordinated to prevent missegregation or damage to the genetic material during cellular division. Here, we show that Sds23, an inhibitor of protein phosphatases, contributes to proper positioning of elongating spindles in fission yeast cells. We found that sds23∆ mutant cells exhibit asymmetric spindles that initially elongate asymmetrically toward one end of the dividing cell. Spindle asymmetry in sds23∆ cells results from a defect that is distinct from previously identified mechanisms, including MT protrusions and enlarged vacuoles. Combined with our previous work, this study demonstrates that Sds23, an inhibitor of PP2A-family protein phosphatases, promotes proper positioning of both the bipolar spindle and cytokinetic ring during fission yeast cell division. These two steps ensure the overall symmetry and fidelity of the cell division process.
Collapse
Affiliation(s)
- Katherine L. Schutt
- Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - James B. Moseley
- Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| |
Collapse
|
48
|
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
|
49
|
BUBR1 Pseudokinase Domain Promotes Kinetochore PP2A-B56 Recruitment, Spindle Checkpoint Silencing, and Chromosome Alignment. Cell Rep 2020; 33:108397. [PMID: 33207204 DOI: 10.1016/j.celrep.2020.108397] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 10/13/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
The balance of phospho-signaling at the outer kinetochore is critical for forming accurate attachments between kinetochores and the mitotic spindle and timely exit from mitosis. A major player in determining this balance is the PP2A-B56 phosphatase, which is recruited to the kinase attachment regulatory domain (KARD) of budding uninhibited by benzimidazole 1-related 1 (BUBR1) in a phospho-dependent manner. This unleashes a rapid, switch-like phosphatase relay that reverses mitotic phosphorylation at the kinetochore, extinguishing the checkpoint and promoting anaphase. Here, we demonstrate that the C-terminal pseudokinase domain of human BUBR1 is required to promote KARD phosphorylation. Mutation or removal of the pseudokinase domain results in decreased PP2A-B56 recruitment to the outer kinetochore attenuated checkpoint silencing and errors in chromosome alignment as a result of imbalance in Aurora B activity. Our data, therefore, elucidate a function for the BUBR1 pseudokinase domain in ensuring accurate and timely exit from mitosis.
Collapse
|
50
|
Jacquet K, Rodrigue MA, Richard DE, Lavoie JN. The adenoviral protein E4orf4: a probing tool to decipher mechanical stress-induced nuclear envelope remodeling in tumor cells. Cell Cycle 2020; 19:2963-2981. [PMID: 33103553 DOI: 10.1080/15384101.2020.1836441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The human adenovirus (Ad) type 2/5 early region 4 (E4) ORF4 protein (E4orf4) exerts a remarkable tumor cell-selective killing activity in mammalian cells. This indicates that E4orf4 can target tumor cell-defining features and is a unique tool to probe cancer cell vulnerabilities. Recently, we found that E4orf4, through an interaction with the polarity protein PAR3, subverts nuclear envelope (NE) remodeling processes in a tumor cell-selective manner. In this Perspective, we outline mechanical signals that modify nuclear dynamics and tumor cell behavior to highlight potential mechanisms for E4orf4's tumoricidal activity. Through an analysis of E4orf4's cellular targets, we define a protein subnetwork that comprises phosphatase systems interconnected to polarity protein hubs, which could contribute to enhanced NE plasticity. We infer that elucidating E4orf4's protein network at a functional level could uncover key mechanisms of NE remodeling that define the tumor cell phenotype.
Collapse
Affiliation(s)
- Kévin Jacquet
- Centre de Recherche sur le Cancer de l'Université Laval , Québec, Canada.,Oncology, Centre de Recherche du CHU de Québec-Université Laval , Québec, Canada
| | - Marc-Antoine Rodrigue
- Centre de Recherche sur le Cancer de l'Université Laval , Québec, Canada.,Oncology, Centre de Recherche du CHU de Québec-Université Laval , Québec, Canada
| | - Darren E Richard
- Centre de Recherche sur le Cancer de l'Université Laval , Québec, Canada.,Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Université Laval , Québec, Canada.,Endocrinology and Nephrology, Centre de Recherche du CHU de Québec-Université Laval , Québec, Canada
| | - Josée N Lavoie
- Centre de Recherche sur le Cancer de l'Université Laval , Québec, Canada.,Oncology, Centre de Recherche du CHU de Québec-Université Laval , Québec, Canada.,Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Université Laval , Québec, Canada
| |
Collapse
|